1
|
Mihaltan F, Csoma Z, Pauk N, Irás B, Baukienė J, Teodorescu G, Malakauskas K, Staevska M. Benralizumab Outcomes in Patients with Severe Eosinophilic Asthma Treated in Real-Life Settings: Results of the BREEZE Study in 5 Countries From Central Eastern Europe and Baltics. J Asthma Allergy 2025; 18:195-210. [PMID: 39990056 PMCID: PMC11844207 DOI: 10.2147/jaa.s503048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 01/22/2025] [Indexed: 02/25/2025] Open
Abstract
Purpose To describe real-world clinical characteristics, treatment patterns and outcomes of severe eosinophilic asthma (SEA) patients initiated on benralizumab after treatment authorization in Central Eastern Europe and Baltic Area. Patients and Methods BREEZE was a retrospective, medical chart review with a pre-post design conducted in 42 clinical centers from Bulgaria, Czech Republic, Hungary, Lithuania and Romania. Eligibility included diagnosis of SEA and at least one dose of benralizumab administered in real-life settings. Descriptive statistics were used in the full analysis set and key subgroups stratified by blood eosinophils (bEOS) number, maintenance oral corticosteroids (mOCS) use and prior biologics exposure and included calculation of the annualized exacerbation rate (AER) at baseline, and weeks (W) 16 and 48. Results Of 381 patients included, 66% were female with overall mean age 56 ±12 years at benralizumab start. At baseline: median bEOS 580 cells/μL (74% bEOS>400), forced expiratory volume in 1 second (FEV1) 1660 mL, mOCS use in 25% of patients (10 mg/day prednisone equivalent, 68% >5 mg/day), AER 3.05 (95% CI 2.9-3.2), and poorly controlled asthma (Asthma Control Test [ACT] <16) in 63% of patients. Median duration of exposure to benralizumab was 11.5 (95% CI 7.7-12.3) months, and discontinuation rate was 1.3% (95% CI 0.4%-3.0%). Median bEOS decreased to 0 at W16 and maintained thereafter; FEV1 increases of +240 mL at W16 and +335 mL at W48 were reported (p <0.001 for both). Overall relative reduction in AER at W16 and W48 was 92% and 93%, respectively, and 82-94% across key subgroups. mOCS dose reduction was 50%, and proportion of patients requiring >5 mg/day decreased constantly (25% at W16, 28% at W48). ACT scores increased from W16 to W56 (p <0.001 for all). Conclusion Our findings indicate clinically meaningful benefits of benralizumab across multiple geographies and various subgroups of patients with SEA.
Collapse
Affiliation(s)
- Florin Mihaltan
- Department of Pneumology, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| | - Zsuzsanna Csoma
- Department of Allergology, National Korányi Institute for Pulmonology, Budapest, Hungary
| | - Norbert Pauk
- Department of Pneumology, Third Faculty of Medicine, Charles University and University Hospital Bulovka, Prague, Czech Republic
| | - Béla Irás
- Medical Affairs, AstraZeneca Hungary, Budapest, Hungary
| | | | | | - Kęstutis Malakauskas
- Department of Pulmonology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Maria Staevska
- Department of Allergology, Medical University Sofia, Sofia, Bulgaria
| |
Collapse
|
2
|
Li B, Dong B, Xie L, Li Y. Exploring Advances in Natural Plant Molecules for Allergic Rhinitis Immunomodulation in Vivo and in Vitro. Int J Gen Med 2025; 18:529-565. [PMID: 39911299 PMCID: PMC11796455 DOI: 10.2147/ijgm.s493021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 12/20/2024] [Indexed: 02/07/2025] Open
Abstract
Allergic rhinitis (AR) is a prevalent allergic disease that imposes significant economic burdens and life pressures on individuals, families, and society, particularly in the context of accelerating globalization and increasing pathogenic factors. Current clinical therapies for AR include antihistamines, glucocorticoids administered via various routes, leukotriene receptor antagonists, immunotherapy, and several decongestants. These treatments have demonstrated efficacy in alleviating clinical symptoms and pathological states. However, with the growing awareness of AR and rising expectations for improvements in quality of life, these treatments have become associated with a higher incidence of side effects and an elevated risk of drug resistance. Furthermore, the development of AR is intricately associated with dysregulation of the immune system, yet the underlying pathogenetic mechanisms remain incompletely understood. In contrast, widely available natural plant molecules offer multiple targeting pathways that uniquely modify the typical pathophysiology of AR through immunomodulatory processes. This review presents a comprehensive analysis of both in vivo and in vitro studies on natural plant molecules that modulate immunity for treating AR. Additionally, we examine their specific mechanisms of action in animal models to provide new insights for developing safe and effective targeted therapies while guiding experimental and clinical applications against AR.
Collapse
Affiliation(s)
- Bingquan Li
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| | - Boyang Dong
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| | - Liangzhen Xie
- Ear-Nose-Throat Department, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| | - Yan Li
- Ear-Nose-Throat Department, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| |
Collapse
|
3
|
FitzPatrick RD, Noone JR, Cartwright RA, Gatti DM, Brosschot TP, Lane JM, Jensen EL, Kroker Kimber I, Reynolds LA. Eosinophils respond to, but are not essential for control of an acute Salmonella enterica serovar Typhimurium infection in mice. Infect Immun 2024; 92:e0032524. [PMID: 39248486 PMCID: PMC11475665 DOI: 10.1128/iai.00325-24] [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: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 09/10/2024] Open
Abstract
Eosinophils are a highly abundant cell type in the gastrointestinal tract during homeostatic conditions, where they have recently been reported to take on an activated phenotype following colonization by the bacterial microbiota. To date, there have been few studies investigating whether eosinophils respond to infection with enteric bacterial pathogens and/or investigating the requirements for eosinophils for effective bacterial pathogen control. In this study, we investigated the response of eosinophils to an acute enteric infection of mice with the bacterial pathogen Salmonella enterica serovar Typhimurium. We also assessed whether eosinophil deficiency impacted Salmonella burdens in the intestinal tract or impacted the systemic dissemination of Salmonella following an oral infection of littermate wild-type BALB/cJ and eosinophil-deficient ΔdblGATA BALB/cJ mice. We found comparable Salmonella burdens in the intestinal tract of wild-type and eosinophil-deficient mice and no significant differences in the levels of Salmonella disseminating to systemic organs within 3 days of infection. Despite our evidence suggesting that eosinophils are not an essential cell type for controlling bacterial burdens in this acute infection setting, we found higher levels of eosinophils in gut-draining lymph nodes following infection, indicating that eosinophils do respond to Salmonella infection. Our data contribute to the growing evidence that eosinophils are responsive to bacterial stimuli, yet the influence of and requirements for eosinophils during bacterial infection appear to be highly context-dependent.
Collapse
Affiliation(s)
- Rachael D. FitzPatrick
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Jonathan R. Noone
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Richard A. Cartwright
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Dominique M. Gatti
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Tara P. Brosschot
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Jenna M. Lane
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Erik L. Jensen
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Isabella Kroker Kimber
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Lisa A. Reynolds
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| |
Collapse
|
4
|
Chang LA, Schotsaert M. Ally, adversary, or arbitrator? The context-dependent role of eosinophils in vaccination for respiratory viruses and subsequent breakthrough infections. J Leukoc Biol 2024; 116:224-243. [PMID: 38289826 PMCID: PMC11288382 DOI: 10.1093/jleuko/qiae010] [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: 10/29/2023] [Revised: 12/12/2023] [Accepted: 12/26/2023] [Indexed: 02/01/2024] Open
Abstract
Eosinophils are a critical type of immune cell and central players in type 2 immunity. Existing literature suggests that eosinophils also can play a role in host antiviral responses, typically type 1 immune events, against multiple respiratory viruses, both directly through release of antiviral mediators and indirectly through activation of other effector cell types. One way to prime host immune responses toward effective antiviral responses is through vaccination, where typically a type 1-skewed immunity is desirable in the context of intracellular pathogens like respiratory viruses. In the realm of breakthrough respiratory viral infection in vaccinated hosts, an event in which virus can still establish productive infection despite preexisting immunity, eosinophils are most prominently known for their link to vaccine-associated enhanced respiratory disease upon natural respiratory syncytial virus infection. This was observed in a pediatric cohort during the 1960s following vaccination with formalin-inactivated respiratory syncytial virus. More recent research has unveiled additional roles of the eosinophil in respiratory viral infection and breakthrough infection. The specific contribution of eosinophils to the quality of vaccine responses, vaccine efficacy, and antiviral responses to infection in vaccinated hosts remains largely unexplored, especially regarding their potential roles in protection. On the basis of current findings, we will speculate upon the suggested function of eosinophils and consider the many potential ways by which eosinophils may exert protective and pathological effects in breakthrough infections. We will also discuss how to balance vaccine efficacy with eosinophil-related risks, as well as the use of eosinophils and their products as potential biomarkers of vaccine efficacy or adverse events.
Collapse
Affiliation(s)
- Lauren A Chang
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, United States
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, United States
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Box 1630, New York, NY 10029, United States
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States
| |
Collapse
|
5
|
Lopez-Perez D, Prados-Lopez B, Galvez J, Leon J, Carazo A. Eosinophils in Colorectal Cancer: Emerging Insights into Anti-Tumoral Mechanisms and Clinical Implications. Int J Mol Sci 2024; 25:6098. [PMID: 38892286 PMCID: PMC11172675 DOI: 10.3390/ijms25116098] [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: 04/19/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Eosinophils are myeloid effector cells whose main homing is the gastrointestinal tract. There, they take part in type I and type II immune responses. They also contribute to other non-immunological homeostatic functions like mucus production, tissue regeneration, and angiogenesis. In colorectal cancer (CRC), eosinophils locate in the center of the tumor and in the front of invasion and play an anti-tumoral role. They directly kill tumor cells by releasing cytotoxic compounds and eosinophil extracellular traps or indirectly by activating other immune cells via cytokines. As CRC progresses, the number of infiltrating eosinophils decreases. Although this phenomenon is not fully understood, it is known that some changes in the microenvironmental milieu and microbiome can affect eosinophil infiltration. Importantly, a high number of intratumoral eosinophils is a favorable prognostic factor independent from the tumor stage. Moreover, after immunotherapy, responding patients usually display eosinophilia, so eosinophils could be a good biomarker candidate to monitor treatment outcomes. Finally, even though eosinophils seem to play an interesting anti-tumoral role in CRC, much more research is needed to fully understand their interactions in the CRC microenvironment. This review explores the multifaceted roles of eosinophils in colorectal cancer, highlighting their anti-tumoral effects, prognostic significance, and potential as a biomarker for treatment outcomes.
Collapse
Affiliation(s)
- David Lopez-Perez
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, 18012 Granada, Spain
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
| | - Belen Prados-Lopez
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
| | - Julio Galvez
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, 18012 Granada, Spain
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
- Centro de Investigación Biomédica en Red para Enfermedades Hepáticas y Digestivas (CIBER-EHD), Center for Biomedical Research, University of Granada, 18012 Granada, Spain
| | - Josefa Leon
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario San Cecilio de Granada, 18016 Granada, Spain
| | - Angel Carazo
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
- Unidad de Gestión de Microbiología, Hospital Universitario San Cecilio de Granada, 18016 Granada, Spain
| |
Collapse
|
6
|
Brand CL, Hunger RE, Seyed Jafari SM. Eosinophilic granulocytes as a potential prognostic marker for cancer progression and therapeutic response in malignant melanoma. Front Oncol 2024; 14:1366081. [PMID: 38756652 PMCID: PMC11096470 DOI: 10.3389/fonc.2024.1366081] [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: 01/05/2024] [Accepted: 04/17/2024] [Indexed: 05/18/2024] Open
Abstract
The importance of eosinophilic granulocytes in cancer has been widely discussed in recent years. The current study reviews the evidence on the role of eosinophilic granulocytes in melanoma as a prognostic marker for cancer progression and the efficacy of treatment with modern immune checkpoint inhibitors. A total of 33 human clinical studies were included in the review, with heterogeneous data due to differences in patients populations, study design and inclusion of small study groups. However, 28 of the 33 studies suggested that eosinophilic granulocytes could be used as a prognostic biomarker for outcome and/or potential response to systemic treatment and/or occurrence of adverse events in melanoma patients. Nevertheless, the exact role of eosinophils remains to be elucidated. Further prospective, larger and better controlled studies are warranted to clarify the significance of eosinophilic granulocytes in patients with melanoma, in more details.
Collapse
Affiliation(s)
| | | | - Seyed Morteza Seyed Jafari
- Department of Dermatology and Venerology, University Hospital of Bern, University Bern, Bern, Switzerland
| |
Collapse
|
7
|
Jahanbani F, Sing JC, Maynard RD, Jahanbani S, Dafoe J, Dafoe W, Jones N, Wallace KJ, Rastan A, Maecker HT, Röst HL, Snyder MP, Davis RW. Longitudinal cytokine and multi-modal health data of an extremely severe ME/CFS patient with HSD reveals insights into immunopathology, and disease severity. Front Immunol 2024; 15:1369295. [PMID: 38650940 PMCID: PMC11033372 DOI: 10.3389/fimmu.2024.1369295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Introduction Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) presents substantial challenges in patient care due to its intricate multisystem nature, comorbidities, and global prevalence. The heterogeneity among patient populations, coupled with the absence of FDA-approved diagnostics and therapeutics, further complicates research into disease etiology and patient managment. Integrating longitudinal multi-omics data with clinical, health,textual, pharmaceutical, and nutraceutical data offers a promising avenue to address these complexities, aiding in the identification of underlying causes and providing insights into effective therapeutics and diagnostic strategies. Methods This study focused on an exceptionally severe ME/CFS patient with hypermobility spectrum disorder (HSD) during a period of marginal symptom improvements. Longitudinal cytokine profiling was conducted alongside the collection of extensive multi-modal health data to explore the dynamic nature of symptoms, severity, triggers, and modifying factors. Additionally, an updated severity assessment platform and two applications, ME-CFSTrackerApp and LexiTime, were introduced to facilitate real-time symptom tracking and enhance patient-physician/researcher communication, and evaluate response to medical intervention. Results Longitudinal cytokine profiling revealed the significance of Th2-type cytokines and highlighted synergistic activities between mast cells and eosinophils, skewing Th1 toward Th2 immune responses in ME/CFS pathogenesis, particularly in cognitive impairment and sensorial intolerance. This suggests a potentially shared underlying mechanism with major ME/CFS comorbidities such as HSD, Mast cell activation syndrome, postural orthostatic tachycardia syndrome (POTS), and small fiber neuropathy. Additionally, the data identified potential roles of BCL6 and TP53 pathways in ME/CFS etiology and emphasized the importance of investigating adverse reactions to medication and supplements and drug interactions in ME/CFS severity and progression. Discussion Our study advocates for the integration of longitudinal multi-omics with multi-modal health data and artificial intelligence (AI) techniques to better understand ME/CFS and its major comorbidities. These findings highlight the significance of dysregulated Th2-type cytokines in patient stratification and precision medicine strategies. Additionally, our results suggest exploring the use of low-dose drugs with partial agonist activity as a potential avenue for ME/CFS treatment. This comprehensive approach emphasizes the importance of adopting a patient-centered care approach to improve ME/CFS healthcare management, disease severity assessment, and personalized medicine. Overall, these findings contribute to our understanding of ME/CFS and offer avenues for future research and clinical practice.
Collapse
Affiliation(s)
- Fereshteh Jahanbani
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Justin Cyril Sing
- Department of Molecular Genetics, Donnelly Center, University of Toronto, Toronto, ON, Canada
| | - Rajan Douglas Maynard
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Shaghayegh Jahanbani
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Veterans Affairs (VA) Palo Alto Health Care System, Palo Alto, CA, United States
| | - Janet Dafoe
- ME/CFS Collaborative Research Center at Stanford, Stanford Genome Technology Center, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Whitney Dafoe
- ME/CFS Collaborative Research Center at Stanford, Stanford Genome Technology Center, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Nathan Jones
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Kelvin J. Wallace
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Azuravesta Rastan
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Holden T. Maecker
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Pulmonary and Critical Care Medicine, Institute of Immunity, Transplantation, and Infectious Diseases, Stanford University, Palo Alto, CA, United States
| | - Hannes L. Röst
- Department of Molecular Genetics, Donnelly Center, University of Toronto, Toronto, ON, Canada
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Ronald W. Davis
- ME/CFS Collaborative Research Center at Stanford, Stanford Genome Technology Center, Stanford University School of Medicine, Palo Alto, CA, United States
| |
Collapse
|
8
|
Wang F, Shang Y. Radical S-adenosyl methionine domain-containing 2, a potential target of D-tryptophan in asthma treatment, regulates T helper cell type 1/2 balance. Int Immunopharmacol 2024; 129:111581. [PMID: 38310765 DOI: 10.1016/j.intimp.2024.111581] [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: 09/12/2023] [Revised: 01/20/2024] [Accepted: 01/21/2024] [Indexed: 02/06/2024]
Abstract
Asthma is a common chronic respiratory disease. D-tryptophan (D-TRP) can inhibit allergic airway inflammation and T helper cell type 2 (Th2) immune response. RNA-sequencing results have indicated that radical S-adenosyl methionine domain-containing 2 (RSAD2) might be a potential molecular target of D-TRP in asthma treatment. Herein, we established a mouse model of asthma using ovalbumin (OVA) via intraperitoneal injection and inhalational challenge. Gain- and loss-of-function studies of RSAD2 were performed in mice following the intratracheal delivery of lentiviral vectors (3 × 106 TU/mL). Naïve CD-4+ T cells were isolated from the spleen and used to explore the effects of RSAD2 on Th2 cell differentiation. RSAD2 expression was higher in the asthma group than in the control group. RSAD2 knockdown alleviated inflammatory cell infiltration and reduced the number of goblet cells. Low RSAD2 expression decreased the levels of IgE, IL-25, IL-33, and TSLP, and it reduced the number of inflammatory cells in the bronchoalveolar lavage fluid. RSAD2 silencing suppressed Th2-related cytokine levels (such as IL-4, IL-5, and IL-13) and increased Th1-related cytokine levels (such as IFN-γ). Additionally, RSAD2 knockdown inhibited the phosphorylation of JAK1, JAK3, and STAT6, and downregulated GATA-3 expression. RSAD2 overexpression increased inflammatory cell infiltration and mucus secretion in the lung tissues of mice pretreated with D-TRP. D-TRP pretreatment reduced OVA-specific IgE content and IL-4 and IL-5 levels, and it increased the IFN-γ levels; however, RSAD2 overexpression reversed these effects. In conclusion, RSAD2 knockdown can mitigate OVA-induced asthma by regulating the Th2 immune response via JAK/STAT6 pathway inhibition.
Collapse
Affiliation(s)
- Fei Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yunxiao Shang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
9
|
Koranteng J, Chung KF, Michaeloudes C, Bhavsar P. The role of mitochondria in eosinophil function: implications for severe asthma pathogenesis. Front Cell Dev Biol 2024; 12:1360079. [PMID: 38495619 PMCID: PMC10940389 DOI: 10.3389/fcell.2024.1360079] [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: 12/22/2023] [Accepted: 02/13/2024] [Indexed: 03/19/2024] Open
Abstract
Mitochondria are key metabolic hubs involved in cellular energy production and biosynthesis. ATP is generated primarily by glucose and fatty acid oxidation through the tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OXPHOS) in the mitochondria. During OXPHOS there is also production of reactive oxygen species (ROS), which are involved in the regulation of cellular function. Mitochondria are also central in the regulating cell survival and death, particularly in the intrinsic apoptosis pathway. Severe asthma is a heterogeneous disease driven by various immune mechanisms. Severe eosinophilic asthma entails a type 2 inflammatory response and peripheral and lung eosinophilia, associated with severe airflow obstruction, frequent exacerbations and poor response to treatment. Mitochondrial dysfunction and altered metabolism have been observed in airway epithelial and smooth muscle cells from patients with asthma. However, the role of mitochondria in the development of eosinophilia and eosinophil-mediated inflammation in severe asthma is unknown. In this review, we discuss the currently limited literature on the role of mitochondria in eosinophil function and how it is regulated by asthma-relevant cytokines, including interleukin (IL)-5 and granulocyte-macrophage colony-stimulating factor (GM-CSF), as well as by corticosteroid drugs. Moreover, we summarise the evidence on the role of mitochondria in the regulation of eosinophils apoptosis and eosinophil extracellular trap formation. Finally, we discuss the possible role of altered mitochondrial function in eosinophil dysfunction in severe asthma and suggest possible research avenues in order to better understand their role in disease pathogenesis, and identify novel therapeutic targets.
Collapse
Affiliation(s)
- Janice Koranteng
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Royal Brompton & Harefield NHS Trust, London, United Kingdom
| | | | - Pankaj Bhavsar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Royal Brompton & Harefield NHS Trust, London, United Kingdom
| |
Collapse
|
10
|
Macchia I, La Sorsa V, Urbani F, Moretti S, Antonucci C, Afferni C, Schiavoni G. Eosinophils as potential biomarkers in respiratory viral infections. Front Immunol 2023; 14:1170035. [PMID: 37483591 PMCID: PMC10358847 DOI: 10.3389/fimmu.2023.1170035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/30/2023] [Indexed: 07/25/2023] Open
Abstract
Eosinophils are bone marrow-derived granulocytes that, under homeostatic conditions, account for as much as 1-3% of peripheral blood leukocytes. During inflammation, eosinophils can rapidly expand and infiltrate inflamed tissues, guided by cytokines and alarmins (such as IL-33), adhesion molecules and chemokines. Eosinophils play a prominent role in allergic asthma and parasitic infections. Nonetheless, they participate in the immune response against respiratory viruses such as respiratory syncytial virus and influenza. Notably, respiratory viruses are associated with asthma exacerbation. Eosinophils release several molecules endowed with antiviral activity, including cationic proteins, RNases and reactive oxygen and nitrogen species. On the other hand, eosinophils release several cytokines involved in homeostasis maintenance and Th2-related inflammation. In the context of SARS-CoV-2 infection, emerging evidence indicates that eosinophils can represent possible blood-based biomarkers for diagnosis, prognosis, and severity prediction of disease. In particular, eosinopenia seems to be an indicator of severity among patients with COVID-19, whereas an increased eosinophil count is associated with a better prognosis, including a lower incidence of complications and mortality. In the present review, we provide an overview of the role and plasticity of eosinophils focusing on various respiratory viral infections and in the context of viral and allergic disease comorbidities. We will discuss the potential utility of eosinophils as prognostic/predictive immune biomarkers in emerging respiratory viral diseases, particularly COVID-19. Finally, we will revisit some of the relevant methods and tools that have contributed to the advances in the dissection of various eosinophil subsets in different pathological settings for future biomarker definition.
Collapse
Affiliation(s)
- Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Valentina La Sorsa
- Research Coordination and Support Service, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Sonia Moretti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy
| | - Caterina Antonucci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Claudia Afferni
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
11
|
Wiese AV, Duhn J, Korkmaz RÜ, Quell KM, Osman I, Ender F, Schröder T, Lewkowich I, Hogan S, Huber-Lang M, Gumprecht F, König P, Köhl J, Laumonnier Y. C5aR1 activation in mice controls inflammatory eosinophil recruitment and functions in allergic asthma. Allergy 2023. [PMID: 36757006 DOI: 10.1111/all.15670] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 12/14/2022] [Accepted: 01/02/2023] [Indexed: 02/10/2023]
Abstract
BACKGROUND Pulmonary eosinophils comprise at least two distinct populations of resident eosinophils (rEOS) and inflammatory eosinophils (iEOS), the latter recruited in response to pulmonary inflammation. Here, we determined the impact of complement activation on rEOS and iEOS trafficking and function in two models of pulmonary inflammation. METHODS BALB/c wild-type and C5ar1-/- mice were exposed to different allergens or IL-33. Eosinophil populations in the airways, lung, or mediastinal lymph nodes (mLN) were characterized by FACS or immunohistochemistry. rEOS and iEOS functions were determined in vivo and in vitro. RESULTS HDM and IL-33 exposure induced a strong accumulation of iEOS but not rEOS in the airways, lungs, and mLNs. rEOS and iEOS expressed C3/C5 and C5aR1, which were significantly higher in iEOS. Initial pulmonary trafficking of iEOS was markedly reduced in C5ar1-/- mice and associated with less IL-5 production from ILC2 cells. Functionally, adoptively transferred pulmonary iEOS from WT but not from C5ar1-/- mice-induced airway hyperresponsiveness (AHR), which was associated with significantly reduced C5ar1-/- iEOS degranulation. Pulmonary iEOS but not rEOS were frequently associated with T cells in lung tissue. After HDM or IL-33 exposure, iEOS but not rEOS were found in mLNs, which were significantly reduced in C5ar1-/- mice. C5ar1-/- iEOS expressed less costimulatory molecules, associated with a decreased potency to drive antigen-specific T cell proliferation and differentiation into memory T cells. CONCLUSIONS We uncovered novel roles for C5aR1 in iEOS trafficking and activation, which affects key aspects of allergic inflammation such as AHR, ILC2, and T cell activation.
Collapse
Affiliation(s)
- Anna V Wiese
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Jannis Duhn
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Rabia Ülkü Korkmaz
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Katharina M Quell
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Ibrahim Osman
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Torsten Schröder
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany.,Institute of Nutritional Medicine, University Hospital of Schleswig-Holstein & University of Lübeck, Lübeck, Germany
| | - Ian Lewkowich
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Simon Hogan
- Mary H. Weiser Food Allergy Center, Experimental Pathology, Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology (ITI), University of Ulm, Ulm, Germany
| | | | - Peter König
- Institute for Anatomy, University of Lübeck, Lübeck, Germany.,Airway Research Center North, Member of the German Center for Lung Research (DZL), Lübeck, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Airway Research Center North, Member of the German Center for Lung Research (DZL), Lübeck, Germany
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany.,Institute of Nutritional Medicine, University Hospital of Schleswig-Holstein & University of Lübeck, Lübeck, Germany.,Airway Research Center North, Member of the German Center for Lung Research (DZL), Lübeck, Germany
| |
Collapse
|
12
|
Blomberg OS, Kos K, Spagnuolo L, Isaeva OI, Garner H, Wellenstein MD, Bakker N, Duits DE, Kersten K, Klarenbeek S, Hau CS, Kaldenbach D, Raeven EA, Vrijland K, Kok M, de Visser KE. Neoadjuvant immune checkpoint blockade triggers persistent and systemic T reg activation which blunts therapeutic efficacy against metastatic spread of breast tumors. Oncoimmunology 2023; 12:2201147. [PMID: 37089449 PMCID: PMC10114978 DOI: 10.1080/2162402x.2023.2201147] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023] Open
Abstract
The clinical successes of immune checkpoint blockade (ICB) in advanced cancer patients have recently spurred the clinical implementation of ICB in the neoadjuvant and perioperative setting. However, how neoadjuvant ICB therapy affects the systemic immune landscape and metastatic spread remains to be established. Tumors promote both local and systemic expansion of regulatory T cells (Tregs), which are key orchestrators of tumor-induced immunosuppression, contributing to immune evasion, tumor progression and metastasis. Tregs express inhibitory immune checkpoint molecules and thus may be unintended targets for ICB therapy counteracting its efficacy. Using ICB-refractory models of spontaneous primary and metastatic breast cancer that recapitulate the poor ICB response of breast cancer patients, we observed that combined anti-PD-1 and anti-CTLA-4 therapy inadvertently promotes proliferation and activation of Tregs in the tumor, tumor-draining lymph node and circulation. Also in breast cancer patients, Treg levels were elevated upon ICB. Depletion of Tregs during neoadjuvant ICB in tumor-bearing mice not only reshaped the intratumoral immune landscape into a state favorable for ICB response but also induced profound and persistent alterations in systemic immunity, characterized by elevated CD8+ T cells and NK cells and durable T cell activation that was maintained after treatment cessation. While depletion of Tregs in combination with neoadjuvant ICB did not inhibit primary tumor growth, it prolonged metastasis-related survival driven predominantly by CD8+ T cells. This study demonstrates that neoadjuvant ICB therapy of breast cancer can be empowered by simultaneous targeting of Tregs, extending metastasis-related survival, independent of a primary tumor response.
Collapse
Affiliation(s)
- Olga S. Blomberg
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Kevin Kos
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lorenzo Spagnuolo
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Olga I. Isaeva
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hannah Garner
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Max D. Wellenstein
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Noor Bakker
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Danique E.M. Duits
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Kelly Kersten
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sjoerd Klarenbeek
- Experimental Animal Pathology Facility, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Cheei-Sing Hau
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Daphne Kaldenbach
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Elisabeth A.M. Raeven
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Kim Vrijland
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Marleen Kok
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Karin E. de Visser
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
- CONTACT Karin E. de Visser Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam1066 CX, The Netherlands
| |
Collapse
|
13
|
Luo C, Wang Y, He B, He Y, Yan Y, Wang J, Gu X. Exploring the Core Prescription and Underlying Mechanism of Traditional Chinese Medicine in Treating Allergic Rhinitis in Children: A Real- World Study Based on an Illustrious Senior Traditional Chinese Medicine Practitioner. Comb Chem High Throughput Screen 2023; 26:207-223. [PMID: 35388748 DOI: 10.2174/1386207325666220406105633] [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: 08/26/2021] [Revised: 12/09/2021] [Accepted: 01/17/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Traditional Chinese medicine (TCM) is widely used to treat allergic rhinitis (AR) in China, especially in children. However, due to the complicated composition rules and unclear underlying mechanisms, effective herbal prescriptions' popularization and application are limited. PURPOSE This study tried to detect the core prescription of herbs in treating AR in children, reveal its mechanism based on the ingredients' network, and explore the main signaling pathways. METHODS We screened medical records of children patients with AR who were treated by TCM in DongZhiMen Hospital from Aug 2009 to Jan 2020 and adopted a descriptive analysis method on herbal characteristics. We used association rules to mine core prescriptions and used network pharmacology to establish the ingredient-target-pathway network through online databases and TCMSP, Genecards, KEGG pathway, Excel, R-Studio, and Cytoscape software. RESULTS The analysis of 1,092 clinical visits highlighted that the principle of formulating prescription was as follows: 'pungent and warm herbs were used more frequently while cold-natured herbs were paid equal attention as warm-natured herbs.' The core prescription was formed by FangFeng, BaiZhi, CangErzi, and ChanTui. These herbs covered 130 underlying targets and 141 signaling pathways of AR, which mainly had an effect on signal transduction and immunoregulation. CONCLUSION The core prescription based on these real-world clinical records includes FangFeng, BaiZhi, CangErzi, and ChanTui. It principally acts on targets of signal transduction pathways and immune pathways.
Collapse
Affiliation(s)
- Changyong Luo
- Beijing University of Traditional Chinese Medicine, Beijing, China
- Dongfang Hospital of Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Yuhan Wang
- Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Bing He
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Yu He
- Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Yurou Yan
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Junhong Wang
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Xiaohong Gu
- Beijing University of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
14
|
Bao Y, Zhu X. Role of Chemokines and Inflammatory Cells in Respiratory Allergy. J Asthma Allergy 2022; 15:1805-1822. [PMID: 36575714 PMCID: PMC9790160 DOI: 10.2147/jaa.s395490] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
The idea of "one airway, one disease" has been gaining importance in the last decade. In the upper and lower airways, allergic mechanisms interact with each other. In the initial stage of respiratory allergic inflammation, allergens contact the respiratory epithelium, which produces chemokines and inflammatory factors, which cause allergic reactions by binding to the corresponding receptors and chemotactic various inflammatory cells to reach the epithelium and tissues. It also drives inflammatory cells to activate and produce more inflammatory factors, thus producing a cascade amplification effect. Inflammatory cell aggregation and activation are very complex and interact with each other in a lattice structure. By blocking the action of various chemokines, inflammatory cell aggregation is reduced, and ultimately the symptoms of respiratory allergy are alleviated. Chemokines can serve as cues for coordinated recruitment of immune cells into and out of tissues, as well as directing the spatial organization of immune cells within tissues and cellular interactions. Chemokines are critical in directing immune cell migration and thus have an important role in the direction of respiratory allergy: however, chemokines are also involved in the production and recruitment of immune cells that contribute to respiratory allergy. In this article, linking the upper and lower respiratory tracts. We review the role of the chemokine system in the respiratory immune response and discuss how respiratory disease modulates overall chemokines to shape the type and outcome of the immune response to the treatment of respiratory allergic disease so that we can further deepen our knowledge of chemokines in the direction of respiratory allergy. In the future, we can do drug research and development based on this network structure and explore new research directions.
Collapse
Affiliation(s)
- Youwei Bao
- Department of Otolaryngology Head & Neck Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Xinhua Zhu
- Department of Otolaryngology Head & Neck Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China,Correspondence: Xinhua Zhu, Email
| |
Collapse
|
15
|
Grisaru-Tal S, Rothenberg ME, Munitz A. Eosinophil-lymphocyte interactions in the tumor microenvironment and cancer immunotherapy. Nat Immunol 2022; 23:1309-1316. [PMID: 36002647 PMCID: PMC9554620 DOI: 10.1038/s41590-022-01291-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 07/18/2022] [Indexed: 12/15/2022]
Abstract
Eosinophils are important effector cells and therapeutic targets in allergic diseases. Emerging data indicate that eosinophils infiltrate a variety of solid tumor types and have pleiotropic activities by at least two non-mutually exclusive mechanisms: direct interactions with tumor cells, and intricate cross-talk with lymphocytes. In light of the immune checkpoint inhibition revolution in cancer therapy, we review eosinophil-lymphocyte interactions in the tumor microenvironment. We also analyze potential interactions between eosinophils and lymphocyte subsets, including T cells, natural killer cells and innate lymphoid cells. We provide perspectives on the consequences of these interactions and how eosinophils are accessory cells that can affect the response to various forms of T cell-mediated immunotherapies and might be therapeutically targeted to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Sharon Grisaru-Tal
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Marc E Rothenberg
- Division of Allergy/Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ariel Munitz
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
16
|
Zeng Q, Zeng Y, Tang Y, Liu W, Sun C. Effect of IL-35 on apoptosis, adhesion, migration, and activation of eosinophils in allergic rhinitis. Pediatr Allergy Immunol 2022; 33:e13717. [PMID: 34902176 DOI: 10.1111/pai.13717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 11/14/2021] [Accepted: 12/09/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Eosinophils play critical roles in the development of allergic rhinitis (AR) by releasing toxic substance. Interleukin-35 (IL-35), a newly identified anti-inflammatory cytokine, had potent inhibitive role for eosinophil infiltration in allergic disease. However, the direct effect of IL-35 on eosinophil was not clear. METHODS Twenty AR children and sixteen controls were recruited. The correlation between IL-35 protein expression and blood eosinophil counts and activation was analyzed. The effect of IL-35 on eosinophil apoptosis and adhesion was analyzed by flow cytometry. Transwell system was used for the migration assay. The eosinophil cationic protein (ECP) from supernatant of eosinophils after IL-35 stimulation was detected by enzyme-linked immunosorbent assay kits. RESULTS The IL-35 protein levels were negatively correlated with eosinophil counts (p < .01) and ECP concentration (p < .01) in AR children. IL-35 promotes apoptosis and inhibits adhesion, migration, and activation of eosinophils. Moreover, the mRNA expression of IL-12 receptor β2 and glycoprotein 130 were significantly enhanced by eosinophils after IL-35 stimulation. The apoptosis induced by IL-35 was mediated by phosphoinositide 3-kinase (PI3K) pathway. IL-35 inhibits adhesion of eosinophils through extracellular regulated protein kinases (ERK) and PI3K pathways. The eosinophil chemotaxis and activation affected by IL-35 were mediated by PI3K and p38 mitogen-activated protein kinase (MAPK) pathways. CONCLUSION Our results confirmed that IL-35 played inhibitive roles in apoptosis, adhesion, migration, and activation of eosinophils in AR, implying that IL-35 may be used as treatment target in future.
Collapse
Affiliation(s)
- Qingxiang Zeng
- Department of Otolaryngology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yinhui Zeng
- Department of Otolaryngology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yiquan Tang
- Department of Otolaryngology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wenlong Liu
- Department of Otolaryngology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Changzhi Sun
- Department of Otolaryngology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
17
|
Virtanen T. Inhalant Mammal-Derived Lipocalin Allergens and the Innate Immunity. FRONTIERS IN ALLERGY 2022; 2:824736. [PMID: 35387007 PMCID: PMC8974866 DOI: 10.3389/falgy.2021.824736] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/30/2021] [Indexed: 12/03/2022] Open
Abstract
A major part of important mammalian respiratory allergens belongs to the lipocalin family of proteins. By this time, 19 respiratory mammalian lipocalin allergens have been registered in the WHO/IUIS Allergen Nomenclature Database. Originally, lipocalins, small extracellular proteins (molecular mass ca. 20 kDa), were characterized as transport proteins but they are currently known to exert a variety of biological functions. The three-dimensional structure of lipocalins is well-preserved, and lipocalin allergens can exhibit high amino acid identities, in several cases more than 50%. Lipocalins contain an internal ligand-binding site where they can harbor small principally hydrophobic molecules. Another characteristic feature is their capacity to bind to specific cell-surface receptors. In all, the physicochemical properties of lipocalin allergens do not offer any straightforward explanations for their allergenicity. Allergic sensitization begins at epithelial barriers where diverse insults through pattern recognition receptors awaken innate immunity. This front-line response is manifested by epithelial barrier-associated cytokines which together with other components of immunity can initiate the sensitization process. In the following, the crucial factor in allergic sensitization is interleukin (IL)-4 which is needed for stabilizing and promoting the type 2 immune response. The source for IL-4 has been searched widely. Candidates for it may be non-professional antigen-presenting cells, such as basophils or mast cells, as well as CD4+ T cells. The synthesis of IL-4 by CD4+ T cells requires T cell receptor engagement, i.e., the recognition of allergen peptides, which also provides the specificity for sensitization. Lipocalin and innate immunity-associated cell-surface receptors are implicated in facilitating the access of lipocalin allergens into the immune system. However, the significance of this for allergic sensitization is unclear, as the recognition by these receptors has been found to produce conflicting results. As to potential adjuvants associated with mammalian lipocalin allergens, the hydrophobic ligands transported by lipocalins have not been reported to enhance sensitization while it is justified to suppose that lipopolysaccharide plays a role in it. Taken together, type 2 immunity to lipocalin allergens appears to be a harmful immune response resulting from a combination of signals involving both the innate and adaptive immunities.
Collapse
Affiliation(s)
- Tuomas Virtanen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
18
|
Felton JM, Bouffi C, Schwartz JT, Schollaert KL, Malik A, Vallabh S, Wronowski B, Magier AZ, Merlin L, Barski A, Weirauch MT, Fulkerson PC, Rothenberg ME. Aiolos regulates eosinophil migration into tissues. Mucosal Immunol 2021; 14:1271-1281. [PMID: 34341502 PMCID: PMC8542574 DOI: 10.1038/s41385-021-00416-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 02/04/2023]
Abstract
Expression of Ikaros family transcription factor IKZF3 (Aiolos) increases during murine eosinophil lineage commitment and maturation. Herein, we investigated Aiolos expression and function in mature human and murine eosinophils. Murine eosinophils deficient in Aiolos demonstrated gene expression changes in pathways associated with granulocyte-mediated immunity, chemotaxis, degranulation, ERK/MAPK signaling, and extracellular matrix organization; these genes had ATAC peaks within 1 kB of the TSS that were enriched for Aiolos-binding motifs. Global Aiolos deficiency reduced eosinophil frequency within peripheral tissues during homeostasis; a chimeric mouse model demonstrated dependence on intrinsic Aiolos expression by eosinophils. Aiolos deficiency reduced eosinophil CCR3 surface expression, intracellular ERK1/2 signaling, and CCL11-induced actin polymerization, emphasizing an impaired functional response. Aiolos-deficient eosinophils had reduced tissue accumulation in chemokine-, antigen-, and IL-13-driven inflammatory experimental models, all of which at least partially depend on CCR3 signaling. Human Aiolos expression was associated with active chromatin marks enriched for IKZF3, PU.1, and GATA-1-binding motifs within eosinophil-specific histone ChIP-seq peaks. Furthermore, treating the EOL-1 human eosinophilic cell line with lenalidomide yielded a dose-dependent decrease in Aiolos. These collective data indicate that eosinophil homing during homeostatic and inflammatory allergic states is Aiolos-dependent, identifying Aiolos as a potential therapeutic target for eosinophilic disease.
Collapse
Affiliation(s)
- Jennifer M Felton
- Division of Allergy and Immunology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Carine Bouffi
- Division of Allergy and Immunology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Justin T Schwartz
- Division of Allergy and Immunology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kaila L Schollaert
- Division of Allergy and Immunology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Astha Malik
- Division of Allergy and Immunology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sushmitha Vallabh
- Division of Allergy and Immunology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Benjamin Wronowski
- Division of Allergy and Immunology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Adam Z Magier
- Division of Allergy and Immunology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Li Merlin
- Division of Allergy and Immunology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Artem Barski
- Division of Allergy and Immunology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Human Genetics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Matthew T Weirauch
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Center for Autoimmune Genomics and Etiology, Division of Biomedical Informatics and Division of Developmental Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Patricia C Fulkerson
- Division of Allergy and Immunology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Marc E Rothenberg
- Division of Allergy and Immunology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
19
|
Dagher R, Kumar V, Copenhaver AM, Gallagher S, Ghaedi M, Boyd J, Newbold P, Humbles AA, Kolbeck R. Novel mechanisms of action contributing to Benralizumab's potent anti-eosinophilic activity. Eur Respir J 2021; 59:13993003.04306-2020. [PMID: 34289975 PMCID: PMC8923056 DOI: 10.1183/13993003.04306-2020] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 07/07/2021] [Indexed: 11/05/2022]
Abstract
Benralizumab is a humanised, anti-IL-5Rα monoclonal antibody with anti-eosinophilic activity. Lack of fucose (afucosylation) increases its affinity to CD16a and significantly enhances antibody-dependent cell-mediated cytotoxicity (ADCC) by NK cells. Although benralizumab proved clinically efficacious in clinical trials for patients with severe asthma and hypereosinophilic syndrome, in-depth characterisation of its anti-eosinophilic mechanisms of action remain elusive. Here, we further investigated the mechanisms involved in benralizumab's anti-eosinophilic activities. In the presence of NK cells benralizumab induced potent eosinophil apoptosis as demonstrated by the upstream induction of caspase 3/7 and upregulation of cytochrome C. In addition, we uncovered a previously unrecognised mechanism whereby benralizumab can induce eosinophil phagocytosis/efferocytosis by macrophages, a process called antibody-dependent cell phagocytosis (ADCP). Using live cell imaging we unravel the stepwise processes leading to eosinophil apoptosis and uptake by activated macrophages. Through careful observations of cellular co-culture assays we identified a novel role for macrophage derived TNF to further enhance benralizumab-mediated eosinophil apoptosis through activation of TNF-receptor 1 on eosinophils. TNF-induced eosinophil apoptosis was associated with Cytochrome C upregulation, mitochondrial membrane depolarisation, and increased caspase 3/7 activity. Moreover, activated NK cells were found to amplify this axis through the secretion of IFNγ, subsequently driving TNF expression by macrophages. Our data provide insights into the timely appearance of events leading to benralizumab-induced eosinophil apoptosis and suggest that additional mechanisms may contribute to the potent anti-eosinophilic activity of benralizumab in vivo Importantly, afucosylation of benralizumab strongly enhanced its potency for all mechanisms investigated.
Collapse
Affiliation(s)
- Rania Dagher
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Varsha Kumar
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Alan M Copenhaver
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Sandra Gallagher
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Mahboobe Ghaedi
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Jonathan Boyd
- Imaging Core, ADPE, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Paul Newbold
- Late Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Alison A Humbles
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Roland Kolbeck
- Bioscience COPD/IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| |
Collapse
|
20
|
Lee LY, Hew GSY, Mehta M, Shukla SD, Satija S, Khurana N, Anand K, Dureja H, Singh SK, Mishra V, Singh PK, Gulati M, Prasher P, Aljabali AAA, Tambuwala MM, Thangavelu L, Panneerselvam J, Gupta G, Zacconi FC, Shastri M, Jha NK, Xenaki D, MacLoughlin R, Oliver BG, Chellappan DK, Dua K. Targeting eosinophils in respiratory diseases: Biological axis, emerging therapeutics and treatment modalities. Life Sci 2021; 267:118973. [PMID: 33400932 DOI: 10.1016/j.lfs.2020.118973] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023]
Abstract
Eosinophils are bi-lobed, multi-functional innate immune cells with diverse cell surface receptors that regulate local immune and inflammatory responses. Several inflammatory and infectious diseases are triggered with their build up in the blood and tissues. The mobilization of eosinophils into the lungs is regulated by a cascade of processes guided by Th2 cytokine generating T-cells. Recruitment of eosinophils essentially leads to a characteristic immune response followed by airway hyperresponsiveness and remodeling, which are hallmarks of chronic respiratory diseases. By analysing the dynamic interactions of eosinophils with their extracellular environment, which also involve signaling molecules and tissues, various therapies have been invented and developed to target respiratory diseases. Having entered clinical testing, several eosinophil targeting therapeutic agents have shown much promise and have further bridged the gap between theory and practice. Moreover, researchers now have a clearer understanding of the roles and mechanisms of eosinophils. These factors have successfully assisted molecular biologists to block specific pathways in the growth, migration and activation of eosinophils. The primary purpose of this review is to provide an overview of the eosinophil biology with a special emphasis on potential pharmacotherapeutic targets. The review also summarizes promising eosinophil-targeting agents, along with their mechanisms and rationale for use, including those in developmental pipeline, in clinical trials, or approved for other respiratory disorders.
Collapse
Affiliation(s)
- Li-Yen Lee
- School of Pharmacy, International Medical University (IMU), Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Geena Suet Yin Hew
- School of Pharmacy, International Medical University (IMU), Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Meenu Mehta
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, Newcastle, NSW 2305, Australia
| | - Saurabh Satija
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Navneet Khurana
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Krishnan Anand
- Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences and National Health Laboratory Service, University of the Free State, Bloemfontein, South Africa
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana 124001, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Dehradun 248007, India
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Jithendra Panneerselvam
- Department of Pharmaceutical Technology, International Medical University (IMU), Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Jaipur 302017, India
| | - Flavia C Zacconi
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile; Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Madhur Shastri
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart 7005, Australia
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida 201310, India
| | - Dikaia Xenaki
- Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Ronan MacLoughlin
- Aerogen, IDA Business Park, Dangan, H91 HE94 Galway, Ireland; School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; School of Pharmacy and Pharmaceutical Sciences, Trinity College, D02 PN40 Dublin, Ireland
| | - Brian G Oliver
- Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia; School of Life Sciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia.
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University (IMU), Bukit Jalil, 57000 Kuala Lumpur, Malaysia.
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia; Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, Newcastle, NSW 2305, Australia; School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh 173229, India.
| |
Collapse
|
21
|
Tiwary M, LeMessurier KS, Samarasinghe AE. Murine Models of Eosinophil Function in Fungal and Viral Infections. Methods Mol Biol 2021; 2241:99-112. [PMID: 33486731 PMCID: PMC8142467 DOI: 10.1007/978-1-0716-1095-4_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Eosinophils are granulocytes that were historically considered to be terminally differentiated at the time of bone marrow egress. However, more recent evidence provides a new outlook on these cells as complex immunomodulators that are involved in host defense and homeostasis. Our work established a role for eosinophils as mediators of antiviral immune responses during influenza in hosts that were sensitized and challenged with fungal allergens. Herein, we describe methods for working with murine eosinophils in the context of influenza A virus.
Collapse
Affiliation(s)
- Meenakshi Tiwary
- Department of Pediatrics and Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
- Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN, USA
| | - Kim S LeMessurier
- Department of Pediatrics and Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
- Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN, USA
| | - Amali E Samarasinghe
- Department of Pediatrics and Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA.
- Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN, USA.
| |
Collapse
|
22
|
Abstract
The eosinophil is an enigmatic cell with a continuing ability to fascinate. A considerable history of research endeavor on eosinophil biology stretches from the present time back to the nineteenth century. Perhaps one of the most fascinating aspects of the eosinophil is how accumulating knowledge has changed the perception of its function from passive bystander, modulator of inflammation, to potent effector cell loaded with histotoxic substances through to more recent recognition that it can act as both a positive and negative regulator of complex events in both innate and adaptive immunity. This book consists of chapters written by experts in the field of eosinophil biology that provide comprehensive clearly written protocols for techniques designed to underpin research into the function of the eosinophil in health and disease.
Collapse
Affiliation(s)
- Paige Lacy
- Alberta Respiratory Centre (ARC) Research, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Helene F Rosenberg
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Garry M Walsh
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
23
|
Disrupting Bordetella Immunosuppression Reveals a Role for Eosinophils in Coordinating the Adaptive Immune Response in the Respiratory Tract. Microorganisms 2020; 8:microorganisms8111808. [PMID: 33212993 PMCID: PMC7698589 DOI: 10.3390/microorganisms8111808] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 12/20/2022] Open
Abstract
Recent findings revealed pivotal roles for eosinophils in protection against parasitic and viral infections, as well as modulation of adaptive immune responses in the gastric mucosa. However, the known effects of eosinophils within the respiratory tract remain predominantly pathological, associated with allergy and asthma. Simulating natural respiratory infections in mice, we examined how efficient and well-adapted pathogens can block eosinophil functions that contribute to the immune response. Bordetella bronchiseptica, a natural pathogen of the mouse, uses the sigma factor btrS to regulate expression of mechanisms that interfere with eosinophil recruitment and function. When btrS is disrupted, immunomodulators are dysregulated, and eosinophils are recruited to the lungs, suggesting they may contribute to much more efficient generation of adaptive immunity induced by this mutant. Eosinophil-deficient mice failed to produce pro-inflammatory cytokines, to recruit lymphocytes, to organize lymphoid aggregates that resemble Bronchus Associated Lymphoid Tissue (BALT), to generate an effective antibody response, and to clear bacterial infection from the respiratory tract. Importantly, the failure of eosinophil-deficient mice to produce these lymphoid aggregates indicates that eosinophils can mediate the generation of an effective lymphoid response in the lungs. These data demonstrate that efficient respiratory pathogens can block eosinophil recruitment, to inhibit the generation of robust adaptive immune responses. They also suggest that some post-infection sequelae involving eosinophils, such as allergy and asthma, might be a consequence of bacterial mechanisms that manipulate their accumulation and/or function within the respiratory tract.
Collapse
|
24
|
LeMessurier KS, Rooney R, Ghoneim HE, Liu B, Li K, Smallwood HS, Samarasinghe AE. Influenza A virus directly modulates mouse eosinophil responses. J Leukoc Biol 2020; 108:151-168. [PMID: 32386457 DOI: 10.1002/jlb.4ma0320-343r] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 03/09/2020] [Accepted: 03/16/2020] [Indexed: 12/14/2022] Open
Abstract
Allergic asthma and influenza are common respiratory diseases with a high probability of co-occurrence. During the 2009 influenza pandemic, hospitalized patients with influenza experienced lower morbidity if asthma was an underlying condition. We have previously demonstrated that acute allergic asthma protects mice from severe influenza and have implicated eosinophils in the airways of mice with allergic asthma as participants in the antiviral response. However, very little is known about how eosinophils respond to direct exposure to influenza A virus (IAV) or the microenvironment in which the viral burden is high. We hypothesized that eosinophils would dynamically respond to the presence of IAV through phenotypic, transcriptomic, and physiologic changes. Using our mouse model of acute fungal asthma and influenza, we showed that eosinophils in lymphoid tissues were responsive to IAV infection in the lungs and altered surface expression of various markers necessary for cell activation in a niche-specific manner. Siglec-F expression was altered in a subset of eosinophils after virus exposure, and those expressing high Siglec-F were more active (IL-5Rαhi CD62Llo ). While eosinophils exposed to IAV decreased their overall transcriptional activity and mitochondrial oxygen consumption, transcription of genes encoding viral recognition proteins, Ddx58 (RIG-I), Tlr3, and Ifih1 (MDA5), were up-regulated. CD8+ T cells from IAV-infected mice expanded in response to IAV PB1 peptide-pulsed eosinophils, and CpG methylation in the Tbx21 promoter was reduced in these T cells. These data offer insight into how eosinophils respond to IAV and help elucidate alternative mechanisms by which they regulate antiviral immune responses during IAV infection.
Collapse
Affiliation(s)
- Kim S LeMessurier
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Children's Foundation Research Institute, Memphis, Tennessee, USA
| | - Robert Rooney
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Genetics, Genomics & Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Children's Foundation Research Institute, Memphis, Tennessee, USA
| | - Hazem E Ghoneim
- Department of Immunology, St Jude Children's Research Hospital, Memphis, Tennessee, USA.,Department of Microbial Infection and Immunity, College of Medicine, Ohio State University, Columbus, Ohio, USA
| | - Baoming Liu
- Department of Microbiology, Immunology & Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Pathology, Division of Medical Microbiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kui Li
- Department of Microbiology, Immunology & Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Heather S Smallwood
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Children's Foundation Research Institute, Memphis, Tennessee, USA
| | - Amali E Samarasinghe
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Microbiology, Immunology & Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Children's Foundation Research Institute, Memphis, Tennessee, USA
| |
Collapse
|
25
|
Jackson DJ, Korn S, Mathur SK, Barker P, Meka VG, Martin UJ, Zangrilli JG. Safety of Eosinophil-Depleting Therapy for Severe, Eosinophilic Asthma: Focus on Benralizumab. Drug Saf 2020; 43:409-425. [PMID: 32242310 PMCID: PMC7165132 DOI: 10.1007/s40264-020-00926-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Eosinophils play a pivotal role in the inflammatory pathology of asthma and have been the target of new biologic treatments for patients with eosinophilic asthma. Given the central role of interleukin (IL)-5 in the eosinophil lifecycle, several therapies directed against the IL-5 pathway have been developed, including the anti-IL-5 antibodies mepolizumab and reslizumab and the IL-5 receptor α (IL-5Rα)-directed cytolytic antibody benralizumab. Eosinophil-depleting therapies represent a relatively new class of asthma treatment, and it is important to understand their long-term efficacy and safety. Eosinophils have been associated with host protection and tumor growth, raising potential concerns about the consequences of long-term therapies that deplete eosinophils. However, evidence for these associations in humans is conflicting and largely indirect or based on mouse models. Substantial prospective clinical trial and postmarketing data have accrued, providing insight into the potential risks associated with eosinophil depletion. In this review, we explore the current safety profile of eosinophil-reducing therapies, with particular attention to the potential risks of malignancies and severe infections and a focus on benralizumab. Benralizumab is an IL-5Rα-directed cytolytic monoclonal antibody that targets and efficiently depletes blood and tissue eosinophils through antibody-dependent cell-mediated cytotoxicity. Benralizumab is intended to treat patients with severe, uncontrolled asthma with eosinophilic inflammation. The integrated analyses of benralizumab safety data from the phase III SIROCCO and CALIMA trials and subsequent BORA extension trial for patients with asthma, and the phase III GALATHEA and TERRANOVA trials for patients with chronic obstructive pulmonary disease, form the principal basis for this review.
Collapse
Affiliation(s)
- David J Jackson
- Guy's Severe Asthma Centre, Guy's & St Thomas' NHS Trust, London, UK
- Asthma UK Centre, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Stephanie Korn
- Universitätsmedizin Mainz, Langenbeckstr, Mainz, Germany
| | - Sameer K Mathur
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Peter Barker
- Research and Development, AstraZeneca, Gaithersburg, MD, USA
| | | | - Ubaldo J Martin
- Research and Development, AstraZeneca, Gaithersburg, MD, USA
| | - James G Zangrilli
- Global Medical Affairs, BioPharmaceuticals Medical, AstraZeneca, One MedImmune Way, Gaithersburg, MD, 20878, USA.
| |
Collapse
|
26
|
Hossain FMA, Choi JY, Uyangaa E, Park SO, Eo SK. The Interplay between Host Immunity and Respiratory Viral Infection in Asthma Exacerbation. Immune Netw 2019; 19:e31. [PMID: 31720042 PMCID: PMC6829071 DOI: 10.4110/in.2019.19.e31] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/22/2019] [Accepted: 08/25/2019] [Indexed: 12/16/2022] Open
Abstract
Asthma is one of the most common and chronic diseases characterized by multidimensional immune responses along with poor prognosis and severity. The heterogeneous nature of asthma may be attributed to a complex interplay between risk factors (either intrinsic or extrinsic) and specific pathogens such as respiratory viruses, and even bacteria. The intrinsic risk factors are highly correlated with asthma exacerbation in host, which may be mediated via genetic polymorphisms, enhanced airway epithelial lysis, apoptosis, and exaggerated viral replication in infected cells, resulting in reduced innate immune response and concomitant reduction of interferon (types I, II, and III) synthesis. The canonical features of allergic asthma include strong Th2-related inflammation, sensitivity to non-steroidal anti-inflammatory drugs (NSAIDs), eosinophilia, enhanced levels of Th2 cytokines, goblet cell hyperplasia, airway hyper-responsiveness, and airway remodeling. However, the NSAID-resistant non-Th2 asthma shows a characteristic neutrophilic influx, Th1/Th17 or even mixed (Th17-Th2) immune response and concurrent cytokine streams. Moreover, inhaled corticosteroid-resistant asthma may be associated with multifactorial innate and adaptive responses. In this review, we will discuss the findings of various in vivo and ex vivo models to establish the critical heterogenic asthmatic etiologies, host-pathogen relationships, humoral and cell-mediated immune responses, and subsequent mechanisms underlying asthma exacerbation triggered by respiratory viral infections.
Collapse
Affiliation(s)
- Ferdaus Mohd Altaf Hossain
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan 54596, Korea.,Faculty of Veterinary, Animal and Biomedical Sciences, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Jin Young Choi
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan 54596, Korea
| | - Erdenebileg Uyangaa
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan 54596, Korea
| | - Seong Ok Park
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan 54596, Korea
| | - Seong Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan 54596, Korea
| |
Collapse
|
27
|
Abstract
Among the monoclonal antibodies (mAbs) developed for severe asthma treatment, three have already been marketed. Omalizumab was the first, more than 10 years ago; today, mepolizumab and reslizumab are also available in the European Union and the US. Omalizumab blocks free immunoglobulin E (IgE), mepolizumab and reslizumab block an interleukin (IL-5). In the near future, dupilumab and benralizumab are expected to emerge as two new alternatives. Benralizumab blocks the receptor for IL-5 (IL5-Rα) and has a direct cytotoxic effect on eosinophils, and dupilumab blocks the α-unit of the heterodimeric receptor for IL-4 and IL-13 (IL-4Rα); as a result, dupilumab can block both IL-4 and IL-13. The purpose of this manuscript is to present the pathophysiology of some immunological aspects of severe asthma, describe the adaptive and innate immunity arms as well as their interrelations (stressing the subordination of the adaptive arm to the innate arm), outline the pharmacologic effects of these mAbs, clarify the overlapping effects of the different mAbs, and discuss the differences between mAbs based on their target molecules. Based on the data presented, I propose omalizumab for patients with an allergic phenotype regardless of their peripheral eosinophilic count, and anti-IL-5 as an alternative in allergic patients with blood eosinophilia in which omalizumab has failed; anti-IL5 for patients with an eosinophilic phenotype and omalizumab as an alternative in patients in whom anti-IL5 fails and IgE ≥30 IU/mL (compassionate use). Omalizumab is also proposed for patients with severe chronic asthma allergic to seasonal allergens.
Collapse
|
28
|
Leiferman KM, Peters MS. Eosinophil-Related Disease and the Skin. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2018; 6:1462-1482.e6. [DOI: 10.1016/j.jaip.2018.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/04/2018] [Accepted: 06/04/2018] [Indexed: 02/06/2023]
|
29
|
Park EJ, Khaliullin TO, Shurin MR, Kisin ER, Yanamala N, Fadeel B, Chang J, Shvedova AA. Fibrous nanocellulose, crystalline nanocellulose, carbon nanotubes, and crocidolite asbestos elicit disparate immune responses upon pharyngeal aspiration in mice. J Immunotoxicol 2018; 15:12-23. [PMID: 29237319 DOI: 10.1080/1547691x.2017.1414339] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
With the rapid development of synthetic alternatives to mineral fibers, their possible effects on the environment and human health have become recognized as important issues worldwide. This study investigated effects of four fibrous materials, i.e. nanofibrillar/nanocrystalline celluloses (NCF and CNC), single-walled carbon nanotubes (CNTs), and crocidolite asbestos (ASB), on pulmonary inflammation and immune responses found in the lungs, as well as the effects on spleen and peripheral blood immune cell subsets. BALB/c mice were given NCF, CNC, CNT, and ASB on Day 1 by oropharyngeal aspiration. At 14 days post-exposure, the animals were evaluated. Total cell number, mononuclear phagocytes, polymorphonuclear leukocytes, lymphocytes, and LDH levels were significantly increased in ASB and CNT-exposed mice. Expression of cytokines and chemokines in bronchoalveolar lavage (BAL) was quite different in mice exposed to four particle types, as well as expression of antigen presentation-related surface proteins on BAL cells. The results revealed that pulmonary exposure to fibrous materials led to discrete local immune cell polarization patterns with a TH2-like response caused by ASB and TH1-like immune reaction to NCF, while CNT and CNC caused non-classical or non-uniform responses. These alterations in immune response following pulmonary exposure should be taken into account when testing the applicability of new nanosized materials with fibrous morphology.
Collapse
Affiliation(s)
- Eun-Jung Park
- a Department of Brain Science , Ajou University School of Medicine , Suwon , Republic of Korea
| | - Timur O Khaliullin
- b Exposure Assessment Branch , NIOSH/CDC , Morgantown , WV , USA.,c Department of Physiology, Pharmacology and Neuroscience , West Virginia University , Morgantown , WV , USA
| | - Michael R Shurin
- d Department of Pathology and Immunology , University of Pittsburgh , Pittsburgh , PA , USA
| | - Elena R Kisin
- b Exposure Assessment Branch , NIOSH/CDC , Morgantown , WV , USA
| | - Naveena Yanamala
- b Exposure Assessment Branch , NIOSH/CDC , Morgantown , WV , USA
| | - Bengt Fadeel
- e Division of Molecular Toxicology, Institute of Environmental Medicine , Karolinska Institute , Stockholm , Sweden
| | - Jaerak Chang
- a Department of Brain Science , Ajou University School of Medicine , Suwon , Republic of Korea.,f Graduate School of Biomedical Sciences , Ajou University School of Medicine , Suwon , Republic of Korea
| | - Anna A Shvedova
- b Exposure Assessment Branch , NIOSH/CDC , Morgantown , WV , USA.,c Department of Physiology, Pharmacology and Neuroscience , West Virginia University , Morgantown , WV , USA
| |
Collapse
|
30
|
Xenakis JJ, Howard ED, Smith KM, Olbrich CL, Huang Y, Anketell D, Maldonado S, Cornwell EW, Spencer LA. Resident intestinal eosinophils constitutively express antigen presentation markers and include two phenotypically distinct subsets of eosinophils. Immunology 2018; 154:298-308. [PMID: 29281125 PMCID: PMC5980140 DOI: 10.1111/imm.12885] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/13/2017] [Accepted: 12/14/2017] [Indexed: 02/05/2023] Open
Abstract
Intestinal eosinophils are implicated in homeostatic and disease-associated processes, yet the phenotype of intestinal tissue-dwelling eosinophils is poorly defined and their roles in intestinal health or disease remain enigmatic. Here we probed the phenotype and localization of eosinophils constitutively homed to the small intestine of naive mice at baseline, and of antigen-sensitized mice following intestinal challenge. Eosinophils homed to the intestinal lamina propria of naive mice were phenotypically distinguished from autologous blood eosinophils, and constitutively expressed antigen-presenting cell markers, suggesting that intestinal eosinophils, unlike blood eosinophils, may be primed for antigen presentation. We further identified a previously unrecognized resident population of CD11chi eosinophils that are recovered with intraepithelial leucocytes, and that are phenotypically distinct from both lamina propria and blood eosinophils. To better visualize intestinal eosinophils in situ, we generated eosinophil reporter mice wherein green fluorescent protein expression is targeted to both granule-delimiting and plasma membranes. Analyses of deconvolved fluorescent z-section image stacks of intestinal tissue sections from eosinophil reporter mice revealed eosinophils within intestinal villi exhibited dendritic morphologies with cellular extensions that often contacted the basement membrane. Using an in vivo model of antigen acquisition in antigen-sensitized mice, we demonstrate that both lamina propria-associated and intraepithelium-associated eosinophils encounter, and are competent to acquire, lumen-derived antigen. Taken together these data provide new foundational insights into the organization and functional potential of intestinal tissue-dwelling eosinophils, including the recognition of different subsets of resident intestinal eosinophils, and constitutive expression of antigen-presenting cell markers.
Collapse
Affiliation(s)
- Jason J. Xenakis
- Division of Allergy and InflammationDepartment of MedicineBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
| | - Emily D. Howard
- Division of Allergy and InflammationDepartment of MedicineBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
| | - Kalmia M. Smith
- Division of Allergy and InflammationDepartment of MedicineBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
| | - Courtney L. Olbrich
- Division of Allergy and InflammationDepartment of MedicineBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
- Department of BiologyGordon CollegeWenhamMAUSA
| | - Yanjun Huang
- Division of Allergy and InflammationDepartment of MedicineBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
- Department of BiologyGordon CollegeWenhamMAUSA
| | | | - Samuel Maldonado
- Division of Allergy and InflammationDepartment of MedicineBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
| | - Evangeline W. Cornwell
- Division of Allergy and InflammationDepartment of MedicineBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
- Department of BiologyGordon CollegeWenhamMAUSA
| | - Lisa A. Spencer
- Division of Allergy and InflammationDepartment of MedicineBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
| |
Collapse
|
31
|
Esnault S, Hebert AS, Jarjour NN, Coon JJ, Mosher DF. Proteomic and Phosphoproteomic Changes Induced by Prolonged Activation of Human Eosinophils with IL-3. J Proteome Res 2018; 17:2102-2111. [PMID: 29706072 DOI: 10.1021/acs.jproteome.8b00057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Purified human eosinophils treated for 18-24 h with IL-3 adopt a unique activated phenotype marked by increased reactivity to aggregated immunoglobulin-G (IgG). To characterize this phenotype, we quantified protein abundance and phosphorylation by multiplexed isobaric labeling combined with high-resolution mass spectrometry. Purified blood eosinophils of five individuals were treated with IL-3 or no cytokine for 20 h, and comparative data were obtained on abundance of 5385 proteins and phosphorylation at 7330 sites. The 1150 proteins that were significantly up-regulated ( q < 0.05, pairwise t test with Benjamini-Hochberg correction) by IL-3 included the IL3RA and CSF2RB subunits of the IL-3 receptor, the low-affinity receptor for IgG (FCGR2B), 96 proteins involved in protein translation, and 55 proteins involved in cytoskeleton organization. Among the 703 proteins that decreased were 78 mitochondrial proteins. Dynamic regulation of protein phosphorylation was detected at 4218 sites. These included multiple serines in CSF2RB; Y694 of STAT5, a key site of activating phosphorylation downstream of IL3RA/CSF2RB; and multiple sites in RPS6KA1, RPS6, and EIF4B, which are responsible for translational initiation. We conclude that IL-3 up-regulates overall protein synthesis and targets specific proteins for up-regulation, including its own receptor.
Collapse
Affiliation(s)
- Stephane Esnault
- Department of Medicine , University of Wisconsin , Madison , Wisconsin 53792 , United States
| | - Alexander S Hebert
- Department of Chemistry , University of Wisconsin , Madison , Wisconsin 53706 , United States
| | - Nizar N Jarjour
- Department of Medicine , University of Wisconsin , Madison , Wisconsin 53792 , United States
| | - Joshua J Coon
- Department of Chemistry , University of Wisconsin , Madison , Wisconsin 53706 , United States.,Department of Biomolecular Chemistry , University of Wisconsin , Madison , Wisconsin 53706 , United States.,Morgridge Institute for Research , Madison , Wisconsin 53715 , United States.,Genome Center of Wisconsin , Madison , Wisconsin 53706 , United States
| | - Deane F Mosher
- Department of Medicine , University of Wisconsin , Madison , Wisconsin 53792 , United States.,Department of Biomolecular Chemistry , University of Wisconsin , Madison , Wisconsin 53706 , United States
| |
Collapse
|
32
|
Goldmann O, Beineke A, Medina E. Identification of a Novel Subset of Myeloid-Derived Suppressor Cells During Chronic Staphylococcal Infection That Resembles Immature Eosinophils. J Infect Dis 2017; 216:1444-1451. [PMID: 29029332 DOI: 10.1093/infdis/jix494] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 09/14/2017] [Indexed: 12/13/2022] Open
Abstract
We have previously reported that myeloid-derived suppressor cells (MDSC), which are a heterogeneous population of immunosuppressive immature myeloid cells, expanded during chronic Staphylococcus aureus infection and promoted bacterial persistence by inhibiting effector T cells. Two major MDSC subsets, including monocytic MDSC and granulocytic MDSC, have been described to date. Here, we identified a new subset of MDSC (Eo-MDSC) in S. aureus-infected mice that phenotypically resembles eosinophils. Eo-MDSC exhibit eosinophilic cytoplasmic granules and express CD11b, the eosinophil marker Syglec-F, variable levels of CCR3, and low levels of interleukin-5Rα. Furthermore, Eo-MDSC accumulated at the site of infection and exerted a potent immunosuppressive effect on T-cell responses that was mediated by nitric oxide-dependent depletion of l-arginine. Increases in the number of Eo-MDSC by adoptive transfer caused a significant exacerbation of infection in S. aureus-infected mice. This study sheds new light on the heterogeneity and complexity of MDSC during chronic infection.
Collapse
Affiliation(s)
- Oliver Goldmann
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Braunschweig
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, Germany
| | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Braunschweig
| |
Collapse
|
33
|
McBrien CN, Menzies-Gow A. The Biology of Eosinophils and Their Role in Asthma. Front Med (Lausanne) 2017; 4:93. [PMID: 28713812 PMCID: PMC5491677 DOI: 10.3389/fmed.2017.00093] [Citation(s) in RCA: 246] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/13/2017] [Indexed: 12/22/2022] Open
Abstract
This review will describe the structure and function of the eosinophil. The roles of several relevant cell surface molecules and receptors will be discussed. We will also explore the systemic and local processes triggering eosinophil differentiation, maturation, and migration to the lungs in asthma, as well as the cytokine-mediated pathways that result in eosinophil activation and degranulation, i.e., the release of multiple pro-inflammatory substances from eosinophil-specific granules, including cationic proteins, cytokines, chemokines growth factors, and enzymes. We will discuss the current understanding of the roles that eosinophils play in key asthma processes such as airway hyperresponsiveness, mucus hypersecretion, and airway remodeling, in addition to the evidence relating to eosinophil–pathogen interactions within the lungs.
Collapse
Affiliation(s)
| | - Andrew Menzies-Gow
- Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
34
|
Stadejek T, Larsen LE, Podgórska K, Bøtner A, Botti S, Dolka I, Fabisiak M, Heegaard PMH, Hjulsager CK, Huć T, Kvisgaard LK, Sapierzyński R, Nielsen J. Pathogenicity of three genetically diverse strains of PRRSV Type 1 in specific pathogen free pigs. Vet Microbiol 2017; 209:13-19. [PMID: 28554869 PMCID: PMC7127113 DOI: 10.1016/j.vetmic.2017.05.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 05/11/2017] [Indexed: 01/28/2023]
Abstract
The pathogenicity of two PRRSV-1 strains BOR59, ILI6 and 18794 was compared specific pathogen free pigs. The most pronounced clinical signs, the highest viremia and the earliest acute phase proteins response were in pigs infected with BOR59 strain. BOR59 strain can be considered a highly virulent strain, while the virulence of strain ILI6 was intermediate between BOR59 and 18794.
Studies from Eastern European countries proved that porcine reproductive and respiratory syndrome virus Type 1 (PRRSV-1) harbours high genetic diversity and that genetically divergent subtypes 2–4 circulate in this area. In the present study, we compared the pathogenicity of two different PRRSV-1 subtype 2 strains and a strain representing PRRSV-1 subtype 1. Four groups of 8-week-old specific pathogen free pigs were either infected with subtype 2 strain ILI6, subtype 2 strain or BOR59, subtype 1 strain 18794, or mock inoculated. The most pronounced clinical signs were observed in pigs infected with BOR59. Pigs from both subtype 2 strain infected groups exhibited significantly elevated mean body temperatures on DPI 2 compared to the other two groups, the difference remaining significant up to DPI 13 for the BOR59 group, only. The pigs in the latter group also displayed significantly highest levels of early viremia together with the most rapid APP response. Overall, the results indicated that BOR59 strain can be considered a highly pathogenic strain, similarly to subtype 3 strains Lena and SU1-bel, while the virulence of the other subtype 2 strain ILI6 was intermediate between BOR59 and subtype 1 strain.
Collapse
Affiliation(s)
- Tomasz Stadejek
- Warsaw University of Life Sciences, Faculty of Veterinary Medicine, Department of Pathology and Veterinary Diagnostics, Nowoursynowska 159C, 02-776 Warsaw, Poland.
| | - Lars E Larsen
- National Veterinary Institute, Technical University of Denmark, Bülowsvej 27, 1870 Frederiksberg C, Denmark
| | - Katarzyna Podgórska
- National Veterinary Research Institute, Swine Diseases Department, Partyzantów 57, 24-100 Puławy, Poland
| | - Anette Bøtner
- National Veterinary Institute, Technical University of Denmark, Lindholm, Kalvehave 4771, Denmark
| | - Sara Botti
- Parco Tecnologico Padano, Polo Universitario, Via Einstein 26900 Lodi, Italy
| | - Izabella Dolka
- Warsaw University of Life Sciences, Faculty of Veterinary Medicine, Department of Pathology and Veterinary Diagnostics, Nowoursynowska 159C, 02-776 Warsaw, Poland
| | - Michał Fabisiak
- Warsaw University of Life Sciences, Faculty of Veterinary Medicine, Department of Pathology and Veterinary Diagnostics, Nowoursynowska 159C, 02-776 Warsaw, Poland
| | - Peter M H Heegaard
- National Veterinary Institute, Technical University of Denmark, Bülowsvej 27, 1870 Frederiksberg C, Denmark
| | - Charlotte K Hjulsager
- National Veterinary Institute, Technical University of Denmark, Bülowsvej 27, 1870 Frederiksberg C, Denmark
| | - Tomasz Huć
- Warsaw University of Life Sciences, Faculty of Veterinary Medicine, Department of Pathology and Veterinary Diagnostics, Nowoursynowska 159C, 02-776 Warsaw, Poland
| | - Lise K Kvisgaard
- National Veterinary Institute, Technical University of Denmark, Bülowsvej 27, 1870 Frederiksberg C, Denmark
| | - Rafał Sapierzyński
- Warsaw University of Life Sciences, Faculty of Veterinary Medicine, Department of Pathology and Veterinary Diagnostics, Nowoursynowska 159C, 02-776 Warsaw, Poland
| | - Jens Nielsen
- National Veterinary Research Institute, Swine Diseases Department, Partyzantów 57, 24-100 Puławy, Poland
| |
Collapse
|
35
|
Pesce S, Thoren FB, Cantoni C, Prato C, Moretta L, Moretta A, Marcenaro E. The Innate Immune Cross Talk between NK Cells and Eosinophils Is Regulated by the Interaction of Natural Cytotoxicity Receptors with Eosinophil Surface Ligands. Front Immunol 2017; 8:510. [PMID: 28503177 PMCID: PMC5408020 DOI: 10.3389/fimmu.2017.00510] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 04/13/2017] [Indexed: 01/21/2023] Open
Abstract
Previous studies suggested that the cross talk between NK cells and other cell types is crucial for the regulation of both innate and adaptive immune responses. In the present study, we analyzed the phenotypic and functional outcome of the interaction between resting or cytokine-activated NK cells and eosinophils derived from non-atopic donors. Our results provide the first evidence that a natural cytotoxicity receptor (NCR)/NCR ligand-dependent cross talk between NK cells and eosinophils may be important to upregulate the activation state and the effector function of cytokine-primed NK cells. This interaction also promotes the NK-mediated editing process of dendritic cells that influence the process of Th1 polarization. In turn, this cross talk also resulted in eosinophil activation and acquisition of the characteristic features of antigen-presenting cells. At higher NK/eosinophil ratios, cytokine-primed NK cells were found to kill eosinophils via NKp46 and NKp30, thus suggesting a potential immunoregulatory role for NK cells in dampening inflammatory responses involving eosinophils.
Collapse
Affiliation(s)
- Silvia Pesce
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genova, Italy
| | - Fredrik B Thoren
- Sahlgrenska Cancer Center, University of Gothenburg, Göteborg, Sweden
| | - Claudia Cantoni
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genova, Italy.,Istituto Giannina Gaslini, Genova, Italy.,Centro di Eccellenza per le Ricerche Biomediche, Università degli Studi di Genova, Genova, Italy
| | - Carola Prato
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genova, Italy
| | - Lorenzo Moretta
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Alessandro Moretta
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genova, Italy.,Centro di Eccellenza per le Ricerche Biomediche, Università degli Studi di Genova, Genova, Italy.,Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Emanuela Marcenaro
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genova, Italy.,Centro di Eccellenza per le Ricerche Biomediche, Università degli Studi di Genova, Genova, Italy
| |
Collapse
|
36
|
Rosenberg HF, Masterson JC, Furuta GT. Eosinophils, probiotics, and the microbiome. J Leukoc Biol 2016; 100:881-888. [PMID: 27549754 PMCID: PMC6608069 DOI: 10.1189/jlb.3ri0416-202r] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/24/2016] [Accepted: 07/26/2016] [Indexed: 12/17/2022] Open
Abstract
There is currently substantial interest in the therapeutic properties of probiotic microorganisms as recent research suggests that oral administration of specific bacterial strains may reduce inflammation and alter the nature of endogenous microflora in the gastrointestinal tract. Eosinophils are multifunctional tissue leukocytes, prominent among the resident cells of the gastrointestinal mucosa that promote local immunity. Recent studies with genetically altered mice indicate that eosinophils not only participate in maintaining gut homeostasis, but that the absence of eosinophils may have significant impact on the nature of the endogenous gut microflora and responses to gut pathogens, notably Clostridium difficile Furthermore, in human subjects, there is an intriguing relationship between eosinophils, allergic inflammation, and the nature of the lung microflora, notably a distinct association between eosinophil infiltration and detection of bacteria of the phylum Actinobacteria. Among topics for future research, it will be important to determine whether homeostatic mechanisms involve direct interactions between eosinophils and bacteria or whether they involve primarily eosinophil-mediated responses to cytokine signaling in the local microenvironment. Likewise, although is it clear that eosinophils can and do interact with bacteria in vivo, their ability to discern between pathogenic and probiotic species in various settings remains to be explored.
Collapse
Affiliation(s)
- Helene F Rosenberg
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA;
| | - Joanne C Masterson
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics and Digestive Health Institute, Children's Hospital Colorado, Aurora, Colorado, USA; and
- Department of Medicine, Mucosal Inflammation Program, University of Colorado School of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Glenn T Furuta
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics and Digestive Health Institute, Children's Hospital Colorado, Aurora, Colorado, USA; and
- Department of Medicine, Mucosal Inflammation Program, University of Colorado School of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
37
|
Farhan RK, Vickers MA, Ghaemmaghami AM, Hall AM, Barker RN, Walsh GM. Effective antigen presentation to helper T cells by human eosinophils. Immunology 2016; 149:413-422. [PMID: 27502559 DOI: 10.1111/imm.12658] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/29/2016] [Accepted: 08/01/2016] [Indexed: 12/20/2022] Open
Abstract
Although eosinophils are inflammatory cells, there is increasing attention on their immunomodulatory roles. For example, murine eosinophils can present antigen to CD4+ T helper (Th) cells, but it remains unclear whether human eosinophils also have this ability. This study determined whether human eosinophils present a range of antigens, including allergens, to activate Th cells, and characterized their expression of MHC class II and co-stimulatory molecules required for effective presentation. Human peripheral blood eosinophils purified from non-allergic donors were pulsed with the antigens house dust mite extract (HDM), Timothy Grass extract (TG) or Mycobacterium tuberculosis purified protein derivative (PPD), before co-culture with autologous CD4+ Th cells. Proliferative and cytokine responses were measured, with eosinophil expression of HLA-DR/DP/DQ and the co-stimulatory molecules CD40, CD80 and CD86 determined by flow cytometry. Eosinophils pulsed with HDM, TG or PPD drove Th cell proliferation, with the response strength dependent on antigen concentration. The cytokine responses varied with donor and antigen, and were not biased towards any particular Th subset, often including combinations of pro- and anti-inflammatory cytokines. Eosinophils up-regulated surface expression of HLA-DR/DP/DQ, CD80, CD86 and CD40 in culture, increases that were sustained over 5 days when incubated with antigens, including HDM, or the major allergens it contains, Der p I or Der p II. Human eosinophils can, therefore, act as effective antigen-presenting cells to stimulate varied Th cell responses against a panel of antigens including HDM, TG or PPD, an ability that may help to determine the development of allergic disease.
Collapse
Affiliation(s)
- Ruhaifah K Farhan
- Immunity, Infection and Inflammation Programme, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Mark A Vickers
- Immunity, Infection and Inflammation Programme, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Amir M Ghaemmaghami
- Cellular Immunology & Allergy Group, Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, Queen's Medical Centre, Nottingham, UK
| | - Andrew M Hall
- Immunity, Infection and Inflammation Programme, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK.
| | - Robert N Barker
- Immunity, Infection and Inflammation Programme, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Garry M Walsh
- Immunity, Infection and Inflammation Programme, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
38
|
Fu CH, Tsai WC, Lee TJ, Huang CC, Chang PH, Su Pang JH. Simvastatin Inhibits IL-5-Induced Chemotaxis and CCR3 Expression of HL-60-Derived and Human Primary Eosinophils. PLoS One 2016; 11:e0157186. [PMID: 27275740 PMCID: PMC4898827 DOI: 10.1371/journal.pone.0157186] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 05/25/2016] [Indexed: 12/27/2022] Open
Abstract
IL-5-induced chemotaxis of eosinophils is an important feature of allergic airway inflammatory diseases. Simvastatin, a lipid lowering agent, has been shown to exhibit anti-inflammatory and anti-allergic effects. Our aim was to investigate the effect of simvastatin on IL-5-induced eosinophil chemotaxis and its regulatory mechanisms. Eosinophils were derived by treating HL-60 clone 15 (HC15) cells with butyric acid (BA) in an alkaline condition or through direct isolation from human peripheral blood. The expressions of CC chemokine receptor 3 (CCR3) and interleukin (IL)-5 receptors (IL5Rα and β) were analyzed using RT/real-time PCR. The granular proteins were stained using fast green. Eotaxin-induced chemotaxis was measured using a transwell migration assay. CCR3 protein expression was revealed by immunocytochemistry. An animal model of allergic rhinitis was established by challenging Sprague-Dawley® rats repeatedly with ovalbumin. Butyric acid significantly increased the expression of IL5Rα and IL5Rβ, CCR3 and granular proteins in HC15 cells, indicating the maturation of eosinophils (BA-E cells). IL-5 further enhanced the CCR3 expression at both the mRNA and protein levels and the eotaxin-induced chemotaxis of BA-E cells. Simvastatin inhibited the effects of IL-5 on BA-E cells, but not in the presence of mevalonate. Similar results were also exhibited in human primary eosinophils. In vivo animal studies further confirmed that oral simvastatin could significantly suppress the infiltration of eosinophils into turbinate tissues of allergic rats. Therefore, simvastatin was demonstrated to inhibit IL-5-induced CCR3 expression and chemotaxis of eosinophils mediated via the mevalonate pathway. We confirmed that simvastatin also reduced eosinophilic infiltration in allergic rhinitis.
Collapse
Affiliation(s)
- Chia-Hsiang Fu
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Tao-Yuan City, Taiwan, ROC
- Department of Otolaryngology, Chang Gung Memorial Hospital, Tao-Yuan City, Taiwan, ROC
| | - Wan-Chun Tsai
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Tao-Yuan City, Taiwan, ROC
- Department of Respiratory Therapy, College of Medicine, Chang Gung University, Tao-Yuan City, Taiwan, ROC
| | - Ta-Jen Lee
- Department of Otolaryngology, Chang Gung Memorial Hospital, Tao-Yuan City, Taiwan, ROC
| | - Chi-Che Huang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Tao-Yuan City, Taiwan, ROC
- Department of Otolaryngology, Chang Gung Memorial Hospital, Tao-Yuan City, Taiwan, ROC
| | - Po-Hung Chang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Tao-Yuan City, Taiwan, ROC
- Department of Otolaryngology, Chang Gung Memorial Hospital, Tao-Yuan City, Taiwan, ROC
| | - Jong-Hwei Su Pang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Tao-Yuan City, Taiwan, ROC
- Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Linkou, Taoyuan City, Taiwan, ROC
- * E-mail:
| |
Collapse
|
39
|
Gangwar RS, Friedman S, Seaf M, Levi-Schaffer F. Mast cells and eosinophils in allergy: Close friends or just neighbors. Eur J Pharmacol 2016; 778:77-83. [DOI: 10.1016/j.ejphar.2015.10.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/21/2015] [Accepted: 10/21/2015] [Indexed: 12/15/2022]
|
40
|
Lotfi R, Kaltenmeier C, Lotze MT, Bergmann C. Until Death Do Us Part: Necrosis and Oxidation Promote the Tumor Microenvironment. Transfus Med Hemother 2016; 43:120-32. [PMID: 27226794 DOI: 10.1159/000444941] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 02/23/2016] [Indexed: 12/12/2022] Open
Abstract
Tumor proliferation is concomitant with autophagy, limited apoptosis, and resultant necrosis. Necrosis is associated with the release of damage-associated molecular pattern molecules (DAMPs), which act as 'danger signals', recruiting inflammatory cells, inducing immune responses, and promoting wound healing. Most of the current treatment strategies for cancer (chemotherapy, radiation therapy, hormonal therapy) promote DAMP release following therapy-induced tumor death by necroptosis and necrosis. Myeloid cells (monocytes, dendritic cells (DCs), and granulocytes), as well as mesenchymal stromal cells (MSCs) belong to the early immigrants in response to unscheduled cell death, initiating and modulating the subsequent inflammatory response. Responding to DAMPs, MSCs, and DCs promote an immunosuppressive milieu, while eosinophils induce oxidative conditions limiting the biologic activity of DAMPs over time and distance. Regulatory T cells are strongly affected by pattern recognition receptor signaling in the tumor microenvironment and limit immune reactivity coordinately with myeloid-derived suppressor cells. Means to 'aerobically' oxidize DAMPs provide a novel strategy for limiting tumor progression. The present article summarizes our current understanding of the impact of necrosis on the tumor microenvironment and the influence of oxidative conditions found within this setting.
Collapse
Affiliation(s)
- Ramin Lotfi
- Institute for Transfusion Medicine, University Hospital Ulm, Ulm, Germany; Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Services Baden-Württemberg-Hessen, Ulm, Germany
| | - Christof Kaltenmeier
- University of Pittsburgh Schools of the Health Sciences G.27A Hillman Cancer Center, Pittsburgh, PA, USA
| | - Michael T Lotze
- University of Pittsburgh Schools of the Health Sciences G.27A Hillman Cancer Center, Pittsburgh, PA, USA
| | - Christoph Bergmann
- Department of Otorhinolaryngology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
41
|
Wang W, Xian M, Xie Y, Zheng J, Li J. Aggravation of airway inflammation and hyper-responsiveness following nasal challenge with Dermatophagoides pteronyssinus in perennial allergic rhinitis without symptoms of asthma. Allergy 2016; 71:378-86. [PMID: 26595454 DOI: 10.1111/all.12808] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2015] [Indexed: 01/01/2023]
Abstract
BACKGROUND House dust mites are the most prevalent allergen causing sensitizations in patients with rhinitis and asthma in China. We aimed to investigate the changes in both upper and lower airway inflammation and responsiveness following Dermatophagoides pteronyssinus (Der-p) nasal provocation test (NPT) in rhinitis patients. METHODS Study subjects included 15 nonasthmatic Der-p-sensitized rhinitis (AR) patients with airway hyper-responsiveness (AHR) (AR+AHR+), 15 AR patients without AHR (AR+AHR-), 15 healthy controls (HCs) with Der-p sensitization (HC+DP+), and 15 HC without Der-p sensitization (HC+DP-). All subjects underwent Der-p NPT. Visual analogue scale (VAS) scores of nasal symptoms, nasal lavage and nasal airway resistance (NAR) measurement, sputum induction, and forced expiratory volume in 1 second (FEV1 ) were performed. Airway responsiveness to histamine bronchoprovocation (PD20 -FEV1 ) and exhaled nitric oxide (FeNO) was determined. RESULTS NAR increased significantly in all subjects with the greatest effect seen in AR+AHR+ individuals. VAS increased in all subjects at 30 min and returned to baseline at 6 h, with significantly higher levels in AR+AHR+ and AR+AHR- subjects (P < 0.05). Eosinophils in nasal lavage fluid and sputum increased significantly after NPT in AR+AHR+ and AR+AHR- subjects (P < 0.001). FEV1 % and PD20 -FEV1 decreased and FeNO increased significantly after NPT only in AR+AHR+ subjects (P < 0.05). Nasal lavage eosinophil count was positively correlated with sputum eosinophil count and the level of FeNO and negatively correlated with FEV1 and PD20 . CONCLUSIONS House dust mite nasal provocation test induces and aggravates both upper and lower airway inflammation and hyper-responsiveness in patients with persistent allergic rhinitis without asthmatic symptoms.
Collapse
Affiliation(s)
- W. Wang
- State Key Laboratory of Respiratory Disease; Department of Allergy and Clinical Immunology; The First Affiliated Hospital; Guangzhou Medical University; Guangzhou China
| | - M. Xian
- State Key Laboratory of Respiratory Disease; Department of Allergy and Clinical Immunology; The First Affiliated Hospital; Guangzhou Medical University; Guangzhou China
| | - Y. Xie
- State Key Laboratory of Respiratory Disease; Department of Allergy and Clinical Immunology; The First Affiliated Hospital; Guangzhou Medical University; Guangzhou China
| | - J. Zheng
- State Key Laboratory of Respiratory Disease; Department of Allergy and Clinical Immunology; The First Affiliated Hospital; Guangzhou Medical University; Guangzhou China
| | - J. Li
- State Key Laboratory of Respiratory Disease; Department of Allergy and Clinical Immunology; The First Affiliated Hospital; Guangzhou Medical University; Guangzhou China
- Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology; Guangzhou Medical University; Guangzhou China
| |
Collapse
|
42
|
Dam V, Sikder T, Santosa S. From neutrophils to macrophages: differences in regional adipose tissue depots. Obes Rev 2016; 17:1-17. [PMID: 26667065 DOI: 10.1111/obr.12335] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 09/01/2015] [Accepted: 09/23/2015] [Indexed: 12/27/2022]
Abstract
Currently, we do not fully understand the underlying mechanisms of how regional adiposity promotes metabolic dysregulation. As adipose tissue expands, there is an increase in chronic systemic low-grade inflammation due to greater infiltration of immune cells and production of cytokines. This chronic inflammation is thought to play a major role in the development of metabolic complications and disease such as insulin resistance and diabetes. We know that different adipose tissue depots contribute differently to the risk of metabolic disease. People who have an upper body fat distribution around the abdomen are at greater risk of disease than those who tend to store fat in their lower body around the hips and thighs. Thus, it is conceivable that adipose tissue depots contribute differently to the inflammatory milieu as a result of varied infiltration of immune cell types. In this review, we describe the role and function of major resident immune cells in the development of adipose tissue inflammation and discuss their regional differences in the context of metabolic disease risk. We find that although initial studies have found regional differences, a more comprehensive understanding of how immune cells interrupt adipose tissue homeostasis is needed.
Collapse
Affiliation(s)
- V Dam
- Department of Exercise Science, Concordia University, Montreal, QC, Canada.,Nutrition, Obesity, and Metabolism Lab, PERFORM Centre, Concordia University, Montreal, QC, Canada
| | - T Sikder
- Department of Exercise Science, Concordia University, Montreal, QC, Canada.,Nutrition, Obesity, and Metabolism Lab, PERFORM Centre, Concordia University, Montreal, QC, Canada
| | - S Santosa
- Department of Exercise Science, Concordia University, Montreal, QC, Canada.,Nutrition, Obesity, and Metabolism Lab, PERFORM Centre, Concordia University, Montreal, QC, Canada
| |
Collapse
|
43
|
Finlay CM, Stefanska AM, Walsh KP, Kelly PJ, Boon L, Lavelle EC, Walsh PT, Mills KHG. Helminth Products Protect against Autoimmunity via Innate Type 2 Cytokines IL-5 and IL-33, Which Promote Eosinophilia. THE JOURNAL OF IMMUNOLOGY 2015; 196:703-14. [PMID: 26673140 DOI: 10.4049/jimmunol.1501820] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 11/08/2015] [Indexed: 01/15/2023]
Abstract
Epidemiologic studies in humans have demonstrated that infection with helminth parasites is associated with a reduced risk of developing autoimmune diseases. Mechanistic studies in mice have linked the protective effect of helminths on autoimmunity to the suppressive activity of helminth-induced regulatory T cells (Tregs) or Th2 cells. In this study, we demonstrate that treatment of mice with Fasciola hepatica excretory-secretory products (FHES) attenuated the clinical signs of experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis. Protection was associated with a significant reduction in the infiltration of pathogenic Th1 and Th17 cells into the brain. Although FHES enhanced anti-inflammatory cytokine and Th2 responses, protection against EAE was independent of IL-4, IL-10, and Tregs. However, administration of FHES induced production of the type 2 cytokines IL-33 and IL-5, which promoted accumulation of eosinophils. FHES-induced expansion of eosinophils and protection against EAE was lost in IL-33(-/-) mice and upon neutralization of IL-5. Furthermore, transfer of FHES-induced or IL-33-induced eosinophils conferred protection against EAE. In addition, treatment of mice with recombinant IL-33 attenuated autoimmunity, and this was dependent on IL-5. To our knowledge, this study is the first to report a role for helminth-induced IL-5 and IL-33 in protection against autoimmunity.
Collapse
Affiliation(s)
- Conor M Finlay
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Anna M Stefanska
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Kevin P Walsh
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Patrick J Kelly
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Louis Boon
- EPIRUS Biopharmaceuticals Netherlands BV, 3584 CM Utrecht, the Netherlands
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; and
| | - Patrick T Walsh
- Tolerance and Autoimmunity Research Group, Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Kingston H G Mills
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland;
| |
Collapse
|
44
|
Jacobsen EA, Lee NA, Lee JJ. Re-defining the unique roles for eosinophils in allergic respiratory inflammation. Clin Exp Allergy 2015; 44:1119-36. [PMID: 24961290 DOI: 10.1111/cea.12358] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The role of eosinophils in the progression and resolution of allergic respiratory inflammation is poorly defined despite the commonality of their presence and in some cases their use as a biomarker for disease severity and/or symptom control. However, this ambiguity belies the wealth of insights that have recently been gained through the use of eosinophil-deficient/attenuated strains of mice that have demonstrated novel immunoregulatory and remodelling/repair functions for these cells in the lung following allergen provocation. Specifically, studies of eosinophil-deficient mice suggest that eosinophils contribute to events occurring in the lungs following allergen provocation at several key moments: (i) the initiating phase of events leading to Th2-polarized pulmonary inflammation, (ii) the suppression Th1/Th17 pathways in lung-draining lymph nodes, (iii) the recruitment of effector Th2 T cells to the lung, and finally, (iv) mechanisms of inflammatory resolution that re-establish pulmonary homoeostasis. These suggested functions have recently been confirmed and expanded upon using allergen provocation of an inducible eosinophil-deficient strain of mice (iPHIL) that demonstrated an eosinophil-dependent mechanism(s) leading to Th2 dominated immune responses in the presence of eosinophils in contrast to neutrophilic as well as mixed Th1/Th17/Th2 variant phenotypes in the absence of eosinophils. These findings highlighted that eosinophils are not exclusively downstream mediators controlled by T cells, dendritic cells (DC) and/or innate lymphocytic cells (ILC2). Instead, eosinophils appear to be more aptly described as significant contributors in complex interrelated pathways that lead to pulmonary inflammation and subsequently promote resolution and the re-establishment of homoeostatic baseline. In this review, we summarize and put into the context the evolving hypotheses that are now expanding our understanding of the roles eosinophils likely have in the lung following allergen provocation.
Collapse
Affiliation(s)
- E A Jacobsen
- Division of Pulmonary Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | | | | |
Collapse
|
45
|
Travers J, Rothenberg ME. Eosinophils in mucosal immune responses. Mucosal Immunol 2015; 8:464-75. [PMID: 25807184 PMCID: PMC4476057 DOI: 10.1038/mi.2015.2] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 12/28/2014] [Indexed: 02/06/2023]
Abstract
Eosinophils, multifunctional cells that contribute to both innate and adaptive immunity, are involved in the initiation, propagation, and resolution of immune responses, including tissue repair. They achieve this multifunctionality by expression of a diverse set of activation receptors, including those that directly recognize pathogens and opsonized targets, and by their ability to store and release preformed cytotoxic mediators that participate in host defense, to produce a variety of de novo pleotropic mediators and cytokines, and to interact directly and indirectly with diverse cell types, including adaptive and innate immunocytes and structural cells. Herein, we review the basic biology of eosinophils and then focus on new emerging concepts about their role in mucosal immune homeostasis, particularly maintenance of intestinal IgA. We review emerging data about their development and regulation and describe new concepts concerning mucosal eosinophilic diseases. We describe recently developed therapeutic strategies to modify eosinophil levels and function and provide collective insight about the beneficial and detrimental functions of these enigmatic cells.
Collapse
|
46
|
|
47
|
Abstract
Leishmania spp. are parasitic protozoa endemic in tropical and subtropical regions and the causative agent of leishmaniasis, a collection of syndromes whose clinical manifestations vary according to host and pathogen factors. Leishmania spp. are inoculated into the mammalian host by the bite of an infected sand fly, whereupon they are taken up by phagocytosis, convert into the replicative amastigote stage within macrophages, reproduce, spread to new macrophages and cause disease manifestations. A curative response against leishmaniasis depends in the classical activation of macrophages and the IL-12-dependent onset of an adaptive type 1 response characterized by the production of IFN-γ. Emerging evidence suggests that neutrophils, dendritic cells and other immune cells can serve as either temporary or stable hosts for Leishmania spp. Furthermore, it is becoming apparent that the initial interactions of the parasite with resident or early recruited immune cells can shape both the macrophage response and the type of adaptive immune response being induced. In this review, we compile a growing number of studies demonstrating how the earliest interactions of Leishmania spp. with eosinophils and mast cells influence the macrophage response to infection and the development of the adaptive immune response, hence, determining the ultimate outcome of infection.
Collapse
|
48
|
Chu DK, Jimenez-Saiz R, Verschoor CP, Walker TD, Goncharova S, Llop-Guevara A, Shen P, Gordon ME, Barra NG, Bassett JD, Kong J, Fattouh R, McCoy KD, Bowdish DM, Erjefält JS, Pabst O, Humbles AA, Kolbeck R, Waserman S, Jordana M. Indigenous enteric eosinophils control DCs to initiate a primary Th2 immune response in vivo. ACTA ACUST UNITED AC 2014; 211:1657-72. [PMID: 25071163 PMCID: PMC4113937 DOI: 10.1084/jem.20131800] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Eosinophils natively inhabit the small intestine, but a functional role for them there has remained elusive. Here, we show that eosinophil-deficient mice were protected from induction of Th2-mediated peanut food allergy and anaphylaxis, and Th2 priming was restored by reconstitution with il4(+/+) or il4(-/-) eosinophils. Eosinophils controlled CD103(+) dendritic cell (DC) activation and migration from the intestine to draining lymph nodes, events necessary for Th2 priming. Eosinophil activation in vitro and in vivo led to degranulation of eosinophil peroxidase, a granule protein whose enzymatic activity promoted DC activation in mice and humans in vitro, and intestinal and extraintestinal mouse DC activation and mobilization to lymph nodes in vivo. Further, eosinophil peroxidase enhanced responses to ovalbumin seen after immunization. Thus, eosinophils can be critical contributors to the intestinal immune system, and granule-mediated shaping of DC responses can promote both intestinal and extraintestinal adaptive immunity.
Collapse
Affiliation(s)
- Derek K Chu
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Rodrigo Jimenez-Saiz
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Christopher P Verschoor
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Tina D Walker
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Susanna Goncharova
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Alba Llop-Guevara
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Pamela Shen
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Melissa E Gordon
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Nicole G Barra
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Jennifer D Bassett
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Joshua Kong
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Ramzi Fattouh
- Clinical Microbiology, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario M5S 1A8, Canada
| | - Kathy D McCoy
- Maurice Müller Laboratories, Universitätsklinik für Viszerale Chirurgie und Medizin (UVCM), University of Bern, 3008 Bern, Switzerland
| | - Dawn M Bowdish
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Jonas S Erjefält
- Department of Experimental Medical Science, Lund University, SE-22184 Lund, Sweden Department of Respiratory Medicine and Allergology, Lund University Hospital, SE-22185 Lund, Sweden
| | - Oliver Pabst
- Institute of Molecular Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Alison A Humbles
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MA 20878
| | - Roland Kolbeck
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MA 20878
| | - Susan Waserman
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Manel Jordana
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| |
Collapse
|
49
|
Philpott H, Nandurkar S, Thien F, Gibson PR, Royce SG. Eosinophilic esophagitis: a clinicopathological review. Pharmacol Ther 2014; 146:12-22. [PMID: 25200122 DOI: 10.1016/j.pharmthera.2014.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 08/28/2014] [Indexed: 12/12/2022]
Abstract
Eosinophilic esophagitis (EoE) is considered to be a chronic antigen-driven disease whereby food and/or aeroallergens induce a chronic inflammatory infiltrate in the esophagus, resulting in pathological hyperplasia of the epithelia and muscular layers, and fibrosis of the lamina propria (referred to collectively as remodelling) and the symptoms of dysphagia and food impaction. EoE shares features with other atopic conditions of asthma and atopic dermatitis, such as a TH2 cytokine milieu and a mixed inflammatory infiltrate of eosinophils, mast cells and lymphocytes. Relatively distinct features include the strong male predominance amongst adult patients, and the expression of the eosinophil chemokine eotaxin 3. Current first line treatments such as strict dietary modification and corticosteroids fail many patients. Looking forward, clarification of distinct genotype/phenotype associations, determining the reversibility of remodelling following treatment, and the development of new pharmacotherapies that target fibrotic pathways (as opposed to eosinophilic inflammation per se) or specifically improve barrier integrity appear relevant.
Collapse
Affiliation(s)
- Hamish Philpott
- Department of Gastroenterology Eastern Health, Monash University Melbourne, Australia.
| | - Sanjay Nandurkar
- Department of Gastroenterology Eastern Health, Monash University Melbourne, Australia
| | - Francis Thien
- Department of Respiratory and Sleep Medicine Eastern Health, Monash University Melbourne, Australia
| | - Peter R Gibson
- Department of Gastroenterology The Alfred Hospital, Monash University Melbourne, Australia
| | - Simon G Royce
- Department of Pharmacology Clayton Campus, Monash University Melbourne, Australia
| |
Collapse
|
50
|
Gaurav R, Agrawal DK. Clinical view on the importance of dendritic cells in asthma. Expert Rev Clin Immunol 2014; 9:899-919. [PMID: 24128155 DOI: 10.1586/1744666x.2013.837260] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Allergic asthma is characterized by airway hyperresponsiveness and inflammation and may lead to airway remodeling in uncontrolled cases. Genetic predisposition to an atopic phenotype plays a major component in the pathophysiology of asthma. However, with tremendous role of epigenetic factors and environmental stimuli in precipitating an immune response, the underlying pathophysiological mechanisms are complicated. Dendritic cells are principal antigen-presenting cells and initiators of the immune response in allergic asthma. Their phenotype, guided by multiple factors may dictate the immune reaction to an allergic or tolerogenic response. Involvement of the local cytokine milieu, microbiome and interplay between immune cells add dimension to the fate of immune response. In addition to allergen exposure, these factors modulate DC phenotype and function. In this article, integration of many factors and pathways associated with the recruitment and activation of DCs in the pathophysiology of allergic asthma is presented in a clinical and translational manner.
Collapse
Affiliation(s)
- Rohit Gaurav
- Department of Biomedical Sciences and Center for Clinical and Translational Science, Creighton University School of Medicine, CRISS II Room 510, 2500 California Plaza Omaha, NE 68178, USA
| | | |
Collapse
|