1
|
Lee D, Jo MG, Min KY, Choi MY, Kim YM, Kim HS, Choi WS. IL-10 + regulatory B cells mitigate atopic dermatitis by suppressing eosinophil activation. Sci Rep 2024; 14:18164. [PMID: 39107352 PMCID: PMC11303538 DOI: 10.1038/s41598-024-68660-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Atopic dermatitis (AD) presents significant therapeutic challenges due to its poorly understood etiology. Eosinophilia, a hallmark of allergic inflammation, is implicated in AD pathogenesis. Interleukin-10 (IL-10)-producing regulatory B (Breg) cells exhibit potent anti-inflammatory effects. However, their role in controlling AD-related eosinophilia is not well understood. To investigate the impact of eosinophils on AD, we employed IL-5Rα-deficient (Il5ra-/-) mice, which lack functional eosinophils. Induction of AD in these mice resulted in attenuated disease symptoms, underscoring the critical role of eosinophils in AD development. Additionally, the adoptive transfer of purified Breg cells into mice with AD significantly alleviated disease severity. Mechanistic studies revealed that IL-10 produced by Breg cells directly inhibits eosinophil activation and infiltration into the skin. In vitro experiments further confirmed that Breg cells inhibited eosinophil peroxidase secretion in an IL-10-dependent manner. Our collective findings demonstrate that IL-10 from Breg cells alleviates AD by suppressing eosinophil activation and tissue infiltration. This study elucidates a novel regulatory mechanism of Breg cells, providing a foundation for future Breg-mediated therapeutic strategies for AD.
Collapse
Affiliation(s)
- Dajeong Lee
- School of Medicine, Konkuk University, Chungju, 27478, Korea
| | - Min Geun Jo
- School of Medicine, Konkuk University, Chungju, 27478, Korea
| | - Keun Young Min
- School of Medicine, Konkuk University, Chungju, 27478, Korea
| | - Min Yeong Choi
- School of Medicine, Konkuk University, Chungju, 27478, Korea
| | - Young Mi Kim
- College of Pharmacy, Duksung Women's University, Seoul, 01369, Korea
| | - Hyuk Soon Kim
- Department of Biomedical Sciences, College of Natural Science and Department of Health Sciences, The Graduate School of Dong-A University, Busan, 49315, Korea.
| | - Wahn Soo Choi
- School of Medicine, Konkuk University, Chungju, 27478, Korea.
- Institute of Biomedical Sciences & Technology, Konkuk University, Seoul, 05029, Korea.
| |
Collapse
|
2
|
Hargitai R, Parráková L, Szatmári T, Monfort-Lanzas P, Galbiati V, Audouze K, Jornod F, Staal YCM, Burla S, Chary A, Gutleb AC, Lumniczky K, Vandebriel RJ, Gostner JM. Chemical respiratory sensitization-Current status of mechanistic understanding, knowledge gaps and possible identification methods of sensitizers. FRONTIERS IN TOXICOLOGY 2024; 6:1331803. [PMID: 39135743 PMCID: PMC11317441 DOI: 10.3389/ftox.2024.1331803] [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: 11/01/2023] [Accepted: 05/27/2024] [Indexed: 08/15/2024] Open
Abstract
Respiratory sensitization is a complex immunological process eventually leading to hypersensitivity following re-exposure to the chemical. A frequent consequence is occupational asthma, which may occur after long latency periods. Although chemical-induced respiratory hypersensitivity has been known for decades, there are currently no comprehensive and validated approaches available for the prospective identification of chemicals that induce respiratory sensitization, while the expectations of new approach methodologies (NAMs) are high. A great hope is that due to a better understanding of the molecular key events, new methods can be developed now. However, this is a big challenge due to the different chemical classes to which respiratory sensitizers belong, as well as because of the complexity of the response and the late manifestation of symptoms. In this review article, the current information on respiratory sensitization related processes is summarized by introducing it in the available adverse outcome pathway (AOP) concept. Potentially useful models for prediction are discussed. Knowledge gaps and gaps of regulatory concern are identified.
Collapse
Affiliation(s)
- Rita Hargitai
- Unit of Radiation Medicine, Department of Radiobiology and Radiohygiene, National Centre for Public Health and Pharmacy (NCPHP), Budapest, Hungary
| | - Lucia Parráková
- Biochemical Immunotoxicology Group, Institute of Medical Biochemistry, Medical University of Innsbruck (MUI), Innsbruck, Austria
| | - Tünde Szatmári
- Unit of Radiation Medicine, Department of Radiobiology and Radiohygiene, National Centre for Public Health and Pharmacy (NCPHP), Budapest, Hungary
| | - Pablo Monfort-Lanzas
- Biochemical Immunotoxicology Group, Institute of Medical Biochemistry, Medical University of Innsbruck (MUI), Innsbruck, Austria
- Institute of Bioinformatics, Medical University of Innsbruck (MUI), Innsbruck, Austria
| | - Valentina Galbiati
- Laboratory of Toxicology, Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università Degli Studi di Milano (UNIMI), Milano, Italy
| | | | | | - Yvonne C. M. Staal
- Centre for Health Protection, National Institute of Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Sabina Burla
- Luxembourg Institute of Science and Technology (LIST), Belvaux, Luxembourg
| | - Aline Chary
- Luxembourg Institute of Science and Technology (LIST), Belvaux, Luxembourg
| | - Arno C. Gutleb
- Luxembourg Institute of Science and Technology (LIST), Belvaux, Luxembourg
| | - Katalin Lumniczky
- Unit of Radiation Medicine, Department of Radiobiology and Radiohygiene, National Centre for Public Health and Pharmacy (NCPHP), Budapest, Hungary
| | - Rob J. Vandebriel
- Centre for Health Protection, National Institute of Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Johanna M. Gostner
- Biochemical Immunotoxicology Group, Institute of Medical Biochemistry, Medical University of Innsbruck (MUI), Innsbruck, Austria
| |
Collapse
|
3
|
Du L, Tang L, Xiao L, Tang K, Zeng Z, Liang Y, Guo Y. Increased expression of CSF1 in patients with eosinophilic asthma. Immun Inflamm Dis 2023; 11:e847. [PMID: 37249291 PMCID: PMC10170305 DOI: 10.1002/iid3.847] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/29/2023] [Accepted: 04/10/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND The link between colony-stimulating factor 1 (CSF1) and asthma was reported recently. However, the role and mechanism of CSF1 in asthma remain poorly understood. In this study, we aimed to explore the expression and its potential mechanism of CSF1 in asthma. METHODS CSF1 expression in the airway samples from asthmatics and healthy controls were examined, then the correlations between CSF1 and eosinophilic indicators were analyzed. Subsequently, bronchial epithelial cells (BEAS-2B) with CSF1 overexpression and knockdown were constructed to investigate the potential molecular mechanism of CSF1. Finally, the effect of CSF1R inhibitor on STAT1 was investigated. RESULTS The expression of CSF1 was significantly increased in patients with asthma compared to healthy controls, especially in patients with severe and eosinophilic asthma. Upregulated CSF1 positively correlated with airway-increased eosinophil inflammation. In vitro, cytokines interleukin 13 (IL-13) and IL-33 can stimulate the upregulation of CSF1 expression. CSF1 overexpression enhanced p-CSF1R/CSF1R and p-STAT1/STAT1 expression, while knockdown CSF1 using anti-CSF1 siRNAs decreased p-CSF1R/CSF1R and p-STAT1/STAT1 expression. Furthermore, the inhibitor of CSF1R significantly decreased p-STAT1/STAT1 expression. CONCLUSIONS Sputum CSF1 may be involved in asthmatic airway eosinophil inflammation by interacting with CSF1R and further activating the STAT1 signaling. Interfering this potential pathway could serve as an anti-inflammatory therapy for asthma.
Collapse
Affiliation(s)
- Lijuan Du
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Department of Respiratory and Critical Care MedicineThe Affiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
| | - Lu Tang
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Lisha Xiao
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Kun Tang
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Zhimin Zeng
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Yuxia Liang
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Yubiao Guo
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
4
|
Badolati I, van der Heiden M, Brodin D, Zuurveld M, Szilágyi S, Björkander S, Sverremark-Ekström E. Staphylococcus aureus-derived factors promote human Th9 cell polarization and enhance a transcriptional program associated with allergic inflammation. Eur J Immunol 2023; 53:e2250083. [PMID: 36550071 DOI: 10.1002/eji.202250083] [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: 07/11/2022] [Revised: 12/09/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
T helper (Th) 9 cells, characterized by robust secretion of IL-9, have been increasingly associated with allergic diseases. However, whether and how Th9 cells are modulated by environmental stimuli remains poorly understood. In this study, we show that in vitro exposure of human PBMCs or isolated CD4 T-cells to Staphylococcus (S.) aureus-derived factors, including its toxins, potently enhances Th9 cell frequency and IL-9 secretion. Furthermore, as revealed by RNA sequencing analysis, S. aureus increases the expression of Th9-promoting factors at the transcriptional level, such as FOXO1, miR-155, and TNFRSF4. The addition of retinoic acid (RA) dampens the Th9 responses promoted by S. aureus and substantially changes the transcriptional program induced by this bacterium, while also altering the expression of genes associated with allergic inflammation. Together, our results demonstrate a strong influence of microbial and dietary factors on Th9 cell polarization, which may be important in the context of allergy development and treatment.
Collapse
Affiliation(s)
- Isabella Badolati
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Marieke van der Heiden
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - David Brodin
- Bioinformatics and Expression Analysis Core Facility, Karolinska Institutet, Huddinge, Sweden
| | - Marit Zuurveld
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Szilvia Szilágyi
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Sophia Björkander
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Eva Sverremark-Ekström
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
5
|
Pant H, Hercus TR, Tumes DJ, Yip KH, Parker MW, Owczarek CM, Lopez AF, Huston DP. Translating the biology of β common receptor-engaging cytokines into clinical medicine. J Allergy Clin Immunol 2023; 151:324-344. [PMID: 36424209 DOI: 10.1016/j.jaci.2022.09.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/16/2022] [Accepted: 09/29/2022] [Indexed: 11/23/2022]
Abstract
The family of cytokines that comprises IL-3, IL-5, and GM-CSF was discovered over 30 years ago, and their biological activities and resulting impact in clinical medicine has continued to expand ever since. Originally identified as bone marrow growth factors capable of acting on hemopoietic progenitor cells to induce their proliferation and differentiation into mature blood cells, these cytokines are also recognized as key mediators of inflammation and the pathobiology of diverse immunologic diseases. This increased understanding of the functional repertoire of IL-3, IL-5, and GM-CSF has led to an explosion of interest in modulating their functions for clinical management. Key to the successful clinical translation of this knowledge is the recognition that these cytokines act by engaging distinct dimeric receptors and that they share a common signaling subunit called β-common or βc. The structural determination of how IL-3, IL-5, and GM-CSF interact with their receptors and linking this to their differential biological functions on effector cells has unveiled new paradigms of cell signaling. This knowledge has paved the way for novel mAbs and other molecules as selective or pan inhibitors for use in different clinical settings.
Collapse
Affiliation(s)
- Harshita Pant
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia; Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Timothy R Hercus
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia
| | - Damon J Tumes
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia
| | - Kwok Ho Yip
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia
| | - Michael W Parker
- Bio 21 Institute, The University of Melbourne, Melbourne, Australia; St Vincent's Institute of Medical Research, Melbourne, Australia
| | | | - Angel F Lopez
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia; Adelaide Medical School, University of Adelaide, Adelaide, Australia.
| | - David P Huston
- Texas A&M University School of Medicine, Houston, Tex; Houston Methodist Hospital and Research Institute, Houston, Tex.
| |
Collapse
|
6
|
Dai Y, Wu S, Cao C, Xue R, Luo X, Wen Z, Xu J. Csf1rb regulates definitive hematopoiesis in zebrafish. Development 2022; 149:276084. [DOI: 10.1242/dev.200534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 07/07/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
In vertebrates, hematopoietic stem and progenitor cells (HSPCs) are capable of self-renewal and continuously replenishing all mature blood lineages throughout life. However, the molecular signaling regulating the maintenance and expansion of HSPCs remains incompletely understood. Colony-stimulating factor 1 receptor (CSF1R) is believed to be the primary regulator for the myeloid lineage but not HSPC development. Here, we show a surprising role of Csf1rb, a zebrafish homolog of mammalian CSF1R, in preserving the HSPC pool by maintaining the proliferation of HSPCs. Deficiency of csf1rb leads to a reduction in both HSPCs and their differentiated progenies, including myeloid, lymphoid and erythroid cells at early developmental stages. Likewise, the absence of csf1rb conferred similar defects upon HSPCs and leukocytes in adulthood. Furthermore, adult hematopoietic cells from csf1rb mutants failed to repopulate immunodeficient zebrafish. Interestingly, loss-of-function and gain-of-function assays suggested that the canonical ligands for Csf1r in zebrafish, including Csf1a, Csf1b and Il34, were unlikely to be ligands of Csf1rb. Thus, our data indicate a previously unappreciated role of Csf1r in maintaining HSPCs, independently of known ligands.
Collapse
Affiliation(s)
- Yimei Dai
- School of Medicine, South China University of Technology 1 Laboratory of Immunology & Regeneration , , Guangzhou 510006, China
| | - Shuting Wu
- State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, the Hong Kong University of Science and Technology 2 Division of Life Science , , Clear Water Bay, Kowloon, Hong Kong , People's Republic of China
| | - Canran Cao
- School of Medicine, South China University of Technology 1 Laboratory of Immunology & Regeneration , , Guangzhou 510006, China
| | - Rongtao Xue
- Nanfang Hospital, Southern Medical University 3 Department of Hematology , , Guangzhou, Guangdong 510515 , China
| | - Xuefen Luo
- State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, the Hong Kong University of Science and Technology 2 Division of Life Science , , Clear Water Bay, Kowloon, Hong Kong , People's Republic of China
| | - Zilong Wen
- State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, the Hong Kong University of Science and Technology 2 Division of Life Science , , Clear Water Bay, Kowloon, Hong Kong , People's Republic of China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen Peking University−Hong Kong University of Science and Technology Medical Center 4 , Shenzhen 518055 , China
| | - Jin Xu
- School of Medicine, South China University of Technology 1 Laboratory of Immunology & Regeneration , , Guangzhou 510006, China
| |
Collapse
|
7
|
Evaluation of Phage Display Biopanning Strategies for the Selection of Anti-Cell Surface Receptor Antibodies. Int J Mol Sci 2022; 23:ijms23158470. [PMID: 35955604 PMCID: PMC9369378 DOI: 10.3390/ijms23158470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/28/2022] [Accepted: 07/28/2022] [Indexed: 12/04/2022] Open
Abstract
Monoclonal antibodies (mAbs) are one of the most successful and versatile protein-based pharmaceutical products used to treat multiple pathological conditions. The remarkable specificity of mAbs and their affinity for biological targets has led to the implementation of mAbs in the therapeutic regime of oncogenic, chronic inflammatory, cardiovascular, and infectious diseases. Thus, the discovery of novel mAbs with defined functional activities is of crucial importance to expand our ability to address current and future clinical challenges. In vitro, antigen-driven affinity selection employing phage display biopanning is a commonly used technique to isolate mAbs. The success of biopanning is dependent on the quality and the presentation format of the antigen, which is critical when isolating mAbs against membrane protein targets. Here, we provide a comprehensive investigation of two established panning strategies, surface-tethering of a recombinant extracellular domain and cell-based biopanning, to examine the impact of antigen presentation on selection outcomes with regards to the isolation of positive mAbs with functional potential against a proof-of-concept type I cell surface receptor. Based on the higher sequence diversity of the resulting antibody repertoire, presentation of a type I membrane protein in soluble form was more advantageous over presentation in cell-based format. Our results will contribute to inform and guide future antibody discovery campaigns against cell surface proteins.
Collapse
|
8
|
Cahill KM, Johnson TK, Perveen Z, Schexnayder M, Xiao R, Heffernan LM, Langohr IM, Paulsen DB, Penn AL, Noël A. In utero exposures to mint-flavored JUUL aerosol impair lung development and aggravate house dust mite-induced asthma in adult offspring mice. Toxicology 2022; 477:153272. [PMID: 35878681 DOI: 10.1016/j.tox.2022.153272] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/04/2022] [Accepted: 07/21/2022] [Indexed: 11/18/2022]
Abstract
There are few reports concerning electronic nicotine delivery system (ENDS) use during pregnancy and no studies on asthma in prenatally JUUL-exposed offspring. Here, we tested the hypothesis that in utero JUUL exposure causes unfavorable birth outcomes and lasting pulmonary health effects in adult offspring. BALB/c dams were exposed to either air or mint-flavored JUUL aerosol, 1-hr/d, 20 consecutive days during gestation. Offspring were sacrificed on post-natal day (PND) 0 or at 11-week of age, following house dust mite (HDM) challenge. Gene expression was assessed in the uterine/placental tissue of the dams and lung responses were assessed in offspring at PND0 and at 11 weeks of age. JUUL-exposed offspring exhibited decreased body weights and lengths at PND0. These birth outcomes were accompanied by dysregulation of 54 genes associated with hypoxia and oxidative stress in the uterine/placental tissues of JUUL-exposed dams, as well as 24 genes in the lungs of the offspring related to Wnt signaling, plus 9 genes related to epigenetics, and 7 genes related to inflammation. At 11 weeks of age, JUUL + HDM exposed mice exhibited pulmonary inflammation when compared to their respective air + HDM controls. Additionally, the JUUL + HDM exposure dysregulated several genes associated with allergies and asthma. Further, the JUUL + HDM females showed decreased methylation of the promoter region of the Il10ra gene. Taken together, our mouse model shows that inhalation of JUUL aerosols during pregnancy affects the intrauterine environment, impairs lung development, and heightens the effects of allergic airway responses later in life.
Collapse
Affiliation(s)
- Kerin M Cahill
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Trenton K Johnson
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Zakia Perveen
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Matthew Schexnayder
- Lincoln Memorial University, College of Veterinary Medicine, 6965 Cumberland Gap Parkway, Harrogate, TN, USA
| | - Rui Xiao
- Department of Anesthesiology, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032, USA
| | - Linda M Heffernan
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Ingeborg M Langohr
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Daniel B Paulsen
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Arthur L Penn
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Alexandra Noël
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
9
|
Martin KE, Kalelkar PP, Coronel MM, Theriault HS, Schneider RS, García AJ. Host type 2 immune response to xenogeneic serum components impairs biomaterial-directed osteo-regenerative therapies. Biomaterials 2022; 286:121601. [PMID: 35660823 PMCID: PMC11458135 DOI: 10.1016/j.biomaterials.2022.121601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/20/2022] [Accepted: 05/22/2022] [Indexed: 11/21/2022]
Abstract
The transformative potential of cells as therapeutic agents is being realized in a wide range of applications, from regenerative medicine to cancer therapy to autoimmune disorders. The majority of these therapies require ex vivo expansion of the cellular product, often utilizing fetal bovine serum (FBS) in the culture media. However, the impact of residual FBS on immune responses to cell therapies and the resulting cell therapy outcomes remains unclear. Here, we show that hydrogel-delivered FBS elicits a robust type 2 immune response characterized by infiltration of eosinophils and CD4+ T cells. Host secretion of cytokines associated with type 2 immunity, including IL-4, IL-5, and IL-13, is also increased in FBS-containing hydrogels. We demonstrate that the immune response to xenogeneic serum components dominates the local environment and masks the immunomodulatory effects of biomaterial-delivered mesenchymal stromal/stem cells. Importantly, delivery of relatively small amounts of FBS (3.2% by volume) within BMP-2-containing biomaterial constructs dramatically reduces the ability of these constructs to promote de novo bone formation in a radial defect model in immunocompetent mice. These results urge caution when interpreting the immunological and tissue repair outcomes in immunocompetent pre-clinical models from cells and biomaterial constructs that have come in contact with xenogeneic serum components.
Collapse
Affiliation(s)
- Karen E Martin
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Pranav P Kalelkar
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - María M Coronel
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hannah S Theriault
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Rebecca S Schneider
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
10
|
Huang Y, Chen Y, Liu Y, Mi R, Han X, Gong H, Cheng L, Chen Z. Isolation and identification of sporozoite membrane protein of Cryptosporidium parvum and evaluation of calmodulin-like protein immune protection. Parasite Immunol 2022; 44:e12937. [PMID: 35652261 DOI: 10.1111/pim.12937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/20/2022] [Accepted: 05/26/2022] [Indexed: 11/30/2022]
Abstract
Until now, no completely effective parasite-specific drugs or vaccines have been approved for the treatment of cryptosporidiosis. Through the separation and identification of the sporozoite membrane protein of Cryptosporidium parvum (C. parvum), 20 related proteins were obtained. Among them, a calmodulin-like protein (CML) has a similar functional domain-exchange factor hand (EF-hand) motif as calmodulin proteins (CaMs), so it may play a similarly important role in the invasion process. A 663 bp full gene encoding the C. parvum calmodulin-like protein (CpCML) was inserted in pET28a vector and expressed in Escherichia coli. An immunofluorescence assay showed that CpCML was mainly located on the surface of the sporozoites. Three-week-old female BALB/c mice were used for modeling the immunoreactions and immunoprotection of recombinant CpCML (rCpCML) against artificial Cryptosporidium tyzzeri (C. tyzzeri) infections. The results indicated a significantly increased in anti-CpCML antibody response, which was induced by the immunized recombinant protein. Compared to rP23(recombinant P23), GST6P-1(expressed by pGEX-6P-1 transfected E. coli) , GST4T-1(expressed by pGEX-4T-1 transfected E. coli) , glutathione (GSH), adjuvant, and blank control groups, rCpCML-immunized mice produced specific spleen cell proliferation in addition to different production levels of IL-2, IFN-γ, TNF-α, IL-4 and IL-5. Additionally, immunization with rCpCML led to 34.08% reduction of oocyst shedding in C. tyzzeri infected mice faeces which was similart to rP23. These results suggest that CpCML may be developed as a potential vaccine candidate antigen against cryptosporidiosis.
Collapse
Affiliation(s)
- Yan Huang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yu Chen
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yuxuan Liu
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Rongsheng Mi
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Xiangan Han
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Haiyan Gong
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Long Cheng
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Zhaoguo Chen
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| |
Collapse
|
11
|
Targeted deletion of Interleukin-3 results in asthma exacerbations. iScience 2022; 25:104440. [PMID: 35707726 PMCID: PMC9189047 DOI: 10.1016/j.isci.2022.104440] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 03/28/2022] [Accepted: 05/17/2022] [Indexed: 11/21/2022] Open
|
12
|
Grosche S, Marenholz I, Esparza-Gordillo J, Arnau-Soler A, Pairo-Castineira E, Rüschendorf F, Ahluwalia TS, Almqvist C, Arnold A, Baurecht H, Bisgaard H, Bønnelykke K, Brown SJ, Bustamante M, Curtin JA, Custovic A, Dharmage SC, Esplugues A, Falchi M, Fernandez-Orth D, Ferreira MAR, Franke A, Gerdes S, Gieger C, Hakonarson H, Holt PG, Homuth G, Hubner N, Hysi PG, Jarvelin MR, Karlsson R, Koppelman GH, Lau S, Lutz M, Magnusson PKE, Marks GB, Müller-Nurasyid M, Nöthen MM, Paternoster L, Pennell CE, Peters A, Rawlik K, Robertson CF, Rodriguez E, Sebert S, Simpson A, Sleiman PMA, Standl M, Stölzl D, Strauch K, Szwajda A, Tenesa A, Thompson PJ, Ullemar V, Visconti A, Vonk JM, Wang CA, Weidinger S, Wielscher M, Worth CL, Xu CJ, Lee YA. Rare variant analysis in eczema identifies exonic variants in DUSP1, NOTCH4 and SLC9A4. Nat Commun 2021; 12:6618. [PMID: 34785669 PMCID: PMC8595373 DOI: 10.1038/s41467-021-26783-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 10/21/2021] [Indexed: 11/10/2022] Open
Abstract
Previous genome-wide association studies revealed multiple common variants involved in eczema but the role of rare variants remains to be elucidated. Here, we investigate the role of rare variants in eczema susceptibility. We meta-analyze 21 study populations including 20,016 eczema cases and 380,433 controls. Rare variants are imputed with high accuracy using large population-based reference panels. We identify rare exonic variants in DUSP1, NOTCH4, and SLC9A4 to be associated with eczema. In DUSP1 and NOTCH4 missense variants are predicted to impact conserved functional domains. In addition, five novel common variants at SATB1-AS1/KCNH8, TRIB1/LINC00861, ZBTB1, TBX21/OSBPL7, and CSF2RB are discovered. While genes prioritized based on rare variants are significantly up-regulated in the skin, common variants point to immune cell function. Over 20% of the single nucleotide variant-based heritability is attributable to rare and low-frequency variants. The identified rare/low-frequency variants located in functional protein domains point to promising targets for novel therapeutic approaches to eczema. Genetic studies of eczema to date have mostly explored common genetic variation. Here, the authors perform a large meta-analysis for common and rare variants and discover 8 loci associated with eczema. Over 20% of the heritability of the condition is attributable to rare variants.
Collapse
Affiliation(s)
- Sarah Grosche
- Max-Delbrück-Center (MDC) for Molecular Medicine, Berlin, Germany.,Clinic for Pediatric Allergy, Experimental and Clinical Research Center, Charité University Medical Center, Berlin, Germany.,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Ingo Marenholz
- Max-Delbrück-Center (MDC) for Molecular Medicine, Berlin, Germany.,Clinic for Pediatric Allergy, Experimental and Clinical Research Center, Charité University Medical Center, Berlin, Germany
| | - Jorge Esparza-Gordillo
- Max-Delbrück-Center (MDC) for Molecular Medicine, Berlin, Germany.,Clinic for Pediatric Allergy, Experimental and Clinical Research Center, Charité University Medical Center, Berlin, Germany.,GlaxoSmithKline, Stevenage, UK
| | - Aleix Arnau-Soler
- Max-Delbrück-Center (MDC) for Molecular Medicine, Berlin, Germany.,Clinic for Pediatric Allergy, Experimental and Clinical Research Center, Charité University Medical Center, Berlin, Germany
| | - Erola Pairo-Castineira
- Roslin Institute, University of Edinburgh, Edinburgh, UK.,MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | | | - Tarunveer S Ahluwalia
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark.,Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Catarina Almqvist
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden.,Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Andreas Arnold
- Clinic and Polyclinic of Dermatology, University Medicine Greifswald, Greifswald, Germany
| | | | - Hansjörg Baurecht
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Kiel, Germany.,Department of Epidemiology and Preventive Medicine, University Regensburg, Regensburg, Germany
| | - Hans Bisgaard
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Sara J Brown
- Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Mariona Bustamante
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
| | - John A Curtin
- Division of Infection Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester Academic Health Science Centre and Manchester University NHS Foundation Trust, Manchester, UK
| | - Adnan Custovic
- National Lung and Heart Institute, Imperial College London, London, UK
| | - Shyamali C Dharmage
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Ana Esplugues
- Nursing School, University of Valencia, FISABIO-University Jaume I-University of Valencia Joint Research Unit of Epidemiology and Environmental Health, CIBERESP, Valencia, Spain
| | - Mario Falchi
- Department of Twins Research and Genetic Epidemiology, King's College London, London, UK
| | | | - Manuel A R Ferreira
- Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Sascha Gerdes
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Christian Gieger
- Research Unit Molecular Epidemiology, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Germany
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, and Division of Human Genetics, Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick G Holt
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Georg Homuth
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Norbert Hubner
- Max-Delbrück-Center (MDC) for Molecular Medicine, Berlin, Germany
| | - Pirro G Hysi
- Department of Twins Research and Genetic Epidemiology, King's College London, London, UK
| | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment & Health, School of Public Health, Imperial College London, London, UK.,Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Robert Karlsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology, University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, the Netherlands
| | - Susanne Lau
- Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité University Medical Center, Berlin, Germany
| | - Manuel Lutz
- Institute of Genetic Epidemiology, Helmholtz Center Munich-German Research Center for Environmental Health, Neuherberg, Germany
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Guy B Marks
- Woolcock Institute of Medical Research, University of Sydney, Sydney, Australia
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Center Munich-German Research Center for Environmental Health, Neuherberg, Germany.,Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Faculty of Medicine, LMU Munich, Munich, Germany.,Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Craig E Pennell
- School of Medicine and Public Health, Faculty of Medicine and Health, The University of Newcastle, Newcastle, Australia
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Center Munich-German Research Center for Environmental Health, Neuherberg, Germany
| | - Konrad Rawlik
- Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Colin F Robertson
- Respiratory Research, Murdoch Children's Research Institute, Melbourne, Australia
| | - Elke Rodriguez
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sylvain Sebert
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment & Health, School of Public Health, Imperial College London, London, UK.,Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Angela Simpson
- Division of Infection Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester Academic Health Science Centre and Manchester University NHS Foundation Trust, Manchester, UK
| | - Patrick M A Sleiman
- Center for Applied Genomics, Children's Hospital of Philadelphia, and Division of Human Genetics, Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marie Standl
- Institute of Epidemiology, Helmholtz Center Munich-German Research Center for Environmental Health, Neuherberg, Germany
| | - Dora Stölzl
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Center Munich-German Research Center for Environmental Health, Neuherberg, Germany.,Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Faculty of Medicine, LMU Munich, Munich, Germany.,Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Agnieszka Szwajda
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Albert Tenesa
- Roslin Institute, University of Edinburgh, Edinburgh, UK.,MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK.,Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, Edinburgh, UK
| | - Philip J Thompson
- Institute for Respiratory Health and Centre for Respiratory Health, School of Biomedical Sciences, University of Western Australia, Nedlands, Australia
| | - Vilhelmina Ullemar
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Alessia Visconti
- Department of Twins Research and Genetic Epidemiology, King's College London, London, UK
| | - Judith M Vonk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, the Netherlands
| | - Carol A Wang
- School of Medicine and Public Health, Faculty of Medicine and Health, The University of Newcastle, Newcastle, Australia
| | - Stephan Weidinger
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Matthias Wielscher
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment & Health, School of Public Health, Imperial College London, London, UK
| | | | - Chen-Jian Xu
- Department of Pediatric Pulmonology and Pediatric Allergology, University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, the Netherlands.,Department of Gastroenterology, Hepatology and Endocrinology, Centre for individualized infection medicine (CIIM), Hannover Medical School, Hannover, Germany
| | - Young-Ae Lee
- Max-Delbrück-Center (MDC) for Molecular Medicine, Berlin, Germany. .,Clinic for Pediatric Allergy, Experimental and Clinical Research Center, Charité University Medical Center, Berlin, Germany.
| |
Collapse
|
13
|
Targeting the Human β c Receptor Inhibits Contact Dermatitis in a Transgenic Mouse Model. J Invest Dermatol 2021; 142:1103-1113.e11. [PMID: 34537191 DOI: 10.1016/j.jid.2021.07.183] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/15/2021] [Accepted: 07/21/2021] [Indexed: 12/23/2022]
Abstract
Allergic contact dermatitis (ACD) is a prevalent and poorly controlled inflammatory disease caused by skin infiltration of T cells and granulocytes. The beta common (βc) cytokines GM-CSF, IL-3, and IL-5 are powerful regulators of granulocyte function that signal through their common receptor subunit βc, a property that has made βc an attractive target to simultaneously inhibit these cytokines. However, the species specificity of βc has precluded testing of inhibitors of human βc in mouse models. To overcome this problem, we developed a human βc receptor transgenic mouse strain with a hematopoietic cell‒specific expression of human βc instead of mouse βc. Human βc receptor transgenic cells responded to mouse GM-CSF and IL-5 but not to IL-3 in vitro and developed tissue pathology and cellular inflammation comparable with those in wild-type mice in a model of ACD. Similarly, Il3-/- mice developed ACD pathology comparable with that of wild-type mice. Importantly, the blocking anti-human βc antibody CSL311 strongly suppressed ear pinna thickening and histopathological changes typical of ACD and reduced accumulation of neutrophils, mast cells, and eosinophils in the skin. These results show that GM-CSF and IL-5 but not IL-3 are major mediators of ACD and define the human βc receptor transgenic mouse as a unique platform to test the inhibitors of βc in vivo.
Collapse
|
14
|
Komlósi ZI, van de Veen W, Kovács N, Szűcs G, Sokolowska M, O'Mahony L, Akdis M, Akdis CA. Cellular and molecular mechanisms of allergic asthma. Mol Aspects Med 2021; 85:100995. [PMID: 34364680 DOI: 10.1016/j.mam.2021.100995] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 12/21/2022]
Abstract
Asthma is a chronic disease of the airways, which affects more than 350 million people worldwide. It is the most common chronic disease in children, affecting at least 30 million children and young adults in Europe. Asthma is a complex, partially heritable disease with a marked heterogeneity. Its development is influenced both by genetic and environmental factors. The most common, as well as the most well characterized subtype of asthma is allergic eosinophilic asthma, which is characterized by a type 2 airway inflammation. The prevalence of asthma has substantially increased in industrialized countries during the last 60 years. The mechanisms underpinning this phenomenon are incompletely understood, however increased exposure to various environmental pollutants probably plays a role. Disease inception is thought to be enabled by a disadvantageous shift in the balance between protective and harmful lifestyle and environmental factors, including exposure to protective commensal microbes versus infection with pathogens, collectively leading to airway epithelial cell damage and disrupted barrier integrity. Epithelial cell-derived cytokines are one of the main drivers of the type 2 immune response against innocuous allergens, ultimately leading to infiltration of lung tissue with type 2 T helper (TH2) cells, type 2 innate lymphoid cells (ILC2s), M2 macrophages and eosinophils. This review outlines the mechanisms responsible for the orchestration of type 2 inflammation and summarizes the novel findings, including but not limited to dysregulated epithelial barrier integrity, alarmin release and innate lymphoid cell stimulation.
Collapse
Affiliation(s)
- Zsolt I Komlósi
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad Sqr. 4, 1089, Budapest, Hungary.
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, CH7265, Davos Wolfgand, Switzerland; Christine Kühne - Center for Allergy Research and Education, Davos, Switzerland
| | - Nóra Kovács
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad Sqr. 4, 1089, Budapest, Hungary; Lung Health Hospital, Munkácsy Mihály Str. 70, 2045, Törökbálint, Hungary
| | - Gergő Szűcs
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad Sqr. 4, 1089, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Tömő Str. 25-29, 1083, Budapest, Hungary
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, CH7265, Davos Wolfgand, Switzerland; Christine Kühne - Center for Allergy Research and Education, Davos, Switzerland
| | - Liam O'Mahony
- Department of Medicine and School of Microbiology, APC Microbiome Ireland, University College Cork, Ireland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, CH7265, Davos Wolfgand, Switzerland; Christine Kühne - Center for Allergy Research and Education, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, CH7265, Davos Wolfgand, Switzerland; Christine Kühne - Center for Allergy Research and Education, Davos, Switzerland
| |
Collapse
|
15
|
Du Y, Luan J, Jiang RP, Liu J, Ma Y. Myrcene exerts anti-asthmatic activity in neonatal rats via modulating the matrix remodeling. Int J Immunopathol Pharmacol 2021; 34:2058738420954948. [PMID: 32962470 PMCID: PMC7517990 DOI: 10.1177/2058738420954948] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Myrcene (MC), an organic hydrocarbon, was found to exert anti-inflammatory, analgesic, antimutagenic and antioxidant properties. However, the protective role of MC has not been reported against neonatal asthma. Wistar rats induced with asthma were administered with MC; while asthma control and vehicle control were maintained without MC administration. At the end of the experimental period, lung histology, inflammatory cell counts, cytokine analysis, matrix protein expressions were elucidated. Rats administered with MC exerted significant (P < 0.05) defense in protecting the lung tissue with the evidenced restoration of alveolar thickening of the lung tissues. Also, the present study elicited the anti-asthmatic activity of MC, especially via modulating the extracellular matrix protein expression in the asthma-induced animals, while a significant reduction (P < 0.05) in the fibrotic markers were found in MC treated animals. Moreover, the protective effect of MC was evidenced with reduced leukocyte infiltration in BALF, hypersensitive specific IgE levels with a profound decrease in the inflammatory cytokines such as IL-2, IL-4, IL-18, and IL-21 in MC administered animals compared to the asthma-induced group. To an extent, the markers of asthmatic inflammation such as CD14, MCP-1, and TARC were also found to be attenuated in MC exposed animals. The possible application of MC is a promising drug for the treatment of asthma-mediated complications.
Collapse
Affiliation(s)
- Yanhui Du
- Department of Pediatrics, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jie Luan
- Department of Pediatrics, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ren Peng Jiang
- Department of Pediatrics, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Juan Liu
- Department of Pediatrics, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yan Ma
- Department of Pediatrics, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
16
|
Cusack RP, Whetstone CE, Xie Y, Ranjbar M, Gauvreau GM. Regulation of Eosinophilia in Asthma-New Therapeutic Approaches for Asthma Treatment. Cells 2021; 10:cells10040817. [PMID: 33917396 PMCID: PMC8067385 DOI: 10.3390/cells10040817] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/31/2021] [Accepted: 04/04/2021] [Indexed: 02/07/2023] Open
Abstract
Asthma is a complex and chronic inflammatory disease of the airways, characterized by variable and recurring symptoms, reversible airflow obstruction, bronchospasm, and airway eosinophilia. As the pathophysiology of asthma is becoming clearer, the identification of new valuable drug targets is emerging. IL-5 is one of these such targets because it is the major cytokine supporting eosinophilia and is responsible for terminal differentiation of human eosinophils, regulating eosinophil proliferation, differentiation, maturation, migration, and prevention of cellular apoptosis. Blockade of the IL-5 pathway has been shown to be efficacious for the treatment of eosinophilic asthma. However, several other inflammatory pathways have been shown to support eosinophilia, including IL-13, the alarmin cytokines TSLP and IL-33, and the IL-3/5/GM-CSF axis. These and other alternate pathways leading to airway eosinophilia will be described, and the efficacy of therapeutics that have been developed to block these pathways will be evaluated.
Collapse
|
17
|
Nobs SP, Pohlmeier L, Li F, Kayhan M, Becher B, Kopf M. GM-CSF instigates a dendritic cell-T-cell inflammatory circuit that drives chronic asthma development. J Allergy Clin Immunol 2021; 147:2118-2133.e3. [PMID: 33440200 DOI: 10.1016/j.jaci.2020.12.638] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 11/06/2020] [Accepted: 12/04/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Steroid-resistant asthma is often characterized by high levels of neutrophils and mixed TH2/TH17 immune profiles. Indeed, neutrophils are key drivers of chronic lung inflammation in multiple respiratory diseases. Their numbers correlate strongly with disease severity, and their presence is often associated with exacerbation of chronic lung inflammation. OBJECTIVE What factors drive development of neutrophil-mediated chronic lung disease remains largely unknown, and we sought to study the role of GM-CSF as a potential regulator in chronic asthma. METHODS Different experimental animal models of chronic asthma were used in combination with alveolar macrophage-reconstitution of global GM-CSF receptor knockout mice as well as cell-type-specific knockout animals to elucidate the role of GM-CSF signaling in chronic airway inflammation. RESULTS We identify GM-CSF signaling as a critical factor regulating pulmonary accumulation of neutrophils. We show that although being not required for intrinsically regulating neutrophil migration, GM-CSF controls lung dendritic cell function, which in turn promotes T-cell-dependent recruitment of neutrophils to the airways. We demonstrate that GM-CSF regulates lung dendritic cell antigen uptake, transport, and TH2/TH17 cell priming in an intrinsic fashion, which in turn drives pulmonary granulocyte recruitment and contributes to development of airway hyperresponsiveness in chronic disease. CONCLUSIONS We identify GM-CSF as a potentially novel therapeutic target in chronic lung inflammation, describing a GM-CSF-dependent lung conventional dendritic cell-T-cell-neutrophil axis that drives chronic lung disease.
Collapse
Affiliation(s)
- Samuel Philip Nobs
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland; Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Lea Pohlmeier
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Fengqi Li
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Merve Kayhan
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Manfred Kopf
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
18
|
Kim BH, Lee S. Sophoricoside from Sophora japonica ameliorates allergic asthma by preventing mast cell activation and CD4 + T cell differentiation in ovalbumin-induced mice. Biomed Pharmacother 2021; 133:111029. [PMID: 33254020 DOI: 10.1016/j.biopha.2020.111029] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/04/2020] [Accepted: 11/15/2020] [Indexed: 12/24/2022] Open
Abstract
Asthma is a chronic inflammatory lung disorder with continuously increasing prevalence worldwide. Novel strategies are needed to prevent or improve asthma. The aim of this study was to investigate the effects of sophoricoside from Sophora japonica on allergic asthma. The mature seeds of S. japonica contain a large amount of sophoricoside. Sophoricoside reduced allergic and asthmatic symptoms by suppressing airway inflammation and antibody-antigen reaction in mouse models. In particular, sophoricoside suppressed immune cell recruitment into the airway lumens of the lungs and production of pro-inflammatory cytokines in the bronchoalveolar lavage fluid (BALF) of ovalbumin (OVA)-induced mice. It also decreased the amounts of histamine and arachidonic acid metabolites released in OVA-induced mice and antibody-antigen stimulated mast cells. In addition, sophoricoside decreased differentiation of naïve CD4+ T cells into T helper type 1 (Th1), Th2, and Th17 cells. Overall, we demonstrated that sophoricoside improved allergic asthma by suppressing mast cell activation and CD4+ T cell differentiation.
Collapse
Affiliation(s)
- Byung-Hak Kim
- Korea Institute of Science and Technology for Eastern Medicine (KISTEM), NEUMED Inc., Seoul 02440, Republic of Korea.
| | - Sanghyun Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea.
| |
Collapse
|
19
|
Hung CH, Wang KY, Liou YH, Wang JP, Huang AYS, Lee TL, Jiang ST, Liao NS, Shyu YC, Shen CKJ. Negative Regulation of the Differentiation of Flk2 - CD34 - LSK Hematopoietic Stem Cells by EKLF/KLF1. Int J Mol Sci 2020; 21:E8448. [PMID: 33182781 PMCID: PMC7697791 DOI: 10.3390/ijms21228448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/30/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
Erythroid Krüppel-like factor (EKLF/KLF1) was identified initially as a critical erythroid-specific transcription factor and was later found to be also expressed in other types of hematopoietic cells, including megakaryocytes and several progenitors. In this study, we have examined the regulatory effects of EKLF on hematopoiesis by comparative analysis of E14.5 fetal livers from wild-type and Eklf gene knockout (KO) mouse embryos. Depletion of EKLF expression greatly changes the populations of different types of hematopoietic cells, including, unexpectedly, the long-term hematopoietic stem cells Flk2- CD34- Lin- Sca1+ c-Kit+ (LSK)-HSC. In an interesting correlation, Eklf is expressed at a relatively high level in multipotent progenitor (MPP). Furthermore, EKLF appears to repress the expression of the colony-stimulating factor 2 receptor β subunit (CSF2RB). As a result, Flk2- CD34- LSK-HSC gains increased differentiation capability upon depletion of EKLF, as demonstrated by the methylcellulose colony formation assay and by serial transplantation experiments in vivo. Together, these data demonstrate the regulation of hematopoiesis in vertebrates by EKLF through its negative regulatory effects on the differentiation of the hematopoietic stem and progenitor cells, including Flk2- CD34- LSK-HSCs.
Collapse
Affiliation(s)
- Chun-Hao Hung
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Keh-Yang Wang
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Yae-Huei Liou
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Jing-Ping Wang
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Anna Yu-Szu Huang
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Tung-Liang Lee
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Si-Tse Jiang
- Department of Research and Development, National Laboratory Animal Center, National Applied Research Laboratories, Tainan 74147, Taiwan;
| | - Nah-Shih Liao
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Yu-Chiau Shyu
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
- Department of Nursing, Chang Gung University of Science and Technology, Taoyuan City 333, Taiwan
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung Branch, Keelung 204, Taiwan
| | - Che-Kun James Shen
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
- The PhD Program for Neural Regenerative Medicine, Taipei Medical University, Taipei 115, Taiwan
| |
Collapse
|
20
|
Zhao CC, Xie QM, Xu J, Yan XB, Fan XY, Wu HM. TLR9 mediates the activation of NLRP3 inflammasome and oxidative stress in murine allergic airway inflammation. Mol Immunol 2020; 125:24-31. [PMID: 32623292 DOI: 10.1016/j.molimm.2020.06.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 05/21/2020] [Accepted: 06/12/2020] [Indexed: 01/13/2023]
Abstract
Toll-like receptor 9 (TLR9) has been reported to mediate airway inflammation, however, the underlying mechanism is poorly understood. In the present study, our objective was to reveal whether TLR9 regulates NLRP3 inflammasome and oxidative stress in murine allergic airway inflammation and Raw264.7 cells. Female wild type(WT)and TLR9-/-mice on C57BL/6 background were used to induce allergic airway inflammation by challenge of OVA, and Raw264.7 cells with or without TLR9 knockdown by small interfering RNA (siRNA) were stimulated by S.aureus. The results demonstrated that deletion of TLR9 effectively attenuated OVA-induced allergic airway inflammation including inflammatory cells infiltration and goblet cell hyperplasia. Meanwhile, OVA-induced protein expression of NLRP3, caspase-1(p20) and mature IL-1β, as well as secretion of IL-1β and IL-18 in wild type mice (WT) was obviously suppressed by TLR9 deficiency. Concomitantly, the expression of oxidative markers 8-OhDG and nitrotyrosine was increased in OVA-challenged WT mice, while TLR9 deficiency significantly inhibited such increase. Similarly, in the in vitro study, we found that knockdown of TLR9 markedly suppressed S.aureus-induced activation of NLRP3 inflammasome and oxidative stress in Raw264.7 cells. Collectively, our findings indicated that TLR9 may mediate allergic airway inflammation via activating NLRP3 inflammasome and oxidative stress.
Collapse
Affiliation(s)
- Cui-Cui Zhao
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| | - Qiu-Meng Xie
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| | - Juan Xu
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| | - Xue-Bo Yan
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| | - Xiao-Yun Fan
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| | - Hui-Mei Wu
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China.
| |
Collapse
|
21
|
Yin L, Yang Z, Wu Y, Denslin V, Yu CC, Tee CA, Lim CT, Han J, Lee EH. Label-free separation of mesenchymal stem cell subpopulations with distinct differentiation potencies and paracrine effects. Biomaterials 2020; 240:119881. [PMID: 32092592 DOI: 10.1016/j.biomaterials.2020.119881] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/23/2020] [Accepted: 02/11/2020] [Indexed: 12/23/2022]
Abstract
Mesenchymal stem cells (MSCs) have the capability to differentiate into multiple cell lineages, and produce trophic factors to facilitate tissue repair and regeneration, and disease regression. However, the heterogeneity of MSCs, whether inherent or developed during culture expansion, has a significant impact on their therapeutic efficacy. Therefore, the ability to identify and select an efficacious subpopulation of MSCs targeting specific tissue damage or disease holds great clinical significance. In this study, we separated three subpopulations from culture expanded human bone marrow derived MSCs according to cell size, using a high-throughput label-free microfluidic cell sorting technology. The size-sorted MSC subpopulations varied in tri-lineage differentiation potencies. The large MSCs showed the strongest osteogenesis, medium-size MSCs were advantageous in chondrogenesis and adipogenesis, and the small MSCs showed the weakest tri-lineage differentiation. The size-sorted MSC subpopulations also exhibited different secretome profiles. The large MSC secretome possessed highest levels of osteogenic promotor proteins and senescence-associated factors, but lower levels of osteogenic inhibitor proteins compared to the medium-size MSC secretome. The medium-size MSC secretome had high levels of chondrogenic promotor proteins, and contained lower levels of chondrogenic inhibitor proteins compared to the large MSC secretome. The secretome of size-sorted MSC subpopulations showed differences in paracrine effects. We found that the secretome of large MSCs enhanced osteogenic and adipogenic potencies during MSC culture expansion, but also induced cell senescence; and the secretome of medium-size MSCs promoted chondrogenesis. This study demonstrates size-dependent differentiation potency and secretome profile of MSC subpopulations, and provides an effective and practical technology to isolate the respective subpopulations, which may be used for more targeted tissue repair and regeneration.
Collapse
Affiliation(s)
- Lu Yin
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore
| | - Zheng Yang
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore; NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore 27 Medical Drive1, DSO (Kent Bridge) Building, Level 4, Singapore, 11751, Singapore; Department of Orthopaedic Surgery, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block 11, Singapore, 119288, Singapore
| | - Yingnan Wu
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore 27 Medical Drive1, DSO (Kent Bridge) Building, Level 4, Singapore, 11751, Singapore; Department of Orthopaedic Surgery, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block 11, Singapore, 119288, Singapore
| | - Vinitha Denslin
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore 27 Medical Drive1, DSO (Kent Bridge) Building, Level 4, Singapore, 11751, Singapore; Department of Orthopaedic Surgery, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block 11, Singapore, 119288, Singapore
| | - Chia Chen Yu
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Ching Ann Tee
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore; Department of Orthopaedic Surgery, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block 11, Singapore, 119288, Singapore
| | - Chwee Teck Lim
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore; Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, T-Lab, #10-01, Singapore, 117411, Singapore; Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Engineering Block 4, #04-08, Singapore, 117583, Singapore; Institute for Health Innovation and Technology, National University of Singapore, MD6, 14 Medical Drive, #14-01, Singapore, 117599, Singapore
| | - Jongyoon Han
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore; Department of Electrical Engineering and Computer Science, Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.
| | - Eng Hin Lee
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore; NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore 27 Medical Drive1, DSO (Kent Bridge) Building, Level 4, Singapore, 11751, Singapore; Department of Orthopaedic Surgery, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block 11, Singapore, 119288, Singapore.
| |
Collapse
|
22
|
Yip KH, Wilson NJ, Pant H, Brown CL, Busfield S, Ng M, Alhamdoosh M, Woodman N, Schembri M, Tumes DJ, Vairo G, Lopez AF, Nash AD, Wilson MJ, Grimbaldeston MA, Owczarek CM. Anti-β c mAb CSL311 inhibits human nasal polyp pathophysiology in a humanized mouse xenograft model. Allergy 2020; 75:475-478. [PMID: 31505024 DOI: 10.1111/all.14041] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Kwok Ho Yip
- Centre for Cancer Biology SA Pathology and the University of South Australia UniSA CRI Adelaide SA Australia
| | - Nicholas J. Wilson
- Research and Development CSL Limited Bio21 Institute Parkville Vic. Australia
| | - Harshita Pant
- Centre for Cancer Biology SA Pathology and the University of South Australia UniSA CRI Adelaide SA Australia
- Department of Otolaryngology, Head and Neck Surgery University of Adelaide Adelaide SA Australia
| | - Christopher L. Brown
- Rhinology Clinic Royal Victorian Eye and Ear Hospital East Melbourne Vic. Australia
| | - Samantha Busfield
- Research and Development CSL Limited Bio21 Institute Parkville Vic. Australia
| | - Milica Ng
- Research and Development CSL Limited Bio21 Institute Parkville Vic. Australia
| | - Monther Alhamdoosh
- Research and Development CSL Limited Bio21 Institute Parkville Vic. Australia
| | - Naomi Woodman
- Research and Development CSL Limited Bio21 Institute Parkville Vic. Australia
| | - Mark Schembri
- Department of Otolaryngology and Head & Neck Surgery Women's and Children's Hospital Adelaide SA Australia
| | - Damon J. Tumes
- Centre for Cancer Biology SA Pathology and the University of South Australia UniSA CRI Adelaide SA Australia
| | - Gino Vairo
- Research and Development CSL Limited Bio21 Institute Parkville Vic. Australia
| | - Angel F. Lopez
- Centre for Cancer Biology SA Pathology and the University of South Australia UniSA CRI Adelaide SA Australia
| | - Andrew D. Nash
- Research and Development CSL Limited Bio21 Institute Parkville Vic. Australia
| | - Michael J. Wilson
- Research and Development CSL Limited Bio21 Institute Parkville Vic. Australia
| | - Michele A. Grimbaldeston
- Centre for Cancer Biology SA Pathology and the University of South Australia UniSA CRI Adelaide SA Australia
| | | |
Collapse
|
23
|
Samarasinghe AE, Rosch JW. Convergence of Inflammatory Pathways in Allergic Asthma and Sickle Cell Disease. Front Immunol 2020; 10:3058. [PMID: 32038616 PMCID: PMC6992560 DOI: 10.3389/fimmu.2019.03058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 12/16/2019] [Indexed: 01/19/2023] Open
Abstract
The underlying pathologies of sickle cell disease and asthma share many characteristics in terms of respiratory inflammation. The principal mechanisms of pulmonary inflammation are largely distinct, but activation of common pathways downstream of the initial inflammatory triggers may lead to exacerbation of both disease states. The altered inflammatory landscape of these respiratory pathologies can differentially impact respiratory pathogen susceptibility in patients with sickle cell disease and asthma. How these two distinct diseases behave in a comorbid setting can further exacerbate pulmonary complications associated with both disease states and impact susceptibility to respiratory infection. This review will provide a concise overview of how asthma distinctly affects individuals with sickle cell disease and how pulmonary physiology and inflammation are impacted during comorbidity.
Collapse
Affiliation(s)
- Amali E Samarasinghe
- Division of Pulmonology, Allergy-Immunology, and Sleep, Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Department of Microbiology Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Children's Foundation Research Institute, Memphis, TN, United States
| | - Jason W Rosch
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, United States
| |
Collapse
|
24
|
Damiani G, McCormick TS, Leal LO, Ghannoum MA. Recombinant human granulocyte macrophage-colony stimulating factor expressed in yeast (sargramostim): A potential ally to combat serious infections. Clin Immunol 2020; 210:108292. [DOI: 10.1016/j.clim.2019.108292] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/09/2019] [Accepted: 10/23/2019] [Indexed: 12/27/2022]
|
25
|
The Enhanced Adhesion of Eosinophils Is Associated with Their Prolonged Viability and Pro-Proliferative Effect in Asthma. J Clin Med 2019; 8:jcm8091274. [PMID: 31443410 PMCID: PMC6780628 DOI: 10.3390/jcm8091274] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 12/24/2022] Open
Abstract
Before eosinophils migrate into the bronchial lumen, they promote airway structural changes after contact with pulmonary cells and extracellular matrix components. We aimed to investigate the impact of eosinophil adhesion to their viability and pro-proliferative effect on airway smooth muscle (ASM) cells and pulmonary fibroblasts during different asthma phenotypes. A total of 39 individuals were included: 14 steroid-free non-severe allergic asthma (AA) patients, 10 severe non-allergic eosinophilic asthma (SNEA) patients, and 15 healthy control subjects (HS). For AA patients and HS groups, a bronchial allergen challenge with Dermatophagoides pteronysinnus was performed. Individual combined cells cultures were prepared between isolated peripheral blood eosinophils and ASM cells or pulmonary fibroblasts. Eosinophil adhesion was measured by evaluating their peroxidase activity, cell viability was performed by annexin V and propidium iodide staining, and proliferation by Alamar blue assay. We found that increased adhesion of eosinophils was associated with prolonged viability (p < 0.05) and an enhanced pro-proliferative effect on ASM cells and pulmonary fibroblasts in asthma (p < 0.05). However, eosinophils from SNEA patients demonstrated higher viability and inhibition of pulmonary structural cell apoptosis, compared to the AA group (p < 0.05), while their adhesive and pro-proliferative properties were similar. Finally, in the AA group, in vivo allergen-activated eosinophils demonstrated a higher adhesion, viability, and pro-proliferative effect on pulmonary structural cells compared to non-activated eosinophils (p < 0.05).
Collapse
|
26
|
Nelson RK, Brickner H, Panwar B, Ramírez-Suástegui C, Herrera-de la Mata S, Liu N, Diaz D, Alexander LEC, Ay F, Vijayanand P, Seumois G, Akuthota P. Human Eosinophils Express a Distinct Gene Expression Program in Response to IL-3 Compared with Common β-Chain Cytokines IL-5 and GM-CSF. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:329-337. [PMID: 31175163 PMCID: PMC6616007 DOI: 10.4049/jimmunol.1801668] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 05/15/2019] [Indexed: 12/31/2022]
Abstract
Despite recent advances in asthma management with anti-IL-5 therapies, many patients have eosinophilic asthma that remains poorly controlled. IL-3 shares a common β subunit receptor with both IL-5 and GM-CSF but, through α-subunit-specific properties, uniquely influences eosinophil biology and may serve as a potential therapeutic target. We aimed to globally characterize the transcriptomic profiles of GM-CSF, IL-3, and IL-5 stimulation on human circulating eosinophils and identify differences in gene expression using advanced statistical modeling. Human eosinophils were isolated from the peripheral blood of healthy volunteers and stimulated with either GM-CSF, IL-3, or IL-5 for 48 h. RNA was then extracted and bulk sequencing performed. DESeq analysis identified differentially expressed genes and weighted gene coexpression network analysis independently defined modules of genes that are highly coexpressed. GM-CSF, IL-3, and IL-5 commonly upregulated 252 genes and downregulated 553 genes, producing a proinflammatory and survival phenotype that was predominantly mediated through TWEAK signaling. IL-3 stimulation yielded the most numbers of differentially expressed genes that were also highly coexpressed (n = 119). These genes were enriched in pathways involving JAK/STAT signaling. GM-CSF and IL-5 stimulation demonstrated redundancy in eosinophil gene expression. In conclusion, IL-3 produces a distinct eosinophil gene expression program among the β-chain receptor cytokines. IL-3-upregulated genes may provide a foundation for research into therapeutics for patients with eosinophilic asthma who do not respond to anti-IL-5 therapies.
Collapse
Affiliation(s)
- Ryan K Nelson
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, La Jolla, CA 92037
- La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Howard Brickner
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, La Jolla, CA 92037
- La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Bharat Panwar
- La Jolla Institute for Immunology, La Jolla, CA 92037
| | | | | | - Neiman Liu
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, La Jolla, CA 92037
- La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Damaris Diaz
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, La Jolla, CA 92037
- La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Laura E Crotty Alexander
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, La Jolla, CA 92037
- Veterans Affairs San Diego Healthcare System, La Jolla, CA 92161; and
| | - Ferhat Ay
- La Jolla Institute for Immunology, La Jolla, CA 92037
- University of California San Diego School of Medicine, La Jolla, CA 92093
| | | | | | - Praveen Akuthota
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, La Jolla, CA 92037;
- La Jolla Institute for Immunology, La Jolla, CA 92037
| |
Collapse
|
27
|
Peptide Mapping, In Silico and In Vivo Analysis of Allergenic Sorghum Profilin Peptides. ACTA ACUST UNITED AC 2019; 55:medicina55050178. [PMID: 31117233 PMCID: PMC6571906 DOI: 10.3390/medicina55050178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/16/2019] [Accepted: 05/17/2019] [Indexed: 01/21/2023]
Abstract
Background and objectives: Nearly 20–30% of the world’s population suffers from allergic rhinitis, among them 15% are progressing to asthma conditions. Sorghum bicolor profilin (Sorb PF), one of the panallergens, was identified, but the allergen specificity is not yet characterized. Materials and Methods: To map the antigenic determinants responsible for IgE binding, the present study is focused on in silico modeling, simulation of Sorb PF and docking of the Sorb PF peptides (PF1-6) against IgG and IgE, followed by in vivo evaluation of the peptides for its allergenicity in mice. Results: Peptide PF3 and PF4 displayed high docking G-scores (−9.05) against IgE only. The mice sensitized with PF3 peptide showed increased levels of IL5, IL12, TNF-alpha, and GMCSF when compared to other peptides and controls, signifying a strong, Th2-based response. Concurrently, the Th1 pathway was inhibited by low levels of cytokine IL2, IFN-γ, and IL-10 justifying the role of PF3 in allergenic IgE response. Conclusions: Based on the results of overlapping peptides PF3 and PF4, the N-terminal part of the PF3 peptide (TGQALVI) plays a crucial role in allergenic response of Sorghum profilin.
Collapse
|
28
|
Gasiuniene E, Janulaityte I, Zemeckiene Z, Barkauskiene D, Sitkauskiene B. Elevated levels of interleukin-33 are associated with allergic and eosinophilic asthma. Scand J Immunol 2019; 89:e12724. [PMID: 30303258 DOI: 10.1111/sji.12724] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 10/01/2018] [Accepted: 10/01/2018] [Indexed: 12/30/2022]
Abstract
IL-33 is a recently discovered cytokine which plays an important role in asthma pathogenesis. AIM To evaluate serum IL-33 in patients with asthma and healthy controls, and to evaluate the association of IL-33 with different asthma phenotypes. METHODS Patients with asthma (n = 115) and healthy subjects (n = 85) were included in the study. Subjects with asthma were divided into groups according to their phenotype: allergic/non-allergic, eosinophilic/non-eosinophilic, obese/non-obese and severity according to GINA (mild, moderate and severe). The concentration of IL-33 in serum was measured by standardized enzyme-linked immunosorbent assay. RESULTS The level of IL-33 was significantly higher in patients with asthma when compared to healthy subjects (672.73 ± 104.47 pg/mL vs 268.52 ± 27.56 pg/mL, P < 0.05). IL-33 was also higher in the allergic asthma group patients when compared to non-allergic asthmatics (844.61 ± 152.08 pg/mL vs 369.56 ± 77.94 pg/mL, P < 0.05). There was a significantly higher serum IL-33 level in the eosinophilic asthma group when compared to the group of non-eosinophilic asthma patients (1001.10 ± 199.11 pg/mL vs 337.49 ± 72.68 pg/mL, P < 0.01). We did not find a significant difference in serum IL-33 level between different asthma severity groups, obese and non-obese asthmatics. CONCLUSION IL-33 is increased in asthma patients, particularly in some phenotypes: allergic asthma and eosinophilic asthma.
Collapse
Affiliation(s)
- Edita Gasiuniene
- Department of Immunology and Allergology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Ieva Janulaityte
- Department of Pulmonology, Laboratory of Pulmonology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Zivile Zemeckiene
- Department of Laboratory Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Diana Barkauskiene
- Department of Pulmonology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Brigita Sitkauskiene
- Department of Immunology and Allergology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
29
|
Nobs SP, Kayhan M, Kopf M. GM-CSF intrinsically controls eosinophil accumulation in the setting of allergic airway inflammation. J Allergy Clin Immunol 2018; 143:1513-1524.e2. [PMID: 30244025 DOI: 10.1016/j.jaci.2018.08.044] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/09/2018] [Accepted: 08/24/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Eosinophils are a therapeutic target in asthmatic patients, and GM-CSF has been suggested to control various aspects of eosinophil biology, including development, function, and survival. However, to date, the role of GM-CSF signaling in eosinophils in vivo is largely unclear. OBJECTIVE We sought to elucidate the role of GM-CSF signaling in asthmatic inflammation. METHODS Wild-type and GM-CSF receptor α (Csf2ra)-deficient mice reconstituted with Csf2ra-proficient alveolar macrophages were subjected to different models of airway inflammation to evaluate the effect of GM-CSF signaling deficiency on asthmatic inflammation in general and on eosinophils in particular. RESULTS We demonstrate that GM-CSF signaling, although being largely dispensable for eosinophil development at steady state, intrinsically promotes accumulation of eosinophils in the lung during allergic airway inflammation. In contrast, chitin-induced eosinophil accumulation in the peritoneal cavity occurs independent of GM-CSF, indicating organ specificity. We show that GM-CSF induces chemokinesis and promotes eosinophil survival in vitro, which likely contribute to eosinophil accumulation in the airways in vivo. CONCLUSION GM-CSF intrinsically promotes eosinophil accumulation in the setting of pulmonary allergic inflammation.
Collapse
Affiliation(s)
- Samuel Philip Nobs
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland; the Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Merve Kayhan
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Manfred Kopf
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
30
|
Role of the β Common (βc) Family of Cytokines in Health and Disease. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a028514. [PMID: 28716883 DOI: 10.1101/cshperspect.a028514] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The β common ([βc]/CD131) family of cytokines comprises granulocyte macrophage colony-stimulating factor (GM-CSF), interleukin (IL)-3, and IL-5, all of which use βc as their key signaling receptor subunit. This is a prototypic signaling subunit-sharing cytokine family that has unveiled many biological paradigms and structural principles applicable to the IL-2, IL-4, and IL-6 receptor families, all of which also share one or more signaling subunits. Originally identified for their functions in the hematopoietic system, the βc cytokines are now known to be truly pleiotropic, impacting on multiple cell types, organs, and biological systems, and thereby controlling the balance between health and disease. This review will focus on the emerging biological roles for the βc cytokines, our progress toward understanding the mechanisms of receptor assembly and signaling, and the application of this knowledge to develop exciting new therapeutic approaches against human disease.
Collapse
|
31
|
Foster PS, Maltby S, Rosenberg HF, Tay HL, Hogan SP, Collison AM, Yang M, Kaiko GE, Hansbro PM, Kumar RK, Mattes J. Modeling T H 2 responses and airway inflammation to understand fundamental mechanisms regulating the pathogenesis of asthma. Immunol Rev 2018; 278:20-40. [PMID: 28658543 DOI: 10.1111/imr.12549] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 02/25/2017] [Indexed: 12/12/2022]
Abstract
In this review, we highlight experiments conducted in our laboratories that have elucidated functional roles for CD4+ T-helper type-2 lymphocytes (TH 2 cells), their associated cytokines, and eosinophils in the regulation of hallmark features of allergic asthma. Notably, we consider the complexity of type-2 responses and studies that have explored integrated signaling among classical TH 2 cytokines (IL-4, IL-5, and IL-13), which together with CCL11 (eotaxin-1) regulate critical aspects of eosinophil recruitment, allergic inflammation, and airway hyper-responsiveness (AHR). Among our most important findings, we have provided evidence that the initiation of TH 2 responses is regulated by airway epithelial cell-derived factors, including TRAIL and MID1, which promote TH 2 cell development via STAT6-dependent pathways. Further, we highlight studies demonstrating that microRNAs are key regulators of allergic inflammation and potential targets for anti-inflammatory therapy. On the background of TH 2 inflammation, we have demonstrated that innate immune cells (notably, airway macrophages) play essential roles in the generation of steroid-resistant inflammation and AHR secondary to allergen- and pathogen-induced exacerbations. Our work clearly indicates that understanding the diversity and spatiotemporal role of the inflammatory response and its interactions with resident airway cells is critical to advancing knowledge on asthma pathogenesis and the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Paul S Foster
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Hunter Medical Research Institute, The University of Newcastle, Callaghan, NSW, Australia
| | - Steven Maltby
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Hunter Medical Research Institute, The University of Newcastle, Callaghan, NSW, Australia
| | - Helene F Rosenberg
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Hock L Tay
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Hunter Medical Research Institute, The University of Newcastle, Callaghan, NSW, Australia
| | - Simon P Hogan
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Adam M Collison
- Paediatric Respiratory and Sleep Medicine Unit, Priority Research Centre for Healthy Lungs and GrowUpWell, University of Newcastle and Hunter Medical Research Institute, John Hunter Children's Hospital, Newcastle, NSW, Australia
| | - Ming Yang
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Hunter Medical Research Institute, The University of Newcastle, Callaghan, NSW, Australia
| | - Gerard E Kaiko
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Hunter Medical Research Institute, The University of Newcastle, Callaghan, NSW, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Hunter Medical Research Institute, The University of Newcastle, Callaghan, NSW, Australia
| | - Rakesh K Kumar
- Pathology, UNSW Sydney, School of Medical Sciences, Sydney, NSW, Australia
| | - Joerg Mattes
- Paediatric Respiratory and Sleep Medicine Unit, Priority Research Centre for Healthy Lungs and GrowUpWell, University of Newcastle and Hunter Medical Research Institute, John Hunter Children's Hospital, Newcastle, NSW, Australia
| |
Collapse
|
32
|
Alteration of Inflammatory Mediators in the Upper and Lower Airways under Chronic Intermittent Hypoxia: Preliminary Animal Study. Mediators Inflamm 2017; 2017:4327237. [PMID: 29038619 PMCID: PMC5606044 DOI: 10.1155/2017/4327237] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 06/20/2017] [Accepted: 08/02/2017] [Indexed: 11/17/2022] Open
Abstract
Purpose We hypothesized that CIH may affect the upper airway immune system and aimed to verify whether CIH can induce airway inflammation in a murine obstructive sleep apnea (OSA) model. Methods C57BL6 male mice were exposed to intermittent hypoxia (CIH group; 5 ~ 21% FiO2, 120 sec cycles, 12 h/d, n = 6) or room air (Sham group, n = 6) for up to 4 weeks in identical chambers. Nasal and lung tissues and lavage fluid were collected and analyzed by multiplex assay. Lung lavage fluid was also utilized for FACS analysis to determine eosinophil count. Results We determined the protein level of 24 different cytokines, chemokines, and inflammatory mediators. Among various cytokines, levels of IL-1α, IL-1β, IL-4, IL-6, and IL-13 were significantly elevated in nose or lung tissue from the CIH group. In addition, MCP-1 and periostin were elevated in nose and lung tissue and lavage fluid from the CIH group. Conclusions CIH for 4 weeks altered the levels of inflammatory mediators in both the nose and lungs of mouse model. We suggest that the airway immune system may be deteriorated by CIH and allergic inflammation in the upper or lower airway could be worsened by sleep apnea.
Collapse
|
33
|
Obesity alters the lung myeloid cell landscape to enhance breast cancer metastasis through IL5 and GM-CSF. Nat Cell Biol 2017; 19:974-987. [PMID: 28737771 DOI: 10.1038/ncb3578] [Citation(s) in RCA: 195] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/20/2017] [Indexed: 12/13/2022]
Abstract
Obesity is associated with chronic, low-grade inflammation, which can disrupt homeostasis within tissue microenvironments. Given the correlation between obesity and relative risk of death from cancer, we investigated whether obesity-associated inflammation promotes metastatic progression. We demonstrate that obesity causes lung neutrophilia in otherwise normal mice, which is further exacerbated by the presence of a primary tumour. The increase in lung neutrophils translates to increased breast cancer metastasis to this site, in a GM-CSF- and IL5-dependent manner. Importantly, weight loss is sufficient to reverse this effect, and reduce serum levels of GM-CSF and IL5 in both mouse models and humans. Our data indicate that special consideration of the obese patient population is critical for effective management of cancer progression.
Collapse
|
34
|
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: 232] [Impact Index Per Article: 33.1] [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
|
35
|
Abstract
ABSTRACT
Asthma is a heterogeneous chronic inflammatory disorder of the airways, and not surprisingly, many myeloid cells play a crucial role in pathogenesis. Antigen-presenting dendritic cells are the first to recognize the allergens, pollutants, and viruses that are implicated in asthma pathogenesis, and subsequently initiate the adaptive immune response by migrating to lymph nodes. Eosinophils are the hallmark of type 2 inflammation, releasing toxic compounds in the airways and contributing to airway remodeling. Mast cells and basophils control both the early- and late-phase allergic response and contribute to alterations in smooth muscle reactivity. Finally, relatively little is known about neutrophils and macrophages in this disease. Although many of these myeloid cells respond well to treatment with inhaled steroids, there is now an increasing armamentarium of targeted biologicals that can specifically eliminate only one myeloid cell population, like eosinophils. It is only with those new tools that we will be able to fully understand the role of myeloid cells in chronic asthma in humans.
Collapse
|
36
|
Anti-colony-stimulating factor therapies for inflammatory and autoimmune diseases. Nat Rev Drug Discov 2016; 16:53-70. [DOI: 10.1038/nrd.2016.231] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
37
|
Ming M, Luo Z, Lv S, Li C. Inhalation of inactivated‑Mycobacterium phlei prevents asthma‑mediated airway hyperresponsiveness and airway eosinophilia in mice by reducing IL‑5 and IL‑13 levels. Mol Med Rep 2016; 14:5343-5349. [PMID: 27779664 DOI: 10.3892/mmr.2016.5865] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 10/04/2016] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate whether inhalation of inactivated‑Mycobacterium phlei could prevent airway hyperresponsiveness and airway eosinophilia. A total of 24 male Balb/c mice were randomly divided into three groups: Normal control group (group A), asthma model group (group B) and the intervention group (group C), (8 mice/group). Group A mice were sensitized and with challenged saline and group B with ovalbumin (OVA). Group C mice were administered with aerosol Mycobacterium phlei once daily prior to the allergen challenge. Airway responsiveness in each group was assessed. All the animals were sacrificed and lung tissues, blood samples and bronchoalveolar lavage fluid (BALF) were harvested. Cell fractionation and differential cells were counted in serum and BALF. HE staining and alcian blue/periodic acid Schiff staining were used to measure airway eosinophilic inflammation and mucus production. The levels of the cytokines IL‑5, IL‑13 and IgE were measured in lung and BALF as determined by ELISA and reverse transcription‑quantitative polymerase chain reaction assays. The results indicated that inactivated‑Mycobacterium phlei suppressed the airway hyperresponsiveness and mitigated airway eosinophilia induced by a methacholine challenge, and significantly reduced the levels of cytokines IL‑5 and IL‑13 in lung tissue and IgE level in BALF when compared with the OVA‑sensitized mice. In conclusion, inhalation of inactivated‑Mycobacterium phlei could reduce OVA‑induced airway hyperresponsiveness and may be a potential alternative therapy for allergic airway diseases.
Collapse
Affiliation(s)
- Moyu Ming
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zhixi Luo
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Shengqiu Lv
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chaoqian Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
38
|
Broughton SE, Hercus TR, Nero TL, Dottore M, McClure BJ, Dhagat U, Taing H, Gorman MA, King-Scott J, Lopez AF, Parker MW. Conformational Changes in the GM-CSF Receptor Suggest a Molecular Mechanism for Affinity Conversion and Receptor Signaling. Structure 2016; 24:1271-1281. [PMID: 27396825 DOI: 10.1016/j.str.2016.05.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 05/02/2016] [Accepted: 05/11/2016] [Indexed: 10/21/2022]
Abstract
The GM-CSF, IL-3, and IL-5 receptors constitute the βc family, playing important roles in inflammation, autoimmunity, and cancer. Typical of heterodimeric type I cytokine receptors, signaling requires recruitment of the shared subunit to the initial cytokine:α subunit binary complex through an affinity conversion mechanism. This critical process is poorly understood due to the paucity of crystal structures of both binary and ternary receptor complexes for the same cytokine. We have now solved the structure of the binary GM-CSF:GMRα complex at 2.8-Å resolution and compared it with the structure of the ternary complex, revealing distinct conformational changes. Guided by these differences we performed mutational and functional studies that, importantly, show GMRα interactions playing a major role in receptor signaling while βc interactions control high-affinity binding. These results support the notion that conformational changes underlie the mechanism of GM-CSF receptor activation and also suggest how related type I cytokine receptors signal.
Collapse
Affiliation(s)
- Sophie E Broughton
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Timothy R Hercus
- The Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA 5000, Australia
| | - Tracy L Nero
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Mara Dottore
- The Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA 5000, Australia
| | - Barbara J McClure
- The Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA 5000, Australia
| | - Urmi Dhagat
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Houng Taing
- The Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA 5000, Australia
| | - Michael A Gorman
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Jack King-Scott
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Angel F Lopez
- The Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA 5000, Australia.
| | - Michael W Parker
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia; Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
39
|
Forkuo GS, Guthrie ML, Yuan NY, Nieman AN, Kodali R, Jahan R, Stephen MR, Yocum GT, Treven M, Poe MM, Li G, Yu OB, Hartzler BD, Zahn NM, Ernst M, Emala CW, Stafford DC, Cook JM, Arnold LA. Development of GABAA Receptor Subtype-Selective Imidazobenzodiazepines as Novel Asthma Treatments. Mol Pharm 2016; 13:2026-38. [PMID: 27120014 DOI: 10.1021/acs.molpharmaceut.6b00159] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent studies have demonstrated that subtype-selective GABAA receptor modulators are able to relax precontracted human airway smooth muscle ex vivo and reduce airway hyper-responsiveness in mice upon aerosol administration. Our goal in this study was to investigate systemic administration of subtype-selective GABAA receptor modulators to alleviate bronchoconstriction in a mouse model of asthma. Expression of GABAA receptor subunits was identified in mouse lungs, and the effects of α4-subunit-selective GABAAR modulators, XHE-III-74EE and its metabolite XHE-III-74A, were investigated in a murine model of asthma (ovalbumin sensitized and challenged BALB/c mice). We observed that chronic treatment with XHE-III-74EE significantly reduced airway hyper-responsiveness. In addition, acute treatment with XHE-III-74A but not XHE-III-74EE decreased airway eosinophilia. Immune suppressive activity was also shown in activated human T-cells with a reduction in IL-2 expression and intracellular calcium concentrations [Ca(2+)]i in the presence of GABA or XHE-III-74A, whereas XHE-III-74EE showed only partial reduction of [Ca(2+)]i and no inhibition of IL-2 secretion. However, both compounds significantly relaxed precontracted tracheal rings ex vivo. Overall, we conclude that the systemic delivery of a α4-subunit-selective GABAAR modulator shows good potential for a novel asthma therapy; however, the pharmacokinetic properties of this class of drug candidates have to be improved to enable better beneficial systemic pharmacodynamic effects.
Collapse
Affiliation(s)
- Gloria S Forkuo
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee , Milwaukee, Wisconsin 53201, United States
| | - Margaret L Guthrie
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee , Milwaukee, Wisconsin 53201, United States
| | - Nina Y Yuan
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee , Milwaukee, Wisconsin 53201, United States
| | - Amanda N Nieman
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee , Milwaukee, Wisconsin 53201, United States
| | - Revathi Kodali
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee , Milwaukee, Wisconsin 53201, United States
| | - Rajwana Jahan
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee , Milwaukee, Wisconsin 53201, United States
| | - Michael R Stephen
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee , Milwaukee, Wisconsin 53201, United States
| | - Gene T Yocum
- Department of Anesthesiology, Columbia University , New York, New York 10032, United States
| | - Marco Treven
- Department of Molecular Neurosciences, Medical University of Vienna , 1090 Vienna, Austria
| | - Michael M Poe
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee , Milwaukee, Wisconsin 53201, United States
| | - Guanguan Li
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee , Milwaukee, Wisconsin 53201, United States
| | - Olivia B Yu
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee , Milwaukee, Wisconsin 53201, United States
| | - Benjamin D Hartzler
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee , Milwaukee, Wisconsin 53201, United States
| | - Nicolas M Zahn
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee , Milwaukee, Wisconsin 53201, United States
| | - Margot Ernst
- Department of Molecular Neurosciences, Medical University of Vienna , 1090 Vienna, Austria
| | - Charles W Emala
- Department of Anesthesiology, Columbia University , New York, New York 10032, United States
| | - Douglas C Stafford
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee , Milwaukee, Wisconsin 53201, United States
| | - James M Cook
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee , Milwaukee, Wisconsin 53201, United States
| | - Leggy A Arnold
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee , Milwaukee, Wisconsin 53201, United States
| |
Collapse
|
40
|
McLeod O, Silveira A, Valdes-Marquez E, Björkbacka H, Almgren P, Gertow K, Gådin JR, Bäcklund A, Sennblad B, Baldassarre D, Veglia F, Humphries SE, Tremoli E, de Faire U, Nilsson J, Melander O, Hopewell JC, Clarke R, Björck HM, Hamsten A, Öhrvik J, Strawbridge RJ. Genetic loci on chromosome 5 are associated with circulating levels of interleukin-5 and eosinophil count in a European population with high risk for cardiovascular disease. Cytokine 2016; 81:1-9. [PMID: 26821299 PMCID: PMC4837217 DOI: 10.1016/j.cyto.2016.01.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 12/18/2015] [Accepted: 01/18/2016] [Indexed: 12/30/2022]
Abstract
IL-5 is a Th2 cytokine which activates eosinophils and is suggested to have an atheroprotective role. Genetic variants in the IL5 locus have been associated with increased risk of CAD and ischemic stroke. In this study we aimed to identify genetic variants associated with IL-5 concentrations and apply a Mendelian randomisation approach to assess IL-5 levels for causal effect on intima-media thickness in a European population at high risk of coronary artery disease. We analysed SNPs within robustly associated candidate loci for immune, inflammatory, metabolic and cardiovascular traits. We identified 2 genetic loci for IL-5 levels (chromosome 5, rs56183820, BETA=0.11, P=6.73E(-5) and chromosome 14, rs4902762, BETA=0.12, P=5.76E(-6)) and one for eosinophil count (rs72797327, BETA=-0.10, P=1.41E(-6)). Both chromosome 5 loci were in the vicinity of the IL5 gene, however the association with IL-5 levels failed to replicate in a meta-analysis of 2 independent cohorts (rs56183820, BETA=0.04, P=0.2763, I(2)=24, I(2)-P=0.2516). No significant associations were observed between SNPs associated with IL-5 levels or eosinophil count and IMT measures. Expression quantitative trait analyses indicate effects of the IL-5 and eosinophil-associated SNPs on RAD50 mRNA expression levels (rs12652920 (r2=0.93 with rs56183820) BETA=-0.10, P=8.64E(-6) and rs11739623 (r2=0.96 with rs72797327) BETA=-0.23, P=1.74E(-29), respectively). Our data do not support a role for IL-5 levels and eosinophil count in intima-media thickness, however SNPs associated with IL-5 and eosinophils might influence stability of the atherosclerotic plaque via modulation of RAD50 levels.
Collapse
Affiliation(s)
- Olga McLeod
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Angela Silveira
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Elsa Valdes-Marquez
- CTSU - Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Harry Björkbacka
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Peter Almgren
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Karl Gertow
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Jesper R Gådin
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Alexandra Bäcklund
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Sennblad
- Cardiovascular Medicine Unit, Department of Medicine, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Damiano Baldassarre
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy; Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | | | | | - Elena Tremoli
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy; Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Ulf de Faire
- Division of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jan Nilsson
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Jemma C Hopewell
- CTSU - Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Robert Clarke
- CTSU - Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Hanna M Björck
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Anders Hamsten
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - John Öhrvik
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Centre for Clinical Research Västerås, Uppsala University, SE-72189 Västerås, Sweden
| | - Rona J Strawbridge
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
41
|
Wang HY, Dai Y, Wang JL, Yang XY, Jiang XG. Anti-CD69 monoclonal antibody treatment inhibits airway inflammation in a mouse model of asthma. J Zhejiang Univ Sci B 2016; 16:622-31. [PMID: 26160720 DOI: 10.1631/jzus.b1400285] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Airway inflammation and airway hyper-responsiveness (AHR) are principle pathological manifestations of asthma. Cluster of differentiation 69 (CD69) is a well-known co-stimulatory factor associated with the activation, proliferation as well as apoptosis of immune cells. This study aims to examine the effect of anti-CD69 monoclonal antibody (mAb) on the pathophysiology of a mouse model of asthma. METHODS A murine model of ovalbumin (OVA)-induced allergic airway inflammation was used in this study. Briefly, mice were injected with 20 μg chicken OVA intraperitoneally on Days 0 and 14, followed by aerosol provocation with 1% (0.01 g/ml) OVA on Days 24, 25, and 26. Anti-CD69 mAb or isotype IgG was injected intraperitoneally after OVA challenge; dexamethasone (DXM) was administrated either before or after OVA challenge. AHR, mucus production, and eosinophil infiltration in the peribronchial area were examined. The levels of granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-5 (IL-5) in bronchoalveolar lavage fluid (BALF) were also assayed as indices of airway inflammation on Day 28 following OVA injection. RESULTS Pretreatment with DXM together with anti-CD69 mAb treatment after OVA provocation completely inhibited AHR, eosinophil infiltration and mucus overproduction, and significantly reduced BALF IL-5. However, treatment with DXM alone after OVA challenge only partially inhibited AHR, eosinophil infiltration and mucus overproduction, and did not diminish BALF IL-5. Treatment with either DXM or anti-CD69 mAb did not alter the concentration of BALF GM-CSF. CONCLUSIONS Anti-CD69 mAb treatment inhibits established airway inflammation as effectively as DXM pretreatment. This study provides a potential alternative therapeutic opportunity for the clinical management of asthma and its exacerbation.
Collapse
Affiliation(s)
- Hui-ying Wang
- Department of Allergy, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China; Department of Clinical Laboratory, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China; Department of Respiratory Medicine, Hangzhou First People's Hospital, Hangzhou 310006, China; Department of Rheumatology and Clinical Immunology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China; Department of Medicine, VA Palo Alto Health Care System/Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|
42
|
Panousis C, Dhagat U, Edwards KM, Rayzman V, Hardy MP, Braley H, Gauvreau GM, Hercus TR, Smith S, Sehmi R, McMillan L, Dottore M, McClure BJ, Fabri LJ, Vairo G, Lopez AF, Parker MW, Nash AD, Wilson NJ, Wilson MJ, Owczarek CM. CSL311, a novel, potent, therapeutic monoclonal antibody for the treatment of diseases mediated by the common β chain of the IL-3, GM-CSF and IL-5 receptors. MAbs 2015; 8:436-53. [PMID: 26651396 PMCID: PMC4966837 DOI: 10.1080/19420862.2015.1119352] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 11/06/2015] [Accepted: 11/06/2015] [Indexed: 12/28/2022] Open
Abstract
The β common-signaling cytokines interleukin (IL)-3, granulocyte-macrophage colony stimulating factor (GM-CSF) and IL-5 stimulate pro-inflammatory activities of haematopoietic cells via a receptor complex incorporating cytokine-specific α and shared β common (βc, CD131) receptor. Evidence from animal models and recent clinical trials demonstrate that these cytokines are critical mediators of the pathogenesis of inflammatory airway disease such as asthma. However, no therapeutic agents, other than steroids, that specifically and effectively target inflammation mediated by all 3 of these cytokines exist. We employed phage display technology to identify and optimize a novel, human monoclonal antibody (CSL311) that binds to a unique epitope that is specific to the cytokine-binding site of the human βc receptor. The binding epitope of CSL311 on the βc receptor was defined by X-ray crystallography and site-directed mutagenesis. CSL311 has picomolar binding affinity for the human βc receptor, and at therapeutic concentrations is a highly potent antagonist of the combined activities of IL-3, GM-CSF and IL-5 on primary eosinophil survival in vitro. Importantly, CSL311 inhibited the survival of inflammatory cells present in induced sputum from human allergic asthmatic subjects undergoing allergen bronchoprovocation. Due to its high potency and ability to simultaneously suppress the activity of all 3 β common cytokines, CSL311 may provide a new strategy for the treatment of chronic inflammatory diseases where the human βc receptor is central to pathogenesis. The coordinates for the βc/CSL311 Fab complex structure have been deposited with the RCSB Protein Data Bank (PDB 5DWU).
Collapse
Affiliation(s)
- Con Panousis
- Research and Development, CSL Limited; Bio21 Molecular Science and Biotechnology Institute, Parkville Victoria, 3010, Australia
| | - Urmi Dhagat
- Australian Cancer Research Foundation Rational Drug Discovery Center, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, 3065, Australia
| | - Kirsten M. Edwards
- Research and Development, CSL Limited; Bio21 Molecular Science and Biotechnology Institute, Parkville Victoria, 3010, Australia
| | - Veronika Rayzman
- Research and Development, CSL Limited; Bio21 Molecular Science and Biotechnology Institute, Parkville Victoria, 3010, Australia
| | - Matthew P. Hardy
- Research and Development, CSL Limited; Bio21 Molecular Science and Biotechnology Institute, Parkville Victoria, 3010, Australia
| | - Hal Braley
- Research and Development, CSL Limited; Bio21 Molecular Science and Biotechnology Institute, Parkville Victoria, 3010, Australia
| | | | - Timothy R. Hercus
- Division of Human Immunology, the Center for Cancer Biology, SA Pathology and the University of South Australia, Frome Road, Adelaide, South Australia 5000 and the University of South Australia, Adelaide, South Australia 5001, Australia
| | - Steven Smith
- McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Roma Sehmi
- McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Laura McMillan
- Research and Development, CSL Limited; Bio21 Molecular Science and Biotechnology Institute, Parkville Victoria, 3010, Australia
| | - Mara Dottore
- Division of Human Immunology, the Center for Cancer Biology, SA Pathology and the University of South Australia, Frome Road, Adelaide, South Australia 5000 and the University of South Australia, Adelaide, South Australia 5001, Australia
| | - Barbara J. McClure
- Division of Human Immunology, the Center for Cancer Biology, SA Pathology and the University of South Australia, Frome Road, Adelaide, South Australia 5000 and the University of South Australia, Adelaide, South Australia 5001, Australia
| | - Louis J. Fabri
- Research and Development, CSL Limited; Bio21 Molecular Science and Biotechnology Institute, Parkville Victoria, 3010, Australia
| | - Gino Vairo
- Research and Development, CSL Limited; Bio21 Molecular Science and Biotechnology Institute, Parkville Victoria, 3010, Australia
| | - Angel F Lopez
- Division of Human Immunology, the Center for Cancer Biology, SA Pathology and the University of South Australia, Frome Road, Adelaide, South Australia 5000 and the University of South Australia, Adelaide, South Australia 5001, Australia
| | - Michael W. Parker
- Australian Cancer Research Foundation Rational Drug Discovery Center, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, 3065, Australia
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andrew D. Nash
- Research and Development, CSL Limited; Bio21 Molecular Science and Biotechnology Institute, Parkville Victoria, 3010, Australia
| | - Nicholas J. Wilson
- Research and Development, CSL Limited; Bio21 Molecular Science and Biotechnology Institute, Parkville Victoria, 3010, Australia
| | - Michael J. Wilson
- Research and Development, CSL Limited; Bio21 Molecular Science and Biotechnology Institute, Parkville Victoria, 3010, Australia
| | - Catherine M. Owczarek
- Research and Development, CSL Limited; Bio21 Molecular Science and Biotechnology Institute, Parkville Victoria, 3010, Australia
| |
Collapse
|
43
|
Genome-Wide Association Study of Staphylococcus aureus Carriage in a Community-Based Sample of Mexican-Americans in Starr County, Texas. PLoS One 2015; 10:e0142130. [PMID: 26569114 PMCID: PMC4646511 DOI: 10.1371/journal.pone.0142130] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/16/2015] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus is the number one cause of hospital-acquired infections. Understanding host pathogen interactions is paramount to the development of more effective treatment and prevention strategies. Therefore, whole exome sequence and chip-based genotype data were used to conduct rare variant and genome-wide association analyses in a Mexican-American cohort from Starr County, Texas to identify genes and variants associated with S. aureus nasal carriage. Unlike most studies of S. aureus that are based on hospitalized populations, this study used a representative community sample. Two nasal swabs were collected from participants (n = 858) 11–17 days apart between October 2009 and December 2013, screened for the presence of S. aureus, and then classified as either persistent, intermittent, or non-carriers. The chip-based and exome sequence-based single variant association analyses identified 1 genome-wide significant region (KAT2B) for intermittent and 11 regions suggestively associated with persistent or intermittent S. aureus carriage. We also report top findings from gene-based burden analyses of rare functional variation. Notably, we observed marked differences between signals associated with persistent and intermittent carriage. In single variant analyses of persistent carriage, 7 of 9 genes in suggestively associated regions and all 5 top gene-based findings are associated with cell growth or tight junction integrity or are structural constituents of the cytoskeleton, suggesting that variation in genes associated with persistent carriage impact cellular integrity and morphology.
Collapse
|
44
|
Ethanol Extract of Perilla frutescens Suppresses Allergen-Specific Th2 Responses and Alleviates Airway Inflammation and Hyperreactivity in Ovalbumin-Sensitized Murine Model of Asthma. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:324265. [PMID: 26064160 PMCID: PMC4429230 DOI: 10.1155/2015/324265] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 11/10/2014] [Indexed: 12/11/2022]
Abstract
This study was to investigate the effects of different fractions of Perilla frutescens (Pf)
leaves extracted by water or ethanol on asthma. BALB/c mice sensitized intraperitoneally and
challenged with ovalbumin (OVA) were divided into six groups. Each group of mice was
tube-feeding with 0 (control), 80 μg (PfWL), or 320 μg (PfWH) water extracts or 80 μg
(PfEL) or 320 μg (PfEH) ethanol extracts of perilla leaves daily for 3 weeks. A negative
control group (PBS) was neither sensitized nor treated with Pf. The effects of perilla leave
extracts on allergic immune response were evaluated. The results showed that OVA-specific
IL-5 and IL-13 secretions from OVA-stimulated splenocytes were significantly suppressed in
the ethanol extract groups PfEL and PfEH. Serum level of anti-OVA IgE tended to be lower in
the PfEH group. The inflammatory mediators, such as eotaxin and histamine, and total cells,
particularly eosinophils in bronchoalveolar lavage fluid (BALF), were also decreased in the
PfEL and the PfEH groups. Therefore, the PfEL and the PfEH groups had significantly lower
methacholine-induced hyperresponsiveness (AHR). In conclusion, ethanol extracts, rather than
water extract, of perilla leaves could significantly suppress Th2 responses and airway
inflammation in allergic murine model of asthma.
Collapse
|
45
|
Hammad H. Epithelial Cell Regulation of Immune Responses in the Lung. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00029-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
46
|
Yamamah GA, Abdel Meguid IE, Fatouh AA, Shaaban HH, Kantoush NA, Beharrey SF. Serum Interleukin-5 Changes in Partly Controlled Atopic Asthmatic Children. Open Access Maced J Med Sci 2014. [DOI: 10.3889/oamjms.2014.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Cytokines including Interleukin-5 play a key role in orchestrating the chronic inflammation of asthma. We aimed to determine the level of serum IL-5 in partly controlled atopic asthma in children and to assess the effect of different therapies on their levels.METHODS: The study included 40 children aged 6-12 years with partly controlled asthma. Cases were randomly divided into two groups; group ‘A’ receiving Leukotriene modifiers and group ‘B’ receiving inhaled corticosteroids; each for two months. They were compared to 20 healthy non-asthmatic, matched controls. Serum IL-5 was measured for cases on the first visit and two months after therapy. Absolute eosinophilic count and serum Ig-E were determined. Pulmonary function testing was performed using spirometer at the beginning and two months after regular therapy.RESULTS: Serum Interleukin-5 was significantly increased in asthmatic children during exacerbation and was significantly decreased after treatment. ROC curve analysis showed significant difference of IgE and PEFR after treatment with leukotriene modifier only.CONCLUSION: Serum IL-5 seems to have a role in asthma pathogenesis. Efficiency of the two therapies (ICs & LTA) was similar in this group of patients. Both treatments led to significant decline in serum IL-5, IgE levels and eosinophilic count.
Collapse
|
47
|
Comparative Burkholderia pseudomallei natural history virulence studies using an aerosol murine model of infection. Sci Rep 2014; 4:4305. [PMID: 24603493 PMCID: PMC3945929 DOI: 10.1038/srep04305] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 02/13/2014] [Indexed: 12/24/2022] Open
Abstract
Melioidosis is an endemic disease caused by the bacterium Burkholderia pseudomallei. Concerns exist regarding B. pseudomallei use as a potential bio-threat agent causing persistent infections and typically manifesting as severe pneumonia capable of causing fatal bacteremia. Development of suitable therapeutics against melioidosis is complicated due to high degree of genetic and phenotypic variability among B. pseudomallei isolates and lack of data establishing commonly accepted strains for comparative studies. Further, the impact of strain variation on virulence, disease presentation, and mortality is not well understood. Therefore, this study evaluate and compare the virulence and disease progression of B. pseudomallei strains K96243 and HBPUB10303a, following aerosol challenge in a standardized BALB/c mouse model of infection. The natural history analysis of disease progression monitored conditions such as weight, body temperature, appearance, activity, bacteremia, organ and tissue colonization (pathological and histological analysis) and immunological responses. This study provides a detailed, direct comparison of infection with different B. pseudomallei strains and set up the basis for a standardized model useful to test different medical countermeasures against Burkholderia species. Further, this protocol serves as a guideline to standardize other bacterial aerosol models of infection or to define biomarkers of infectious processes caused by other intracellular pathogens.
Collapse
|
48
|
MicroRNA-155 is essential for T(H)2-mediated allergen-induced eosinophilic inflammation in the lung. J Allergy Clin Immunol 2013; 133:1429-38, 1438.e1-7. [PMID: 24373357 DOI: 10.1016/j.jaci.2013.11.008] [Citation(s) in RCA: 173] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 11/01/2013] [Accepted: 11/06/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Allergic asthma is a chronic disease of the conducting airways characterized by T(H)2 inflammation and tissue remodeling after exposure to inhaled allergens. Although the T(H)2 profile is undisputed, the underlying molecular mechanisms leading to this abnormal T(H)2 profile remain largely unclear. MicroRNAs (miRNAs) are short noncoding RNAs that are important regulators of gene expression in the immune system. However, the role of miRNAs, specifically miR-155, in the regulation of allergic airway inflammation is unexplored. OBJECTIVES We sought to assess the contribution of miR-155 in a mouse model of allergic airway inflammation. METHODS To investigate a role for miR-155 in the regulation of allergic inflammation in vivo, we used miR-155 knockout (KO) and wild-type (WT) mice sensitized and exposed to ovalbumin. RESULTS miR-155 deficiency resulted in diminished eosinophilic inflammation and mucus hypersecretion in the lungs of allergen-sensitized and allergen-challenged mice compared with WT control animals. This was supported by a reduction in T(H)2 cell numbers and airway T(H)2 cytokine levels and complete abrogation of allergen-induced airway eotaxin-2/CCL24 and periostin levels in miR-155 KO mice. Intranasal instillation of eotaxin-2/CCL24 before allergen challenge partially restored airway eosinophilia in miR-155 KO mice, and adoptive transfer of CD4(+) T cells resulted in a similar degree of airway eosinophilia in miR-155 KO and WT mice. Furthermore, the transcription factor PU.1, a negative regulator of T(H)2 cytokine production, was upregulated in the airways of allergen-challenged miR-155 KO mice compared with WT mice. CONCLUSIONS Our data provides evidence that miR-155 contributes to the regulation of allergic airway inflammation by modulating T(H)2 responses through the transcription factor PU.1.
Collapse
|
49
|
Kim S, Karasuyama H, Lopez AF, Ouyang W, Li X, Le Gros G, Min B. IL-4 Derived from Non-T Cells Induces Basophil- and IL-3-independent Th2 Immune Responses. Immune Netw 2013; 13:249-56. [PMID: 24385943 PMCID: PMC3875783 DOI: 10.4110/in.2013.13.6.249] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 11/26/2013] [Accepted: 11/27/2013] [Indexed: 12/19/2022] Open
Abstract
How Th2 immunity develops in vivo remains obscure. Basophils have been considered key innate cells producing IL-4, a cytokine essential for Th2 immunity. Increasing evidence suggests that basophils are dispensable for the initiation of Th2 immunity. In this study, we revisited the role of basophils in Th2 immune responses induced by various types of adjuvants. Mice deficient in IL-3 or IL-3 receptor, in which basophil lymph node recruitment is completely abolished, fully developed wild type level Th2 CD4 T cell responses in response to parasite antigen or papain immunization. Similar finding was also observed in mice where basophils are inducibly ablated. Interestingly, IL-4-derived from non-T cells appeared to be critical for the generation of IL-4-producing CD4 T cells. Other Th2 promoting factors including IL-25 and thymic stromal lymphopoietin (TSLP) were dispensable. Therefore, our results suggest that IL-3- and basophil-independent in vivo Th2 immunity develops with the help of non-T cell-derived IL-4, offering an additional mechanism by which Th2 type immune responses arise in vivo.
Collapse
Affiliation(s)
- Sohee Kim
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Hajime Karasuyama
- Department of Immune Regulation and JST, CREST, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - Angel F Lopez
- Division of Human Immunology, Center for Cancer Biology, Adelaide, South Australia, Australia
| | | | - Xiaoxia Li
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Graham Le Gros
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Booki Min
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| |
Collapse
|
50
|
Lambrecht BN, Hammad H. Asthma: the importance of dysregulated barrier immunity. Eur J Immunol 2013; 43:3125-37. [PMID: 24165907 DOI: 10.1002/eji.201343730] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 10/09/2013] [Accepted: 10/24/2013] [Indexed: 12/15/2022]
Abstract
Chronic asthma is an inflammatory disease of the airway wall that leads to bronchial smooth muscle hyperreactivity and airway obstruction, caused by inflammation, goblet cell metaplasia, and airway wall remodeling. In response to allergen presentation by airway DCs, T-helper lymphocytes of the adaptive immune system control many aspects of the disease through secretion of IL-4, IL-5, IL-13, IL-17, and IL-22, and these are counterbalanced by cytokines produced by Treg cells. Many cells of the innate immune system such as mast cells, basophils, neutrophils, eosinophils, and innate lymphoid cells also play an important role in disease pathogenesis. Barrier epithelial cells are being ever more implicated in disease pathogenesis than previously thought, as these cells have in recent years been shown to sense exposure to allergens via pattern recognition receptors and to activate conventional and inflammatory-type DCs and other innate immune cells through the secretion of thymic stromal lymphopoietin, granulocyte-macrophage colony stimulating factor, IL-1, IL-33, and IL-25. Understanding this cytokine crosstalk between barrier epithelial cells, DCs, and immune cells provides important insights into the mechanisms of allergic sensitization and asthma progression as discussed in this review.
Collapse
Affiliation(s)
- Bart N Lambrecht
- VIB-Inflammation Research Center, Gent, Belgium; Department of Respiratory Medicine, University of Gent, Gent, Belgium; Department of Pulmonary Medicine, ErasmusMC, Rotterdam, The Netherlands
| | | |
Collapse
|