1
|
Kulkarni DH, Starick M, Aponte Alburquerque R, Kulkarni HS. Local complement activation and modulation in mucosal immunity. Mucosal Immunol 2024; 17:739-751. [PMID: 38838816 DOI: 10.1016/j.mucimm.2024.05.006] [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: 01/01/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024]
Abstract
The complement system is an evolutionarily conserved arm of innate immunity, which forms one of the first lines of host response to pathogens and assists in the clearance of debris. A deficiency in key activators/amplifiers of the cascade results in recurrent infection, whereas a deficiency in regulating the cascade predisposes to accelerated organ failure, as observed in colitis and transplant rejection. Given that there are over 60 proteins in this system, it has become an attractive target for immunotherapeutics, many of which are United States Food and Drug Administration-approved or in multiple phase 2/3 clinical trials. Moreover, there have been key advances in the last few years in the understanding of how the complement system operates locally in tissues, independent of its activities in circulation. In this review, we will put into perspective the abovementioned discoveries to optimally modulate the spatiotemporal nature of complement activation and regulation at mucosal surfaces.
Collapse
Affiliation(s)
- Devesha H Kulkarni
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, MO, USA
| | - Marick Starick
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Rafael Aponte Alburquerque
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Hrishikesh S Kulkarni
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
2
|
Tang J, Maihemuti N, Fang Y, Tan J, Jia M, Mu Q, Huang K, Gan H, Zhao J. JR14a: A novel antagonist of C3aR attenuates neuroinflammation in cerebral ischemia-reperfusion injury. Brain Res Bull 2024; 213:110986. [PMID: 38810789 DOI: 10.1016/j.brainresbull.2024.110986] [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: 02/08/2024] [Revised: 05/09/2024] [Accepted: 05/25/2024] [Indexed: 05/31/2024]
Abstract
Cerebral ischemia-reperfusion injury (CIRI), a prevalent stroke-related complication, can lead to severe brain damage. Inflammation is a crucial factor in CIRI pathogenesis, and the complement component 3a receptor (C3aR) could be a key mediator in the post-CIRI inflammatory cascade. In this study, the role of C3aR in CIRI was investigated utilizing a middle cerebral artery occlusion (MCAO) model in C3aR knockout (KO) mice. Magnetic resonance imaging (MRI) and neurofunctional assessments revealed that C3aR KO mice exhibited significantly diminished cerebral infarction and improved neurological impairments. Consequently, the focus shifted to searching for a small molecule antagonist of C3aR. JR14a, a new potent thiophene antagonist of C3aR, was injected intraperitoneally into mice 1-h post-MCAO model implementation. The mass spectrometry (MS) results indicated the ability of JR14a to penetrate the blood-brain barrier. Subsequent TTC staining and neurofunctional assessments revealed the efficacy of JR14a in reducing cerebral infarct volume and neurological impairment following MCAO. In addition, immunofluorescence (IF) and immunohistochemistry (IHC) demonstrated attenuated microglial activation, neutrophil infiltration, and blood-brain barrier disruption by JR14a in the MCAO model. Furthermore, enzyme-linked immunosorbent assay (ELISA) and Western blotting supported the role of JR14a in downregulating the expression levels of C3aR, tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6), as well as the phosphorylation of p65. In conclusion, the findings suggested that C3aR could be a potential therapeutic target for CIRI, and JR14a emerged as a promising treatment candidate.
Collapse
Affiliation(s)
- Jiutang Tang
- Center for Neuroscience Research, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China; Chongqing Traditional Chinese Medicine Hospital, Chongqing 400021, China
| | - Nueraili Maihemuti
- Center for Neuroscience Research, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yu Fang
- Chongqing Traditional Chinese Medicine Hospital, Chongqing 400021, China
| | - Junyi Tan
- Center for Neuroscience Research, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Mengjie Jia
- Center for Neuroscience Research, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Qinglan Mu
- Center for Neuroscience Research, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Keli Huang
- Center for Neuroscience Research, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Hui Gan
- Center for Neuroscience Research, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Jing Zhao
- Center for Neuroscience Research, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
3
|
Shah SA, Kobayashi M. Pathogenesis of chronic rhinosinusitis with nasal polyp and a prominent T2 endotype. Heliyon 2023; 9:e19249. [PMID: 37674852 PMCID: PMC10477494 DOI: 10.1016/j.heliyon.2023.e19249] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 06/29/2023] [Accepted: 08/16/2023] [Indexed: 09/08/2023] Open
Abstract
Chronic rhinosinusitis is a heterogenous and multifactorial disease, characterized by persistent inflammation of the nose and paranasal sinuses, which causes nasal obstruction, nasal discharge, facial pain, and smell disturbance. Chronic rhinosinusitis is divided into two phenotypes: chronic rhinosinusitis with nasal polyp and chronic rhinosinusitis without nasal polyp. Nasal polyps can be associated with many inflammatory cells including eosinophil cells, neutrophil cells, plasma cells, and lymphocytes. T2 endotype is characterized by the type-2 immune response and nasal polyps are associated with eosinophilic dominant infiltration. In contrast, in the T1 and T3 endotypes, chronic rhinosinusitis can be associated with neutrophilic dominant infiltration. In addition, there are mixed types of inflammation with different proportions of eosinophils-neutrophils in chronic rhinosinusitis. In the T2 endotype, there is an increase in the production of Th2 cytokines, including interleukin-4, interleukin-5, and interleukin-13, high levels of immunoglobulin-E in polyp tissue, and eosinophilia. Stimulation of Th2 cells, type-2 innate lymphoid cells, epithelial cell damage, Staphylococcus aureus enterotoxins, and autoimmune antibodies have important roles in the enhancement of Th2 cytokines and pathogenesis of chronic rhinosinusitis with nasal polyp. Monoclonal antibodies target type-2 inflammation, decrease nasal polyp size, and improve the clinical symptoms of CRSwNP patients. The present review will focus on factors involved in the pathogenesis of chronic rhinosinusitis and its treatment.
Collapse
Affiliation(s)
- Said Ahmad Shah
- Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, Mie, Japan
| | - Masayoshi Kobayashi
- Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, Mie, Japan
| |
Collapse
|
4
|
Liu JX, Chen AN, Yu Q, Shi KT, Liu YB, Guo CL, Wang ZZ, Yao Y, Pan L, Lu X, Xu K, Wang H, Zeng M, Liu C, Schleimer RP, Wu N, Liao B, Liu Z. MEX3B inhibits collagen production in eosinophilic nasal polyps by downregulating epithelial cell TGFBR3 mRNA stability. JCI Insight 2023; 8:e159058. [PMID: 36976645 PMCID: PMC10243817 DOI: 10.1172/jci.insight.159058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Although the expression of Mex3 RNA-binding family member B (MEX3B) is upregulated in human nasal epithelial cells (HNECs) predominately in the eosinophilic chronic rhinosinusitis (CRS) with nasal polyps (CRSwNP) subtype, its functions as an RNA binding protein in airway epithelial cells remain unknown. Here, we revealed the role of MEX3B based on different subtypes of CRS and demonstrated that MEX3B decreased the TGF-β receptor III (TGFBR3) mRNA level by binding to its 3' UTR and reducing its stability in HNECs. TGF-βR3 was found to be a TGF-β2-specific coreceptor in HNECs. Knocking down or overexpressing MEX3B promoted or inhibited TGF-β2-induced phosphorylation of SMAD2 in HNECs, respectively. TGF-βR3 and phosphorylated SMAD2 levels were downregulated in CRSwNP compared with controls and CRS without nasal polyps with a more prominent downregulation in the eosinophilic CRSwNP. TGF-β2 promoted collagen production in HNECs. Collagen abundance decreased and edema scores increased in CRSwNP compared with control, again more prominently in the eosinophilic type. Collagen expression in eosinophilic CRSwNP was negatively correlated with MEX3B but positively correlated with TGF-βR3. These results suggest that MEX3B inhibits tissue fibrosis in eosinophilic CRSwNP by downregulating epithelial cell TGFBR3 expression; consequently, MEX3B might be a valuable therapeutic target against eosinophilic CRSwNP.
Collapse
Affiliation(s)
- Jin-Xin Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital; and
| | - Ao-Nan Chen
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital; and
| | - Qihong Yu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Ke-Tai Shi
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital; and
| | - Yi-Bo Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital; and
| | - Cui-Lian Guo
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital; and
| | - Zhe-Zheng Wang
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital; and
| | - Yin Yao
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital; and
| | - Li Pan
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital; and
| | - Xiang Lu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital; and
| | - Kai Xu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital; and
| | - Heng Wang
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital; and
| | - Ming Zeng
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital; and
| | - Chaohong Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital; and
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Robert P. Schleimer
- Division of Allergy-Immunology, Department of Medicine; and
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ning Wu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital; and
- Department of Immunology, School of Basic Medicine, Tongji Medical College, and
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Liao
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital; and
| | - Zheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital; and
| |
Collapse
|
5
|
Zhu Y, Sun X, Tan S, Luo C, Zhou J, Zhang S, Li Z, Lin H, Zhang W. M2 macrophage-related gene signature in chronic rhinosinusitis with nasal polyps. Front Immunol 2022; 13:1047930. [PMID: 36466903 PMCID: PMC9712459 DOI: 10.3389/fimmu.2022.1047930] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/02/2022] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Chronic rhinosinusitis with nasal polyps (CRSwNP) is a common sinonasal inflammatory disorder with high heterogeneity. Increasing evidence have indicated that the infiltration of macrophages especially M2 macrophages play pivotal roles in the pathogenesis of CRSwNP, but the underlying mechanisms remain undetermined. This study sought to identify potential biomarkers related to M2 macrophages in CRSwNP. METHODS The expression datasets of GSE136825 and GSE179265 were download from Gene Expression Omnibus (GEO) database and merged. Then, CIBERSORT and weighted gene co-expression network analysis (WGCNA) algorithms were applied to identify M2 macrophage-related gene modules. Thereafter, differentially expressed genes (DEGs) related to M2 macrophages were selected to perform functional enrichment analyses. A protein-protein interaction (PPI) network was built to identify hub genes and quantitative real-time reverse transcriptions PCR was used to verify the bioinformatics results. RESULTS A total of 92 DEGs associated with M2 macrophages were identified for further analysis. The results of Gene ontology (GO) and Kyoto Encyclopedia of genes and genomes (KEGG) analyses illustrated that M2 macrophage-associated DEGs primarily enriched in immune responses and extracellular matrix structure. PPI network analysis identified 18 hub genes related to M2 macrophages that might be pivotal in the pathogenesis of CRSwNP. After verification, AIF1, C1QA, C1QB, C3AR1, CCR1, CD163, CD4, CD53, CD86, CSF1R, CYBB, FCER1G, FCGR3A, IL10RA, ITGB2, LAPTM5, PLEK, TYROBP were identified as potential M2 macrophage-related biomarkers for CRSwNP. CONCLUSION These findings yield new insights into the hub genes and mechanisms related to M2 macrophages in the pathogenesis of CRSwNP. Further studies of these hub genes would help better understand the disease progression and identify potential treatment targets.
Collapse
Affiliation(s)
- Ying Zhu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Otolaryngological Institute, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University, Shanghai, China
| | - Xiwen Sun
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Otolaryngological Institute, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University, Shanghai, China
| | - Shaolin Tan
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Otolaryngological Institute, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University, Shanghai, China
| | - Chunyu Luo
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Otolaryngological Institute, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University, Shanghai, China
| | - Jiayao Zhou
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Otolaryngological Institute, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University, Shanghai, China
| | - Shiyao Zhang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Otolaryngological Institute, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University, Shanghai, China
| | - Zhipeng Li
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Otolaryngological Institute, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University, Shanghai, China
| | - Hai Lin
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Otolaryngological Institute, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University, Shanghai, China
| | - Weitian Zhang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Otolaryngological Institute, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
6
|
Parra-Ferro M, Justice JM, Lobo BC, Munger SD, Schlosser RJ, Mulligan JK. Utilization of Nasal Mucus to Investigate the Pathophysiology of Chronic Rhinosinusitis. Am J Rhinol Allergy 2022; 36:872-883. [PMID: 35848564 DOI: 10.1177/19458924221111830] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Nasal mucus is proving to be a useful means by which to study the pathogenesis of chronic rhinosinusitis (CRS). Given the increase in publications examining nasal mucus and the lack of a review on this topic, we will focus on this noninvasive approach to studying CRS. Particular attention will be drawn towards inflammatory cytokines and biomarkers and their influence on disease severity. METHODS A literature review of papers published in English pertaining to nasal mucus was performed using the PubMed database. The search utilized combinations of the following keywords: sinusitis, polyps, sample collection, nasal mucus, or nasal secretion. Studies solely on acute or bacterial sinusitis, allergic rhinitis, or cystic fibrosis were not included. RESULTS A wide variety of materials and methods have been used to collect nasal mucus. Numerous assay types have been performed with the most common being ELISA, cytometric bead array, and proteomics. Most studies have focused on examining the levels of Th1/Th2 cytokines along with chemokines associated with type 2 immunity. Other factors identified include growth factors, senescence-associated proteins, complement, and antimicrobial defenses have also been identified. Nasal mucus cytokines have proven useful in cluster analysis and predicting postoperative improvement in Sino-nasal Outcome Test (SNOT-22) scores. One limitation of the use of nasal mucus is that some studies have suggested that nasal mucus does not always reflect the tissue microenvironment. CONCLUSIONS Nasal mucus represents a critical tool by which to examine the sinonasal microenvironment in a noninvasive manner. Unlike studies of tissue, it can be utilized in both surgically and medically managed patients and avoids the trauma of biopsies. However, studies are still needed to determine the most effective method for nasal mucus collection. Studies should also take care to confirm that nasal mucus markers do, in fact, reflect the levels of the product studied in the tissue.
Collapse
Affiliation(s)
- Mauricio Parra-Ferro
- Department of Otolaryngology-Head & Neck Surgery, 3463University of Florida, Gainesville, Florida
| | - Jeb M Justice
- Department of Otolaryngology-Head & Neck Surgery, 3463University of Florida, Gainesville, Florida.,Center for Smell and Taste, 3463University of Florida, Gainesville, Florida
| | - Brian C Lobo
- Department of Otolaryngology-Head & Neck Surgery, 3463University of Florida, Gainesville, Florida
| | - Steven D Munger
- Department of Otolaryngology-Head & Neck Surgery, 3463University of Florida, Gainesville, Florida.,Department of Pharmacology and Therapeutics, 440202University of Florida College of Medicine, Gainesville, Florida.,Center for Smell and Taste, 3463University of Florida, Gainesville, Florida.,Training Program in Chemosensory Science, 3463University of Florida, Gainesville, Florida.,Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, 3463University of Florida College of Medicine, Gainesville, Florida
| | - Rodney J Schlosser
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Jennifer K Mulligan
- Center for Smell and Taste, 3463University of Florida, Gainesville, Florida.,Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, 3463University of Florida, Gainesville, Florida
| |
Collapse
|
7
|
Mulligan JK, Nord D, Villanueva MV, Justice J, Lobo B, Schlosser RJ, Atkinson C. Role of C3a as a Novel Regulator of 25(OH)D 3 to 1α,25-Dihydroxyvitamin D 3 Metabolism in Upper Airway Epithelial Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:262-269. [PMID: 35793909 PMCID: PMC9432430 DOI: 10.4049/jimmunol.2000726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 04/29/2022] [Indexed: 05/21/2023]
Abstract
In patients with chronic rhinosinusitis with nasal polyps, primary human sinonasal epithelial cell (HSNEC) 1α-hydroxylase levels are reduced, as is their ability to metabolize 25-hydroxycholecalciferol [25(OH)D3] to its active metabolite, 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3]. In this study, we sought to identify the factor responsible for the regulation of HSNEC metabolism of 25(OH)D3, focusing on C3 and C3a. Multiple inhaled irritants trigger the release of complement components, C3 and C3a, leading to suppression of 1α-hydroxylase levels in HSNECs. Recombinant C3a was able to decrease 1α-hydroxylase and impair 25(OH)D3 to 1,25(OH)2D3 metabolism, while addition of a C3a receptor antagonist restored conversion. Conversely, 1,25(OH)2D3 suppressed Aspergillus fumigatus-induced C3 and C3a levels in HSNEC supernatant. Given the ability of 1,25(OH)2D3 to modulate LL37 in other cell types, we examined its regulation in HSNECs and relationship to C3a. 1,25(OH)2D3 stimulated the secretion of LL37, whereas A. fumigatus and C3a suppressed it. Conversely, LL37 reduced the release of C3/C3a by HSNECs. Lastly, oral steroid use and in vitro dexamethasone application both failed to increase 1α-hydroxylase or reduce C3a levels. In summary, in this article, we describe for the first time a novel relationship between complement activation and local vitamin D metabolism in airway epithelial cells. The presence of elevated C3/C3a in patients with asthma and/or chronic rhinosinusitis with nasal polyps may account for their impaired HSNEC 25(OH)D3 to 1,25(OH)2D3 metabolism and explain why they receive limited therapeutic benefit from oral vitamin D3 supplementation.
Collapse
Affiliation(s)
- Jennifer K Mulligan
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL
- Department of Otolaryngology, University of Florida, Gainesville, FL; and
| | - Dianna Nord
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL
| | - Maria V Villanueva
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL
| | - Jeb Justice
- Department of Otolaryngology, University of Florida, Gainesville, FL; and
| | - Brian Lobo
- Department of Otolaryngology, University of Florida, Gainesville, FL; and
| | - Rodney J Schlosser
- Department of Otolaryngology, Medical University of South Carolina, Charleston, SC
| | - Carl Atkinson
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL;
| |
Collapse
|
8
|
Vizuet-de-Rueda JC, Montero-Vargas JM, Galván-Morales MÁ, Porras-Gutiérrez-de-Velasco R, Teran LM. Current Insights on the Impact of Proteomics in Respiratory Allergies. Int J Mol Sci 2022; 23:ijms23105703. [PMID: 35628512 PMCID: PMC9144092 DOI: 10.3390/ijms23105703] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 12/11/2022] Open
Abstract
Respiratory allergies affect humans worldwide, causing extensive morbidity and mortality. They include allergic rhinitis (AR), asthma, pollen food allergy syndrome (PFAS), aspirin-exacerbated respiratory disease (AERD), and nasal polyps (NPs). The study of respiratory allergic diseases requires new technologies for early and accurate diagnosis and treatment. Omics technologies provide the tools required to investigate DNA, RNA, proteins, and other molecular determinants. These technologies include genomics, transcriptomics, proteomics, and metabolomics. However, proteomics is one of the main approaches to studying allergic disorders' pathophysiology. Proteins are used to indicate normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention. In this field, the principal goal of proteomics has been to discover new proteins and use them in precision medicine. Multiple technologies have been applied to proteomics, but that most used for identifying, quantifying, and profiling proteins is mass spectrometry (MS). Over the last few years, proteomics has enabled the establishment of several proteins for diagnosing and treating respiratory allergic diseases.
Collapse
|
9
|
Time-dependent effect of desensitization with wasp venom on selected parameters of the immune system. Sci Rep 2022; 12:7206. [PMID: 35504938 PMCID: PMC9064979 DOI: 10.1038/s41598-022-11155-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/12/2022] [Indexed: 11/08/2022] Open
Abstract
The emergence of tolerance during Hymenoptera venom immunotherapy (VIT) is a complex process. The main goal of VIT is to induce a change from proinflammatory Th2 response to the Th1 response. However, the immune mechanism of acquiring rapid tolerance during VIT has not yet been fully understood. Therefore, we have analyzed (in 4-time points: 0, 2, 6, and 24 weeks after the initiation phase of VIT) the concentration of complement C3, C4, and C5 components, lymphocyte subpopulations (flow cytometry), as well as histamine and tryptase serum concentrations of 43 patients with wasp venom allergy (III and IV Müller grade) classified to ultra-rush treatment and 18 volunteers as the control group (CG). We observed that VIT affected the immune system by inducing changes in the complement system (decreased C3 and C4 compartment protein concentrations) and "normalized" the percentage of lymphocytes and neutrophils in the peripheral blood. Moreover, a significant increase in the percentage of nTreg in the blood of patients treated with VIT was observed. On the other hand, there were no changes in histamine or tryptase concentrations in the blood. Increased percentage of nTreg cells is a well-known mechanism by which VIT affects the immune system. Finally, VIT also modulated the concentrations of the complement components, which may be a previously unknown VIT mechanism of action.
Collapse
|
10
|
Li K, Liu F. Analysis of competing endogenous RNA (ceRNA) crosstalk in eosinophilic chronic rhinosinusitis with nasal polyps. Int Forum Allergy Rhinol 2022; 12:1468-1479. [PMID: 35385217 PMCID: PMC10084371 DOI: 10.1002/alr.23008] [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: 01/27/2022] [Revised: 03/11/2022] [Accepted: 04/01/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Chronic rhinosinusitis with nasal polyps (CRSwNP) is one of the most common chronic inflammatory diseases, and has various phenotypes. Although its pathophysiology remains obscure, evidence has shown that dysregulation of noncoding RNAs (ncRNAs) is associated with CRSwNP. ncRNAs in the cytoplasm can act as competing endogenous RNAs (ceRNAs), which are involved in many inflammatory processes. However, the ceRNA crosstalk in CRSwNP is still unclear METHODS: We investigated expression profiles of messenger RNA (mRNA), microRNAs (miRNAs), and long noncoding RNAs (lncRNAs) in eosinophilic CRSwNP and constructed a global triple ceRNA network. RESULTS As a result, 964 differentially expressed mRNAs (DEmRs), 207 differentially expressed miRNAs (DEmiRs), and 15 differentially expressed lncRNAs (DElncRs) were identified, and a ceRNA network containing 598 miRNA-mRNA pairs and 70 lncRNA-miRNA pairs was finally constructed. Gene set enrichment analysis (GSEA) results indicated these DEmRs were mainly enriched in "cytokine-cytokine receptor interaction," "salivary secretion," "hematopoietic cell lineage," and "chemokine signaling pathway." Moreover, we also predicted the subcellular localization of the DElncRs identified in the network via bioinformatics approaches CONCLUSION: In summary, the present study provided the first comprehensive assessment of the ceRNA crosstalk in eosinophilic CRSwNP. These findings will be of interest to the understanding of the potential pathophysiology of this disease.
Collapse
Affiliation(s)
- Ke Li
- Department of Blood TransfusionTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Fang‐Fang Liu
- Department of PathologyThe Central Hospital of WuhanTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
11
|
Dutta K, Friscic J, Hoffmann MH. Targeting the tissue-complosome for curbing inflammatory disease. Semin Immunol 2022; 60:101644. [PMID: 35902311 DOI: 10.1016/j.smim.2022.101644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/07/2022] [Accepted: 07/12/2022] [Indexed: 01/15/2023]
Abstract
Hyperactivated local tissue is a cardinal feature of immune-mediated inflammatory diseases of various organs such as the joints, the gut, the skin, or the lungs. Tissue-resident structural and stromal cells, which get primed during repeated or long-lasting bouts of inflammation form the basis of this sensitization of the tissue. During priming, cells change their metabolism to make them fit for the heightened energy demands that occur during persistent inflammation. Epigenetic changes and, curiously, an activation of intracellularly expressed parts of the complement system drive this metabolic invigoration and enable tissue-resident cells and infiltrating immune cells to employ an arsenal of inflammatory functions, including activation of inflammasomes. Here we provide a current overview on complement activation and inflammatory transformation in tissue-occupying cells, focusing on fibroblasts during arthritis, and illustrate ways how therapeutics directed at complement C3 could potentially target the complosome to unprime cells in the tissue and induce long-lasting abatement of inflammation.
Collapse
Affiliation(s)
- Kuheli Dutta
- Department of Dermatology, Allergology, and Venereology, University of Lübeck, Lübeck, Germany
| | - Jasna Friscic
- Department of Dermatology, Allergology, and Venereology, University of Lübeck, Lübeck, Germany
| | - Markus H Hoffmann
- Department of Dermatology, Allergology, and Venereology, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
12
|
Chen S, Zhou A, Emmanuel B, Garcia D, Rosta E. Systematic literature review of humanistic and economic burdens of chronic rhinosinusitis with nasal polyposis. Curr Med Res Opin 2020; 36:1913-1926. [PMID: 32851882 DOI: 10.1080/03007995.2020.1815683] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES We conducted a systematic literature review (SLR) of randomized controlled trials and real-world evidence (RWE) studies to determine the humanistic (e.g. health-related/disease-specific quality of life [QOL]) and economic (e.g. direct and indirect costs) burdens of chronic rhinosinusitis with nasal polyposis (CRSwNP). METHODS The SLR adhered to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Embase, MEDLINE and Evidence-Based Medicine Reviews databases were searched using OVID. Relevant studies involving adult patients with CRSwNP published between 1 January 2008 and 16 February 2019 were included, with relevant conference abstracts from 1 January 2017, onward. RESULTS Sino-Nasal Outcomes Test (SNOT)-22 was the most frequently used disease-specific health-related QOL/patient-reported outcomes instrument for patients with CRSwNP. Baseline SNOT-22 scores ranged from 25 to 73 for surgical candidates and from 14 to 56 for medically managed patients with CRSwNP. Mean baseline EuroQol-5 Dimensions (EQ-5D) index for patients with CRSwNP ranged from 0.81 to 0.86, and mean baseline Short Form-6 Dimensions (SF-6D) ranged from 0.67 to 0.75. Three months (EQ-5D) and 5 years (SF-6D) post-endoscopic sinus surgery (ESS), rates increased from 0.81 to 0.89 and from 0.69 to 0.80, respectively. One year post-diagnosis, patients with CRSwNP had significantly more systemic prescriptions, underwent significantly more medical procedures, demonstrated greater health care resource utilization and had significantly greater mean health care costs compared with matched controls (all p < .001). Overall, for patients with initial ESS, CRSwNP was associated with higher disease-related expenditures compared with CRS without nasal polyposis (NP), even for patients who did not undergo revision surgery. CONCLUSIONS This SLR identified substantial humanistic burden among surgery candidates. RWE shows that surgeries were used to treat relatively more severe CRSwNP patients as recommended by guidelines. Patient QOL is improved significantly after surgery; however, there is a lack of evidence on patients with revision surgery. Surgery is also associated with higher costs, and the presence of NP was a predictor of revision surgery. Patients with CRSwNP demonstrate greater health care resource utilization and costs compared to those with CRS without NP. Costs associated with different severity of CRSwNP and revision surgery need to be assessed further.
Collapse
|
13
|
Smole U, Kratzer B, Pickl WF. Soluble pattern recognition molecules: Guardians and regulators of homeostasis at airway mucosal surfaces. Eur J Immunol 2020; 50:624-642. [PMID: 32246830 PMCID: PMC7216992 DOI: 10.1002/eji.201847811] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 02/25/2020] [Accepted: 03/31/2020] [Indexed: 01/08/2023]
Abstract
Maintenance of homeostasis at body barriers that are constantly challenged by microbes, toxins and potentially bioactive (macro)molecules requires complex, highly orchestrated mechanisms of protection. Recent discoveries in respiratory research have shed light on the unprecedented role of airway epithelial cells (AEC), which, besides immune cells homing to the lung, also significantly contribute to host defence by expressing membrane‐bound and soluble pattern recognition receptors (sPRR). Recent evidence suggests that distinct, evolutionary ancient, sPRR secreted by AEC might become activated by usually innocuous proteins, commonly referred to as allergens. We here provide a systematic overview on sPRR detectable in the mucus lining of AEC. Some of them become actively produced and secreted by AECs (like the pentraxins C‐reactive protein and pentraxin 3; the collectins mannose binding protein and surfactant proteins A and D; H‐ficolin; serum amyloid A; and the complement components C3 and C5). Others are elaborated by innate and adaptive immune cells such as monocytes/macrophages and T cells (like the pentraxins C‐reactive protein and pentraxin 3; L‐ficolin; serum amyloid A; and the complement components C3 and C5). Herein we discuss how sPRRs may contribute to homeostasis but sometimes also to overt disease (e.g. airway hyperreactivity and asthma) at the alveolar–air interface.
Collapse
Affiliation(s)
- Ursula Smole
- Institute of ImmunologyCenter for PathophysiologyInfectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Bernhard Kratzer
- Institute of ImmunologyCenter for PathophysiologyInfectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Winfried F. Pickl
- Institute of ImmunologyCenter for PathophysiologyInfectiology and ImmunologyMedical University of ViennaViennaAustria
| |
Collapse
|
14
|
Linking Complement C3 and B Cells in Nasal Polyposis. J Immunol Res 2020; 2020:4832189. [PMID: 32724828 PMCID: PMC7366218 DOI: 10.1155/2020/4832189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/16/2020] [Indexed: 12/30/2022] Open
Abstract
Nasal polyposis often is characterized by a persistent inflammation of the sinonasal mucosa, disease recurrence after medical or surgical intervention, and asthma comorbidity. Dysregulated complement activation may contribute to immunologic alterations and disease. To date, there is only scattered knowledge on the source and regulation of the central complement factors in the pathogenesis of nasal polyps. Here, we aim to study complement signatures, especially the C3-C3aR axis, and focus on cellular sources and targets in nasal polyps. Expression of complement factors, including C3, C5, and the anaphylatoxin receptors, was analyzed in nasal polyp tissue samples, the corresponding inferior turbinates, and healthy controls using transcriptomic methods and protein measurements. Distinct patterns of complement expression were found in nasal polyps compared to controls, characterized by an increased C3 activation and an increase in C3aR-bearing cells. In contrast, no difference was shown for epithelial-dependent C3 production. Besides low intracellular C3-expression levels for lymphocytes in general, we could identify an enlarged B lymphocyte population in nasal polyps displaying high amounts of intracellular C3. Our data suggest a prominent role for the C3-C3aR-axis in nasal polyps and, for the first time, describe a B cell population containing high levels of intracellular C3, suggesting a new role of B cells in the maintenance of the inflammation by complement.
Collapse
|
15
|
Parente R, Doni A, Bottazzi B, Garlanda C, Inforzato A. The complement system in Aspergillus fumigatus infections and its crosstalk with pentraxins. FEBS Lett 2020; 594:2480-2501. [PMID: 31994174 DOI: 10.1002/1873-3468.13744] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/19/2019] [Accepted: 01/16/2020] [Indexed: 12/13/2022]
Abstract
Aspergillosis is a life-threatening infection mostly affecting immunocompromised individuals and primarily caused by the saprophytic fungus Aspergillus fumigatus. At the host-pathogen interface, both cellular and humoral components of the innate immune system are increasingly acknowledged as essential players in the recognition and disposal of this opportunistic mold. Fundamental hereof is the contribution of the complement system, which deploys all three activation pathways in the battle against A. fumigatus, and functionally cooperates with other soluble pattern recognition molecules, including pentraxins. In particular, preclinical and clinical observations point to the long pentraxin PTX3 as a nonredundant and complement-dependent effector with protective functions against A. fumigatus. Based on past and current literature, here we discuss how the complement participates in the immune response to this fungal pathogen, and illustrate its crosstalk with the pentraxins, with a focus on PTX3. Emphasis is placed on the molecular mechanisms underlying such processes, the genetic evidence from human epidemiology, and the translational potential of the currently available knowledge.
Collapse
Affiliation(s)
- Raffaella Parente
- Department of Immunology and Inflammation, Humanitas Clinical and Research Institute - IRCCS, Milan, Italy
| | - Andrea Doni
- Department of Immunology and Inflammation, Humanitas Clinical and Research Institute - IRCCS, Milan, Italy
| | - Barbara Bottazzi
- Department of Immunology and Inflammation, Humanitas Clinical and Research Institute - IRCCS, Milan, Italy
| | - Cecilia Garlanda
- Department of Immunology and Inflammation, Humanitas Clinical and Research Institute - IRCCS, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Antonio Inforzato
- Department of Immunology and Inflammation, Humanitas Clinical and Research Institute - IRCCS, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
16
|
Wynne M, Atkinson C, Schlosser RJ, Mulligan JK. Contribution of Epithelial Cell Dysfunction to the Pathogenesis of Chronic Rhinosinusitis with Nasal Polyps. Am J Rhinol Allergy 2019; 33:782-790. [PMID: 31382760 DOI: 10.1177/1945892419868588] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background In the past, the airway epithelium was thought to be primarily an inert physical barrier. We now know that the upper airway epithelium plays a critical role in both innate and adaptive immunity, and that epithelial dysfunction is strongly associated with inflammatory airway disease. The pathogenesis of chronic rhinosinusitis is poorly understood, but growing evidence supports a key role for the airway epithelium in the pathophysiology of the disease. Objective The purpose of this study is to explore our current understanding of how dysfunction in human sinonasal epithelial cells (HSNECs) contributes to the pathogenesis of chronic rhinosinusitis with nasal polyps (CRSwNP) and to examine how current and developing therapies affect epithelial cell functions. Methods A literature review of papers published in English pertaining to epithelial cell dysfunction in patients with CRSwNP was performed using the PubMed database. The search utilized combinations of the following key words: sinusitis, polyps, epithelium, pathophysiology, barrier function, dendritic cells, eosinophils, T cells, complement, mucociliary clearance, vitamin D, cytokines, chemokines, taste receptors, steroids, saline, and therapy. Results HSNEC mucociliary clearance, barrier function, secretion of cytokines, influence on dendritic cells, influence on T-cells, regulation of eosinophils, vitamin D metabolism, complement production, and taste receptor function are altered in patients with CRSwNP and contribute to the pathogenesis of the disease. Current therapies utilized to manage CRSwNP counteract the effects of HSNEC dysfunction and relieve key symptoms of the disease. Conclusion HSNECs are key players in both innate and adaptive immunity, and altered epithelial functions are closely intertwined with the pathogenesis of CRSwNP. Our review supports further investigation of altered HSNEC function in patients with CRSwNP and supports development of novel epithelial-targeted therapies for its management.
Collapse
Affiliation(s)
- Michael Wynne
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Carl Atkinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina.,Department of Surgery, Medical University of South Carolina, Lee Patterson Allen Transplant Immunobiology Laboratory, Charleston, South Carolina
| | - Rodney J Schlosser
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Jennifer K Mulligan
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina.,Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
17
|
Frantzi M, Latosinska A, Mischak H. Proteomics in Drug Development: The Dawn of a New Era? Proteomics Clin Appl 2019; 13:e1800087. [DOI: 10.1002/prca.201800087] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/13/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Maria Frantzi
- Mosaiques Diagnostics GmbHRotenburger Straße 20 D‐30659 Hannover Germany
| | | | - Harald Mischak
- Mosaiques Diagnostics GmbHRotenburger Straße 20 D‐30659 Hannover Germany
- BHF Glasgow Cardiovascular Research CentreUniversity of Glasgow G12 8TA Glasgow UK
| |
Collapse
|