1
|
Ishikawa K, Suzuki H, Ohishi T, Li G, Tanaka T, Kawada M, Ohkoshi A, Kaneko MK, Katori Y, Kato Y. Anti-CD44 Variant 10 Monoclonal Antibody Exerts Antitumor Activity in Mouse Xenograft Models of Oral Squamous Cell Carcinomas. Int J Mol Sci 2024; 25:9190. [PMID: 39273139 PMCID: PMC11395228 DOI: 10.3390/ijms25179190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
CD44 regulates cell adhesion, proliferation, survival, and stemness and has been considered a tumor therapy target. CD44 possesses the shortest CD44 standard (CD44s) and a variety of CD44 variant (CD44v) isoforms. Since the expression of CD44v is restricted in epithelial cells and carcinomas compared to CD44s, CD44v has been considered a promising target for monoclonal antibody (mAb) therapy. We previously developed an anti-CD44v10 mAb, C44Mab-18 (IgM, kappa), to recognize the variant exon 10-encoded region. In the present study, a mouse IgG2a version of C44Mab-18 (C44Mab-18-mG2a) was generated to evaluate the antitumor activities against CD44-positive cells compared with the previously established anti-pan CD44 mAb, C44Mab-46-mG2a. C44Mab-18-mG2a exhibited higher reactivity compared with C44Mab-46-mG2a to CD44v3-10-overexpressed CHO-K1 (CHO/CD44v3-10) and oral squamous cell carcinoma cell lines (HSC-2 and SAS) in flow cytometry. C44Mab-18-mG2a exerted a superior antibody-dependent cellular cytotoxicity (ADCC) against CHO/CD44v3-10. In contrast, C44Mab-46-mG2a showed a superior complement-dependent cytotoxicity (CDC) against CHO/CD44v3-10. A similar tendency was observed in ADCC and CDC against HSC-2 and SAS. Furthermore, administering C44Mab-18-mG2a or C44Mab-46-mG2a significantly suppressed CHO/CD44v3-10, HSC-2, and SAS xenograft tumor growth compared with the control mouse IgG2a. These results indicate that C44Mab-18-mG2a could be a promising therapeutic regimen for CD44v10-positive tumors.
Collapse
Affiliation(s)
- Kenichiro Ishikawa
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan
| | - Hiroyuki Suzuki
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, 18-24 Miyamoto, Numazu-shi 410-0301, Shizuoka, Japan
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Oncology, Microbial Chemistry Research Foundation, 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Guanjie Li
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan
| | - Manabu Kawada
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Oncology, Microbial Chemistry Research Foundation, 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Akira Ohkoshi
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan
| | - Yukio Katori
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan
| |
Collapse
|
2
|
Li W, You L, Lin J, Zhang J, Zhou Z, Wang T, Wu Y, Zheng C, Gao Y, Kong X, Sun X. An herbal formula Shenlian decoction upregulates M1/M2 macrophage proportion in hepatocellular carcinoma by suppressing complement cascade. Biomed Pharmacother 2024; 177:116943. [PMID: 38878636 DOI: 10.1016/j.biopha.2024.116943] [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: 01/03/2024] [Revised: 06/04/2024] [Accepted: 06/10/2024] [Indexed: 07/28/2024] Open
Abstract
The immunosuppressive microenvironment is a vital factor for the hepatocellular carcinoma (HCC) progression. However, effective treatment is lacking at current. Shenlian decoction (SLD) is a registered herbal therapy for the HCC treatment, but the underlying mechanism of SLD remains largely elusive. Here, we aimed to explore the anti-tumor effect of SLD in the treatment of HCC. SLD was intragastrically given after the tumor initiation in β-catenin/C-Met or DEN and CCl4 induced HCC mouse model. The tumor growth levels were evaluated by liver weight and histological staining. The tumor-infiltrating immune cells were detected by immunological staining and flow cytometry. The mechanism of the SLD was detected by non-targeted proteomics and verified by a cell co-culture system. The result showed that SLD significantly attenuated HCC progression. SLD promoted macrophage infiltration and increased the M1/M2 macrophage ratio within the tumor tissues. Non-targeted proteomics showed the inhibition of complement C5/C5a signaling is the key mechanism of SLD. Immunological staining showed SLD inhibited C5/C5a expression and C5aR1+ macrophage infiltration. The suggested mechanism was demonstrated by application of C5aR1 inhibitor, PMX-53 in mouse HCC model. Hepatoma cell-macrophage co-culture showed SLD targeted hepatoma cells and inhibited the supernatant-induced macrophage M2 polarization. SLD inhibited AMPK/p38 signaling which is an upstream mechanism of C5 transcription. In conclusion, we found SLD relieved immune-suppressive environment by inhibiting C5 expression. SLD could suppress the C5 secretion in hepatoma cells via inhibition of AMPK/p38 signaling. We suggested that SLD is a potential herbal therapy for the treatment of HCC by alleviating immune-suppressive status.
Collapse
Affiliation(s)
- Wenxuan Li
- Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China; Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liping You
- Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China; Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiacheng Lin
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jinghao Zhang
- Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhijia Zhou
- Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China; Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Wang
- Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China; Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuelan Wu
- Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China; Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chao Zheng
- Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yueqiu Gao
- Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China; Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xiaoni Kong
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xuehua Sun
- Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
3
|
Suzuki H, Ohishi T, Tanaka T, Kaneko MK, Kato Y. Anti-HER2 Cancer-Specific mAb, H 2Mab-250-hG 1, Possesses Higher Complement-Dependent Cytotoxicity than Trastuzumab. Int J Mol Sci 2024; 25:8386. [PMID: 39125956 PMCID: PMC11313270 DOI: 10.3390/ijms25158386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Cancer-specific monoclonal antibodies (CasMabs) that recognize cancer-specific antigens with in vivo antitumor efficacy are innovative therapeutic strategies for minimizing adverse effects. We previously established a cancer-specific anti-human epidermal growth factor receptor 2 (HER2) monoclonal antibody (mAb), H2Mab-250/H2CasMab-2. In flow cytometry and immunohistochemistry, H2Mab-250 reacted with HER2-positive breast cancer cells but did not show reactivity to normal epithelial cells. In contrast, a clinically approved anti-HER2 mAb, trastuzumab, strongly recognizes both breast cancer and normal epithelial cells in flow cytometry. The human IgG1 version of H2Mab-250 (H2Mab-250-hG1) possesses compatible in vivo antitumor effects against breast cancer xenografts to trastuzumab despite the lower affinity and effector activation than trastuzumab in vitro. This study compared the antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cellular cytotoxicity (CDC) between H2Mab-250-hG1 and trastuzumab. Both H2Mab-250-hG1 and trastuzumab showed ADCC activity against HER2-overexpressed Chinese hamster ovary -K1 and breast cancer cell lines (BT-474 and SK-BR-3) in the presence of human natural killer cells. Some tendency was observed where trastuzumab showed a more significant ADCC effect compared to H2Mab-250-hG1. Importantly, H2Mab-250-hG1 exhibited superior CDC activity in these cells compared to trastuzumab. Similar results were obtained in the mouse IgG2a types of both H2Mab-250 and trastuzumab. These results suggest the different contributions of ADCC and CDC activities to the antitumor effects of H2Mab-250-hG1 and trastuzumab, and indicate a future direction for the clinical development of H2Mab-250-hG1 against HER2-positive tumors.
Collapse
Affiliation(s)
- Hiroyuki Suzuki
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (H.S.); (T.T.); (M.K.K.)
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, 18-24 Miyamoto, Numazu-shi, Shizuoka 410-0301, Japan;
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Oncology, Microbial Chemistry Research Foundation, 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (H.S.); (T.T.); (M.K.K.)
| | - Mika K. Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (H.S.); (T.T.); (M.K.K.)
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (H.S.); (T.T.); (M.K.K.)
| |
Collapse
|
4
|
Gao P, Tang K, Lu Y, Wang M, Wang W, Wang T, Sun Y, Zhao J, Mao Y. Increased expression of ficolin-1 is associated with airway obstruction in asthma. BMC Pulm Med 2023; 23:470. [PMID: 37996869 PMCID: PMC10668451 DOI: 10.1186/s12890-023-02772-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 11/17/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND The activated complement cascade is involved in asthmatic airway inflammation. Ficolins are essential for innate immunity and can activate the complement lectin pathway. Despite this, the significance of ficolins in asthma has yet to be determined. This study aimed to explore the presence of ficolins in individuals with asthma and to determine the relationship between ficolins and clinical characteristics. METHODS For the study, 68 asthmatic patients and 30 healthy control subjects were recruited. Enzyme-linked immunosorbent assay was used to determine plasma ficolin-1, ficolin-2, and ficolin-3 concentrations both before and after inhaled corticosteroid (ICS) therapy. Further, the associations of plasma ficolin-1 level with pulmonary function and asthma control questionnaire (ACQ) score were examined in the asthma patients. RESULTS Patients with asthma exhibited significantly elevated plasma ficolin-1 levels (median, 493.9 ng/mL; IQR, 330.2-717.8 ng/mL) in comparison to healthy controls (median, 330.6 ng/mL; IQR, 233.8-371.1 ng/mL). After ICS treatment, plasma ficolin-1 (median, 518.1 ng/mL; IQR, 330.2-727.0 ng/mL) in asthmatic patients was significantly reduced (median, 374.7 ng/mL; IQR, 254.8-562.5 ng/mL). Additionally, ficolin-1 expressions in plasma were significantly correlated with pulmonary function parameters and ACQ score in asthmatic patients. Asthma patients with higher plasma ficolin-1 levels demonstrated poorer lung function than those with lower plasma ficolin-1 levels. CONCLUSIONS The results revealed that asthmatic patients had higher plasma ficolin-1 concentrations, which decreased after ICS treatment and were linked to their lung function, implying a potential involvement of ficolin-1 in asthma pathogenesis.
Collapse
Affiliation(s)
- Pengfei Gao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, China.
- Department of Respiratory and Critical Care Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Kun Tang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yanjiao Lu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Meijia Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, China
| | - Tongsheng Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, China
| | - Yuxia Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, China
| | - Jianping Zhao
- Department of Respiratory and Critical Care Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Yimin Mao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, China.
| |
Collapse
|
5
|
Li H, Ma L, Li W, Zheng B, Wang J, Chen S, Wang Y, Ge F, Qin B, Zheng X, Deng Y, Zeng R. Proline metabolism reprogramming of trained macrophages induced by early respiratory infection combined with allergen sensitization contributes to development of allergic asthma in childhood of mice. Front Immunol 2022; 13:977235. [PMID: 36211408 PMCID: PMC9533174 DOI: 10.3389/fimmu.2022.977235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/02/2022] [Indexed: 11/13/2022] Open
Abstract
Background Infants with respiratory syncytial virus (RSV)-associated bronchiolitis are at increased risk of childhood asthma. Recent studies demonstrated that certain infections induce innate immune memory (also termed trained immunity), especially in macrophages, to respond more strongly to future stimuli with broad specificity, involving in human inflammatory diseases. Metabolic reprogramming increases the capacity of the innate immune cells to respond to a secondary stimulation, is a crucial step for the induction of trained immunity. We hypothesize that specific metabolic reprogramming of lung trained macrophages induced by neonatal respiratory infection is crucial for childhood allergic asthma. Objective To address the role of metabolic reprogramming in lung trained macrophages induced by respiratory virus infection in allergic asthma. Methods Neonatal mice were infected and sensitized by the natural rodent pathogen Pneumonia virus of mice (PVM), a mouse equivalent strain of human RSV, combined with ovalbumin (OVA). Lung CD11b+ macrophages in the memory phase were re-stimulated to investigate trained immunity and metabonomics. Adoptive transfer, metabolic inhibitor and restore experiments were used to explore the role of specific metabolic reprogramming in childhood allergic asthma. Results PVM infection combined with OVA sensitization in neonatal mice resulted in non-Th2 (Th1/Th17) type allergic asthma following OVA challenge in childhood of mice. Lung CD11b+ macrophages in the memory phage increased, and showed enhanced inflammatory responses following re-stimulation, suggesting trained macrophages. Adoptive transfer of the trained macrophages mediated the allergic asthma in childhood. The trained macrophages showed metabolic reprogramming after re-stimulation. Notably, proline biosynthesis remarkably increased. Inhibition of proline biosynthesis suppressed the development of the trained macrophages as well as the Th1/Th17 type allergic asthma, while supplement of proline recovered the trained macrophages as well as the allergic asthma. Conclusion Proline metabolism reprogramming of trained macrophages induced by early respiratory infection combined with allergen sensitization contributes to development of allergic asthma in childhood. Proline metabolism could be a well target for prevention of allergic asthma in childhood.
Collapse
Affiliation(s)
- Hanglin Li
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
| | - Linyan Ma
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
| | - Wenjian Li
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Boyang Zheng
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Junhai Wang
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
| | - Shunyan Chen
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
| | - Yang Wang
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
| | - Fei Ge
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
| | - Beibei Qin
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Clinical Lab, Hebei Provincial People’s Hospital, Shijiazhuang, China
| | - Xiaoqing Zheng
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Yuqing Deng
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Ruihong Zeng
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Shijiazhuang, China
- *Correspondence: Ruihong Zeng,
| |
Collapse
|
6
|
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: 0.5] [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
|
7
|
Hosseini B, Berthon BS, Starkey MR, Collison A, McLoughlin RF, Williams EJ, Nichol K, Wark PA, Jensen ME, Da Silva Sena CR, Baines KJ, Mattes J, Wood LG. Children With Asthma Have Impaired Innate Immunity and Increased Numbers of Type 2 Innate Lymphoid Cells Compared With Healthy Controls. Front Immunol 2021; 12:664668. [PMID: 34220812 PMCID: PMC8248177 DOI: 10.3389/fimmu.2021.664668] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/17/2021] [Indexed: 12/20/2022] Open
Abstract
Background Asthma is the most frequent cause of hospitalisation among children; however, little is known regarding the effects of asthma on immune responses in children. Objective The present study aimed to evaluate cytokine responses of peripheral blood mononuclear cells (PBMCs), PBMC composition and lung function in children with and without asthma. Methods Using a case-control design, we compared 48 children with asthma aged 3-11 years with 14 age-matched healthy controls. PBMC composition and cytokine production including interferon (IFN)-γ, interleukin (IL)-1β, IL-5 and lL-6 following stimulation with rhinovirus-1B (RV1B), house dust mite (HDM) and lipopolysaccharide (LPS) were measured. Lung function was assessed using impulse oscillometry and nitrogen multiple breath washout. Results The frequency of group 2 innate lymphoid cells were significantly higher in asthmatics and PBMCs from asthmatics had deficient IFN-γ production in response to both RV1B and LPS compared with controls (P<0.01). RV1B-induced IL-1β response and HDM-stimulated IL-5 production was higher in asthmatics than controls (P<0.05). In contrast, IL-1β and IL-6 were significantly reduced in response to HDM and LPS in asthmatics compared to controls (P<0.05). Children with asthma also had reduced pulmonary function, indicated by lower respiratory reactance as well as higher area of-reactance and lung clearance index values compared with controls (P<0.05). Conclusion Our study indicates that children with asthma have a reduced lung function in concert with impaired immune responses and altered immune cell subsets. Improving our understanding of immune responses to viral and bacterial infection in childhood asthma can help to tailor management of the disease.
Collapse
Affiliation(s)
- Banafshe Hosseini
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Bronwyn S Berthon
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Malcolm R Starkey
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia.,Priority Research Centre GrowUpWell, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia.,Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Adam Collison
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia.,Priority Research Centre GrowUpWell, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Rebecca F McLoughlin
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Evan J Williams
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Kristy Nichol
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Peter Ab Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Megan E Jensen
- Priority Research Centre GrowUpWell, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Carla Rebeca Da Silva Sena
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Katherine J Baines
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Joerg Mattes
- Priority Research Centre GrowUpWell, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Lisa G Wood
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia.,Priority Research Centre GrowUpWell, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
8
|
Lu P, Ma Y, Wei S, Liang X. The dual role of complement in cancers, from destroying tumors to promoting tumor development. Cytokine 2021; 143:155522. [PMID: 33849765 DOI: 10.1016/j.cyto.2021.155522] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 12/30/2022]
Abstract
Complement is an important branch of innate immunity; however, its biological significance goes far beyond the scope of simple nonspecific defense and involves a variety of physiological functions, including the adaptive immune response. In this review, to unravel the complex relationship between complement and tumors, we reviewed the high diversity of complement components in cancer and the heterogeneity of their production and activation pathways. In the tumor microenvironment, complement plays a dual regulatory role in the occurrence and development of tumors, affecting the outcomes of the immune response. We explored the differential expression levels of various complement components in human cancers via the Oncomine database. The gene expression profiling interactive analysis (GEPIA) tool and Kaplan-Meier plotter (K-M plotter) confirmed the correlation between differentially expressed complement genes and tumor prognosis. The tumor immune estimation resource (TIMER) database was used to statistically analyze the effect of complement on tumor immune infiltration. Finally, with a view to the role of complement in regulating T cell metabolism, complement could be a potential target for immunotherapies. Targeting complement to regulate the antitumor immune response seems to have potential for future treatment strategies. However, there are still many complex problems, such as who will benefit from this therapy and how to select the right therapeutic target and determine the appropriate drug concentration. The solutions to these problems depend on a deeper understanding of complement generation, activation, and regulatory and control mechanisms.
Collapse
Affiliation(s)
- Ping Lu
- Department of Medical Oncology, Hubei Cancer Hospital, the Seventh Clinical School Affiliated of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Colorectal Cancer Clinical Research Center of HuBei Province, Wuhan, China; Colorectal Cancer Clinical Research Center of Wuhan, Wuhan, China
| | - Yifei Ma
- Department of Gastrointestinal Oncology Surgery, Hubei Cancer Hospital, the Seventh Clinical School Affiliated with Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Colorectal Cancer Clinical Research Center of HuBei Province, Wuhan, China; Colorectal Cancer Clinical Research Center of Wuhan, Wuhan, China
| | - Shaozhong Wei
- Department of Gastrointestinal Oncology Surgery, Hubei Cancer Hospital, the Seventh Clinical School Affiliated with Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Colorectal Cancer Clinical Research Center of HuBei Province, Wuhan, China; Colorectal Cancer Clinical Research Center of Wuhan, Wuhan, China.
| | - Xinjun Liang
- Department of Medical Oncology, Hubei Cancer Hospital, the Seventh Clinical School Affiliated of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Colorectal Cancer Clinical Research Center of HuBei Province, Wuhan, China; Colorectal Cancer Clinical Research Center of Wuhan, Wuhan, China.
| |
Collapse
|
9
|
Gadwa J, Bickett TE, Darragh LB, Knitz MW, Bhatia S, Piper M, Van Court B, Bhuvane S, Nguyen D, Nangia V, Kleczko EK, Nemenoff RA, Karam SD. Complement C3a and C5a receptor blockade modulates regulatory T cell conversion in head and neck cancer. J Immunother Cancer 2021; 9:e002585. [PMID: 33789881 PMCID: PMC8016081 DOI: 10.1136/jitc-2021-002585] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Resistance to therapy is a major problem in treating head and neck squamous cell carcinomas (HNSCC). Complement system inhibition has been shown to reduce tumor growth, metastasis, and therapeutic resistance in other tumor models, but has yet to be explored in the context of HNSCC. Here, we tested the effects of complement inhibition and its therapeutic potential in HNSCC. METHODS We conducted our studies using two Human Papilloma Virus (HPV)-negative HNSCC orthotopic mouse models. Complement C3aR and C5aR1 receptor antagonists were paired with radiation therapy (RT). Tumor growth was measured and immune populations from tumor, lymph node, and peripheral blood were compared among various treatment groups. Genetically engineered mouse models DEREG and C3-/- were used in addition to standard wild type models. Flow cytometry, clinical gene sets, and in vitro assays were used to evaluate the role complement receptor blockade has on the immunological makeup of the tumor microenvironment. RESULTS In contrast to established literature, inhibition of complement C3a and C5a signaling using receptor antagonists accelerated tumor growth in multiple HNSCC cell lines and corresponded with increased frequency of regulatory T cell (Treg) populations. Local C3a and C5a signaling has importance for CD4 T cell homeostasis and eventual development into effector phenotypes. Interruption of this signaling axis drives a phenotypic conversion of CD4+ T cells into Tregs, characterized by enhanced expression of Foxp3. Depletion of Tregs reversed tumor growth, and combination of Treg depletion and C3a and C5a receptor inhibition decreased tumor growth below that of the control groups. Complete knockout of C3 does not harbor the expected effect on tumor growth, indicating a still undetermined compensatory mechanism. Dexamethasone is frequently prescribed to patients undergoing RT and inhibits complement activation. We report no deleterious effects associated with dexamethasone due to complement inhibition. CONCLUSIONS Our data establish Tregs as a pro-tumorigenic driver during complement inhibition and provide evidence that targeted C3a and C5a receptor inhibition may add therapeutic advantage when coupled with anti-Treg therapy.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Complement C3/genetics
- Complement C3/metabolism
- Complement Inactivating Agents/toxicity
- Dexamethasone/toxicity
- Forkhead Transcription Factors/metabolism
- Head and Neck Neoplasms/genetics
- Head and Neck Neoplasms/immunology
- Head and Neck Neoplasms/metabolism
- Head and Neck Neoplasms/pathology
- Humans
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Receptor, Anaphylatoxin C5a/antagonists & inhibitors
- Receptor, Anaphylatoxin C5a/metabolism
- Receptors, Complement/antagonists & inhibitors
- Receptors, Complement/metabolism
- Signal Transduction
- Squamous Cell Carcinoma of Head and Neck/genetics
- Squamous Cell Carcinoma of Head and Neck/immunology
- Squamous Cell Carcinoma of Head and Neck/metabolism
- Squamous Cell Carcinoma of Head and Neck/pathology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Time Factors
- Tumor Burden/drug effects
- Mice
Collapse
Affiliation(s)
- Jacob Gadwa
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Thomas E Bickett
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laurel B Darragh
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Michael William Knitz
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Shilpa Bhatia
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Miles Piper
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Benjamin Van Court
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Shiv Bhuvane
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Diemmy Nguyen
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Varuna Nangia
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Emily K Kleczko
- Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Raphael A Nemenoff
- Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sana D Karam
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
10
|
Nacaroglu HT, Büke Ö, Gayret ÖB, Erol M, Zengi O. Serum osteoprotegerin levels in school-aged children with asthma. Allergol Immunopathol (Madr) 2020; 48:484-489. [PMID: 32284263 DOI: 10.1016/j.aller.2019.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/28/2019] [Accepted: 11/12/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND Various inflammatory biomarkers have been used in asthma cases for evaluating inflammation, however it has been determined that the majority of these biomarkers are insufficient for putting forth the course and severity of the disease. Osteoprotegerin is a glycoprotein mediator in the lung and macrophages. As far as we know, there are no studies about the role played by osteoprotegerin in child patients with asthma. OBJECTIVE It was planned to examine the relationship between osteoprotegerin levels in childhood asthma and respiratory functions and airway inflammation and to assess its use as a biomarker. METHODS The study included patients aged 6-16 years who were diagnosed with asthma at the pediatric allergy outpatient clinic of Bagcilar Training and Research Hospital in Turkey. The correlation analyses for the osteoprotegerin levels of asthma patients and their respiratory functions were examined. RESULTS The age average of asthma cases was 10.61±3.04 years and 51.2 % were female. No statistically significant difference was observed between the osteoprotegerin levels of the groups (p>0.05). A negative and statistically significant correlation was observed between the FEV1 and FVC values and osteoprotegerin levels (p=0.015, p=0.003). CONCLUSIONS This was the first study to examine the relationship between osteoprotegerin levels and airway inflammation in children with asthma. We believe that there is a need for wider scale studies in which clinical symptoms and more parameters are evaluated for defining the role played by osteoprotegerin level in children with asthma and for determining its usability as a biomarker.
Collapse
|
11
|
Shivshankar P, Fekry B, Eckel-Mahan K, Wetsel RA. Circadian Clock and Complement Immune System-Complementary Control of Physiology and Pathology? Front Cell Infect Microbiol 2020; 10:418. [PMID: 32923410 PMCID: PMC7456827 DOI: 10.3389/fcimb.2020.00418] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Mammalian species contain an internal circadian (i.e., 24-h) clock that is synchronized to the day and night cycles. Large epidemiological studies, which are supported by carefully controlled studies in numerous species, support the idea that chronic disruption of our circadian cycles results in a number of health issues, including obesity and diabetes, defective immune response, and cancer. Here we focus specifically on the role of the complement immune system and its relationship to the internal circadian clock system. While still an incompletely understood area, there is evidence that dysregulated proinflammatory cytokines, complement factors, and oxidative stress can be induced by circadian disruption and that these may feed back into the oscillator at the level of circadian gene regulation. Such a feedback cycle may contribute to impaired host immune response against pathogenic insults. The complement immune system including its activated anaphylatoxins, C3a and C5a, not only facilitate innate and adaptive immune response in chemotaxis and phagocytosis, but they can also amplify chronic inflammation in the host organism. Consequent development of autoimmune disorders, and metabolic diseases associated with additional environmental insults that activate complement can in severe cases, lead to accelerated tissue dysfunction, fibrosis, and ultimately organ failure. Because several promising complement-targeted therapeutics to block uncontrolled complement activation and treat autoimmune diseases are in various phases of clinical trials, understanding fully the circadian properties of the complement system, and the reciprocal regulation by these two systems could greatly improve patient treatment in the long term.
Collapse
Affiliation(s)
- Pooja Shivshankar
- Research Center for Immunology and Autoimmune Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Baharan Fekry
- Center for Metabolic and Degenerative Diseases, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Kristin Eckel-Mahan
- Center for Metabolic and Degenerative Diseases, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Rick A Wetsel
- Research Center for Immunology and Autoimmune Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
12
|
Abstract
The recognition of microbial or danger-associated molecular patterns by complement proteins initiates a cascade of events that culminates in the activation of surface complement receptors on immune cells. Such signalling pathways converge with those activated downstream of pattern recognition receptors to determine the type and magnitude of the immune response. Intensive investigation in the field has uncovered novel pathways that link complement-mediated signalling with homeostatic and pathological T cell responses. More recently, the observation that complement proteins also act in the intracellular space to shape T cell fates has added a new layer of complexity. Here, we consider fundamental mechanisms and novel concepts at the interface of complement biology and immunity and discuss how these affect the maintenance of homeostasis and the development of human pathology.
Collapse
|
13
|
Winnicki W, Pichler P, Mechtler K, Imre R, Steinmacher I, Sengölge G, Knafl D, Beilhack G, Wagner L. A novel approach to immunoapheresis of C3a/C3 and proteomic identification of associates. PeerJ 2019; 7:e8218. [PMID: 31871840 PMCID: PMC6921979 DOI: 10.7717/peerj.8218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/15/2019] [Indexed: 02/06/2023] Open
Abstract
Background Complement factor C3 represents the central component of the complement cascade and its activation split product C3a plays an important role in inflammation and disease. Many human disorders are linked to dysregulation of the complement system and alteration in interaction molecules. Therefore, various therapeutic approaches to act on the complement system have been initiated. Methods and Results Aiming to develop a tool to eliminate C3a/C3 from the circulation, in a first step a high affine murine monoclonal antibody (mAb) (3F7E2-mAb) was generated against complement factor C3 and selected for binding to the C3a region to serve as immunoaffinity reagent. Functional testing of the 3F7E2-mAb revealed an inhibition of Zymosan-induced cleavage of C3a from C3. Subsequently, a C3a/C3 specific 3F7E2-immunoaffinity column was developed and apheresis of C3a/C3 and associates was performed. Finally, a proteomic analysis was carried out for identification of apheresis products. C3a/C3 was liberated from the 3F7E2-column together with 278 proteins. C3a/C3 interaction specificity was validated by using a haptoglobin immunoaffinity column as control and biostatistic analysis revealed 39 true C3a/C3 interactants. Conclusion A novel and functionally active mAb was developed against complement factor C3a/C3 and used in a specific immunoaffinity column that allows apheresis of C3a/C3 and associates and their identification by proteomic analysis. This methodological approach of developing specific antibodies that can be used as immunoaffinity reagents to design immunoaffinity columns for elimination and further identification of associated proteins could open new avenues for the development of tailored immunotherapy in various complement-mediated or autoimmune diseases.
Collapse
Affiliation(s)
- Wolfgang Winnicki
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | - Peter Pichler
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | - Karl Mechtler
- ProtChem Facility, Research Institute of Molecular Pathology, Vienna, Austria
| | - Richard Imre
- ProtChem Facility, Research Institute of Molecular Pathology, Vienna, Austria
| | - Ines Steinmacher
- ProtChem Facility, Research Institute of Molecular Pathology, Vienna, Austria
| | - Gürkan Sengölge
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | - Daniela Knafl
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | - Georg Beilhack
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | - Ludwig Wagner
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
14
|
Kordowski A, Reinicke AT, Wu D, Orinska Z, Hagemann P, Huber-Lang M, Lee JB, Wang YH, Hogan SP, Köhl J. C5a receptor 1 -/- mice are protected from the development of IgE-mediated experimental food allergy. Allergy 2019; 74:767-779. [PMID: 30341777 DOI: 10.1111/all.13637] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 09/25/2018] [Accepted: 09/27/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Food-induced anaphylaxis is a serious allergic reaction caused by Fcε-receptor activation on mast cells (MCs). The exact mechanisms breaking oral tolerance and the effector pathways driving food allergy remain elusive. As complement is activated in food-induced anaphylaxis, we aimed to assess the role of C5a in disease pathogenesis. METHODS Oral antigen-induced food-induced anaphylaxis was induced in BALB/c wild-type (wt) and C5ar1-/- mice. Readouts included diarrhea development, changes in rectal temperature, hematocrit, antigen-specific serum IgE, MCPT-1, and intestinal MC numbers, as well as FcεR1-mediated MC functions including C5a receptor 1 (C5aR1) regulation. Further, histamine-mediated hypothermia and regulation of endothelial tight junctions were determined. RESULTS Repeated oral OVA challenge resulted in diarrhea, hypothermia, increased hematocrit, high OVA-specific serum IgE, and MCPT-1 levels in wt mice. Male C5ar1-/- mice were completely whereas female C5ar1-/- mice were partially protected from anaphylaxis development. Serum MCPT-1 levels were reduced gender-independent, whereas IgE levels were reduced in male but not in female C5ar1-/- mice. Mechanistically, IgE-mediated degranulation and IL-6 production from C5ar1-/- BMMCs of both sexes were significantly reduced. Importantly, FcεR1 cross-linking strongly upregulated C5aR1 MC expression in vitro and in vivo. Finally, C5ar1-/- male mice were largely protected from histamine-induced hypovolemic shock, which was associated with protection from histamine-induced barrier dysfunction in vitro following C5aR targeting. CONCLUSIONS Our findings identify C5aR1 activation as an important driver of IgE-mediated food allergy through regulation of allergen-specific IgE production, FcεR1-mediated MC degranulation, and histamine-driven effector functions preferentially in male mice.
Collapse
Affiliation(s)
- Anna Kordowski
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Anna T Reinicke
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - David Wu
- Division of Allergy and Immunology, Cincinnati Children's Hospital and University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA
| | - Zane Orinska
- Division of Experimental Pneumology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Philipp Hagemann
- Division of Experimental Pneumology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Jee-Boong Lee
- Division of Allergy and Immunology, Cincinnati Children's Hospital and University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA
| | - Yui-Hsi Wang
- Division of Allergy and Immunology, Cincinnati Children's Hospital and University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA
| | - Simon P Hogan
- Division of Allergy and Immunology, Cincinnati Children's Hospital and University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
- Division of Immunobiology, Cincinnati Children's Hospital and University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
15
|
Yang X, Wang X, Chi M, Zhang M, Shan H, Zhang QH, Zhang J, Shi J, Zhang JZ, Wu RM, Li YL. Osteoprotegerin mediate RANK/RANKL signaling inhibition eases asthma inflammatory reaction by affecting the survival and function of dendritic cells. Allergol Immunopathol (Madr) 2019; 47:179-184. [PMID: 30292447 DOI: 10.1016/j.aller.2018.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/22/2018] [Accepted: 06/13/2018] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Asthma is a chronic inflammatory, heterogeneous airway disease affecting millions of people around the world. Dendritic cells (DCs) are considered the most important antigen-presenting cell in asthma airway inflammatory reaction. But whether osteoprotegerin (OPG) mediate RANK/RANKL signaling inhibition influences asthma development by affecting the survival and function of DCs remains unclear. In this study, we assessed the effects of OPG on DCs and asthma. MATERIAL AND METHODS BALB/c mice immunized with ovalbumin (OVA) were challenged thrice with an aerosol of OVA every second day for eight days. Dexamethasone (1.0mg/kg) or OPG (50μg/kg) was administered intraperitoneally to OVA-immunized BALB/c mice on day 24 once a day for nine days. Mice were analyzed for effects of OPG on asthma, inflammatory cell infiltration and cytokine levels in lung tissue. The expression of RANK and β-actin was detected by Western Blot. DCs were isolated from mouse bone morrow. Cell survival was assessed by cell counting. The content of IL-12 was detected by ELISA. RESULTS Results showed that OVA increased the number of inflammatory factors in BALF, elevated lung inflammation scores in mice. OPG reversed the alterations induced by OVA in the asthmatic mice. OPG inhibited the survival and function of DC via inhibition of RANK/RANKL signaling. CONCLUSIONS This research proved inhibition of RANK/RANKL signaling by OPG could ease the inflammatory reaction in asthma, providing new evidence for the application of OPG on asthma.
Collapse
Affiliation(s)
- X Yang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - X Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - M Chi
- Department of Pediatrics, BaYi Children's Hospital of the PLA Army General Hospital, Beijing, China; The Clinical Medical College of the PLA Army, Second Military Medical University of People's Liberation Army, Shanghai, China
| | - M Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - H Shan
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Q-H Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - J Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - J Shi
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - J-Z Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - R-M Wu
- Department of Respiratory Medicine, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Y-L Li
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
16
|
Olcina MM, Kim RK, Melemenidis S, Graves EE, Giaccia AJ. The tumour microenvironment links complement system dysregulation and hypoxic signalling. Br J Radiol 2018; 92:20180069. [PMID: 29544344 PMCID: PMC6435069 DOI: 10.1259/bjr.20180069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The complement system is an innate immune pathway typically thought of as part of the first line of defence against “non-self” species. In the context of cancer, complement has been described to have an active role in facilitating cancer-associated processes such as increased proliferation, angiogenesis and migration. Several cellular members of the tumour microenvironment express and/or produce complement proteins locally, including tumour cells. Dysregulation of the complement system has been reported in numerous tumours and increased expression of complement activation fragments in cancer patient specimens correlates with poor patient prognosis. Importantly, genetic or pharmacological targeting of complement has been shown to reduce tumour growth in several cancer preclinical models, suggesting that complement could be an attractive therapeutic target. Hypoxia (low oxygen) is frequently found in solid tumours and has a profound biological impact on cellular and non-cellular components of the tumour microenvironment. In this review, we focus on hypoxia since this is a prevailing feature of the tumour microenvironment that, like increased complement, is typically associated with poor prognosis. Furthermore, interesting links between hypoxia and complement have been recently proposed but never collectively reviewed. Here, we explore how hypoxia alters regulation of complement proteins in different cellular components of the tumour microenvironment, as well as the downstream biological consequences of this regulation.
Collapse
Affiliation(s)
- Monica M Olcina
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Ryan K Kim
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | | | - Edward E Graves
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Amato J Giaccia
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| |
Collapse
|
17
|
Zhang D, Wu Y, Sun G. miR-192 suppresses T follicular helper cell differentiation by targeting CXCR5 in childhood asthma. Scandinavian Journal of Clinical and Laboratory Investigation 2018; 78:236-242. [PMID: 29490514 DOI: 10.1080/00365513.2018.1440628] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The aim of this study was to investigate the role of miR-192 in differentiation of T follicular helper cells in childhood asthma. Blood samples were taken from eighteen children with acute asthma attacks and fifteen healthy children (HC). Quantitative real-time PCR and Western blotting were used to detect the expression levels of miR-192, C-X-C chemokine receptor type 5 (CXCR5), B-cell lymphoma 6 (BCL-6) and inducible T-cell costimulator (ICOS). The flow cytometry was performed to detect the proportion of CD4 + CXCR5+ Tfh cells on CD4 + T lymphocytes. The enzyme-linked immunosorbent assay (ELISA) was carried out to determine the plasma concentrations of total IgE and IL-21. The effect of miR-192 on the T follicular helper cells differentiation by targeting CXCR5 was determined by dual-luciferase reporter assay. Children with asthma had lower levels of miR-192 than HC. The proportion of CD4 + CXCR + Tfh cells was significantly higher in the acute asthma group than HC. Similarly, the plasma concentration of total IgE and IL-21 in the acute group markedly increased compared with the HC, and IgE concentration was positively correlated with the proportion of CD4 + CXCR5 + Tfh cells. Furthermore, the expression levels of CXCR5, Bcl-6 and ICOS were significantly higher in the acute group than in the HC. While the proportion of CD4 + CXCR5 + Tfh cells, IL-21, CXCR5, Bcl-6 and ICOS were obviously lower in the CD4 + T cells transfected with miR-192 plasmid than that in miR-192 + CXCR5 group and control group. In conclusion, miR-192 blocks the activation pathway of Tfh cells by targeting CXCR5, which is a reasonable cellular target for therapeutic intervention.
Collapse
Affiliation(s)
- Defeng Zhang
- a Department of Pediatrics , Anhui Provincial Hospital, Anhui Medical University , Hefei , Anhui , China
| | - Yuanbo Wu
- b Department of Neurology , Anhui Provincial Hospital, Anhui Medical University , Hefei , Anhui , China
| | - Gengyun Sun
- c Department of Respiration , First Affiliated Hospital of Anhui Medical University , Hefei , Anhui , China
| |
Collapse
|
18
|
Reis ES, Mastellos DC, Ricklin D, Mantovani A, Lambris JD. Complement in cancer: untangling an intricate relationship. Nat Rev Immunol 2018; 18:5-18. [PMID: 28920587 PMCID: PMC5816344 DOI: 10.1038/nri.2017.97] [Citation(s) in RCA: 262] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In tumour immunology, complement has traditionally been considered as an adjunctive component that enhances the cytolytic effects of antibody-based immunotherapies, such as rituximab. Remarkably, research in the past decade has uncovered novel molecular mechanisms linking imbalanced complement activation in the tumour microenvironment with inflammation and suppression of antitumour immune responses. These findings have prompted new interest in manipulating the complement system for cancer therapy. This Review summarizes our current understanding of complement-mediated effector functions in the tumour microenvironment, focusing on how complement activation can act as a negative or positive regulator of tumorigenesis. It also offers insight into clinical aspects, including the feasibility of using complement biomarkers for cancer diagnosis and the use of complement inhibitors during cancer treatment.
Collapse
Affiliation(s)
- Edimara S Reis
- Department of Pathology and Laboratory Medicine, School of Medicine, University of Pennsylvania 19104, Philadelphia, Pennsylvania, USA
| | | | - Daniel Ricklin
- Department of Pharmaceutical Sciences, University of Basel, Basel 4056, Switzerland
| | - Alberto Mantovani
- Humanitas Clinical and Research Center and Humanitas University, Rozzano-Milan 20089, Italy
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, School of Medicine, University of Pennsylvania 19104, Philadelphia, Pennsylvania, USA
| |
Collapse
|
19
|
Respiratory Syncytial Virus Exacerbates OVA-mediated asthma in mice through C5a-C5aR regulating CD4 +T cells Immune Responses. Sci Rep 2017; 7:15207. [PMID: 29123203 PMCID: PMC5680322 DOI: 10.1038/s41598-017-15471-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 10/27/2017] [Indexed: 12/15/2022] Open
Abstract
Asthma exacerbation could be induced by respiratory syncytial virus (RSV), and the underlying pathogenic mechanism is related to complement activation. Although complement might regulate CD4+T cells immune responses in asthma model, this regulation existed in RSV-induced asthma model remains incompletely characterrized. In this study, we assessed the contribution of C5a-C5aR to CD4+T cell immune responses in RSV-infected asthma mice. Female BALB/C mice were sensitized and challenged with ovalbumin (OVA) while treated with RSV infection and C5a receptor antagonist (C5aRA) during challenge period. RSV enhanced lung damage, airway hyperresponsiveness, and C5aR expressions in asthma mice, while C5aRA alleviated these pathologic changes. The percentages of Th1, Th2 and Th17 cells were increased, while the percentage of Treg cells was decreased in RSV-infected asthma mice compared with asthma mice. IFN-γ, IL-4, IL-10 and IL-17A levels have similar trend with Th1, Th2, Th17 and Treg cells. Notably, above changes of CD4+T cells and their related cytokines were reversed by C5aRA. Together, the data indicates that RSV infection could apparently increase C5a and C5aR expression in the pathogenesis of RSV-infected asthma mice, meanwhile C5aRA prevents some of the CD4+T cells immune changes that are induced by RSV.
Collapse
|
20
|
Laumonnier Y, Wiese AV, Figge J, Karsten C. Regulation and function of anaphylatoxins and their receptors in allergic asthma. Mol Immunol 2017; 84:51-56. [PMID: 27916272 DOI: 10.1016/j.molimm.2016.11.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 11/18/2016] [Indexed: 02/08/2023]
Abstract
Allergic asthma is a disease of the airways driven by maladaptive T helper 2 (Th2) and Th17 immune response against harmless, airborne substances. The hallmarks of this disease are airway hyperresponsiveness (AHR), eosinophilic and neutrophilic airway inflammation and mucus overproduction. Distinct dendric cell (DC) subsets together with airway epithelial and pulmonary vascular endothelial cells play critical roles in allergen sensing and in driving T cell differentiation towards Th2 and Th17 effector or regulatory T cells (Treg). Previous studies suggested already a pivotal role for the anaphylatoxins (C5a, C3a) in the pathogenesis of allergic asthma. During sensitization for example it is described, that C3a promotes, whereas C5a protects from the development of maladaptive immunity during allergen sensitization. Here we will discuss the role of the anaphylatoxins (C3a, C5a) and their receptors during the pathogenesis of allergic asthma, and specifically in lung DC biology. We will also have a look on canonical and non-canonical complement activation and we will discuss novel concepts on how the adaptive immune system can regulate the function of ATRs also in the context of allergic asthma.
Collapse
Affiliation(s)
- Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany.
| | - Anna V Wiese
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany
| | - Julia Figge
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany
| | - Christian Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany.
| |
Collapse
|
21
|
Verschoor A, Karsten CM, Broadley SP, Laumonnier Y, Köhl J. Old dogs-new tricks: immunoregulatory properties of C3 and C5 cleavage fragments. Immunol Rev 2017; 274:112-126. [PMID: 27782330 DOI: 10.1111/imr.12473] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The activation of the complement system by canonical and non-canonical mechanisms results in the generation of multiple C3 and C5 cleavage fragments including anaphylatoxins C3a and C5a as well as opsonizing C3b/iC3b. It is now well appreciated that anaphylatoxins not only act as pro-inflammatory mediators but as immunoregulatory molecules that control the activation status of cells and tissue at several levels. Likewise, C3b/iC3b is more than the opsonizing fragment that facilitates engulfment and destruction of targets by phagocytes. In the circulation, it also facilitates the transport and delivery of bacteria and immune complexes to phagocytes, through a process known as immune adherence, with consequences for adaptive immunity. Here, we will discuss non-classical immunoregulatory properties of C3 and C5 cleavage fragments. We highlight the influence of anaphylatoxins on Th2 and Th17 cell development during allergic asthma with a particular emphasis on their role in the modulation of CD11b+ conventional dendritic cells and monocyte-derived dendritic cells. Furthermore, we discuss the control of anaphylatoxin-mediated activation of dendritic cells and allergic effector cells by adaptive immune mechanisms that involve allergen-specific IgG1 antibodies and plasma or regulatory T cell-derived IL-10 production. Finally, we take a fresh look at immune adherence with a particular focus on the development of antibacterial cytotoxic T-cell responses.
Collapse
Affiliation(s)
- Admar Verschoor
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany.
| | - Christian M Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Steven P Broadley
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany. .,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
22
|
Sinha M, Lowell CA. Immune Defense Protein Expression in Highly Purified Mouse Lung Epithelial Cells. Am J Respir Cell Mol Biol 2017; 54:802-13. [PMID: 26574781 DOI: 10.1165/rcmb.2015-0171oc] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Lung epithelial cells play critical roles in initiating and modulating immune responses during pulmonary infection or injury. To better understand the spectrum of immune response-related proteins present in lung epithelial cells, we developed an improved method of isolating highly pure primary murine alveolar type (AT) II cells and murine tracheal epithelial cells (mTECs) using negative selection for a variety of lineage markers and positive selection for epithelial cell adhesion molecule (EpCAM), a pan-epithelial cell marker. This method yielded 2-3 × 10(6) ATII cells/mouse lung and 1-2 × 10(4) mTECs/trachea that were highly pure (>98%) and viable (>98%). Using these preparations, we found that both ATII cells and mTECs expressed the Lyn tyrosine kinase, which is best studied as an inhibitory kinase in hematopoietic cells. However, we found little or no expression of Syk in either ATII cells or mTECs, which is in contrast to earlier published reports. Both cell types expressed C-type lectin receptors, anaphylatoxin receptors, and various Toll-like receptors (TLRs). In addition, stimulation of ATII cells with TLR ligands led to secretion of various cytokines and chemokines. Interestingly, lyn(-/-) ATII cells were hyperresponsive to TLR3 stimulation, suggesting that, as in hematopoietic cells, Lyn might be playing an inhibitory role in ATII cells. In conclusion, the improved isolation method reported here, along with expression profiles of various immune defense proteins, will help refocus investigations of immune-related signaling events in pulmonary epithelium.
Collapse
Affiliation(s)
- Meenal Sinha
- Department of Laboratory Medicine and the Program in Immunology, University of California, San Francisco, San Francisco, California
| | - Clifford A Lowell
- Department of Laboratory Medicine and the Program in Immunology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
23
|
Busch C, Annamalai B, Abdusalamova K, Reichhart N, Huber C, Lin Y, Jo EAH, Zipfel PF, Skerka C, Wildner G, Diedrichs-Möhring M, Rohrer B, Strauß O. Anaphylatoxins Activate Ca 2+, Akt/PI3-Kinase, and FOXO1/FoxP3 in the Retinal Pigment Epithelium. Front Immunol 2017; 8:703. [PMID: 28663750 PMCID: PMC5472091 DOI: 10.3389/fimmu.2017.00703] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/31/2017] [Indexed: 12/20/2022] Open
Abstract
PURPOSE The retinal pigment epithelium (RPE) is a main target for complement activation in age-related macular degeneration (AMD). The anaphylatoxins C3a and C5a have been thought to mostly play a role as chemoattractants for macrophages and immune cells; here, we explore whether they trigger RPE alterations. Specifically, we investigated the RPE as a potential immunoregulatory gate, allowing for active changes in the RPE microenvironment in response to complement. DESIGN In vitro and in vivo analysis of signaling pathways. METHODS Individual activities of and interaction between the two anaphylatoxin receptors were tested in cultured RPE cells by fluorescence microscopy, western blot, and immunohistochemistry. MAIN OUTCOME MEASURES Intracellular free calcium, protein phosphorylation, immunostaining of tissues/cells, and multiplex secretion assay. RESULTS Similar to immune cells, anaphylatoxin exposure resulted in increases in free cytosolic Ca2+, PI3-kinase/Akt activation, FoxP3 and FOXO1 phosphorylation, and cytokine/chemokine secretion. Differential responses were elicited depending on whether C3a and C5a were co-administered or applied consecutively, and response amplitudes in co-administration experiments ranged from additive to driven by C5a (C3a + C5a = C5a) or being smaller than those elicited by C3a alone (C3a + C5a < C3a). CONCLUSION We suggest that this combination of integrative signaling between C3aR and C5aR helps the RPE to precisely adopt its immune regulatory function. These data further contribute to our understanding of AMD pathophysiology.
Collapse
Affiliation(s)
- Catharina Busch
- Department of Ophthalmology, Charité University Medicine Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | | | - Khava Abdusalamova
- Department of Ophthalmology, Charité University Medicine Berlin, Berlin, Germany
| | - Nadine Reichhart
- Department of Ophthalmology, Charité University Medicine Berlin, Berlin, Germany
| | - Christian Huber
- Department of Ophthalmology, Charité University Medicine Berlin, Berlin, Germany
- Department of Ophthalmology, University of Heidelberg, Heidelberg, Germany
| | - Yuchen Lin
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Emeraldo A. H. Jo
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Peter F. Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Christine Skerka
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Gerhild Wildner
- Department of Ophthalmology, Section of Immunobiology, Clinic of the LMU Munich, Munich, Germany
| | - Maria Diedrichs-Möhring
- Department of Ophthalmology, Section of Immunobiology, Clinic of the LMU Munich, Munich, Germany
| | - Bärbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, United States
- Ralph H. Johnson VA Medical Center, Division of Research, Charleston, SC, United States
| | - Olaf Strauß
- Department of Ophthalmology, Charité University Medicine Berlin, Berlin, Germany
| |
Collapse
|
24
|
Reinmuth-Selzle K, Kampf CJ, Lucas K, Lang-Yona N, Fröhlich-Nowoisky J, Shiraiwa M, Lakey PSJ, Lai S, Liu F, Kunert AT, Ziegler K, Shen F, Sgarbanti R, Weber B, Bellinghausen I, Saloga J, Weller MG, Duschl A, Schuppan D, Pöschl U. Air Pollution and Climate Change Effects on Allergies in the Anthropocene: Abundance, Interaction, and Modification of Allergens and Adjuvants. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2017; 51:4119-4141. [PMID: 28326768 PMCID: PMC5453620 DOI: 10.1021/acs.est.6b04908] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 03/07/2017] [Accepted: 03/22/2017] [Indexed: 05/13/2023]
Abstract
Air pollution and climate change are potential drivers for the increasing burden of allergic diseases. The molecular mechanisms by which air pollutants and climate parameters may influence allergic diseases, however, are complex and elusive. This article provides an overview of physical, chemical and biological interactions between air pollution, climate change, allergens, adjuvants and the immune system, addressing how these interactions may promote the development of allergies. We reviewed and synthesized key findings from atmospheric, climate, and biomedical research. The current state of knowledge, open questions, and future research perspectives are outlined and discussed. The Anthropocene, as the present era of globally pervasive anthropogenic influence on planet Earth and, thus, on the human environment, is characterized by a strong increase of carbon dioxide, ozone, nitrogen oxides, and combustion- or traffic-related particulate matter in the atmosphere. These environmental factors can enhance the abundance and induce chemical modifications of allergens, increase oxidative stress in the human body, and skew the immune system toward allergic reactions. In particular, air pollutants can act as adjuvants and alter the immunogenicity of allergenic proteins, while climate change affects the atmospheric abundance and human exposure to bioaerosols and aeroallergens. To fully understand and effectively mitigate the adverse effects of air pollution and climate change on allergic diseases, several challenges remain to be resolved. Among these are the identification and quantification of immunochemical reaction pathways involving allergens and adjuvants under relevant environmental and physiological conditions.
Collapse
Affiliation(s)
| | - Christopher J. Kampf
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
- Institute
of Inorganic and Analytical Chemistry, Johannes
Gutenberg University, Mainz, 55128, Germany
| | - Kurt Lucas
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Naama Lang-Yona
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | | | - Manabu Shiraiwa
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
- Department
of Chemistry, University of California, Irvine, California 92697-2025, United States
| | - Pascale S. J. Lakey
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Senchao Lai
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
- South
China University of Technology, School of
Environment and Energy, Guangzhou, 510006, China
| | - Fobang Liu
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Anna T. Kunert
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Kira Ziegler
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Fangxia Shen
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Rossella Sgarbanti
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Bettina Weber
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Iris Bellinghausen
- Department
of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, 55131, Germany
| | - Joachim Saloga
- Department
of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, 55131, Germany
| | - Michael G. Weller
- Division
1.5 Protein Analysis, Federal Institute
for Materials Research and Testing (BAM), Berlin, 12489, Germany
| | - Albert Duschl
- Department
of Molecular Biology, University of Salzburg, 5020 Salzburg, Austria
| | - Detlef Schuppan
- Institute
of Translational Immunology and Research Center for Immunotherapy,
Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, 55131 Germany
- Division
of Gastroenterology, Beth Israel Deaconess
Medical Center and Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Ulrich Pöschl
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| |
Collapse
|
25
|
C5aR contributes to the weak Th1 profile induced by an outbreak strain of Mycobacterium tuberculosis. Tuberculosis (Edinb) 2017; 103:16-23. [DOI: 10.1016/j.tube.2016.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 12/12/2016] [Accepted: 12/19/2016] [Indexed: 01/21/2023]
|
26
|
Leaker BR, Malkov VA, Mogg R, Ruddy MK, Nicholson GC, Tan AJ, Tribouley C, Chen G, De Lepeleire I, Calder NA, Chung H, Lavender P, Carayannopoulos LN, Hansel TT. The nasal mucosal late allergic reaction to grass pollen involves type 2 inflammation (IL-5 and IL-13), the inflammasome (IL-1β), and complement. Mucosal Immunol 2017; 10:408-420. [PMID: 27677865 DOI: 10.1038/mi.2016.74] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 07/21/2016] [Indexed: 02/04/2023]
Abstract
Non-invasive mucosal sampling (nasosorption and nasal curettage) was used following nasal allergen challenge with grass pollen in subjects with allergic rhinitis, in order to define the molecular basis of the late allergic reaction (LAR). It was found that the nasal LAR to grass pollen involves parallel changes in pathways of type 2 inflammation (IL-4, IL-5 and IL-13), inflammasome-related (IL-1β), and complement and circadian-associated genes. A grass pollen nasal spray was given to subjects with hay fever followed by serial sampling, in which cytokines and chemokines were measured in absorbed nasal mucosal lining fluid, and global gene expression (transcriptomics) assessed in nasal mucosal curettage samples. Twelve of 19 subjects responded with elevations in interleukin (IL)-5, IL-13, IL-1β and MIP-1β/CCL4 protein levels in the late phase. In addition, in these individuals whole-genome expression profiling showed upregulation of type 2 inflammation involving eosinophils and IL-4, IL-5 and IL-13; neutrophil recruitment with IL-1α and IL-1β; the alternative pathway of complement (factor P and C5aR); and prominent effects on circadian-associated transcription regulators. Baseline IL-33 mRNA strongly correlated with these late-phase responses, whereas a single oral dose of prednisone dose-dependently reversed most nasal allergen challenge-induced cytokine and transcript responses. This study shows that the LAR to grass pollen involves a range of inflammatory pathways and suggests potential new biomarkers and therapeutic targets. Furthermore, the marked variation in mucosal inflammatory events between different patients suggests that in the future precision mucosal sampling may enable rational specific therapy.
Collapse
Affiliation(s)
- B R Leaker
- Respiratory Clinical Trials Ltd, London, UK
| | - V A Malkov
- Merck Research Laboratories, Rahway, New Jersey, USA
| | - R Mogg
- Merck Research Laboratories, Rahway, New Jersey, USA.,Present address: Celgene (L.N.C. and G.C.); Janssen R & D, Spring House, PA (R.M.); Alnylam (M.K.R.); Novartis (C.T.); GSK (N.A.C.); Otsuka (H.C.)
| | - M K Ruddy
- Merck Research Laboratories, Rahway, New Jersey, USA.,Present address: Celgene (L.N.C. and G.C.); Janssen R & D, Spring House, PA (R.M.); Alnylam (M.K.R.); Novartis (C.T.); GSK (N.A.C.); Otsuka (H.C.)
| | | | - A J Tan
- Imperial Clinical Respiratory Research Unit (ICRRU), St Mary's Hospital, Imperial College, London, UK
| | - C Tribouley
- Merck Research Laboratories, Rahway, New Jersey, USA.,Present address: Celgene (L.N.C. and G.C.); Janssen R & D, Spring House, PA (R.M.); Alnylam (M.K.R.); Novartis (C.T.); GSK (N.A.C.); Otsuka (H.C.)
| | - G Chen
- Merck Research Laboratories, Rahway, New Jersey, USA.,Present address: Celgene (L.N.C. and G.C.); Janssen R & D, Spring House, PA (R.M.); Alnylam (M.K.R.); Novartis (C.T.); GSK (N.A.C.); Otsuka (H.C.)
| | | | - N A Calder
- MSD (Europe) Inc., Brussels, Belgium.,Present address: Celgene (L.N.C. and G.C.); Janssen R & D, Spring House, PA (R.M.); Alnylam (M.K.R.); Novartis (C.T.); GSK (N.A.C.); Otsuka (H.C.)
| | - H Chung
- Present address: Celgene (L.N.C. and G.C.); Janssen R & D, Spring House, PA (R.M.); Alnylam (M.K.R.); Novartis (C.T.); GSK (N.A.C.); Otsuka (H.C.)
| | - P Lavender
- Department of Asthma, Allergy and Respiratory Science, King's College, London, UK
| | - L N Carayannopoulos
- Merck Research Laboratories, Rahway, New Jersey, USA.,Present address: Celgene (L.N.C. and G.C.); Janssen R & D, Spring House, PA (R.M.); Alnylam (M.K.R.); Novartis (C.T.); GSK (N.A.C.); Otsuka (H.C.)
| | - T T Hansel
- Imperial Clinical Respiratory Research Unit (ICRRU), St Mary's Hospital, Imperial College, London, UK
| |
Collapse
|
27
|
Ender F, Wiese AV, Schmudde I, Sun J, Vollbrandt T, König P, Laumonnier Y, Köhl J. Differential regulation of C5a receptor 1 in innate immune cells during the allergic asthma effector phase. PLoS One 2017; 12:e0172446. [PMID: 28231307 PMCID: PMC5322932 DOI: 10.1371/journal.pone.0172446] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 02/03/2017] [Indexed: 12/31/2022] Open
Abstract
C5a drives airway constriction and inflammation during the effector phase of allergic asthma, mainly through the activation of C5a receptor 1 (C5aR1). Yet, C5aR1 expression on myeloid and lymphoid cells during the allergic effector phase is ill-defined. Recently, we generated and characterized a floxed green fluorescent protein (GFP)-C5aR1 knock-in mouse. Here, we used this reporter strain to monitor C5aR1 expression in airway, pulmonary and lymph node cells during the effector phase of OVA-driven allergic asthma. C5aR1 reporter and wildtype mice developed a similar allergic phenotype with comparable airway resistance, mucus production, eosinophilic/neutrophilic airway inflammation and Th2/Th17 cytokine production. During the allergic effector phase, C5aR1 expression increased in lung tissue eosinophils but decreased in airway and pulmonary macrophages as well as in pulmonary CD11b+ conventional dendritic cells (cDCs) and monocyte-derived DCs (moDCs). Surprisingly, expression in neutrophils was not affected. Of note, moDCs but not CD11b+ cDCs from mediastinal lymph nodes (mLN) expressed less C5aR1 than DCs residing in the lung after OVA challenge. Finally, neither CD103+ cDCs nor cells of the lymphoid lineage such as Th2 or Th17-differentiated CD4+ T cells, B cells or type 2 innate lymphoid cells (ILC2) expressed C5aR1 under allergic conditions. Our findings demonstrate a complex regulation pattern of C5aR1 in the airways, lung tissue and mLN of mice, suggesting that the C5a/C5aR1 axis controls airway constriction and inflammation through activation of myeloid cells in all three compartments in an experimental model of allergic asthma.
Collapse
Affiliation(s)
- Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Anna V. Wiese
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Inken Schmudde
- Institute for Anatomy, University of Lübeck, Lübeck, Germany
| | - Jing Sun
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | | | - Peter König
- Institute for Anatomy, University of Lübeck, Lübeck, Germany
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- * E-mail: (JK); (YL)
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- * E-mail: (JK); (YL)
| |
Collapse
|
28
|
Ricklin D, Reis ES, Mastellos DC, Gros P, Lambris JD. Complement component C3 - The "Swiss Army Knife" of innate immunity and host defense. Immunol Rev 2016; 274:33-58. [PMID: 27782325 PMCID: PMC5427221 DOI: 10.1111/imr.12500] [Citation(s) in RCA: 273] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
As a preformed defense system, complement faces a delicate challenge in providing an immediate, forceful response to pathogens even at first encounter, while sparing host cells in the process. For this purpose, it engages a tightly regulated network of plasma proteins, cell surface receptors, and regulators. Complement component C3 plays a particularly versatile role in this process by keeping the cascade alert, acting as a point of convergence of activation pathways, fueling the amplification of the complement response, exerting direct effector functions, and helping to coordinate downstream immune responses. In recent years, it has become evident that nature engages the power of C3 not only to clear pathogens but also for a variety of homeostatic processes ranging from tissue regeneration and synapse pruning to clearing debris and controlling tumor cell progression. At the same time, its central position in immune surveillance makes C3 a target for microbial immune evasion and, if improperly engaged, a trigger point for various clinical conditions. In our review, we look at the versatile roles and evolutionary journey of C3, discuss new insights into the molecular basis for C3 function, provide examples of disease involvement, and summarize the emerging potential of C3 as a therapeutic target.
Collapse
Affiliation(s)
- Daniel Ricklin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Edimara S Reis
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dimitrios C Mastellos
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- National Center for Scientific Research 'Demokritos', Athens, Greece
| | - Piet Gros
- Utrecht University, Utrecht, The Netherlands
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
29
|
Shi J, Zhu X, Xie M, Wang J, He Y, Xu Y, Liu X. MBL2 polymorphisms and the risk of asthma: A meta-analysis. Ann Allergy Asthma Immunol 2016; 117:417-422.e1. [PMID: 27590640 DOI: 10.1016/j.anai.2016.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/20/2016] [Accepted: 08/01/2016] [Indexed: 11/18/2022]
Abstract
BACKGROUND The association between MBL2 gene polymorphisms and the risk of asthma has been evaluated in multiple studies; however, the results are inconsistent. OBJECTIVE To perform a meta-analysis to explore whether MBL2 gene polymorphisms were associated with the risk of asthma. METHODS We searched PubMed, Web of Science, and Cochrane Library to find relevant articles published up to March 2016. Nine studies, including 2066 cases and 2183 controls, were included in the meta-analysis. The strength of association was evaluated by odds ratio (OR) with 95% confidence interval (CI). RESULTS The results reveal that MBL2 gene polymorphisms (codon 54 A/B, -550 H/L or -221 X/Y) were not associated with the risk of asthma (codon 54 A/B: BB+AB vs AA: OR, 1.02; 95% CI, 0.85-1.23; -550 H/L: LL+HL vs HH: OR, 0.81; 95% CI, 0.63-1.03; -221 X/Y: XX+YX vs YY: OR, 0.85; 95% CI, 0.69-1.04). Subgroup analysis by ethnicity implied that the MBL2 codon 54 A/B polymorphism was not significantly associated with the risk of asthma in Asians (BB+AB vs AA: OR, 0.95; 95% CI, 0.70-1.29) or whites (BB+AB vs AA: OR, 1.07; 95% CI, 0.84-1.35). CONCLUSION The results indicated that MBL2 gene polymorphisms (codon 54 A/B, -550 H/L or -221 X/Y) may be not associated with the risk of asthma.
Collapse
Affiliation(s)
- Jing Shi
- Department of Respiration and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianying Zhu
- Department of Respiration and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Xie
- Department of Respiration and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianmiao Wang
- Department of Respiration and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanzhou He
- Department of Respiration and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongjian Xu
- Department of Respiration and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiansheng Liu
- Department of Respiration and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
30
|
Hiemstra PS. Parallel activities and interactions between antimicrobial peptides and complement in host defense at the airway epithelial surface. Mol Immunol 2015; 68:28-30. [DOI: 10.1016/j.molimm.2015.07.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 07/23/2015] [Accepted: 07/24/2015] [Indexed: 01/28/2023]
|
31
|
Morara S, Colangelo AM, Provini L. Microglia-Induced Maladaptive Plasticity Can Be Modulated by Neuropeptides In Vivo. Neural Plast 2015; 2015:135342. [PMID: 26273481 PMCID: PMC4529944 DOI: 10.1155/2015/135342] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 06/25/2015] [Indexed: 02/06/2023] Open
Abstract
Microglia-induced maladaptive plasticity is being recognized as a major cause of deleterious self-sustaining pathological processes that occur in neurodegenerative and neuroinflammatory diseases. Microglia, the primary homeostatic guardian of the central nervous system, exert critical functions both during development, in neural circuit reshaping, and during adult life, in the brain physiological and pathological surveillance. This delicate critical role can be disrupted by neural, but also peripheral, noxious stimuli that can prime microglia to become overreactive to a second noxious stimulus or worsen underlying pathological processes. Among regulators of microglia, neuropeptides can play a major role. Their receptors are widely expressed in microglial cells and neuropeptide challenge can potently influence microglial activity in vitro. More relevantly, this regulator activity has been assessed also in vivo, in experimental models of brain diseases. Neuropeptide action in the central nervous system has been associated with beneficial effects in neurodegenerative and neuroinflammatory pathological experimental models. This review describes some of the mechanisms of the microglia maladaptive plasticity in vivo and how neuropeptide activity can represent a useful therapeutical target in a variety of human brain pathologies.
Collapse
Affiliation(s)
- Stefano Morara
- Neuroscience Institute (CNR), Via Vanvitelli 32, 20129 Milano, Italy
- Department of BIOMETRA, University of Milano, Via Vanvitelli 32, 20129 Milano, Italy
| | - Anna Maria Colangelo
- Laboratory of Neuroscience “R. Levi-Montalcini”, Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milano, Italy
- SYSBIO Centre of Systems Biology, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milano, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, 20126 Milano, Italy
| | - Luciano Provini
- Department of BIOMETRA, University of Milano, Via Vanvitelli 32, 20129 Milano, Italy
| |
Collapse
|
32
|
Shin NR, Shin IS, Jeon CM, Hong JM, Kwon OK, Kim HS, Oh SR, Hahn KW, Ahn KS. Zingiber mioga (Thunb.) Roscoe attenuates allergic asthma induced by ovalbumin challenge. Mol Med Rep 2015; 12:4538-4545. [PMID: 26063513 DOI: 10.3892/mmr.2015.3914] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 03/05/2015] [Indexed: 11/06/2022] Open
Abstract
Zingiber mioga (Thunb.) Roscoe (ZM) is a traditional medicine, used to treat inflammatory diseases. The present study aimed to evaluate the inhibitory effects of ZM on the inflammatory response in lipopolysaccharide (LPS)‑stimulated RAW264.7 murine macrophage cells and in a mouse model of ovalbumin (OVA)‑induced allergic asthma. Mice received OVA sensitization on day 0 and 14, and were challenged with OVA between days 21 and 23. ZM was administered to the mice at a dose of 30 mg/kg, 1 h prior to OVA challenge. In LPS‑stimulated RAW264.7 cells, ZM significantly decreased nitric oxide (NO) and tumor necrosis factor (TNF)‑α production in a concentration‑dependent manner, and mRNA expression of inducible NO synthase (iNOS), TNF‑α and matrix metalloproteinase (MMP)‑9 was reduced. In addition, treatment with ZM decreased the inflammatory cell count in bronchoalveolar lavage fluid from the mice, and reduced the expression of interleukin (IL)‑4, IL‑5, IL‑13, eotaxin and immunoglobulin E. ZM also reduced airway hyperresponsiveness in OVA‑challenged mice, and attenuated the infiltration of inflammatory cells and mucus production in the airways, with a decrease in the expression of iNOS and MMP‑9 in lung tissue. In conclusion, the results of the present study indicate that ZM effectively inhibits inflammatory responses. Therefore, it may be that ZM has potential as a therapeutic agent for use in inflammatory diseases.
Collapse
Affiliation(s)
- Na-Rae Shin
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon‑Gun, Chungbuk 363‑883, Republic of Korea
| | - In-Sik Shin
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon‑Gun, Chungbuk 363‑883, Republic of Korea
| | - Chan-Mi Jeon
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon‑Gun, Chungbuk 363‑883, Republic of Korea
| | - Ju-Mi Hong
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon‑Gun, Chungbuk 363‑883, Republic of Korea
| | - Ok-Kyoung Kwon
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon‑Gun, Chungbuk 363‑883, Republic of Korea
| | - Hui-Seong Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon‑Gun, Chungbuk 363‑883, Republic of Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon‑Gun, Chungbuk 363‑883, Republic of Korea
| | - Kyu-Woung Hahn
- Department of Biological Science and Biotechnology, Hannam University, Yuesong‑Gu, Daejeon 305‑811, Republic of Korea
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon‑Gun, Chungbuk 363‑883, Republic of Korea
| |
Collapse
|
33
|
Surace L, Lysenko V, Fontana AO, Cecconi V, Janssen H, Bicvic A, Okoniewski M, Pruschy M, Dummer R, Neefjes J, Knuth A, Gupta A, van den Broek M. Complement is a central mediator of radiotherapy-induced tumor-specific immunity and clinical response. Immunity 2015; 42:767-77. [PMID: 25888260 DOI: 10.1016/j.immuni.2015.03.009] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 01/16/2015] [Accepted: 03/21/2015] [Indexed: 01/21/2023]
Abstract
Radiotherapy induces DNA damage and cell death, but recent data suggest that concomitant immune stimulation is an integral part of the therapeutic action of ionizing radiation. It is poorly understood how radiotherapy supports tumor-specific immunity. Here we report that radiotherapy induced tumor cell death and transiently activated complement both in murine and human tumors. The local production of pro-inflammatory anaphylatoxins C3a and C5a was crucial to the tumor response to radiotherapy and concomitant stimulation of tumor-specific immunity. Dexamethasone, a drug frequently given during radiotherapy, limited complement activation and the anti-tumor effects of the immune system. Overall, our findings indicate that anaphylatoxins are key players in radiotherapy-induced tumor-specific immunity and the ensuing clinical responses.
Collapse
Affiliation(s)
- Laura Surace
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Veronika Lysenko
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Andrea Orlando Fontana
- Department of Radio-Oncology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Virginia Cecconi
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Hans Janssen
- Division Cell Biology II, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Antonela Bicvic
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Michal Okoniewski
- ID Scientific IT Services, Swiss Federal Institute for Technology (ETH), Weinbergstrasse 11, 8092 Zurich, Switzerland
| | - Martin Pruschy
- Department of Radio-Oncology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Jacques Neefjes
- Division Cell Biology II, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Alexander Knuth
- Clinic of Oncology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Anurag Gupta
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Maries van den Broek
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
34
|
Engelke C, Wiese AV, Schmudde I, Ender F, Ströver HA, Vollbrandt T, König P, Laumonnier Y, Köhl J. Distinct roles of the anaphylatoxins C3a and C5a in dendritic cell-mediated allergic asthma. THE JOURNAL OF IMMUNOLOGY 2014; 193:5387-401. [PMID: 25355927 DOI: 10.4049/jimmunol.1400080] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Conventional dendritic cells (cDC) are necessary and sufficient to drive mixed maladaptive Th2/Th17 immune responses toward aeroallergens in experimental allergy models. Previous studies suggest that the anaphylatoxin C3a promotes, whereas C5a protects from the development of maladaptive immunity during allergen sensitization. However, only limited evidence exists that such effects are directly mediated through anaphylatoxin-receptor signaling in cDCs. In this study, we assessed the impact of C3a and C5a on cDC-mediated induction pulmonary allergy by adoptively transferring house dust mite (HDM)-pulsed bone marrow-derived DCs (BMDC) from wild-type (WT) C3aR(-/-), C5aR1(-/-), or C3aR(-/-)/C5aR1(-/-) into WT mice. Transfer of HDM-pulsed WT BMDCs promoted a strong asthmatic phenotype characterized by marked airway resistance, strong Th2 cytokine, and mucus production, as well as mixed eosinophilic and neurophilic airway inflammation. Surprisingly, C3aR(-/-) cDCs induced a strong allergic phenotype, but no IL-17A production, whereas HDM-pulsed C5aR1(-/-) cDCs failed to drive pulmonary allergy. Transfer of C3aR(-/-)/C5aR1(-/-) cDCs resulted in a slightly reduced allergic phenotype associated with increased IFN-γ production. Mechanistically, C3aR and C5aR1 signaling is required for IL-23 production from HDM-pulsed BMDCs in vitro. Furthermore, C3aR(-/-) BMDCs produced less IL-1β. The mechanisms underlying the failure of C5aR1(-/-) BMDCs to induce experimental allergy include a reduced capability to migrate into the lung tissue and a decreased potency to direct pulmonary homing of effector T cells. Thus, we uncovered a crucial role for C5a, but only a minor role for C3a in BMDC-mediated pulmonary allergy, suggesting that BMDCs inappropriately reflect the impact of complement on lung cDC-mediated allergic asthma development.
Collapse
Affiliation(s)
- Carsten Engelke
- Institute for Systemic Inflammation Research, University of Lübeck and Airway Research Center North, member of the German Center for Lung Research, 23538 Lübeck, Germany
| | - Anna V Wiese
- Institute for Systemic Inflammation Research, University of Lübeck and Airway Research Center North, member of the German Center for Lung Research, 23538 Lübeck, Germany
| | - Inken Schmudde
- Institute for Systemic Inflammation Research, University of Lübeck and Airway Research Center North, member of the German Center for Lung Research, 23538 Lübeck, Germany
| | - Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck and Airway Research Center North, member of the German Center for Lung Research, 23538 Lübeck, Germany
| | - Heike A Ströver
- Institute for Systemic Inflammation Research, University of Lübeck and Airway Research Center North, member of the German Center for Lung Research, 23538 Lübeck, Germany
| | | | - Peter König
- Institute for Anatomy, University of Lübeck and Airway Research Center North, member of the German Center for Lung Research, 23538 Lübeck, Germany; and
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck and Airway Research Center North, member of the German Center for Lung Research, 23538 Lübeck, Germany;
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck and Airway Research Center North, member of the German Center for Lung Research, 23538 Lübeck, Germany; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229
| |
Collapse
|
35
|
Zabel BA, Rott A, Butcher EC. Leukocyte chemoattractant receptors in human disease pathogenesis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2014; 10:51-81. [PMID: 25387059 DOI: 10.1146/annurev-pathol-012513-104640] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Combinations of leukocyte attractant ligands and cognate heptahelical receptors specify the systemic recruitment of circulating cells by triggering integrin-dependent adhesion to endothelial cells, supporting extravasation, and directing specific intratissue localization via gradient-driven chemotaxis. Chemoattractant receptors also control leukocyte egress from lymphoid organs and peripheral tissues. In this article, we summarize the fundamental mechanics of leukocyte trafficking, from the evolution of multistep models of leukocyte recruitment and navigation to the regulation of chemoattractant availability and function by atypical heptahelical receptors. To provide a more complete picture of the migratory circuits involved in leukocyte trafficking, we integrate a number of nonchemokine chemoattractant receptors into our discussion. Leukocyte chemoattractant receptors play key roles in the pathogenesis of autoimmune diseases, allergy, inflammatory disorders, and cancer. We review recent advances in our understanding of chemoattractant receptors in disease pathogenesis, with a focus on genome-wide association studies in humans and the translational implications of mechanistic studies in animal disease models.
Collapse
Affiliation(s)
- Brian A Zabel
- Palo Alto Veterans Institute for Research and Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304;
| | | | | |
Collapse
|
36
|
Athari SS, Omidi R. Report of a patient with complex composites of hepatitis B virus, allergic asthma and diabetes. Asian Pac J Trop Biomed 2014; 4:S59-61. [PMID: 25183147 DOI: 10.12980/apjtb.4.2014c683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 02/27/2014] [Indexed: 01/01/2023] Open
Abstract
HBV is a non-cytopathic virus and cell mediated immune response against this. Humoral mediated immune response are responsible for allergic diseases. Balance between these two subsets of Th CD4+ cells are result of the immune system response. A 56 year old woman presented with chronic HBV infection, allergic asthma, type 2 diabetes mellitus and high blood pressure and high blood lipid. Patients should be followed for the allergic and autoimmune diseases along with their viral reactivation.
Collapse
Affiliation(s)
- Seyyed Shamsadin Athari
- Department of Immunology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Razie Omidi
- Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
37
|
Expression and regulation of complement receptors by human natural killer cells. Immunobiology 2014; 219:671-9. [PMID: 24775270 DOI: 10.1016/j.imbio.2014.03.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 03/27/2014] [Accepted: 03/29/2014] [Indexed: 11/22/2022]
Abstract
Integration of cellular and humoral arms of the innate immune response is fundamental to the development of powerful effector functions in host defence as well as aberrant immune responses. Here, we provide evidence in support of the relationship between complement activation and NK cell functional modulation. We demonstrate that human NK cells and both CD56(bright)CD16(-) and CD56(dim)CD16(+) populations express receptors known to detect the biologically active peptides C3a and C5a (i.e. C3aR, C5aR, C5L2) and the covalently-bound fragments C3b and metabolites iC3b and C3d which serve in immune adhesion (e.g. CR3, CR4). We also show that several pathogen- or tumour/inflammation-related stimuli differentially regulated those complement receptor expression. Furthermore, our results suggest that C3 fragments (C3a, iC3b) have a negative regulatory effect on IFN-γ production in NK cells. This work provides extensive information of human complement receptors relevant to the integrated actions of complement and NK cells which has been suggested by animal studies. The observations may act as a resource that allows further understanding and exploitation of role of complement in human health and immune mediated diseases.
Collapse
|
38
|
Shin NR, Shin IS, Jeon CM, Hong JM, Oh SR, Hahn KW, Ahn KS. Inhibitory effects of Picrasma quassioides (D.Don) Benn. on airway inflammation in a murine model of allergic asthma. Mol Med Rep 2014; 10:1495-500. [PMID: 24927487 DOI: 10.3892/mmr.2014.2322] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 03/12/2014] [Indexed: 11/05/2022] Open
Abstract
Picrasma quassioides (D.Don) Benn. (PQ) is used in traditional medicine for the treatment of inflammatory conditions, including gastritis. This study aimed to evaluate the inhibitory effects of PQ on the inflammatory responses in mice with allergic asthma induced by ovalbumin (OVA) and in lipopolysaccharide (LPS)‑stimulated RAW264.7 cells. To induce allergic asthma, the mice underwent OVA sensitization on days 0 and 14 and then were challenged with OVA from days 21‑23. The mice were administered 15 and 30 mg/kg doses of PQ 1 h prior to the OVA challenge. The PQ treatment decreased the inflammatory cell count in the bronchoalveolar lavage fluid of the mice and reduced the levels of interleukin (IL)‑4, IL‑5, IL‑13 and immunoglobulin (Ig)E when compared with those in the OVA group. The PQ treatment also reduced the airway hyperresponsiveness induced by the OVA challenge, attenuated the recruitment of inflammatory cells and the mucus production in the airways of the mice. In the LPS‑stimulated RAW264.7 cells, the PQ treatment reduced the overexpression of inducible nitric oxide synthase (iNOS). The results indicated that PQ inhibits inflammatory responses in mice with OVA‑sensitized/challenged allergic asthma and in LPS‑stimulated RAW264.7 cells. These effects were considered to be associated with the suppression of iNOS expression. Therefore, PQ may have the potential to treat airway inflammatory diseases, including allergic asthma.
Collapse
Affiliation(s)
- Na-Rae Shin
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon‑gun, Chungbuk 363‑883, Republic of Korea
| | - In-Sik Shin
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon‑gun, Chungbuk 363‑883, Republic of Korea
| | - Chan-Mi Jeon
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon‑gun, Chungbuk 363‑883, Republic of Korea
| | - Ju-Mi Hong
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon‑gun, Chungbuk 363‑883, Republic of Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon‑gun, Chungbuk 363‑883, Republic of Korea
| | - Kyu-Woung Hahn
- Department of Biological Sciences and Biotechnology, College of Life Science and Nano Technology, Hannam University, Daejeon 306‑791, Republic of Korea
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon‑gun, Chungbuk 363‑883, Republic of Korea
| |
Collapse
|
39
|
Treatment with the C5a receptor/CD88 antagonist PMX205 reduces inflammation in a murine model of allergic asthma. Int Immunopharmacol 2014; 21:293-300. [PMID: 24859057 DOI: 10.1016/j.intimp.2014.05.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 05/05/2014] [Accepted: 05/05/2014] [Indexed: 02/03/2023]
Abstract
Allergic asthma is a chronic inflammatory airway disease arising from an aberrant immune response following exposure to environmental stimuli in genetically susceptible persons. The complement component 5 (C5)/C5a Receptor (C5aR/CD88) signaling pathway has been implicated in both experimental allergic asthma and human asthmatic disease. Targeting the C5a/C5aR signaling pathway in rodent models has been shown to either enhance or reduce allergic asthma consequences. Treatment with a recombinant humanized monoclonal antibody directed against C5 has shown unclear results in patients with asthma. The objective of this proof-of-concept animal study was to determine whether the low molecular weight C5aR peptidomimetic antagonist, PMX205, would reduce experimental allergic asthma consequences in mice. PMX205 or vehicle control was administered subcutaneously to BALB/c mice prior to and during standard ovalbumin (OVA) allergen sensitization and aerosolized challenge phases. PMX205 substantially reduced OVA-induced total cell (60%), neutrophil (66%) and eosinophil (65%) influxes in lavage fluid sampling. There were also significant reductions in OVA-induced lavage fluid IL-13 protein and lung Th2 cytokine gene expression with PMX205 administration. PMX205 treatment also diminished OVA-induced lung parenchyma cellular infiltration. PMX205 administration did not reduce OVA-induced serum IgE levels or epithelial mucous/goblet cell generation. There was no evidence of toxicity observed with PMX205 treatment in saline or OVA-challenged animals. These data provide evidence that pharmacologic blockade of C5aR by a low molecular weight antagonist (PMX205) reduces airway inflammatory cell and cytokine responses in experimental allergic asthma, and suggests that PMX205 might represent a novel therapeutic agent for reducing asthmatic outcomes.
Collapse
|