1
|
Tashiro H, Kuwahara Y, Kurihara Y, Takahashi K. Molecular mechanisms and clinical impact of biologic therapies in severe asthma. Respir Investig 2025; 63:50-60. [PMID: 39642687 DOI: 10.1016/j.resinv.2024.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
Severe asthma is a critical condition for patients with asthma, characterized by frequent exacerbations, decreased pulmonary function, and unstable symptoms related to asthma. Consequently, the administration of systemic corticosteroids, which cause secondary damage because of their adverse effects, is considered. Recently, several types of molecular-targeted biological therapies have become available for patients with severe asthma, and they have a capacity to improve the pathophysiology of severe asthma. However, several clinical reports indicate that the effects differ depending on the biological targets of asthma in individual patients. In this review, the molecular mechanisms and clinical impact of biologic therapies in severe asthma are described. In addition, molecules targeted by possible future biologics are also addressed. Better understanding of the mechanistic basis for the role of biologics in severe asthma could lead to new therapeutic options for these patients.
Collapse
Affiliation(s)
- Hiroki Tashiro
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan
| | - Yuki Kuwahara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan
| | - Yuki Kurihara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan
| | - Koichiro Takahashi
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan.
| |
Collapse
|
2
|
Xia Z, Zhao X, Wang L, Huang L, Yang Y, Yin X, He L, Aga Y, Kahaer A, Yang S, Hao L, Chen C. Amelioration of Inflammation in Rats with Experimentally Induced Asthma by Spenceria ramalana Trimen Polyphenols via the PI3K/Akt Signaling Pathway. Int J Mol Sci 2024; 26:165. [PMID: 39796021 PMCID: PMC11720363 DOI: 10.3390/ijms26010165] [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: 11/15/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/13/2025] Open
Abstract
Asthma is a chronic inflammatory respiratory disease that affects millions globally and poses a serious public health challenge. Current therapeutic strategies, including corticosteroids, are constrained by variable patient responses and adverse effects. In this study, a polyphenolic extract derived from the Tibetan medicinal plant Spenceria ramalana Trimen (SRT) was employed and shown to improve experimentally (ovalbumin + cigarette smoke, OVA + CS) induced asthma in rats. Initially, the potential therapeutic mechanism of the polyphenolic components in SRT on OVA + CS-induced asthma was predicated by network pharmacology analysis. Subsequently, in vivo experiments identified that SRT polyphenols exhibit significant anti-asthmatic activities, primarily mediated by lowering inflammatory cell counts such as the WBC (white blood cell), eosinophils, and neutrophils, decreasing the expression of inflammatory cytokines (IL-4, IL-5, IL-13, and TNF-α), alleviating lung histological damage (reduced inflammation, collagen deposition, and mucus secretion), and enhancing the epithelial barrier integrity (upregulation of ZO-1, occludin, and claudin-1). Additionally, SRT polyphenols downregulated the PI3K/Akt (Phosphoinositide 3-kinase/protein kinase B) signaling pathway, improved gut microbiota disruption, and regulated fecal metabolites (glucose-6-glutamate, PS (16:0/0:0), 8-aminocaprylic acid, galactonic acid, Ascr#10, 2,3,4,5,6,7-hexahydroxyheptanoic acid, phosphodimethylethanolamine, muramic acid, 9-oxohexadeca-10e-enoic acid, and sedoheptulose) in asthmatic rats. In conclusion, SRT polyphenols exerted multifaceted protective effects against OVA + CS-induced asthma in rats, highlighting their potential value in preventing asthma via the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Zhaobin Xia
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
| | - Xing Zhao
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
| | - Lu Wang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Chengdu 610041, China
| | - Lin Huang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Chengdu 610041, China
| | - Yanwen Yang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Chengdu 610041, China
| | - Xiangyu Yin
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
| | - Luyu He
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
| | - Yuebumo Aga
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
| | - Ankaer Kahaer
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
| | - Shiyu Yang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
| | - Lili Hao
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Chengdu 610041, China
| | - Chaoxi Chen
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Chengdu 610041, China
| |
Collapse
|
3
|
Sim KH, Lee E, Shrestha P, Choi BH, Hong J, Lee YJ. Isobavachin attenuates FcεRI-mediated inflammatory allergic responses by regulating SHP-1-dependent Fyn/Lyn/Syk/Lck signaling. Biochem Pharmacol 2024; 232:116698. [PMID: 39643121 DOI: 10.1016/j.bcp.2024.116698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 11/30/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Isobavachin, isolated from Psoralea corylifolia L. exhibits therapeutic potential for osteoporosis or skin disease. Here, we evaluated the pharmacological effects of isobavachin on IgE-dependent inflammatory allergic reactions, as well as the underlying mechanisms, in bone marrow-derived mast cells and a mouse model of passive cutaneous anaphylaxis (PCA). Isobavachin reduced IgE/Ag-stimulated degranulation, eicosanoid (leukotriene C4 and prostaglandin D2) generation, and release of pro-inflammatory cytokines (tumor necrosis factor-α (TNF-α) and interleukin (IL)-6). Mechanistic studies revealed that isobavachin suppressed activation of Fyn, Lyn, spleen tyrosine kinase (Syk), and lymphocyte-specific-protein-kinase (Lck), receptor-proximal tyrosine kinases that initiate and play a central role in FcɛRI-mediated mast cell activation, as well as their common downstream signaling molecules including linker for activation of T cells, phospholipase Cγ1, AKT, mitogen-activated protein kinases (MAPKs), and intracellular Ca2+. Additionally, isobavachin increased phosphorylation of Src homology region 2 domain-containing phosphatase-1 (SHP-1), thereby strengthening its interaction with Syk and Lck as well as Fyn and Lyn, resulting in de-phosphorylation of these proximal tyrosine kinases. Genetic knockdown of SHP-1 reversed the inhibitory effects of isobavachin on mast cell activation, as well as the related signaling pathways, indicating that the inhibitory effects of isobavachin are mediated by negative regulation of SHP-1-dependent Fyn, Lyn, Syk and Lck. The anti-inflammatory properties of isobavachin were also examined in macrophages. Isobavachin suppressed production of lipopolysaccharide-stimulated production of pro-inflammatory cytokines and nitric oxide. Furthermore, oral administration of isobavachin attenuated mast cell-mediated PCA reactions in mice. These results suggest that isobavachin is a potential treatment for mast cell-mediated allergic inflammatory diseases.
Collapse
Affiliation(s)
- Kyeong Hwa Sim
- Department of Pharmacology, School of Medicine, Daegu Catholic University, 33 Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea
| | - Eunkyung Lee
- Department of Korean Medicine Development, National Institute for Korean Medicine Development, Gyeongsan 38540, Republic of Korea
| | - Prafulla Shrestha
- Department of Pharmacology, School of Medicine, Daegu Catholic University, 33 Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea
| | - Bo-Hyun Choi
- Department of Pharmacology, School of Medicine, Daegu Catholic University, 33 Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea
| | - Jaewoo Hong
- Department of Physiology, School of Medicine, Daegu Catholic University, 33 Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea; Department of Companion Animal Health, Daegu Catholic University, Gyeongsan, Gyeongbuk 38430, Republic of Korea; Eversummer Lab, Daegu Catholic University, Gyeongsan, Gyeongbuk 38430, Republic of Korea; Department of Research and Development, CaniCatiCare Inc., Daegu 42078, Republic of Korea
| | - Youn Ju Lee
- Department of Pharmacology, School of Medicine, Daegu Catholic University, 33 Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea.
| |
Collapse
|
4
|
Werner R, Carnazza M, Li XM, Yang N. Effect of Small-Molecule Natural Compounds on Pathologic Mast Cell/Basophil Activation in Allergic Diseases. Cells 2024; 13:1994. [PMID: 39682741 DOI: 10.3390/cells13231994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 12/18/2024] Open
Abstract
Pathologic mast cells and basophils, key effector cells in allergic reactions, play pivotal roles in initiating and perpetuating IgE-mediated allergic responses. Conventional therapies for allergies have limitations, prompting exploration into alternative approaches such as small-molecule natural compounds derived from botanical sources. This review synthesizes the existing literature on the effects of these compounds on pathologic mast cells and basophils, highlighting their potential in allergy management, and utilizes the PubMed database for literature acquisition, employing keyword-based searches to identify relevant peer-reviewed sources. Additionally, mechanistic insights were evaluated to contextualize how small-molecule natural compounds can inhibit mast cell/basophil activation, degranulation, and signaling pathways crucial for IgE-mediated allergic reactions. Small-molecule natural compounds exhibit promising anti-allergic effects, yet despite these findings, challenges persist in the development and translation of natural compound-based therapies, including bioavailability and standardization issues. Future research directions include optimizing dosing regimens, exploring synergistic effects with existing therapies, and employing systems pharmacology approaches for a holistic understanding of their mechanisms of action. By harnessing the therapeutic potential of small-molecule natural compounds, effective treatments for allergic diseases may be realized, offering hope for individuals with allergies.
Collapse
Affiliation(s)
- Robert Werner
- Division of R&D, General Nutraceutical Technology LLC, Elmsford, NY 10523, USA
| | - Michelle Carnazza
- Division of R&D, General Nutraceutical Technology LLC, Elmsford, NY 10523, USA
| | - Xiu-Min Li
- Department of Pathology, Microbiology & Immunology, New York Medical College, Valhalla, NY 10595, USA
- Department of Otolaryngology, School of Medicine, New York Medical College, Valhalla, NY 10595, USA
- Department of Dermatology, New York Medical College, Valhalla, NY 10595, USA
| | - Nan Yang
- Division of R&D, General Nutraceutical Technology LLC, Elmsford, NY 10523, USA
| |
Collapse
|
5
|
Poto R, Marone G, Galli SJ, Varricchi G. Mast cells: a novel therapeutic avenue for cardiovascular diseases? Cardiovasc Res 2024; 120:681-698. [PMID: 38630620 PMCID: PMC11135650 DOI: 10.1093/cvr/cvae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/28/2023] [Accepted: 01/08/2024] [Indexed: 04/19/2024] Open
Abstract
Mast cells are tissue-resident immune cells strategically located in different compartments of the normal human heart (the myocardium, pericardium, aortic valve, and close to nerves) as well as in atherosclerotic plaques. Cardiac mast cells produce a broad spectrum of vasoactive and proinflammatory mediators, which have potential roles in inflammation, angiogenesis, lymphangiogenesis, tissue remodelling, and fibrosis. Mast cells release preformed mediators (e.g. histamine, tryptase, and chymase) and de novo synthesized mediators (e.g. cysteinyl leukotriene C4 and prostaglandin D2), as well as cytokines and chemokines, which can activate different resident immune cells (e.g. macrophages) and structural cells (e.g. fibroblasts and endothelial cells) in the human heart and aorta. The transcriptional profiles of various mast cell populations highlight their potential heterogeneity and distinct gene and proteome expression. Mast cell plasticity and heterogeneity enable these cells the potential for performing different, even opposite, functions in response to changing tissue contexts. Human cardiac mast cells display significant differences compared with mast cells isolated from other organs. These characteristics make cardiac mast cells intriguing, given their dichotomous potential roles of inducing or protecting against cardiovascular diseases. Identification of cardiac mast cell subpopulations represents a prerequisite for understanding their potential multifaceted roles in health and disease. Several new drugs specifically targeting human mast cell activation are under development or in clinical trials. Mast cells and/or their subpopulations can potentially represent novel therapeutic targets for cardiovascular disorders.
Collapse
Affiliation(s)
- Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- World Allergy Organization (WAO), Center of Excellence (CoE), Via S. Pansini 5, Naples 80131, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- World Allergy Organization (WAO), Center of Excellence (CoE), Via S. Pansini 5, Naples 80131, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- Institute of Experimental Endocrinology and Oncology ‘G. Salvatore’, National Research Council (CNR), Via S. Pansini 5, Naples 80131, Italy
| | - Stephen J Galli
- Department of Pathology and the Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, 291 Campus Dr, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, 291 Campus Dr, Stanford, CA, USA
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- World Allergy Organization (WAO), Center of Excellence (CoE), Via S. Pansini 5, Naples 80131, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- Institute of Experimental Endocrinology and Oncology ‘G. Salvatore’, National Research Council (CNR), Via S. Pansini 5, Naples 80131, Italy
| |
Collapse
|
6
|
Shaik GM, Khan MS. Betulinic Acid Potentiates Mast Cell Degranulation by Compromising Cell Membrane Integrity and Without Involving Fcεri Receptors. Immunol Invest 2024; 53:695-711. [PMID: 38504489 DOI: 10.1080/08820139.2024.2329990] [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] [Indexed: 03/21/2024]
Abstract
Mast cells play important role in acquired and natural immunity making these favorable therapeutic targets in various inflammatory diseases. Here we observed that, pentacyclic tri terpenoid betulinic acid (BA) treatment resulted in a significantly high number (9%) of cells positive for Hoechst and negative for annexin-V indicating that BA could interfere with plasma membrane integrity. The degranulation of both activated and non-activated mast cells was enhanced upon treatment with BA. The pre-treatment of BA had remarkable effect on calcium response in activated mast cells which showed increased calcium influx relative compared to untreated cells. The results also showed potentially less migration of BA treated mast cells signifying the possible effect of BA on cell membrane. BA treatment resulted in a significant increase in mRNA levels of IL-13 while as mRNA levels of other target cytokines, IL-6 and TNF-α seem to be not affected. Moreover, there was global Increase in phosphorylation of signaling proteins and no significant change in phosphorylation of FcεRI receptors indicating that the effect of BA was independent of signaling cascade or FcεRI receptor mediated mast cell aggregation. Overall, these results portray BA potentiates mast cell effector functions by compromising the membrane integrity and independent of FcεRI involvement.
Collapse
Affiliation(s)
- Gouse M Shaik
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
7
|
Guo X, Bai Y, Jia X, Wu P, Luo L, Wang J, Li H, Guo H, Li J, Guo Z, Yun K, Gao C, Yan J. DNA methylation profiling reveals potential biomarkers of β-lactams induced fatal anaphylactic shock. Forensic Sci Int 2024; 356:111943. [PMID: 38290418 DOI: 10.1016/j.forsciint.2024.111943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 10/30/2023] [Accepted: 01/14/2024] [Indexed: 02/01/2024]
Abstract
Anaphylaxis is a serious reaction of systemic hypersensitivity with that rapid onset and sudden death. Drug hypersensitivity, particularly induced by β-lactams, is one of the most frequent causes of anaphylaxis in adults. But identification of anaphylactic shock, in forensic sciences recently, is difficult, because it mainly depends on nonspecific characteristic morphological changes, as well as exclusion and circumstantial evidence. Here, we detected DNA methylation signatures of β-lactams-induced fatal anaphylactic shock with the Illumina Infinium Human Methylation EPIC BeadChip, to screen potential forensic biomarkers and reveal the molecular mechanisms of drug-induced anaphylaxis with fatal shock and sudden death. Our results indicated that DNA methylation was associated with β-lactams-induced fatal anaphylactic shock, in which the hypomethylation played a vital role. We found that 1459 differentially methylated positions (DMPs) were mainly involved in β-lactams-induced fatal anaphylactic shock by regulating MAPK and other signaling pathways. 18 DNA methylation signatures that could separate β-lactams-induced anaphylactic shock from healthy individuals were identified. The altered methylation of DMPs can affect the transcription of corresponding genes and promote β-lactams-induced fatal anaphylactic shock. The results suggest that DNA methylation can detect forensic identification markers of drug-induced anaphylaxis with fatal shock and sudden death, and it is an effective method for the forensic diagnosis.
Collapse
Affiliation(s)
- Xiangjie Guo
- Department of Forensic Medicine, Shanxi Medical University, Taiyuan, Shanxi, China; Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory of Drug Toxicology and Drug for Radiation Injury, China Institute for Radiation Protection, Taiyuan, ShanXi, China.
| | - Yaqin Bai
- Department of Forensic Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiao Jia
- College of Pharmacy, Nankai University, Tianjin, China
| | - Peng Wu
- Department of Forensic Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Li Luo
- Department of Forensic Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jiaqi Wang
- Department of Forensic Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hao Li
- Institute of Forensic Science of China, Beijing, China
| | - Hualin Guo
- China Astronaut Research and Training Center, Beijing, China
| | - Jianguo Li
- Shanxi Key Laboratory of Drug Toxicology and Drug for Radiation Injury, China Institute for Radiation Protection, Taiyuan, ShanXi, China
| | - Zhongyuan Guo
- Department of Forensic Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Keming Yun
- Department of Forensic Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Cairong Gao
- Department of Forensic Medicine, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Jiangwei Yan
- Department of Forensic Medicine, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
8
|
Okumura M, Yokoyama Y, Yoshida T, Okada Y, Takizawa M, Ikeda O, Kambayashi T. The diacylglycerol kinase ζ inhibitor ASP1570 augments natural killer cell function. Int Immunopharmacol 2023; 125:111145. [PMID: 37935092 DOI: 10.1016/j.intimp.2023.111145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/18/2023] [Accepted: 10/26/2023] [Indexed: 11/09/2023]
Abstract
The enhancement of T cell and NK cell function is an immunotherapeutic strategy for combating cancer. Antibodies that block inhibitory receptors, such as PD-1 and CTLA4, augment T cell function and have been successful in curing patients with some types of cancer. As an alternative approach to targeting specific inhibitory receptors by antibodies, small molecule drugs that inhibit negative regulators of T cell activation have been sought. One potential pharmacological target is diacylglycerol (DAG) kinase (DGK)ζ, which is an enzyme that acts as a negative regulator of DAG by phosphorylating DAG and converting it into phosphatidic acid. DAG-mediated signaling is critical for T cell activation through its T cell receptor and NK cell activation downstream of a variety of activating receptors. Thus, DGKζ-deficient T cells and NK cells display increased function upon activating receptor engagement. Moreover, treatment with the DGKζ-selective inhibitor ASP1570 augments T cell function. In this study, we sought to test whether the acute inhibition of DGKζ by ASP1570 augments NK cell function. We find that ASP1570 enhances DAG-mediated signaling in immunoreceptor-stimulated NK cells. Accordingly, ASP1570 treatment enhanced IFNγ production and degranulation of immunoreceptor-activated NK cells in vitro and NK cell-mediated tumor clearance in vivo. Thus, ASP1570 enhances both T and NK cell function, which could possibly induce more durable anti-tumor responses for immunotherapy.
Collapse
Affiliation(s)
- Mariko Okumura
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuichi Yokoyama
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Taku Yoshida
- Immuno-Oncology, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba 305-8585, Japan
| | - Yohei Okada
- Immuno-Oncology, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba 305-8585, Japan
| | - Masaomi Takizawa
- Research Program Management-Applied Research Management, Astellas Pharma Inc., 2-5-1, Nihonbashi-Honcho, Chuo-ku, Tokyo 103-8411,Japan
| | - Osamu Ikeda
- Immuno-Oncology, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba 305-8585, Japan
| | - Taku Kambayashi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
9
|
Wei C, Wei Y, Cheng J, Tan X, Zhou Z, Lin S, Pang L. Identification and verification of diagnostic biomarkers in recurrent pregnancy loss via machine learning algorithm and WGCNA. Front Immunol 2023; 14:1241816. [PMID: 37691920 PMCID: PMC10485775 DOI: 10.3389/fimmu.2023.1241816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 08/11/2023] [Indexed: 09/12/2023] Open
Abstract
Background Recurrent pregnancy loss defined as the occurrence of two or more pregnancy losses before 20-24 weeks of gestation, is a prevalent and significant pathological condition that impacts human reproductive health. However, the underlying mechanism of RPL remains unclear. This study aimed to investigate the biomarkers and molecular mechanisms associated with RPL and explore novel treatment strategies for clinical applications. Methods The GEO database was utilized to retrieve the RPL gene expression profile GSE165004. This profile underwent differential expression analysis, WGCNA, functional enrichment, and subsequent analysis of RPL gene expression using LASSO regression, SVM-RFE, and RandomForest algorithms for hub gene screening. ANN model were constructed to assess the performance of hub genes in the dataset. The expression of hub genes in both the RPL and control group samples was validated using RT-qPCR. The immune cell infiltration level of RPL was assessed using CIBERSORT. Additionally, pan-cancer analysis was conducted using Sangerbox, and small-molecule drug screening was performed using CMap. Results A total of 352 DEGs were identified, including 198 up-regulated genes and 154 down-regulated genes. Enrichment analysis indicated that the DEGs were primarily associated with Fc gamma R-mediated phagocytosis, the Fc epsilon RI signaling pathway, and various metabolism-related pathways. The turquoise module, which showed the highest relevance to clinical symptoms based on WGCNA results, contained 104 DEGs. Three hub genes, WBP11, ACTR2, and NCSTN, were identified using machine learning algorithms. ROC curves demonstrated a strong diagnostic value when the three hub genes were combined. RT-qPCR confirmed the low expression of WBP11 and ACTR2 in RPL, whereas NCSTN exhibited high expression. The immune cell infiltration analysis results indicated an imbalance of macrophages in RPL. Meanwhile, these three hub genes exhibited aberrant expression in multiple malignancies and were associated with a poor prognosis. Furthermore, we identified several small-molecule drugs. Conclusion This study identifies and validates hub genes in RPL, which may lead to significant advancements in understanding the molecular mechanisms and treatment strategies for this condition.
Collapse
Affiliation(s)
- Changqiang Wei
- Department of Prenatal Diagnosis, The First Afliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yiyun Wei
- Department of Prenatal Diagnosis, The First Afliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Thalassemia Research, Nanning, Guangxi, China
- National Health Commission Key Laboratory of Thalassemia Medicine (Guangxi Medical University), Nanning, Guangxi, China
| | - Jinlian Cheng
- Department of Prenatal Diagnosis, The First Afliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xuemei Tan
- Department of Prenatal Diagnosis, The First Afliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhuolin Zhou
- Department of Prenatal Diagnosis, The First Afliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, China
| | - Shanshan Lin
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, China
| | - Lihong Pang
- Department of Prenatal Diagnosis, The First Afliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Thalassemia Research, Nanning, Guangxi, China
- National Health Commission Key Laboratory of Thalassemia Medicine (Guangxi Medical University), Nanning, Guangxi, China
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, Guangxi, China
| |
Collapse
|
10
|
Bitting K, Hedgespeth B, Ehrhardt-Humbert LC, Arthur GK, Schubert AG, Bradding P, Tilley SL, Cruse G. Identification of redundancy between human FcεRIβ and MS4A6A proteins points toward additional complex mechanisms for FcεRI trafficking and signaling. Allergy 2023; 78:1204-1217. [PMID: 36424895 PMCID: PMC10159887 DOI: 10.1111/all.15595] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 10/19/2022] [Accepted: 10/31/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Allergic diseases are triggered by signaling through the high-affinity IgE receptor, FcεRI. In both mast cells (MCs) and basophils, FcεRI is a tetrameric receptor complex comprising a ligand-binding α subunit (FcεRIα), a tetraspan β subunit (FcεRIβ, MS4A2) responsible for trafficking and signal amplification, and a signal transducing dimer of single transmembrane γ subunits (FcεRIγ). However, FcεRI also exists as presumed trimeric complexes that lack FcεRIβ and are expressed on several cell types outside the MC and basophil lineages. Despite known differences between humans and mice in the presence of the trimeric FcεRI complex, questions remain as to how it traffics and whether it signals in the absence of FcεRIβ. We have previously reported that targeting FcεRIβ with exon-skipping oligonucleotides eliminates IgE-mediated degranulation in mouse MCs, but equivalent targeting in human MCs was not effective at reducing degranulation. RESULTS Here, we report that the FcεRIβ-like protein MS4A6A exists in human MCs and compensates for FcεRIβ in FcεRI trafficking and signaling. Human MS4A6A promotes surface expression of FcεRI complexes and facilitates degranulation. MS4A6A and FcεRIβ are encoded by highly related genes within the MS4A gene family that cluster within the human gene loci 11q12-q13, a region linked to allergy and asthma susceptibility. CONCLUSIONS Our data suggest the presence of either FcεRIβ or MS4A6A is sufficient for degranulation, indicating that MS4A6A could be an elusive FcεRIβ-like protein in human MCs that performs compensatory functions in allergic disease.
Collapse
Affiliation(s)
- Katie Bitting
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, NC State University. Raleigh, NC 27607, USA
| | - Barry Hedgespeth
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, NC State University. Raleigh, NC 27607, USA
| | - Lauren C. Ehrhardt-Humbert
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, NC State University. Raleigh, NC 27607, USA
| | - Greer K. Arthur
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, NC State University. Raleigh, NC 27607, USA
| | - Alicia G. Schubert
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, NC State University. Raleigh, NC 27607, USA
| | - Peter Bradding
- Department of Respiratory Sciences, University of Leicester, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Stephen L. Tilley
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Glenn Cruse
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, NC State University. Raleigh, NC 27607, USA
| |
Collapse
|
11
|
Gimeno R, Ribas‐Llauradó C, Pesque D, Andrades E, Cenni B, Ambros B, Pujol R, Giménez‐Arnau AM. Remibrutinib inhibits hives effector cells stimulated by serum from chronic urticaria patients independently of FcεR1 expression level and omalizumab clinical response. Clin Transl Allergy 2023; 13:e12227. [PMID: 36973953 PMCID: PMC9985467 DOI: 10.1002/clt2.12227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/22/2023] [Accepted: 02/07/2023] [Indexed: 03/07/2023] Open
Abstract
BACKGROUND Despite advances in the treatment of chronic urticaria, in a significant percentage of the patients symptoms are not fully controlled with conventional approaches. New strategies under development include blocking intracellular mediators of mast cell and basophil activation. OBJECTIVE We aim to investigate the effects of the Bruton's tyrosine kinase (BTK) inhibitor remibrutinib on human blood basophils and CD34+ -derived mast cells activation induced by serum obtained from chronic urticaria patients. METHODS Twenty-two patients with chronic spontaneous urticaria (mean age 52 years, 27% women) and 22 patients with chronic inducible urticaria (46 years, 27% women) were included in the study together with a sex-matched control group. Patients were classified as responders or non-responders to anti-IgE therapy on the basis of their clinical data, FcεR1a expression on blood basophils and total IgE levels. Changes on CD63 expression-as an activation marker-, were used to evaluate in vitro the response of basophils and mast cells to serum exposure and the inhibitory effects of remibrutinib. RESULTS Remibrutinib inhibits degranulation induced by IgE cross-linking in mast cells and basophils and also the activation triggered by factors present in the sera of spontaneous and inducible chronic urticaria patients. Patient's serum induces a greater degranulation of effector cells than controls. Activation of mast cells and basophils by patient sera and remibrutinib effects were not related to omalizumab responsiveness. CONCLUSION Remibrutinib inhibits activation of human basophils and mast cells induced in vitro by exposure to the serum of chronic urticaria patients independently of their response to omalizumab.
Collapse
Affiliation(s)
- Ramón Gimeno
- Laboratory of ImmunologyDepartment of PathologyHospital del MarBarcelonaSpain
- Department ImmnologyHospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Department of Medicine and Life SciencesUniversitat Pompeu FabraBarcelonaSpain
| | - Clara Ribas‐Llauradó
- Department ImmnologyHospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
| | - David Pesque
- Department of DermatologyHospital del MarIMIMUniversitat Pompeu FabraBarcelonaSpain
| | - Evelyn Andrades
- Department ImmnologyHospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Department of DermatologyHospital del MarIMIMUniversitat Pompeu FabraBarcelonaSpain
| | - Bruno Cenni
- Department of ResearchBC Novartis Institutes for BioMedical Research, NovartisBaselSwitzerland
| | - Barbara Ambros
- Department of Clinical DevelopmentBA Global Drug Development, NovartisBaselSwitzerland
| | - Ramon Pujol
- Department ImmnologyHospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Department of Medicine and Life SciencesUniversitat Pompeu FabraBarcelonaSpain
- Department of DermatologyHospital del MarIMIMUniversitat Pompeu FabraBarcelonaSpain
| | - Ana M. Giménez‐Arnau
- Department ImmnologyHospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Department of Medicine and Life SciencesUniversitat Pompeu FabraBarcelonaSpain
- Department of DermatologyHospital del MarIMIMUniversitat Pompeu FabraBarcelonaSpain
| |
Collapse
|
12
|
Kitano T, Togawa K, Takemori J, Motoki Y, Kishida K, Itoh S, Takamoto M, Taki S, Hida S. Interleukin-3-dependent potentiation of IgE responsiveness in mouse basophils. Genes Cells 2023; 28:226-236. [PMID: 36637417 DOI: 10.1111/gtc.13007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/14/2023]
Abstract
Basophils produce interleukins (IL)-4 in response to various stimuli and may contribute to type 2 immune responses to various infections and allergens. We found that resting basophils freshly isolated from mice produce IL-4 in response to IL-3 but not to high-affinity Fc receptor (FcεRI) cross-linking (CL), yet both required the immunoreceptor tyrosine-based activation motif (ITAM) containing adaptor Fc receptor γ-chain (FcRγ), while basophils activated in vitro by IL-3 become responsive to FcεRI CL. Acquisition of responsiveness to FcεRI CL occurred upon infection with Trichinella spiralis or administration of superantigen. Because cultured basophils return to a quiescent state upon starvation with IL-3 with surface FcεRI levels unchanged, this acquisition is reversible and probably reflects intracellular events requiring protein synthesis. Interestingly, similar activation-associated acquisition was observed for responsiveness to other stimuli, including CD200R3 CL, which is known to signal via DAP-12, and the allergen protease papain. This acquisition of responsiveness to FcεRI CL was inhibited by Jak inhibitor. Thus, the IL-3 signal bifurcates downstream of Jak, into two distinct pathway, one leading to IL-4 production and the other to render basophils competent to respond to stimuli dependent on ITAM-containing adaptors DAP12 and FcRγ for IL-4 production.
Collapse
Affiliation(s)
- Takuma Kitano
- Department of Molecular and Cellular Health Sciences, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Kaho Togawa
- Department of Molecular and Cellular Health Sciences, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Juri Takemori
- Department of Molecular and Cellular Health Sciences, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Yuya Motoki
- Department of Molecular and Cellular Health Sciences, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Keitaroh Kishida
- Department of Molecular and Cellular Health Sciences, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Saotomo Itoh
- Department of Molecular and Cellular Health Sciences, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Masaya Takamoto
- Department of Infection and Host Defense, Shinshu University School of Medicine, Matsumoto, Japan
| | - Shinsuke Taki
- Department of Molecular and Cellular Immunology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Shigeaki Hida
- Department of Molecular and Cellular Health Sciences, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
13
|
Crozier RWE, Yousef M, Coish JM, Fajardo VA, Tsiani E, MacNeil AJ. Carnosic acid inhibits secretion of allergic inflammatory mediators in IgE-activated mast cells via direct regulation of Syk activation. J Biol Chem 2023; 299:102867. [PMID: 36608933 PMCID: PMC10068559 DOI: 10.1016/j.jbc.2022.102867] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/22/2022] [Accepted: 12/29/2022] [Indexed: 01/09/2023] Open
Abstract
Mast cells are essential regulators of inflammation most recognized for their central role in allergic inflammatory disorders. Signaling via the high-affinity immunoglobulin E (IgE) receptor, FcεRI, leads to rapid degranulation of preformed granules and the sustained release of newly-synthesized pro-inflammatory mediators. Our group recently established rosemary extract (RE) as a potent regulator of mast cell functions, attenuating MAPK and NF-κB signaling. Carnosic acid (CA)-a major polyphenolic constituent of RE-has been shown to exhibit anti-inflammatory effects in other immune cell models, but its role as a potential modulator of mast cell activation is undefined. Therefore, we sought here to determine the modulatory effects of CA in a mast cell model of allergic inflammation. We sensitized bone marrow-derived mast cells (BMMCs) with anti-trinitrophenyl (TNP) IgE and activated with allergen (TNP-BSA) under stem cell factor (SCF) potentiation, in addition to treatment with CA. Our results indicate that CA significantly inhibits allergen-induced early phase responses including Ca2+ mobilization, ROS production, and subsequent degranulation. We also show CA treatment reduced late phase responses, including the release of all cytokines and chemokines examined following IgE stimulation, and corresponding gene expression excepting that of CCL2. Importantly, we determined that CA mediates its inhibitory effects through modulation of tyrosine kinase Syk and downstream effectors TAK1 (Ser412) and Akt (Ser473) as well as NF-κB signaling, while phosphorylation of FcεRI (γ chain) and MAPK proteins remained unaltered. These novel findings establish CA as a potent modulator of mast cell activation, warranting further investigation as a putative anti-allergy therapeutic.
Collapse
Affiliation(s)
- Robert W E Crozier
- Department of Health Sciences, Brock University, St Catharines, Ontario, L2S 3A1, Canada
| | - Michael Yousef
- Department of Health Sciences, Brock University, St Catharines, Ontario, L2S 3A1, Canada
| | - Jeremia M Coish
- Department of Health Sciences, Brock University, St Catharines, Ontario, L2S 3A1, Canada
| | - Val A Fajardo
- Department of Kinesiology, Brock University, St Catharines, Ontario, L2S 3A1, Canada
| | - Evangelia Tsiani
- Department of Health Sciences, Brock University, St Catharines, Ontario, L2S 3A1, Canada
| | - Adam J MacNeil
- Department of Health Sciences, Brock University, St Catharines, Ontario, L2S 3A1, Canada.
| |
Collapse
|
14
|
Crozier RWE, Fajardo VA, MacNeil AJ. Targeting glycogen synthase kinase 3 with CHIR99021 negatively regulates allergen-induced mast cell activation. Eur J Immunol 2023; 53:e2250104. [PMID: 36349527 DOI: 10.1002/eji.202250104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/16/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
Abstract
Mast cells are granulated immune sentinels responsible for allergic inflammation. Allergen-induced FcεRI-signaling leads to rapid degranulation in the early-phase and sustained production and release of pro-inflammatory mediators in the late phase. Glycogen synthase kinase 3 (GSK3) is a constitutively active serine/threonine kinase and a central molecular convergence point for several pro-inflammatory pathways. GSK3 inhibition has been shown to reduce inflammation but has not yet been fully characterized in mast cell activation. Therefore, the objective of this study was to evaluate GSK3 as a putative therapeutic target in allergic inflammation using the GSK3 inhibitor, CHIR99021. Here, we found that GSK3 inhibition impaired ROS production and degranulation. Through modulation of MKK4-JNK, c-jun, and NF-κB signaling, GSK3 inhibition reduced the production/release of IL-6, IL-13, TNF, and CCL1, while only the release of CCL2 and CCL3 was impaired. Furthermore, CHIR99021-mediated GSK3 inhibition altered the pro-inflammatory phenotype of mast cells, reducing c-kit receptor levels. This implicated GSK3 in FcεRI signaling, reducing release of IL-6, TNF, and CCL1 when stimulated through FcεRI, while CCL2 and CCL3 remained unaffected, and were increased when stimulated with SCF only. These results identify GSK3 as a potential therapeutic target of utility warranting further consideration in contexts of pathological mast cell activation.
Collapse
Affiliation(s)
- Robert W E Crozier
- Department of Health Sciences, Brock University, St Catharines, Ontario, Canada
| | - Val A Fajardo
- Department of Kinesiology, Brock University, St Catharines, Ontario, Canada
| | - Adam J MacNeil
- Department of Health Sciences, Brock University, St Catharines, Ontario, Canada
| |
Collapse
|
15
|
Fujisawa S, Nagata Y, Suzuki R. Leukotriene D4 accelerates antigen-mediated mast cell responses via the cysteinyl leukotriene 1 receptor. Cell Immunol 2022; 382:104632. [PMID: 36274438 DOI: 10.1016/j.cellimm.2022.104632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/01/2022] [Accepted: 10/11/2022] [Indexed: 01/13/2023]
Abstract
Cysteinyl leukotrienes (CysLTs), released from mast cells (MCs), are important mediators in allergy. Type 1 receptors for CysLTs (CysLT1R) are involved in accelerating IgE-mediated MC activation. In this study, we aimed to elucidate the mechanisms underlying CysLT1R-mediated MC activation. The CysLT1R agonist/antagonist was applied to two types of major MC models-RBL-2H3 cells and bone marrow-derived MCs (BMMCs). The use of CysLT1R and CysLT2R inhibitors revealed that CysLT1R plays a major role in the acceleration of MC activation. The administration of the CysLT1R agonist leukotriene D4 upregulated IgE-mediated Akt and ERK phosphorylation and subsequently enhanced TNF-α expression, suggesting that CysLT1R regulates the downstream pathway of MC activation. However, these observations were not corroborated by CysLT1R knockdown using shRNA, suggesting a differential regulatory mechanism between the temporal and constitutive inhibitions of CysLT. In conclusion, CysLT1R enhances MC activation by accelerating IgE-induced signal transduction, which enables the co-regulation of rapid degranulation and delayed synthesis of inflammatory mediators in MCs.
Collapse
Affiliation(s)
- Sakura Fujisawa
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Ishikawa 920-1192, Japan
| | - Yuka Nagata
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Ishikawa 920-1192, Japan
| | - Ryo Suzuki
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Ishikawa 920-1192, Japan.
| |
Collapse
|
16
|
Poto R, Criscuolo G, Marone G, Brightling CE, Varricchi G. Human Lung Mast Cells: Therapeutic Implications in Asthma. Int J Mol Sci 2022; 23:14466. [PMID: 36430941 PMCID: PMC9693207 DOI: 10.3390/ijms232214466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Mast cells are strategically located in different compartments of the lung in asthmatic patients. These cells are widely recognized as central effectors and immunomodulators in different asthma phenotypes. Mast cell mediators activate a wide spectrum of cells of the innate and adaptive immune system during airway inflammation. Moreover, these cells modulate the activities of several structural cells (i.e., fibroblasts, airway smooth muscle cells, bronchial epithelial and goblet cells, and endothelial cells) in the human lung. These findings indicate that lung mast cells and their mediators significantly contribute to the immune induction of airway remodeling in severe asthma. Therapies targeting mast cell mediators and/or their receptors, including monoclonal antibodies targeting IgE, IL-4/IL-13, IL-5/IL-5Rα, IL-4Rα, TSLP, and IL-33, have been found safe and effective in the treatment of different phenotypes of asthma. Moreover, agonists of inhibitory receptors expressed by human mast cells (Siglec-8, Siglec-6) are under investigation for asthma treatment. Increasing evidence suggests that different approaches to depleting mast cells show promising results in severe asthma treatment. Novel treatments targeting mast cells can presumably change the course of the disease and induce drug-free remission in bronchial asthma. Here, we provide an overview of current and promising treatments for asthma that directly or indirectly target lung mast cells.
Collapse
Affiliation(s)
- Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Gjada Criscuolo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (CNR), 80131 Naples, Italy
| | - Chris E. Brightling
- Department of Respiratory Sciences, Leicester NIHR BRC, Institute for Lung Health, University of Leicester, Leicester LE1 7RH, UK
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (CNR), 80131 Naples, Italy
| |
Collapse
|
17
|
Alam SB, Wagner A, Willows S, Kulka M. Quercetin and Resveratrol Differentially Decrease Expression of the High-Affinity IgE Receptor (FcεRI) by Human and Mouse Mast Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196704. [PMID: 36235240 PMCID: PMC9573482 DOI: 10.3390/molecules27196704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/24/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022]
Abstract
Mast cells (MC) synthesize and store proinflammatory mediators and are centrally important in atopic diseases such as asthma and atopic dermatitis. Quercetin a and resveratrol are plant derived polyphenolic compounds with anti-inflammatory properties that inhibit MC degranulation and mediator release. However, the underlying mechanism of these inhibitory effects on MC is poorly understood and it is unclear whether this is a general effect on all MC phenotypes. We have characterized and compared the effects of quercetin with resveratrol on human (LAD2) and mouse (MC/9 and BMMC) MC mediator release, receptor expression and FcεRI signaling to better understand the mechanisms involved in quercetin and resveratrol-mediated inhibition of MC activation. Quercetin significantly decreased the expression of FcεRI by BMMC and MC/9, although the effects on MC/9 were associated with a significant reduction in cell viability. Quercetin also inhibited antigen-stimulated TNF release by BMMC. Although neither quercetin nor resveratrol significantly altered antigen-stimulated BMMC degranulation or downstream signaling events such as phosphorylation of spleen tyrosine kinase (SYK) or extracellular signal-regulated kinase 1/2 (ERK), resveratrol inhibited ERK phosphorylation and FcεRI- stimulated degranulation in LAD2. Our data suggests that quercetin and resveratrol inhibit human and mouse MC differentially and that these effects are associated with modification of FcεRI expression, signaling (phosphorylation of SYK and ERK) and mediator release.
Collapse
Affiliation(s)
- Syed Benazir Alam
- Nanotechnology Research Center, National Research Council, Edmonton, AB T6G 2M9, Canada
- Correspondence:
| | - Ashley Wagner
- Nanotechnology Research Center, National Research Council, Edmonton, AB T6G 2M9, Canada
| | - Steven Willows
- Nanotechnology Research Center, National Research Council, Edmonton, AB T6G 2M9, Canada
| | - Marianna Kulka
- Nanotechnology Research Center, National Research Council, Edmonton, AB T6G 2M9, Canada
- Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada
| |
Collapse
|
18
|
Mishima S, Sakamoto M, Kioka H, Nagata Y, Suzuki R. Multifunctional regulation of VAMP3 in exocytic and endocytic pathways of RBL-2H3 cells. Front Immunol 2022; 13:885868. [PMID: 35990647 PMCID: PMC9388853 DOI: 10.3389/fimmu.2022.885868] [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: 02/28/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022] Open
Abstract
Mast cells (MCs) are inflammatory cells involved in allergic reactions. Crosslinking of the high-affinity receptor for IgE (FcϵRI) with multivalent antigens (Ags) induces secretory responses to release various inflammatory mediators. These responses are largely mediated by soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs). Vesicle-associated membrane protein 3 (VAMP3) is a vesicular-SNARE that interacts with targeted SNARE counterparts, driving the fusion of MC secretory granules with the membrane and affecting subsequent assembly of the plasma membrane. However, the role of VAMP3 in FcϵRI-mediated MC function remains unclear. In this study, we comprehensively examined the role of VAMP3 and the molecular mechanisms underlying VAMP3-mediated MC function upon FcϵRI activation. VAMP3 shRNA transduction considerably decreased VAMP3 expression compared with non-target shRNA-transduced (NT) cells. VAMP3 knockdown (KD) cells were sensitized with an anti-DNP IgE antibody and subsequently stimulated with Ag. The VAMP3 KD cells showed decreased degranulation response upon Ag stimulation. Next, we observed intracellular granule formation using CD63-GFP fluorescence. The VAMP3 KD cells were considerably impaired in their capacity to increase the size of granules when compared to NT cells, suggesting that VAMP3 mediates granule fusion and therefore promotes granule exocytosis in MCs. Analysis of FcϵRI-mediated activation of signaling events (FcϵRI, Lyn, Syk, and intracellular Ca2+ response) revealed that signaling molecule activation was enhanced in VAMP3 KD cells. We also found that FcϵRI expression on the cell surface decreased considerably in VAMP3 KD cells, although the amount of total protein did not vary. VAMP3 KD cells also showed dysregulation of plasma membrane homeostasis, such as endocytosis and lipid raft formation. The difference in the plasma membrane environment in VAMP3 KD cells might affect FcϵRI membrane dynamics and the subsequent signalosome formation. Furthermore, IgE/Ag-mediated secretion of TNF-α and IL-6 is oppositely regulated in the absence of VAMP3, which appears to be attributed to both the activation of FcϵRI and defects in VAMP3-mediated membrane fusion. Taken together, these results suggest that enhanced FcϵRI-mediated signal transduction in VAMP3 KD cells occurs due to the disruption of plasma membrane homeostasis. Hence, a multifunctional regulation of VAMP3 is involved in complex secretory responses in MCs.
Collapse
|
19
|
Takemura A, Ohto N, Kuwahara H, Mizuno M. Sphingoid base in pineapple glucosylceramide suppresses experimental allergy by binding leukocyte mono-immunoglobulin-like receptor 3. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:2704-2709. [PMID: 34708420 PMCID: PMC9299474 DOI: 10.1002/jsfa.11610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/13/2021] [Accepted: 10/28/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND The increase in patients suffering from type I hypersensitivity, including hay fever and food allergy, is a serious public health issue around the world. Recent studies have focused on allergy prevention by food factors with fewer side effects. The purpose of this study was to evaluate the effect of dietary glucosylceramide from pineapples (P-GlcCer) on type I hypersensitivity and elucidate mechanisms. RESULTS Oral administration of P-GlcCer inhibited ear edema in passive cutaneous anaphylaxis reaction. In a Caco-2/RBL-2H3 co-culture system, P-GlcCer inhibited β-hexosaminidase release from RBL-2H3 cells. The direct treatment of P-GlcCer on RBL-2H3 did not affect β-hexosaminidase release, but sphingoid base moiety of P-GlcCer did. These results predicted that sphingoid base, a metabolite of P-GlcCer, through the intestine inhibited type I hypersensitivity by inhibiting mast cell degranulation. In addition, the inhibitory effects of P-GlcCer on ear edema and degranulation of RBL-2H3 cells were canceled by pretreatment of leukocyte mono-immunoglobulin-like receptor 3 (LMIR3)-Fc, which can block LMIR3-mediated inhibitory signals. CONCLUSION It was demonstrated that a sphingoid base, one of the metabolites of P-GlcCer, may inhibit mast cell degranulation by binding to LMIR3. The oral administration of P-GlcCer is a novel and attractive food factor that acts directly on mast cells to suppress allergy. © 2021 The Authors. Journal of The Science of Food and Agriculture published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.
Collapse
Affiliation(s)
- Ayumi Takemura
- Department of Agrobioscience, Graduate School of Agricultural ScienceKobe UniversityKobeJapan
| | | | | | - Masashi Mizuno
- Department of Agrobioscience, Graduate School of Agricultural ScienceKobe UniversityKobeJapan
| |
Collapse
|
20
|
Recent Progress in Traditional Chinese Medicines and Their Mechanism in the Treatment of Allergic Rhinitis. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:3594210. [PMID: 35444784 PMCID: PMC9015857 DOI: 10.1155/2022/3594210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/17/2022] [Indexed: 02/06/2023]
Abstract
Objective To conduct a systematic review on the mechanism of action and use of traditional Chinese medicines (TCM) in allergic rhinitis treatment. Background Allergic rhinitis (AR) is a type I allergic disease of the immune system induced by immunoglobulin E mediated inflammation and is characterized by sneezing, nasal itching, paroxysmal nasal obstruction, mucosal edema, cough, and rhinorrhea. More than 500 million people have been affected by rhinitis worldwide in the past 20 years, leading to negative effects on health, quality of life, and social relationships. Currently, the trending medicines used in the case of AR include intranasal corticosteroids and oral H1 antihistamines, which are given as combinatorial medicines supplemented with immune therapy. These medications have been found to be very effective in either the short term or long term; however, they have been found to possess some serious side effects. Search Methodology. The information in this article on classical and traditional Chinese medications used to treat AR was derived from original papers and reviews published in Chinese and English language journals. Two Chinese databases (Wanfang and CNKI) and three English databases (Cochrane Library, PubMed, and Embase) were utilized for data gathering. Results Traditional Chinese remedies have been identified to influence the production of cytokines such as IL-5 and IL-6, which are key mediators of eosinophilic inflammation, TNF-α, which stimulates TH2 cells at the site of inflammation, and NF-кB, which is required for cytokine and IgE antibody production. TCM has also been shown to be successful in lowering histamine levels, preserving histological changes by decreasing the thickness of the lamina propria, and downregulating the expression of Orai1, STIM1, and TRYC1, showing low expression of Ca+2 channel proteins. Conclusion In this review, we discussed a series of classical, traditional Chinese medications, including Centipeda minima, Scutellaria baicalensis, licorice root (Glycyrrhiza uralensis), and others, as potential antiallergic agents and investigate their in vivo effect upon the production of cytokines and release of histamines for allergic rhinitis treatment.
Collapse
|
21
|
Dok-1 regulates mast cell degranulation negatively through inhibiting calcium-dependent F-actin disassembly. Clin Immunol 2022; 238:109008. [PMID: 35421591 DOI: 10.1016/j.clim.2022.109008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022]
Abstract
In food allergies, antigen-induced aggregation of FcεRI on mast cells initiates highly ordered and sequential signaling events. Dok-1(downstream of tyrosine kinase 1), undergoes intense tyrosine phosphorylation upon FcεRI stimulation, which negatively regulates Ras/Erk signaling and the subsequent cytokine release, but it remains unclear whether Dok-1 regulates Fc-mediated degranulation. In this study, we investigated the role of Dok-1 in FcεRI-mediated degranulation. Dok-1 overexpressing RBL-2H3 cells were established. Degranulation, immunoprecipitation, co-immunoprecipitation, immunoblotting and flow cytometry assay were performed to explore the effects of Dok-1 and its underlying mechanisms. We found that, following FcεRI activation, Dok-1 was recruited to the plasma membrane, leading to tyrosine phosphorylation. Phosphorylated Dok-1 inhibits FcεRI-operated calcium influx, and negatively regulated degranulation by inhibiting calcium-dependent disassembly of actin filaments. Our data revealed that Dok-1 is a negative regulator of FcεRI-mediated mast cell degranulation. These findings contribute to the identification of therapeutic targets for food allergies.
Collapse
|
22
|
Nagata Y, Suzuki R. FcεRI: A Master Regulator of Mast Cell Functions. Cells 2022; 11:cells11040622. [PMID: 35203273 PMCID: PMC8870323 DOI: 10.3390/cells11040622] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/29/2022] [Accepted: 02/09/2022] [Indexed: 02/04/2023] Open
Abstract
Mast cells (MCs) perform multiple functions thought to underlie different manifestations of allergies. Various aspects of antigens (Ags) and their interactions with immunoglobulin E (IgE) cause diverse responses in MCs. FcεRI, a high-affinity IgE receptor, deciphers the Ag–IgE interaction and drives allergic responses. FcεRI clustering is essential for signal transduction and, therefore, determines the quality of MC responses. Ag properties precisely regulate FcεRI dynamics, which consequently initiates differential outcomes by switching the intracellular-signaling pathway, suggesting that Ag properties can control MC responses, both qualitatively and quantitatively. Thus, the therapeutic benefits of FcεRI-targeting strategies have long been examined. Disrupting IgE–FcεRI interactions is a potential therapeutic strategy because the binding affinity between IgE and FcεRI is extremely high. Specifically, FcεRI desensitization, due to internalization, is also a potential therapeutic target that is involved in the mechanisms of allergen-specific immunotherapy. Several recent findings have suggested that silent internalization is strongly associated with FcεRI dynamics. A comprehensive understanding of the role of FcεRI may lead to the development of novel therapies for allergies. Here, we review the qualitatively diverse responses of MCs that impact the attenuation/development of allergies with a focus on the role of FcεRI toward Ag exposure.
Collapse
|
23
|
Korver W, Wong A, Gebremeskel S, Negri GL, Schanin J, Chang K, Leung J, Benet Z, Luu T, Brock EC, Luehrsen K, Xu A, Youngblood BA. The Inhibitory Receptor Siglec-8 Interacts With FcεRI and Globally Inhibits Intracellular Signaling in Primary Mast Cells Upon Activation. Front Immunol 2022; 13:833728. [PMID: 35154156 PMCID: PMC8837033 DOI: 10.3389/fimmu.2022.833728] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Immunomodulation of mast cell (MC) activity is warranted in allergic and inflammatory diseases where MCs have a central role in pathogenesis. Targeting Siglec-8, an inhibitory receptor on MCs and eosinophils, has shown promising activity in preclinical and clinical studies. While the intracellular pathways that regulate Siglec-8 activity in eosinophils have been well studied, the signaling mechanisms that lead to MC inhibition have not been fully elucidated. Here, we evaluate the intracellular signaling pathways of Siglec-8-mediated inhibition in primary MCs using an anti-Siglec-8 monoclonal antibody (mAb). Phospho-proteomic profiling of FcεRI-activated MCs revealed Siglec-8 mAb-treatment globally inhibited proximal and downstream kinases, leading to attenuated MC activation and degranulation. In fact, Siglec-8 was found to directly interact with FcεRI signaling molecules. Siglec-8 inhibition was dependent on both cytoplasmic immunoreceptor tyrosine-based inhibitory motifs (ITIMs) that interact with the SH2 containing protein phosphatase Shp-2 upon Siglec-8 phosphorylation. Taken together, these data support a model in which Siglec-8 regulates proximal FcεRI-induced phosphorylation events through phosphatase recruitment and interaction with FcεRIγ, resulting in global inhibition of MCs upon Siglec-8 mAb engagement.
Collapse
Affiliation(s)
| | - Alan Wong
- Allakos Inc., Redwood City, CA, United States
| | | | | | | | | | - John Leung
- Allakos Inc., Redwood City, CA, United States
| | | | - Thuy Luu
- Allakos Inc., Redwood City, CA, United States
| | | | | | - Alan Xu
- Allakos Inc., Redwood City, CA, United States
| | | |
Collapse
|
24
|
Luo F, Wen Y, Zhao L, Su S, Zhao Y, Lei W, Li Z. Chlamydia trachomatis induces lncRNA MIAT upregulation to regulate mitochondria-mediated host cell apoptosis and chlamydial development. J Cell Mol Med 2021; 26:163-177. [PMID: 34859581 PMCID: PMC8742237 DOI: 10.1111/jcmm.17069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 10/27/2021] [Accepted: 11/11/2021] [Indexed: 01/12/2023] Open
Abstract
Chlamydia trachomatis persistent infection is the leading cause of male prostatitis and female genital tract diseases. Inhibition of host cell apoptosis is the key to maintaining Chlamydia survival in vivo, and long noncoding RNAs (lncRNAs) play important roles in its developmental cycle and pathogenesis. However, it is not clear how lncRNAs regulate persistent Chlamydia infection. Here, using a microarray method, we identified 1718 lncRNAs and 1741 mRNAs differentially expressed in IFN-γ-induced persistent C. trachomatis infection. Subsequently, 10 upregulated and 5 downregulated differentially expressed lncRNAs were verified by qRT-PCR to confirm the reliability of the chip data. The GO and KEGG analyses revealed that differentially regulated transcripts were predominantly involved in various signalling pathways related to host immunity and apoptosis response. Targeted silencing of three lncRNAs (MIAT, ZEB1-AS1 and IRF1) resulted in increased apoptosis rates. Furthermore, interference with lncRNA MIAT caused not only an obvious downregulation of the Bcl-2/Bax ratio but also a marked release of cytochrome c, resulting in a significantly elevated level of caspase-3 activation. Meanwhile, MIAT was involved in the regulation of chlamydial development during the persistent infection. Collectively, these observations shed light on the enormous complex lncRNA regulatory networks involved in mitochondria-mediated host cell apoptosis and the growth and development of C. trachomatis.
Collapse
Affiliation(s)
- Fangzhen Luo
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China.,Hunan Polytechnic of Environment and Biology, Hengyang, China
| | - Yating Wen
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Lanhua Zhao
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Shengmei Su
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Yuqi Zhao
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Wenbo Lei
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Zhongyu Li
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| |
Collapse
|
25
|
Dhakal H, Kim MJ, Lee S, Choi YA, Kim N, Kwon TK, Khang D, Kim SH. Ursolic acid inhibits FcεRI-mediated mast cell activation and allergic inflammation. Int Immunopharmacol 2021; 99:107994. [PMID: 34435583 DOI: 10.1016/j.intimp.2021.107994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/14/2021] [Accepted: 07/14/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Mast cells are the primary cells that play a crucial role in the allergic diseases via secretion of diverse allergic mediators. Ursolic acid (UA) is a naturally occurring anti-inflammatory triterpenoid possessing various biological properties such as immune regulation, antioxidant, and anti-fibrotic. The aim of this study was to evaluate the effects of UA in FcεRI-mediated mast cell activation and allergic inflammation. METHODS In this study, mast cells were stimulated with immunoglobulin E (IgE) and the anti-allergic effects of UA were assessed by measuring the levels of allergic mediators. In vivo effects of UA were observed by generating passive cutaneous anaphylaxis (PCA) and active systemic anaphylaxis (ASA) in mouse model. RESULTS We found that UA inhibited the degranulation of mast cell by suppressing the intracellular calcium level in a concentration-dependent manner. UA inhibited the expression and the release of pro-inflammatory cytokines in mast cells. Anti-allergic effects of UA were demonstrated via suppression of FcεRI-mediated signaling molecules. In addition, UA inhibited the IgE-mediated PCA and ovalbumin-induced ASA reactions in a dose-dependent manner. CONCLUSIONS Based on these findings, we suggest that UA might have potential as a therapeutic candidate for the treatment of allergic inflammatory diseases via inhibition of FcεRI-mediated mast cell activation.
Collapse
Affiliation(s)
- Hima Dhakal
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Min-Jong Kim
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Soyoung Lee
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Young-Ae Choi
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Namkyung Kim
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Dongwoo Khang
- Department of Physiology, School of Medicine, Gachon University, Incheon, Republic of Korea.
| | - Sang-Hyun Kim
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
26
|
Xie Z, Xiang C, Li X, Fan C, Chen T, Liu M, Ma Y, Bai F, Tang W, Hu Y. Discovery of Potent Antiallergic Agents Based on an o-Aminopyridinyl Alkynyl Scaffold. J Med Chem 2021; 64:13588-13603. [PMID: 34476950 DOI: 10.1021/acs.jmedchem.1c00976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Effective therapeutic agents are highly desired for immune-mediated allergic diseases. Herein, we report the design, synthesis, and structure-activity relationship of an o-aminopyridinyl alkyne series as novel orally bioavailable antiallergic agents, which was identified through phenotypic screening. Compound optimization yielded a highly potent compound 36, which effectively suppressed mast cell degranulation in a dose-dependent manner (IC50, 2.54 nM for RBL-2H3 cells; 48.28 nM for peritoneal mast cells (PMCs)) with a good therapeutic index. It also regulated the activation of FcεRI-mediated downstream signaling proteins in IgE/Ag-stimulated RBL-2H3 cells. In addition, 36 exhibited excellent in vivo pharmacokinetic properties and antiallergic efficacy in both passive systemic anaphylaxis (PSA) and house dust mite (HDM)-induced murine models of pulmonary allergic inflammation. Furthermore, preliminary analysis of the kinases profile identified Src-family kinases as potential targets for 36. Compound 36 may serve as a new valuable lead compound for future antiallergic drug discovery.
Collapse
Affiliation(s)
- Zhicheng Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zu-Chong-Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Caigui Xiang
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Xin Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zu-Chong-Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Chen Fan
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Taiwen Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zu-Chong-Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Moting Liu
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yanjie Ma
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zu-Chong-Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Fang Bai
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wei Tang
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Youhong Hu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zu-Chong-Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
27
|
Tontini C, Bulfone-Paus S. Novel Approaches in the Inhibition of IgE-Induced Mast Cell Reactivity in Food Allergy. Front Immunol 2021; 12:613461. [PMID: 34456900 PMCID: PMC8387944 DOI: 10.3389/fimmu.2021.613461] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 06/23/2021] [Indexed: 01/21/2023] Open
Abstract
Allergy is an IgE-dependent type-I hypersensitivity reaction that can lead to life-threatening systemic symptoms such as anaphylaxis. In the pathogenesis of the allergic response, the common upstream event is the binding of allergens to specific IgE, inducing cross-linking of the high-affinity FcεRI on mast cells, triggering cellular degranulation and the release of histamine, proteases, lipids mediators, cytokines and chemokines with inflammatory activity. A number of novel therapeutic options to curb mast cell activation are in the pipeline for the treatment of severe allergies. In addition to anti-IgE therapy and allergen-specific immunotherapy, monoclonal antibodies targeted against several key Th2/alarmin cytokines (i.e. IL-4Rα, IL-33, TSLP), active modification of allergen-specific IgE (i.e. inhibitory compounds, monoclonal antibodies, de-sialylation), engagement of inhibitory receptors on mast cells and allergen-specific adjuvant vaccines, are new promising options to inhibit the uncontrolled release of mast cell mediators upon allergen exposure. In this review, we critically discuss the novel approaches targeting mast cells limiting allergic responses and the immunological mechanisms involved, with special interest on food allergy treatment.
Collapse
Affiliation(s)
- Chiara Tontini
- Lydia Becker Institute for Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Silvia Bulfone-Paus
- Lydia Becker Institute for Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
28
|
Lipid-based and protein-based interactions synergize transmembrane signaling stimulated by antigen clustering of IgE receptors. Proc Natl Acad Sci U S A 2021; 118:2026583118. [PMID: 34433665 DOI: 10.1073/pnas.2026583118] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Antigen (Ag) crosslinking of immunoglobulin E-receptor (IgE-FcεRI) complexes in mast cells stimulates transmembrane (TM) signaling, requiring phosphorylation of the clustered FcεRI by lipid-anchored Lyn tyrosine kinase. Previous studies showed that this stimulated coupling between Lyn and FcεRI occurs in liquid ordered (Lo)-like nanodomains of the plasma membrane and that Lyn binds directly to cytosolic segments of FcεRI that it initially phosphorylates for amplified activity. Net phosphorylation above a nonfunctional threshold is achieved in the stimulated state but not in the resting state, and current evidence supports the hypothesis that this relies on Ag crosslinking to disrupt a balance between Lyn and tyrosine phosphatase activities. However, the structural interactions that underlie the stimulation process remain poorly defined. This study evaluates the relative contributions and functional importance of different types of interactions leading to suprathreshold phosphorylation of Ag-crosslinked IgE-FcεRI in live rat basophilic leukemia mast cells. Our high-precision diffusion measurements by imaging fluorescence correlation spectroscopy on multiple structural variants of Lyn and other lipid-anchored probes confirm subtle, stimulated stabilization of the Lo-like nanodomains in the membrane inner leaflet and concomitant sharpening of segregation from liquid disordered (Ld)-like regions. With other structural variants, we determine that lipid-based interactions are essential for access by Lyn, leading to phosphorylation of and protein-based binding to clustered FcεRI. By contrast, TM tyrosine phosphatase, PTPα, is excluded from these regions due to its Ld-preference and steric exclusion of TM segments. Overall, we establish a synergy of lipid-based, protein-based, and steric interactions underlying functional TM signaling in mast cells.
Collapse
|
29
|
Srivastava M, Kaplan MH. Transcription Factors in the Development and Pro-Allergic Function of Mast Cells. FRONTIERS IN ALLERGY 2021; 2:679121. [PMID: 35387064 PMCID: PMC8974754 DOI: 10.3389/falgy.2021.679121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/12/2021] [Indexed: 11/13/2022] Open
Abstract
Mast cells (MCs) are innate immune cells of hematopoietic origin localized in the mucosal tissues of the body and are broadly implicated in the pathogenesis of allergic inflammation. Transcription factors have a pivotal role in the development and differentiation of mast cells in response to various microenvironmental signals encountered in the resident tissues. Understanding the regulation of mast cells by transcription factors is therefore vital for mechanistic insights into allergic diseases. In this review we summarize advances in defining the transcription factors that impact the development of mast cells throughout the body and in specific tissues, and factors that are involved in responding to the extracellular milieu. We will further describe the complex networks of transcription factors that impact mast cell physiology and expansion during allergic inflammation and functions from degranulation to cytokine secretion. As our understanding of the heterogeneity of mast cells becomes more detailed, the contribution of specific transcription factors in mast cell-dependent functions will potentially offer new pathways for therapeutic targeting.
Collapse
Affiliation(s)
- Mansi Srivastava
- Department of BioHealth Informatics, School of Informatics and Computing, Indiana University-Purdue University, Indianapolis, IN, United States
| | - Mark H. Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
- *Correspondence: Mark H. Kaplan
| |
Collapse
|
30
|
Novosad J, Krčmová I. Evolution of our view on the IgE molecule role in bronchial asthma and the clinical effect of its modulation by omalizumab: Where do we stand today? Int J Immunopathol Pharmacol 2021; 34:2058738420942386. [PMID: 32689848 PMCID: PMC7375718 DOI: 10.1177/2058738420942386] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Bronchial asthma is a heterogeneous disease whose definition and treatment are based on evidence of variable airway obstruction and airway inflammation. Despite the enormous increase in the amount of information on the pathogenesis of this disease, diagnosis is still an unresolved problem, as we still lack sensitive and specific biomarkers. On the other hand, at the turn of the 20th and 21st century, there was a rapid development of therapeutic modalities based on the principle of biological therapy. The first authorized drug matching these characteristics was omalizumab – a monoclonal antibody directed against immunoglobulin E (IgE). It has been used for the treatment of severe forms of bronchial asthma for more than 15 years, which is a sufficient time to acquire ways of its effective use and to assess whether the treatment with omalizumab has met our expectations. However, we continue to discover new and surprising facts about the effects of omalizumab treatment which leads to widening of therapeutic indications. In this work, a basic overview of the very complex role of the IgE molecule in the organism (with a special emphasis on allergic asthma) is discussed, and the most important practical and clinical consequences resulting from its modulation by targeted therapy with omalizumab are summarized.
Collapse
Affiliation(s)
- Jakub Novosad
- Institute of Clinical Immunology and Allergology, University Hospital Hradec Králové, Hradec Králové, Czech Republic.,Faculty of Medicine in Hradec Králové, Charles University in Prague, Prague, Czech Republic
| | - Irena Krčmová
- Institute of Clinical Immunology and Allergology, University Hospital Hradec Králové, Hradec Králové, Czech Republic.,Faculty of Medicine in Hradec Králové, Charles University in Prague, Prague, Czech Republic
| |
Collapse
|
31
|
Catalpol exerts antiallergic effects in IgE/ovalbumin-activated mast cells and a murine model of ovalbumin-induced allergic asthma. Int Immunopharmacol 2021; 96:107782. [PMID: 34022666 DOI: 10.1016/j.intimp.2021.107782] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/17/2021] [Accepted: 05/07/2021] [Indexed: 12/22/2022]
Abstract
Immunoglobulin E (IgE) and mast cells play important roles in the pathogenesis of allergic asthma. Catalpol, an iridoid glycoside, exerts many biological functions including anti-inflammatory activities. Herein, we investigated catalpol to determine both its antiallergic effects on IgE/ovalbumin (OVA)-stimulated mouse bone marrow-derived mast cells and its therapeutic actions in murine allergic asthma. We found that catalpol dramatically suppressed IgE/OVA-induced mast cell degranulation. Meanwhile, 5 ~ 100 μM of catalpol neither affected the expression level of the high-affinity receptor of IgE (FcεRI) by mast cells nor induced mast cell apoptosis. In addition, mRNA expression levels of inflammatory enzymes including cyclooxygenase (COX)-1, COX-2, and 5-lipoxygenase were downregulated. Administration of catalpol also suppressed production of prostaglandin D2 (PGD2), interleukin (IL)-6, and IL-13, while not affecting tumor necrosis factor (TNF)-α production. Further, catalpol pretreatment significantly attenuated the FcεRI-mediated Akt signaling pathway. In mice with IgE/OVA-induced asthma, oral administration of catalpol remarkably suppressed the production of OVA-specific IgE, the development of airway hyperresponsiveness (AHR), and the infiltration of eosinophils and neutrophils into the lungs. Histological studies demonstrated that catalpol substantially inhibited the recruitment of mast cells and increased mucus production in lung tissues. Catalpol-treated mice had significantly lower levels of helper T cell type 2 (Th2) cytokines (IL-4, IL-5, and IL-13), PGD2, eotaxin-1, and C-X-C chemokine ligand-1 (CXCL1) in bronchoalveolar lavage fluid (BALF) than did the allergic group. Collectively, these results indicated that the suppressive effects of catalpol on degranulation and mediator generation by mast cells were beneficial in treating allergic asthma.
Collapse
|
32
|
Zhou G, Zeng Q, Wei W, Teng H, Liu C, Zhou Z, Liang B, Long H. A pilot study of differential gene expressions in patients with cough variant asthma and classic bronchial asthma. J Asthma 2021; 59:1070-1078. [PMID: 33878997 DOI: 10.1080/02770903.2021.1917604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Despite extensive exploration of asthma, the mechanism of asthma has not been fully elucidated. Cough variant asthma (CVA) is considered as precursor to classical asthma (CA). Comparative study between CA and CVA may be helpful in further understanding the pathogenesis of asthma. METHODS Peripheral blood mononuclear cells (PBMCs) were isolated from CVA, CA and healthy adults. Each group consisted of five cases. Total RNA was extracted from the PBMCs. Agilent 4 × 44 K human genome oligo microarray was used to detect whole genome expression. Allogeneic clustering, Gene Ontology and KEGG analysis were performed to investigate differentially expressed genes (DEGs). Then, ten candidate genes were screened and verified by real-time PCR. RESULTS Gene expressions were significantly different among the three groups, with 202 DEGs between the CA and the CVA groups. The Gene Ontology analysis suggested that the DEGs were significantly enriched in 'histone H4-K20 demethylation' and 'antigen processing and presentation of endogenous antigens'. HDC, EGR1, DEFA4, LTF, G0S2, IL4, TFF3, CTSG, FCER1A and CAMP were selected as candidate genes. However, the results of real-time PCR showed that the expression levels of FCER1A, IL4 and HDC in the cough variant asthma group were significantly different from those in the other two groups (p < 0.05). CONCLUSIONS The pathogenesis of CVA and CA may be related to genes such as FCER1A, HDC and IL4. Further studies incorporating a larger sample size should be conducted to find more candidate genes and mechanisms.
Collapse
Affiliation(s)
- Guanghong Zhou
- Department of Respiratory and Critical Care Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, Sichuan, P. R. China.,North Sichuan Medical College, Nanchong, Sichuan, P. R. China
| | - Qingcui Zeng
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, Sichuan, P. R. China.,Department of Geriatric intensive care unit, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
| | - Wei Wei
- Department of Geriatric intensive care unit, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China.,Department of Respiratory and Critical Care Medicine, Anyue County People's Hospital, Anyue, Sichuan, P. R. China
| | - Hong Teng
- Department of Respiratory and Critical Care Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
| | - Chuntao Liu
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Zhongwei Zhou
- Department of Respiratory and Critical Care Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
| | - Binmiao Liang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Huaicong Long
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, Sichuan, P. R. China.,Department of Geriatric intensive care unit, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
| |
Collapse
|
33
|
Blank U, Huang H, Kawakami T. The high affinity IgE receptor: a signaling update. Curr Opin Immunol 2021; 72:51-58. [PMID: 33838574 DOI: 10.1016/j.coi.2021.03.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/22/2021] [Indexed: 01/01/2023]
Abstract
Here we update receptor proximal and distant signaling events of the mast cell high affinity IgE receptor (FcεRI) launching immediate type I hypersensitivity and an inflammatory cytokine-chemokine cascade. Different physiologic antigen concentrations, their affinity, and valency for the IgE ligand produce distinct intracellular signaling events with different outcomes. Investigating mast cell degranulation has revealed a complex molecular machinery that relays proximal signaling to cytoskeletal reorganization, granule transport and membrane fusion. Several new phosphorylation- and calcium-responsive effectors have been described. FcεRI signaling also promotes de novo gene transcription. Recent progress has identified enhancers at genes that are upregulated in mast cells after stimulation through FcεRI using next generation sequencing methods. Enhancers at genes that respond to antigenic stimulation in human mast cells revealed Ca2+-dependency. Stimulation-responsive super enhancers in mouse mast cells have also been identified. Mast cell lineage-determining transcription factor GATA2 primes these enhancers to respond to antigenic stimulation.
Collapse
Affiliation(s)
- Ulrich Blank
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Laboratoire d'Excellence Inflamex, Paris, France.
| | - Hua Huang
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO 80206, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Toshiaki Kawakami
- Laboratory of Allergic Diseases, Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Department of Dermatology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
34
|
Cai R, Dong Y, Fang M, Fan Y, Cheng Z, Zhou Y, Gao J, Han F, Guo C, Ma X. Genome-Wide Association Identifies Risk Pathways for SAPHO Syndrome. Front Cell Dev Biol 2021; 9:643644. [PMID: 33816493 PMCID: PMC8012550 DOI: 10.3389/fcell.2021.643644] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/18/2021] [Indexed: 11/13/2022] Open
Abstract
SAPHO syndrome is a rare chronic inflammatory disease which is characterized by the comprehensive manifestations of bone, joint, and skin. However, little is known about the pathogenesis of SAPHO syndrome. A genome-wide association study (GWAS) of 49 patients and 121 control subjects have primarily focused on identification of common genetic variants associated with SAPHO, the data were analyzed by classical multiple logistic regression. Later, GWAS findings were further validated using whole exome sequencing (WES) in 16 patients and 15 controls to identify potentially functional pathways involved in SAPHO pathogenesis. In general, 40588 SNPs in genomic regions were associated with P < 0.05 after filter process, only 9 SNPs meet the expected cut-off P-value, however, none of them had association with SAPHO syndrome based on published literatures. And then, 15 pathways were found involved in SAPHO pathogenesis, of them, 6 pathways including osteoclast differentiation, bacterial invasion of epithelial cells, et al., had strong association with skin, osteoarticular manifestations of SAPHO or inflammatory reaction based published research. This study identified aberrant osteoclast differentiation and other pathways were involved in SAPHO syndrome. This finding may give insight into the understanding of pathogenic genes of SAPHO and provide the basis for SAPHO research and treatment.
Collapse
Affiliation(s)
- Ruikun Cai
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Yichao Dong
- National Research Institute for Family Planning, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Mingxia Fang
- National Research Institute for Family Planning, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Yuxuan Fan
- National Research Institute for Family Planning, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Zian Cheng
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Yue Zhou
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Jianen Gao
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Feifei Han
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Changlong Guo
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Xu Ma
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| |
Collapse
|
35
|
Silva VC, Silva AMGS, Basílio JAD, Xavier JA, do Nascimento TG, Naal RMZG, del Lama MP, Leonelo LAD, Mergulhão NLON, Maranhão FCA, Silva DMW, Owen R, Duarte IFB, Bulhões LCG, Basílio ID, Goulart MOF. New Insights for Red Propolis of Alagoas-Chemical Constituents, Topical Membrane Formulations and Their Physicochemical and Biological Properties. Molecules 2020; 25:E5811. [PMID: 33317120 PMCID: PMC7763695 DOI: 10.3390/molecules25245811] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/15/2022] Open
Abstract
The main objectives of this study were to evaluate the chemical constitution and allergenic potential of red propolis extract (RPE). They were evaluated, using high performance liquid chromatography (HPLC) and the release of β-hexosaminidase, respectively. A plethora of biologically active polyphenols and the absence of allergic responses were evinced. RPE inhibited the release of β-hexosaminidase, suggesting that the extract does not stimulate allergic responses. Additionally, the physicochemical properties and antibacterial activity of hydrogel membranes loaded with RPE were analyzed. Bio-polymeric hydrogel membranes (M) were obtained using 5% carboxymethylcellulose (M1 and M2), 1.0% of citric acid (M3) and 10% RPE (for all). Their characterization was performed using thermal analysis, Fourier transform infrared (FTIR), total phenolic content, phenol release test and, antioxidant activity through 2,2-diphenyl-1-picrylhydrazyl radical (DPPH) and Ferric Reducing Antioxidant Power (FRAP). The latter appointed to the similar antioxidant capacity of the M1, M2 and M3. The degradation profiles showed higher thermostability to M3, followed by M2 and M1. The incorporation of RPE into the matrices and the crosslinking of M3 were evinced by FTIR. There were differences in the release of phenolic compounds, with a higher release related to M1 and lower in the strongly crosslinked M3. The degradation profiles showed higher thermostability to M3, followed by M2 and M1. The antibacterial activity of the membranes was determined using the disc diffusion assay, in comparison with controls, obtained in the same way, without RPE. The membranes elicited antibacterial activity against Staphylococcus aureus and Staphylococcus epidermidis, with superior performance over M3. The hydrogel membranes loaded with RPE promote a physical barrier against bacterial skin infections and may be applied in the wound healing process.
Collapse
Affiliation(s)
- Valdemir C. Silva
- Laboratory of Pharmaceutical Technology, Research Program Post-Graduation in Pharmaceutical Sciences, Institute of Pharmaceutical Sciences, Federal University of Alagoas (UFAL), Maceió, Alagoas 57072-970, Brazil; (V.C.S.); (A.M.G.S.S.); (T.G.d.N.); (N.L.O.N.M.); (I.F.B.D.); (L.C.G.B.)
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Alagoas 57072-970, Brazil; (J.A.D.B.); (J.A.X.)
| | - Abiane M. G. S. Silva
- Laboratory of Pharmaceutical Technology, Research Program Post-Graduation in Pharmaceutical Sciences, Institute of Pharmaceutical Sciences, Federal University of Alagoas (UFAL), Maceió, Alagoas 57072-970, Brazil; (V.C.S.); (A.M.G.S.S.); (T.G.d.N.); (N.L.O.N.M.); (I.F.B.D.); (L.C.G.B.)
| | - Jacqueline A. D. Basílio
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Alagoas 57072-970, Brazil; (J.A.D.B.); (J.A.X.)
| | - Jadriane A. Xavier
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Alagoas 57072-970, Brazil; (J.A.D.B.); (J.A.X.)
| | - Ticiano G. do Nascimento
- Laboratory of Pharmaceutical Technology, Research Program Post-Graduation in Pharmaceutical Sciences, Institute of Pharmaceutical Sciences, Federal University of Alagoas (UFAL), Maceió, Alagoas 57072-970, Brazil; (V.C.S.); (A.M.G.S.S.); (T.G.d.N.); (N.L.O.N.M.); (I.F.B.D.); (L.C.G.B.)
| | - Rose M. Z. G. Naal
- Department of BioMolecular Sciences, Faculty of Pharmaceutical Sciences of Ribeirão Preto. Avenida do Café, s/n, Ribeirão Preto, São Paulo 14040-903, Brazil; (R.M.Z.G.N.); (M.P.d.L.); (L.A.D.L.)
| | - Maria Perpetua del Lama
- Department of BioMolecular Sciences, Faculty of Pharmaceutical Sciences of Ribeirão Preto. Avenida do Café, s/n, Ribeirão Preto, São Paulo 14040-903, Brazil; (R.M.Z.G.N.); (M.P.d.L.); (L.A.D.L.)
| | - Laila A. D. Leonelo
- Department of BioMolecular Sciences, Faculty of Pharmaceutical Sciences of Ribeirão Preto. Avenida do Café, s/n, Ribeirão Preto, São Paulo 14040-903, Brazil; (R.M.Z.G.N.); (M.P.d.L.); (L.A.D.L.)
| | - Naianny L. O. N. Mergulhão
- Laboratory of Pharmaceutical Technology, Research Program Post-Graduation in Pharmaceutical Sciences, Institute of Pharmaceutical Sciences, Federal University of Alagoas (UFAL), Maceió, Alagoas 57072-970, Brazil; (V.C.S.); (A.M.G.S.S.); (T.G.d.N.); (N.L.O.N.M.); (I.F.B.D.); (L.C.G.B.)
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Alagoas 57072-970, Brazil; (J.A.D.B.); (J.A.X.)
| | - Fernanda C. A. Maranhão
- Institute of Biological Science and Health, Federal University of Alagoas, Maceió, Alagoas 57072-970, Brazil; (F.C.A.M.); (D.M.W.S.)
| | - Denise M. W. Silva
- Institute of Biological Science and Health, Federal University of Alagoas, Maceió, Alagoas 57072-970, Brazil; (F.C.A.M.); (D.M.W.S.)
| | - Robert Owen
- Division of Preventive Oncology, German Cancer Research Center, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany;
| | - Ilza F. B. Duarte
- Laboratory of Pharmaceutical Technology, Research Program Post-Graduation in Pharmaceutical Sciences, Institute of Pharmaceutical Sciences, Federal University of Alagoas (UFAL), Maceió, Alagoas 57072-970, Brazil; (V.C.S.); (A.M.G.S.S.); (T.G.d.N.); (N.L.O.N.M.); (I.F.B.D.); (L.C.G.B.)
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Alagoas 57072-970, Brazil; (J.A.D.B.); (J.A.X.)
| | - Laisa C. G. Bulhões
- Laboratory of Pharmaceutical Technology, Research Program Post-Graduation in Pharmaceutical Sciences, Institute of Pharmaceutical Sciences, Federal University of Alagoas (UFAL), Maceió, Alagoas 57072-970, Brazil; (V.C.S.); (A.M.G.S.S.); (T.G.d.N.); (N.L.O.N.M.); (I.F.B.D.); (L.C.G.B.)
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Alagoas 57072-970, Brazil; (J.A.D.B.); (J.A.X.)
| | - Irinaldo D. Basílio
- Laboratory of Pharmaceutical Technology, Research Program Post-Graduation in Pharmaceutical Sciences, Institute of Pharmaceutical Sciences, Federal University of Alagoas (UFAL), Maceió, Alagoas 57072-970, Brazil; (V.C.S.); (A.M.G.S.S.); (T.G.d.N.); (N.L.O.N.M.); (I.F.B.D.); (L.C.G.B.)
| | - Marília O. F. Goulart
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Alagoas 57072-970, Brazil; (J.A.D.B.); (J.A.X.)
| |
Collapse
|
36
|
PGE2 deficiency predisposes to anaphylaxis by causing mast cell hyperresponsiveness. J Allergy Clin Immunol 2020; 146:1387-1396.e13. [DOI: 10.1016/j.jaci.2020.03.046] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/22/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022]
|
37
|
Lee D, Park YH, Lee JE, Kim HS, Min KY, Jo MG, Kim HS, Choi WS, Kim YM. Dasatinib Inhibits Lyn and Fyn Src-Family Kinases in Mast Cells to Suppress Type I Hypersensitivity in Mice. Biomol Ther (Seoul) 2020; 28:456-464. [PMID: 32268657 PMCID: PMC7457176 DOI: 10.4062/biomolther.2020.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 12/21/2022] Open
Abstract
Mast cells (MCs) are systemically distributed and secrete several allergic mediators such as histamine and leukotrienes to cause type I hypersensitivity. Dasatinib is a type of anti-cancer agent and it has also been reported to inhibit human basophils. However, dasatinib has not been reported for its inhibitory effects on MCs or type I hypersensitivity in mice. In this study, we examined the inhibitory effect of dasatinib on MCs and MC-mediated allergic response in vitro and in vivo. in vitro, dasatinib inhibited the degranulation of MCs by antigen stimulation in a dose-dependent manner (IC50, ~34 nM for RBL-2H3 cells; ~52 nM for BMMCs) without any cytotoxicity. It also suppressed the secretion of inflammatory cytokines IL-4 and TNF-α by antigen stimulation. Furthermore, dasatinib inhibited MC-mediated passive cutaneous anaphylaxis (PCA) in mice (ED50, ~29 mg/kg). Notably, dasatinib significantly suppressed the degranulation of MCs in the ear tissue. As the mechanism of its effect, dasatinib inhibited the activation of Syk and Syk-mediated downstream signaling proteins, LAT, PLCγ1, and three typical MAP kinases (Erk1/2, JNK, and p38), which are essential for the activation of MCs. Interestingly, in vitro tyrosine kinase assay, dasatinib directly inhibited the activities of Lyn and Fyn, the upstream tyrosine kinases of Syk in MCs. Taken together, dasatinib suppresses MCs and PCA in vitro and in vivo through the inhibition of Lyn and Fyn Src-family kinases. Therefore, we suggest the possibility of repositioning the anti-cancer drug dasatinib as a treatment for various MC-mediated type I hypersensitive diseases.
Collapse
Affiliation(s)
- Dajeong Lee
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Young Hwan Park
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Ji Eon Lee
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea.,College of Pharmacy, Duksung Women's University, Seoul 01369, Republic of Korea
| | - Hyuk Soon Kim
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Keun Young Min
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Min Geun Jo
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Hyung Sik Kim
- Division of Toxicology, College of Pharmacy, Sungkyunkwan University, Suwon 6419, Republic of Korea
| | - Wahn Soo Choi
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Young Mi Kim
- College of Pharmacy, Duksung Women's University, Seoul 01369, Republic of Korea
| |
Collapse
|
38
|
Kambayashi T, Deshpande DA. The role of diacylglycerol kinases in allergic airway disease. Curr Opin Pharmacol 2020; 51:50-58. [PMID: 32836013 DOI: 10.1016/j.coph.2020.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/28/2022]
Abstract
Asthma is an obstructive inflammatory airway disease. Airway obstruction is mediated by hyperresponsive airway smooth muscle cell contraction, which is induced and compounded by inflammation caused by T lymphocytes. One important signal transduction pathway that is involved in the activation of these cell types involves the generation of a lipid second messenger known as diacylglycerol (DAG). DAG levels are controlled in cells by a negative regulator known as DAG kinase (DGK). In this review, we discuss how the DAG signaling pathway attenuates the pathological function of immune cells and airway smooth muscle cells in allergic airway disease and asthma. Furthermore, we discuss how the enhancement of the DAG signaling pathway through the inhibition of DGK may represent a novel therapeutic strategy for these diseases.
Collapse
Affiliation(s)
- Taku Kambayashi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Deepak A Deshpande
- Center for Translational Medicine, Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
39
|
Hogenkamp A, Ehlers A, Garssen J, Willemsen LEM. Allergy Modulation by N-3 Long Chain Polyunsaturated Fatty Acids and Fat Soluble Nutrients of the Mediterranean Diet. Front Pharmacol 2020; 11:1244. [PMID: 32973501 PMCID: PMC7472571 DOI: 10.3389/fphar.2020.01244] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022] Open
Abstract
The Mediterranean diet, containing valuable nutrients such as n-3 long chain poly-unsaturated fatty acids (LCPUFAs) and other fat-soluble micronutrients, is known for its health promoting and anti-inflammatory effects. Its valuable elements might help in the battle against the rising prevalence of non-communicable diseases (NCD), including the development of allergic diseases and other (chronic) inflammatory diseases. The fat fraction of the Mediterranean diet contains bioactive fatty acids but can also serve as a matrix to dissolve and increase the uptake of fat-soluble vitamins and phytochemicals, such as luteolin, quercetin, resveratrol and lycopene with known immunomodulatory and anti-inflammatory capacities. Especially n-3 LCPUFAs such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) derived from marine oils can target specific receptors or signaling cascades, act as eicosanoid precursors and/or alter membrane fluidity and lipid raft formation, hereby exhibiting anti-inflammatory properties. Beyond n-3 LCPUFAs, fat-soluble vitamins A, D, E, and K1/2 have the potential to affect pro-inflammatory signaling cascades by interacting with receptors or activating/inhibiting signaling proteins or phosphorylation in immune cells (DCs, T-cells, mast cells) involved in allergic sensitization or the elicitation/effector phase of allergic reactions. Moreover, fat-soluble plant-derived phytochemicals can manipulate signaling cascades, mostly by interacting with other receptors or signaling proteins compared to those modified by fat-soluble vitamins, suggesting potential additive or synergistic actions by applying a combination of these nutrients which are all part of the regular Mediterranean diet. Research concerning the effects of phytochemicals such as polyphenols has been hampered due to their poor bio-availability. However, their solubility and uptake are improved by applying them within the dietary fat matrix. Alternatively, they can be prepared for targeted delivery by means of pharmaceutical approaches such as encapsulation within liposomes or even unique nanoparticles. This review illuminates the molecular mechanisms of action and possible immunomodulatory effects of n-3 LCPUFAs and fat-soluble micronutrients from the Mediterranean diet in allergic disease development and allergic inflammation. This will enable us to further appreciate how to make use of the beneficial effects of n-3 LCPUFAs, fat-soluble vitamins and a selection of phytochemicals as active biological components in allergy prevention and/or symptom reduction.
Collapse
Affiliation(s)
- Astrid Hogenkamp
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Anna Ehlers
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Department of Dermatology/Allergology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands.,Global Centre of Excellence Immunology, Danone Nutricia Research B.V., Utrecht, Netherlands
| | - Linette E M Willemsen
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
40
|
Uribe-Querol E, Rosales C. Phagocytosis: Our Current Understanding of a Universal Biological Process. Front Immunol 2020; 11:1066. [PMID: 32582172 PMCID: PMC7280488 DOI: 10.3389/fimmu.2020.01066] [Citation(s) in RCA: 314] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/04/2020] [Indexed: 12/22/2022] Open
Abstract
Phagocytosis is a cellular process for ingesting and eliminating particles larger than 0.5 μm in diameter, including microorganisms, foreign substances, and apoptotic cells. Phagocytosis is found in many types of cells and it is, in consequence an essential process for tissue homeostasis. However, only specialized cells termed professional phagocytes accomplish phagocytosis with high efficiency. Macrophages, neutrophils, monocytes, dendritic cells, and osteoclasts are among these dedicated cells. These professional phagocytes express several phagocytic receptors that activate signaling pathways resulting in phagocytosis. The process of phagocytosis involves several phases: i) detection of the particle to be ingested, ii) activation of the internalization process, iii) formation of a specialized vacuole called phagosome, and iv) maturation of the phagosome to transform it into a phagolysosome. In this review, we present a general view of our current understanding on cells, phagocytic receptors and phases involved in phagocytosis.
Collapse
Affiliation(s)
- Eileen Uribe-Querol
- División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
41
|
Rodríguez-López GM, Soria-Castro R, Campillo-Navarro M, Pérez-Tapia SM, Flores-Borja F, Wong-Baeza I, Muñoz-Cruz S, López-Santiago R, Estrada-Parra S, Estrada-García I, Chávez-Blanco AD, Chacón-Salinas R. The histone deacetylase inhibitor valproic acid attenuates phospholipase Cγ2 and IgE-mediated mast cell activation. J Leukoc Biol 2020; 108:859-866. [PMID: 32480423 DOI: 10.1002/jlb.3ab0320-547rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/30/2020] [Accepted: 04/02/2020] [Indexed: 12/25/2022] Open
Abstract
Mast cell activation through the high-affinity IgE receptor (FcεRI) plays a central role in allergic reactions. FcεRI-mediated activation triggers multiple signaling pathways leading to degranulation and synthesis of different inflammatory mediators. IgE-mediated mast cell activation can be modulated by different molecules, including several drugs. Herein, we investigated the immunomodulatory activity of the histone deacetylase inhibitor valproic acid (VPA) on IgE-mediated mast cell activation. To this end, bone marrow-derived mast cells (BMMC) were sensitized with IgE and treated with VPA followed by FcεRI cross-linking. The results indicated that VPA reduced mast cell IgE-dependent degranulation and cytokine release. VPA also induced a significant reduction in the cell surface expression of FcεRI and CD117, but not other mast cell surface molecules. Interestingly, VPA treatment inhibited the phosphorylation of PLCγ2, a key signaling molecule involved in IgE-mediated degranulation and cytokine secretion. However, VPA did not affect the phosphorylation of other key components of the FcεRI signaling pathway, such as Syk, Akt, ERK1/2, or p38. Altogether, our data demonstrate that VPA affects PLCγ2 phosphorylation, which in turn decreases IgE-mediated mast cell activation. These results suggest that VPA might be a key modulator of allergic reactions and might be a promising therapeutic candidate.
Collapse
Affiliation(s)
- Gloria Mariana Rodríguez-López
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, ENCB-IPN, Mexico City, Mexico
| | - Rodolfo Soria-Castro
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, ENCB-IPN, Mexico City, Mexico
| | - Marcia Campillo-Navarro
- Laboratorio de Inmunología Integrativa, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Sonia Mayra Pérez-Tapia
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, ENCB-IPN, Mexico City, Mexico.,Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, ENCB-IPN, Mexico City, Mexico
| | - Fabián Flores-Borja
- Centre for Immunobiology and Regenerative Medicine, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Isabel Wong-Baeza
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, ENCB-IPN, Mexico City, Mexico
| | - Samira Muñoz-Cruz
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Hospital de Pediatría, Centro Médico Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Rubén López-Santiago
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, ENCB-IPN, Mexico City, Mexico
| | - Sergio Estrada-Parra
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, ENCB-IPN, Mexico City, Mexico
| | - Iris Estrada-García
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, ENCB-IPN, Mexico City, Mexico
| | | | - Rommel Chacón-Salinas
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, ENCB-IPN, Mexico City, Mexico
| |
Collapse
|
42
|
Loratadine, an antihistamine drug, exhibits anti-inflammatory activity through suppression of the NF- kB pathway. Biochem Pharmacol 2020; 177:113949. [PMID: 32251678 DOI: 10.1016/j.bcp.2020.113949] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/01/2020] [Indexed: 12/15/2022]
Abstract
Loratadine is an antihistamine drug that shows promise as an anti-inflammatory drug, but supportive studies are lacking. We elucidated the effects and mechanisms by which loratadine inhibits inflammatory responses. Molecular components were evaluated in macrophages by nitric oxide assay, polymerase chain reaction, luciferase assay, immunoblotting, overexpression strategies and cellular thermal shift assay. At the molecular level, loratadine reduced the levels of nitric oxide, iNOS, IL-1β, TNF-α, IL-6, and COX-2 in RAW264.7 cells treated with lipopolysaccharide. Loratadine also specifically inhibited the NF-kB pathway, targeting the Syk and Src proteins. Furthermore, loratadine bound Src in the bridge between SH2 and SH3, and bound Syk in the protein tyrosine kinase domain. The NF-kB signaling pathway was assessed along with putative binding sites through a docking approach. The anti-inflammatory effect of loratadine was tested using mouse models of gastritis, hepatitis, colitis, and peritonitis. Stomach tissue histopathology, liver morphology, and colon length in the loratadine group were improved over the group without loratadine treatment. Taken together, loratadine inhibited the inflammatory response through the NF-kB pathway by binding with the Syk and Src proteins.
Collapse
|
43
|
Rakhmanova V, Park S, Lee S, Kim YH, Shin J. 3-Benzyl-5-((2-nitrophenoxy) methyl)-dihydrofuran-2(3H)-one suppresses FcεRI-mediated mast cell degranulation via the inhibition of mTORC2-Akt signaling. Biochem Biophys Res Commun 2020; 521:72-76. [PMID: 31629474 DOI: 10.1016/j.bbrc.2019.10.075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022]
Abstract
Mast cells express high-affinity IgE receptor (FcεRI) on their surface, cross-linking of which leads to the immediate release of proinflammatory mediators such as histamine but also late-phase cytokine secretion, which are central to the pathogenesis of allergic diseases. Despite the growing evidences that mammalian target of rapamycin (mTOR) plays important roles in the immune system, it is still unclear how mTOR signaling regulates mast cell function. In this study, we investigated the effects of 3-benzyl-5-((2-nitrophenoxy) methyl)-dihydrofuran-2(3H)-one (3BDO) as an mTOR agonist on FcεRI-mediated allergic responses of mast cells. Our data showed that administration of 3BDO decreased β-hexosaminidase, interleukin 6 (IL-6), and tumor necrosis factor-α (TNF-α) release in murine bone marrow-derived mast cells (BMMCs) after FcεRI cross-linking, which was associated with an increase in mTOR complex 1 (mTORC1) signaling but a decrease in activation of Erk1/2, Jnk, and mTORC2-Akt. In addition, we found that a specific Akt agonist, SC79, is able to fully restore the decrease of β-hexosaminidase release in 3BDO-treated BMMCs but has no effect on IL-6 release in these cells, suggesting that 3BDO negatively regulates FcεRI-mediated degranulation and cytokine release through differential mechanisms in mast cells. The present data demonstrate that proper activation of mTORC1 is crucial for mast cell effector function, suggesting the applicability of the mTORC1 activator as a useful therapeutic agent in mast cell-related diseases.
Collapse
Affiliation(s)
- Valeriya Rakhmanova
- Department of Microbiology, Inha University School of Medicine, Incheon, 22212, South Korea
| | - Sukyoung Park
- Department of Microbiology, Inha University School of Medicine, Incheon, 22212, South Korea
| | - Sungwook Lee
- Division of Tumor Immunology, National Cancer Center, Goyang, 10408, South Korea
| | - Young Hyo Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Inha University School of Medicine, Incheon, 22212, South Korea
| | - Jinwook Shin
- Department of Microbiology, Inha University School of Medicine, Incheon, 22212, South Korea.
| |
Collapse
|
44
|
Pivetta TP, Silva LB, Kawakami CM, Araújo MM, Del Lama MPF, Naal RMZ, Maria-Engler SS, Gaspar LR, Marcato PD. Topical formulation of quercetin encapsulated in natural lipid nanocarriers: Evaluation of biological properties and phototoxic effect. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101148] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
45
|
Yablonski D. Bridging the Gap: Modulatory Roles of the Grb2-Family Adaptor, Gads, in Cellular and Allergic Immune Responses. Front Immunol 2019; 10:1704. [PMID: 31402911 PMCID: PMC6669380 DOI: 10.3389/fimmu.2019.01704] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/08/2019] [Indexed: 01/07/2023] Open
Abstract
Antigen receptor signaling pathways are organized by adaptor proteins. Three adaptors, LAT, Gads, and SLP-76, form a heterotrimeric complex that mediates signaling by the T cell antigen receptor (TCR) and by the mast cell high affinity receptor for IgE (FcεRI). In both pathways, antigen recognition triggers tyrosine phosphorylation of LAT and SLP-76. The recruitment of SLP-76 to phospho-LAT is bridged by Gads, a Grb2 family adaptor composed of two SH3 domains flanking a central SH2 domain and an unstructured linker region. The LAT-Gads-SLP-76 complex is further incorporated into larger microclusters that mediate antigen receptor signaling. Gads is positively regulated by dimerization, which promotes its cooperative binding to LAT. Negative regulation occurs via phosphorylation or caspase-mediated cleavage of the linker region of Gads. FcεRI-mediated mast cell activation is profoundly impaired in LAT- Gads- or SLP-76-deficient mice. Unexpectedly, the thymic developmental phenotype of Gads-deficient mice is much milder than the phenotype of LAT- or SLP-76-deficient mice. This distinction suggests that Gads is not absolutely required for TCR signaling, but may modulate its sensitivity, or regulate a particular branch of the TCR signaling pathway; indeed, the phenotypic similarity of Gads- and Itk-deficient mice suggests a functional connection between Gads and Itk. Additional Gads binding partners include costimulatory proteins such as CD28 and CD6, adaptors such as Shc, ubiquitin regulatory proteins such as USP8 and AMSH, and kinases such as HPK1 and BCR-ABL, but the functional implications of these interactions are not yet fully understood. No interacting proteins or function have been ascribed to the evolutionarily conserved N-terminal SH3 of Gads. Here we explore the biochemical and functional properties of Gads, and its role in regulating allergy, T cell development and T-cell mediated immunity.
Collapse
Affiliation(s)
- Deborah Yablonski
- The Immune Cell Signaling Lab, Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
46
|
Arakawa T, Okubo H, Mae M, Okuno T, Ogino H, Ueno H. Seleno-L-Methionine Suppresses Immunoglobulin E-Mediated Allergic Response in RBL-2H3 Cells. Biol Pharm Bull 2019; 42:1179-1184. [PMID: 30982787 DOI: 10.1248/bpb.b19-00098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The effect of seleno-L-methionine (SeMet) on immunoglobulin (Ig) E-mediated allergic responses were investigated using rat basophilic leukemia RBL-2H3 cells. Cells were first treated with or without SeMet, sensitized with anti-dinitrophenyl IgE and stimulated with the antigen dinitrophenyl-human serum albumin, before the measurement of degranulation, calcium mobilization, mRNA expression and protein secretion of interleukin (IL)-4 and tumor necrosis factor (TNF)-α, and phosphorylation of spleen tyrosine kinase (Syk), Akt, and mitogen-activated protein kinases (MAPKs). The antigen-induced β-hexosaminidase release, a degranulation marker, was significantly inhibited by SeMet treatment. SeMet also significantly suppressed antigen-induced calcium mobilization. Antigen-induced increases in the mRNA expression and protein secretion of IL-4 and TNF-α were both significantly attenuated by SeMet treatment. In addition, SeMet significantly suppressed antigen-induced phosphorylation of Syk, Akt, and MAPKs. These results demonstrate that SeMet suppresses antigen-induced degranulation, and mRNA expression and protein secretion of IL-4 and TNF-α, and inhibits antigen-induced mobilization of calcium and activation of Syk, Akt, and MAPKs. Our study provides valuable information that may be useful in the prevention and treatment of allergic diseases.
Collapse
Affiliation(s)
- Tomohiro Arakawa
- Department of Public Health & Preventive Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University
| | - Haruki Okubo
- Department of Public Health & Preventive Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University
| | - Midori Mae
- Department of Public Health & Preventive Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University
| | - Tomofumi Okuno
- Department of Public Health & Preventive Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University
| | - Hirofumi Ogino
- Department of Public Health & Preventive Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University
| | - Hitoshi Ueno
- Department of Public Health & Preventive Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University
| |
Collapse
|
47
|
Gomez G. Current Strategies to Inhibit High Affinity FcεRI-Mediated Signaling for the Treatment of Allergic Disease. Front Immunol 2019; 10:175. [PMID: 30792720 PMCID: PMC6374298 DOI: 10.3389/fimmu.2019.00175] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/21/2019] [Indexed: 12/11/2022] Open
Abstract
Allergies and asthma are a major cause of chronic disease whose prevalence has been on the rise. Allergic disease including seasonal rhinitis, atopic dermatitis, urticaria, anaphylaxis, and asthma, are associated with activation of tissue-resident mast cells and circulating basophils. Although these cells can be activated in different ways, allergic reactions are normally associated with the crosslinking of the high affinity Fc receptor for Immunoglobulin E, FcεRI, with multivalent antigen. Inflammatory mediators released from cytoplasmic granules, or biosynthesized de novo, following FcεRI crosslinking induce immediate hypersensitivity reactions, including life-threatening anaphylaxis, and contribute to prolonged inflammation leading to chronic diseases like asthma. Thus, inappropriate or unregulated activation of mast cells and basophils through antigenic crosslinking of FcεRI can have deleterious, sometimes deadly, consequences. Accordingly, FcεRI has emerged as a viable target for the development of biologics that act to inhibit or attenuate the activation of mast cells and basophils. At the forefront of these strategies are (1) Anti-IgE monoclonal antibody, namely omalizumab, which has the secondary effect of reducing FcεRI surface expression, (2) Designed Ankyrin Repeat Proteins (DARPins), which take advantage of the most common structural motifs in nature involved in protein-protein interactions, to inhibit FcεRI-IgE interactions, and (3) Fusion proteins to co-aggregate FcεRI with the inhibitory FcγRIIb. This review presents the published research studies that support omalizumab, DARPins, and fusion proteins as, arguably, the three most currently viable strategies for inhibiting the expression and activation of the high affinity FcεRI on mast cells and basophils.
Collapse
Affiliation(s)
- Gregorio Gomez
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| |
Collapse
|
48
|
Jiang S, Da Y, Han S, He Y, Che H. Notch ligand Delta-like1 enhances degranulation and cytokine production through a novel Notch/Dok-1/MAPKs pathway in vitro. Immunol Res 2019; 66:87-96. [PMID: 29181775 DOI: 10.1007/s12026-017-8977-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Food allergy includes sensitization phase and effect phase, and effect cells degranulate and secrete cytokines in the effect phase, causing allergic clinical symptoms. We have demonstrated that Notch signaling plays an important role in the sensitization phase, but its role in effect phases still remains unclear. In this study, we investigated the role of Notch signaling in degranulation and cytokine production of the effect phase response. A RBL-2H3 cell model was used and Notch signaling was induced by priming with Notch ligands. Our results showed after priming with Notch ligand, Delta-like1(Dll1)-Fc, β-hexosaminidase release, and cytokines production, including TGF-β, IL-1β, IL-4, IL-6, and IL-13, were increased significantly, and the enhancement was abolished after DAPT treatment, a γ-secretase inhibitor, indicating that Dll1 Notch signaling enhanced RBL-2H3 cell degranulation and cytokine production. Western blot analysis showed that Dll1 Notch signaling augmented high-affinity IgE receptors-mediated phosphorylation of MAPKs through suppressing the expression of downstream tyrosine kinases 1 (Dok-1). Besides, a passive systemic anaphylaxis mouse model was used to confirm the role of Notch signaling. And our data showed that allergic clinical features of mice were alleviated, and the level of degranulation was decreased significantly after inhibiting Notch signaling in vivo. Therefore, we demonstrated Notch ligand Dll1 enhanced RBL-2H3 cell degranulation and cytokine production through a novel Notch/Dok-1/MAPKs pathway, suggesting Notch signaling played a key role in the effect phase of food allergy.
Collapse
Affiliation(s)
- Songsong Jiang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua Donglu, Haidian District, Beijing, People's Republic of China
| | - Yifan Da
- College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua Donglu, Haidian District, Beijing, People's Republic of China
| | - Shiwen Han
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua Donglu, Haidian District, Beijing, People's Republic of China
| | - Yahong He
- College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua Donglu, Haidian District, Beijing, People's Republic of China
| | - Huilian Che
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua Donglu, Haidian District, Beijing, People's Republic of China.
| |
Collapse
|
49
|
Yablonski D. Bridging the Gap: Modulatory Roles of the Grb2-Family Adaptor, Gads, in Cellular and Allergic Immune Responses. Front Immunol 2019; 10:1704. [PMID: 31402911 DOI: 10.3389/fimmu.2019.01704/full] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/08/2019] [Indexed: 05/22/2023] Open
Abstract
Antigen receptor signaling pathways are organized by adaptor proteins. Three adaptors, LAT, Gads, and SLP-76, form a heterotrimeric complex that mediates signaling by the T cell antigen receptor (TCR) and by the mast cell high affinity receptor for IgE (FcεRI). In both pathways, antigen recognition triggers tyrosine phosphorylation of LAT and SLP-76. The recruitment of SLP-76 to phospho-LAT is bridged by Gads, a Grb2 family adaptor composed of two SH3 domains flanking a central SH2 domain and an unstructured linker region. The LAT-Gads-SLP-76 complex is further incorporated into larger microclusters that mediate antigen receptor signaling. Gads is positively regulated by dimerization, which promotes its cooperative binding to LAT. Negative regulation occurs via phosphorylation or caspase-mediated cleavage of the linker region of Gads. FcεRI-mediated mast cell activation is profoundly impaired in LAT- Gads- or SLP-76-deficient mice. Unexpectedly, the thymic developmental phenotype of Gads-deficient mice is much milder than the phenotype of LAT- or SLP-76-deficient mice. This distinction suggests that Gads is not absolutely required for TCR signaling, but may modulate its sensitivity, or regulate a particular branch of the TCR signaling pathway; indeed, the phenotypic similarity of Gads- and Itk-deficient mice suggests a functional connection between Gads and Itk. Additional Gads binding partners include costimulatory proteins such as CD28 and CD6, adaptors such as Shc, ubiquitin regulatory proteins such as USP8 and AMSH, and kinases such as HPK1 and BCR-ABL, but the functional implications of these interactions are not yet fully understood. No interacting proteins or function have been ascribed to the evolutionarily conserved N-terminal SH3 of Gads. Here we explore the biochemical and functional properties of Gads, and its role in regulating allergy, T cell development and T-cell mediated immunity.
Collapse
Affiliation(s)
- Deborah Yablonski
- The Immune Cell Signaling Lab, Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
50
|
Shi LB, Xu HP, Wu YJ, Li X, Gao JY, Chen HB. The effects of imidacloprid combined with endosulfan on IgE-mediated mouse bone marrow-derived mast cell degranulation and anaphylaxis. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2018; 148:159-165. [PMID: 29891367 DOI: 10.1016/j.pestbp.2018.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 03/14/2018] [Accepted: 04/23/2018] [Indexed: 05/07/2023]
Abstract
Low levels of endosulfan are known to stimulate mast cells to release allergic mediators, while imidacloprid can inhibit IgE-mediated mast cell degranulation. However, little information about the effects of both pesticides together on mast cell degranulation is available. To measure the effects, IgE-activated mouse bone marrow-derived mast cells (BMMCs) were treated with imidacloprid and endosulfan, individually, and simultaneously at equi-molar concentrations in tenfold steps ranging from 10-4 to 10-11 M, followed by measuring several allergy-related parameters expressed in BMMCs: the mediator production and influx of Ca2+, the phosphorylation content of NF-κB in the FcεRI signaling pathway. Then, the effects of the mixtures on IgE-induced passive systemic anaphylaxis (PSA) of BALB/c was detectded. This study clearly showed that the application of equi-molar mixtures of both pesticides with 10-4-10-5 M significantly inhibited the IgE-mediated mouse bone marrow-derived mast cells degranulation in vitro and 10-4 M of them decreased IgE-mediated PSA in vivo, as the application of imidacloprid at the same concentration alone did. Morever endosulfan alone had no remarkable stimulatory effects on any of the factors measured. In conclusion, simultaneous application of equi-molar concentrations of both pesticides generally showed highly similar responses compared to the responses to imidacloprid alone, suggesting that the effects of the mixture could be solely attributed to the effects of imidacloprid.
Collapse
Affiliation(s)
- Lin-Bo Shi
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China; Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| | - Hua-Ping Xu
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yu-Jie Wu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China; Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| | - Xin Li
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China; School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Jin-Yan Gao
- School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Hong-Bing Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China; Sino-German Joint Research Institute, Nanchang University, Nanchang, China.
| |
Collapse
|