1
|
Liu Z, Bai Z, Chen X, Chen Y, Chen Z, Wang L, He Y, Guo Y. Advances and applications of biosensors in pulmonary hypertension. Respir Res 2025; 26:147. [PMID: 40234824 PMCID: PMC11998464 DOI: 10.1186/s12931-025-03221-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/05/2025] [Indexed: 04/17/2025] Open
Abstract
Pulmonary hypertension (PH) is a serious disease characterized by elevated pulmonary artery pressure, with its prevalence and incidence continuously increasing, posing a threat to the lives of many patients worldwide. Due to the complex etiology of PH and the lack of specificity in clinical manifestations, there is currently a lack of effective and specific methods for early diagnosis in clinical practice. Biosensors hold significant promise for the early detection, therapeutic monitoring, prognostic evaluation, and personalized treatment of PH, owing to their rapid, sensitive, and highly selective characteristics. The rapid development of various types of biosensors, such as electrochemical biosensors, optical biosensors, microfluidic biosensors, and wireless biosensors, combined with the use of nanomaterials, makes the rapid and accurate detection of PH-related biomarkers possible. Despite the broad application prospects of biosensors in the field of PH, challenges remain in terms of sensitivity, selectivity, stability, and regulation. This article reviews the main pathophysiological mechanisms and commonly used biomarkers of PH, the types and principles of biosensors, and summarizes the progress of biosensors in PH research as well as the current challenges, in order to promote further in-depth research and the development of biosensor technology, thereby improving the diagnosis and treatment effects of PH. Clinical trial number: Not applicable.
Collapse
Affiliation(s)
- Zhi Liu
- Graduate Collaborative Training Base of Zhuzhou Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- Department of Cardiovascular Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, No. 116 South Changjiang Road, Zhuzhou, 412007, Hunan, China
| | - Zhuojun Bai
- Department of Laboratory, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, No. 116 South Changjiang Road, Zhuzhou, 412007, Hunan, China
| | - Xiang Chen
- Department of Laboratory, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, No. 116 South Changjiang Road, Zhuzhou, 412007, Hunan, China
| | - Yajie Chen
- Graduate Collaborative Training Base of Zhuzhou Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhu Chen
- Graduate Collaborative Training Base of Zhuzhou Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Li Wang
- Department of Laboratory, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, No. 116 South Changjiang Road, Zhuzhou, 412007, Hunan, China.
| | - Yi He
- Department of Cardiovascular Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, No. 116 South Changjiang Road, Zhuzhou, 412007, Hunan, China.
| | - Yuan Guo
- Graduate Collaborative Training Base of Zhuzhou Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- Department of Cardiovascular Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, No. 116 South Changjiang Road, Zhuzhou, 412007, Hunan, China.
| |
Collapse
|
2
|
Hao R, Zhao M, Tayyab M, Lin Z, Zhang Y. The mucosal immunity in crustaceans: Inferences from other species. FISH & SHELLFISH IMMUNOLOGY 2024; 152:109785. [PMID: 39053584 DOI: 10.1016/j.fsi.2024.109785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/10/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024]
Abstract
Crustaceans such as shrimps and crabs, hold significant ecological significance and substantial economic value within marine ecosystems. However, their susceptibility to disease outbreaks and pathogenic infections has posed major challenges to production in recent decades. As invertebrate, crustaceans primarily rely on their innate immune system for defense, lacking the adaptive immune system found in vertebrates. Mucosal immunity, acting as the frontline defense against a myriad of pathogenic microorganisms, is a crucial aspect of their immune repertoire. This review synthesizes insights from comparative immunology, highlighting parallels between mucosal immunity in vertebrates and innate immune mechanisms in invertebrates. Despite lacking classical adaptive immunity, invertebrates, including crustaceans, exhibit immune memory and rely on inherent "innate immunity factors" to combat invading pathogens. Drawing on parallels from mammalian and piscine systems, this paper meticulously explores the complex role of mucosal immunity in regulating immune responses in crustaceans. Through the extrapolation from well-studied models like mammals and fish, this review infers the potential mechanisms of mucosal immunity in crustaceans and provides insights for research on mucosal immunity in crustaceans.
Collapse
Affiliation(s)
- Ruixue Hao
- Guangdong Provincial Key Laboratory of Marine Biology and Department of Biology, Shantou University, Shantou, 515063, China
| | - Mingming Zhao
- Guangdong Provincial Key Laboratory of Marine Biology and Department of Biology, Shantou University, Shantou, 515063, China
| | - Muhammad Tayyab
- Guangdong Provincial Key Laboratory of Marine Biology and Department of Biology, Shantou University, Shantou, 515063, China
| | - Zhongyang Lin
- Guangdong Provincial Key Laboratory of Marine Biology and Department of Biology, Shantou University, Shantou, 515063, China.
| | - Yueling Zhang
- Guangdong Provincial Key Laboratory of Marine Biology and Department of Biology, Shantou University, Shantou, 515063, China.
| |
Collapse
|
3
|
Li Z, Hong L, Li Y, Lin X, Chen M, Bu S, Xu S, Zeng Q, Huang Y, Bian L, Zheng J, Gao B, Liu G, He W, Song X, Ye F, Huang L, Jiang S, Shi J, Tang T. Allergic hyper-carcinoembryonic antigen syndrome: A syndrome summarized by case series. SAGE Open Med Case Rep 2024; 12:2050313X241261152. [PMID: 38887262 PMCID: PMC11181888 DOI: 10.1177/2050313x241261152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 05/24/2024] [Indexed: 06/20/2024] Open
Abstract
Allergic respiratory diseases can increase serum carcinoembryonic antigen levels. We report three cases experiencing allergic symptoms that proved refractory to inhaled corticosteroids but exhibited a positive response to long-term treatment with oral corticosteroids. This response was characterized by a synchronous alteration in serum eosinophil counts and carcinoembryonic antigen levels. Immunofluorescence assays indicated localized carcinoembryonic antigen production within eosinophils. In addition, we conducted a systematic review of patients exhibiting similar characteristics on PubMed. After comprehensively reviewing this unique pathophysiological condition, we herein introduced a novel term "Allergic hyper-carcinoembryonic antigen syndrome," defined by the following criteria: (1) recurrent asthmatic attacks; (2) eosinophilia or pulmonary eosinophilic infiltrations accompanied by elevated serum carcinoembryonic antigen levels; (3) pulmonary lesions determined by imaging or biopsy; (4) exclusion of malignancy and infections; and (5) responsive to systemic corticosteroids. Allergic hyper-carcinoembryonic antigen syndrome suggests systemic corticosteroids should be introduced early when managing allergic patients with both eosinophilia and elevated serum carcinoembryonic antigen levels.
Collapse
Affiliation(s)
- Zhaolin Li
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Luna Hong
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yuewei Li
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaoling Lin
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ming Chen
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shiyi Bu
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shuwan Xu
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Qiaojun Zeng
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yijiao Huang
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lijuan Bian
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jianming Zheng
- Cardiovascular Medicine Department, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Boying Gao
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Guirong Liu
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wenman He
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xueming Song
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fengzhan Ye
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Linjie Huang
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shanping Jiang
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jianting Shi
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Tiantian Tang
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
4
|
Wetten PA, Arismendi Sosa AC, Mariani ML, Vargas PM, Michaut MA, Penissi AB. Dehydroleucodine and xanthatin, two natural anti-inflammatory lactones, inhibit mast cell degranulation by affecting the actin cytoskeleton. Cytoskeleton (Hoboken) 2024; 81:215-226. [PMID: 37929805 DOI: 10.1002/cm.21805] [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: 06/26/2023] [Revised: 10/16/2023] [Accepted: 10/22/2023] [Indexed: 11/07/2023]
Abstract
Actin remodeling is a critical regulator of mast cell secretion. In previous work, we have shown that dehydroleucodine and xanthatin, two natural α,β-unsaturated lactones, exhibit anti-inflammatory and mast cell stabilizing properties. Based on this background, this study aimed to determine whether the mast cell stabilizing action of these lactones is associated with changes in the actin cytoskeleton. Rat peritoneal mast cells were preincubated in the presence of dehydroleucodine or xanthatin before incubation with compound 48/80. Comparative studies with sodium cromoglycate and latrunculin B were also made. After treatments, different assays were performed on mast cell samples: β-hexosaminidase release, cell viability studies, quantification of mast cells and their state of degranulation by light microscopy, transmission electron microscopy, and actin staining for microscopy observation. Results showed that dehydroleucodine and xanthatin inhibited mast cell degranulation, evidenced by the inhibition of β-hexosaminidase release and decreased degranulated mast cell percentage. At the same time, both lactones altered the F-actin cytoskeleton in mast cells resulting, similarly to Latrunculin B, in a higher concentration of nuclear F-actin when activated by compound 48/80. For the first time, this study describes the biological properties of dehydroleucodine and xanthatin concerning to the rearrangement of actin filaments during stimulated exocytosis in mast cells. These data have important implications for developing new anti-inflammatory and mast cell stabilizing drugs and for designing new small molecules that may interact with the actin cytoskeleton.
Collapse
Affiliation(s)
- Paula A Wetten
- Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM), Universidad Nacional de Cuyo-CONICET, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | | | - María Laura Mariani
- Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM), Universidad Nacional de Cuyo-CONICET, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Patricia M Vargas
- Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM), Universidad Nacional de Cuyo-CONICET, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Marcela Alejandra Michaut
- Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM), Universidad Nacional de Cuyo-CONICET, Mendoza, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Alicia Beatriz Penissi
- Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM), Universidad Nacional de Cuyo-CONICET, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
5
|
Martínez M, Mariani ML, García C, Ceñal JP, Penissi AB. A one-pot and eco-friendly synthesis of novel β-substituted-α-halomethyl acrylates and the bioactivity of these compounds in an in vitro model of mast cell degranulation induced by pro-inflammatory stimuli. Biomed Pharmacother 2024; 170:116009. [PMID: 38134632 DOI: 10.1016/j.biopha.2023.116009] [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: 08/28/2023] [Revised: 12/03/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
The goal of the present work was to develop novel β-substituted-α-halomethyl acrylates from a methodology in an aqueous phase and to evaluate their bioactivity as potential inhibitors of mast cell activation. Eleven β-substituted-α-halomethyl acrylates were synthesized through a modified Horner-Wadsworth-Emmons reaction. Compound 48/80 and the calcium ionophore A23187 stimulated the release of β-hexosaminidase from mast cells. The effect induced by compound 48/80 was inhibited by compound 5 (320 µM) and compound 9 (160 and 320 µM) without causing cytotoxic effects. The effect induced by A23187 was inhibited by compound 5 (40, 80, 160, and 320 µM) without affecting cell viability. The inhibitory effects exhibited by compounds 5 and 9 were more potent than those of the reference compound sodium cromoglycate at the same concentrations. The biochemical results were consistent with the morphological findings obtained by light and transmission electron microscopy. This study reports, for the first time, that the new synthetic compounds methyl (Z)- 2-bromo-3-(furan-3-yl)acrylate (compound 5) and methyl (E)- 2-bromo-3-(3-bromophenyl)acrylate (compound 9) strongly inhibit mast cell degranulation, without affecting cell viability. The implications of these results are relevant as a basis for developing new anti-inflammatory and mast cell stabilizing drugs.
Collapse
Affiliation(s)
- Maricel Martínez
- Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM, UNCUYO-CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina; Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina; Instituto de Investigaciones en Tecnología Química (INTEQUI-CONICET), Universidad Nacional de San Luis, San Luis, Argentina
| | - María Laura Mariani
- Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM, UNCUYO-CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina; Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Celina García
- Instituto Universitario de Bio-Organica "Antonio Gonzalez", Universidad de La Laguna, La Laguna, Tenerife, Spain
| | - Juan Pedro Ceñal
- Instituto de Investigaciones en Tecnología Química (INTEQUI-CONICET), Universidad Nacional de San Luis, San Luis, Argentina; Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - Alicia Beatriz Penissi
- Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM, UNCUYO-CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina; Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.
| |
Collapse
|
6
|
Wang Y, Fu Y, Li W, Simpson BK, Rui X. Modulation of soy protein immunoreactivity by different matrix structures of lactic acid bacterium-induced soy protein gels: Epitope destruction during in vitro gastroduodenal digestion and absorption. Food Res Int 2023; 173:113281. [PMID: 37803593 DOI: 10.1016/j.foodres.2023.113281] [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: 04/19/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 10/08/2023]
Abstract
Soy allergy is a common health problem. Food structure may change the gastroduodenal digestion and absorption of soy proteins, thus leading to the modulation of the immunoreactivity of soy proteins. In this study, lactic acid bacterium (LAB)-fermented soy protein isolates (FSPIs) were prepared at four concentrations (0.2 %-5.0 %, w/v) to present various matrix structures (nongel, NG; weak gel, WG; medium gel, MG; and firm gel, FG) and subjected to in vitro dynamic gastroduodenal digestion model. The results of sandwich enzyme-linked immunosorbent and human serum IgE binding capacity assays demonstrated that FSPI gels, especially the FSPI-MG/WG digestates obtained at the early and medium stages of duodenal digestion (D-5 and D-30), possessed greater potency in immunoreactivity reduction than FSPI-NG and reduced to 1.9 %-68.3 %. The transepithelial transport study revealed that the immunoreactivity of FSPI-MG/WG D-5 and D-30 digestates decreased through the stimulation of interferon-γ production and the induction of dominant Th1/Th2 differentiation. Peptidomics and bioinformatics analyses illustrated that compared with FSPI-NG, the FSPI-gel structure promoted the epitope degradation of the major allergens glycinin G2/G5, β-conglycinin α/β subunit, P34, lectin, trypsin inhibitor, and basic 7S globulin. Spatial structure analysis showed that FSPI-gel elicited an overall promotion in the degradation of allergen epitopes located in interior and exterior regions and was dominated by α-helix and β-sheet secondary structures, whereas FSPI-MG/WG promoted the degradation of epitopes located in the interior region of glycinin/β-conglycinin and exterior region of P34/basic 7S globulin. This study suggested that the FSPI-gel structure is a promising food matrix for decreasing the allergenic potential of allergenic epitopes during gastroduodenal digestion and provided basic information on the production of hypoallergenic soy products.
Collapse
Affiliation(s)
- Yaqiong Wang
- College of Food Science and Technology, Nanjing Agricultural University, Jiangsu Province, PR China
| | - Yumeng Fu
- College of Food Science and Technology, Nanjing Agricultural University, Jiangsu Province, PR China
| | - Wei Li
- College of Food Science and Technology, Nanjing Agricultural University, Jiangsu Province, PR China
| | - Benjamin K Simpson
- Department of Food Science and Agricultural Chemistry, McGill University, Macdonald, QC, Canada
| | - Xin Rui
- College of Food Science and Technology, Nanjing Agricultural University, Jiangsu Province, PR China.
| |
Collapse
|
7
|
Moriyama H, Endo J. Pathophysiological Involvement of Mast Cells and the Lipid Mediators in Pulmonary Vascular Remodeling. Int J Mol Sci 2023; 24:6619. [PMID: 37047587 PMCID: PMC10094825 DOI: 10.3390/ijms24076619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/24/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Mast cells are responsible for IgE-dependent allergic responses, but they also produce various bioactive mediators and contribute to the pathogenesis of various cardiovascular diseases, including pulmonary hypertension (PH). The importance of lipid mediators in the pathogenesis of PH has become evident in recent years, as exemplified by prostaglandin I2, the most central therapeutic target in pulmonary arterial hypertension. New bioactive lipids other than eicosanoids have also been identified that are associated with the pathogenesis of PH. However, it remains largely unknown how mast cell-derived lipid mediators are involved in pulmonary vascular remodeling. Recently, it has been demonstrated that mast cells produce epoxidized n-3 fatty acid (n-3 epoxides) in a degranulation-independent manner, and that n-3 epoxides produced by mast cells regulate the abnormal activation of pulmonary fibroblasts and suppress the progression of pulmonary vascular remodeling. This review summarizes the role of mast cells and bioactive lipids in the pathogenesis of PH. In addition, we introduce the pathophysiological role and therapeutic potential of n-3 epoxides, a mast cell-derived novel lipid mediator, in the pulmonary vascular remodeling in PH. Further knowledge of mast cells and lipid mediators is expected to lead to the development of innovative therapies targeting pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Hidenori Moriyama
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku 160-8582, Tokyo, Japan
- Department of Cardiology, Tokyo Dental College Ichikawa General Hospital, 5-11-13 Sugano, Ichikawa 272-8513, Chiba, Japan
| | - Jin Endo
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku 160-8582, Tokyo, Japan
| |
Collapse
|
8
|
Omega-3 fatty acid epoxides produced by PAF-AH2 in mast cells regulate pulmonary vascular remodeling. Nat Commun 2022; 13:3013. [PMID: 35641514 PMCID: PMC9156667 DOI: 10.1038/s41467-022-30621-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/03/2022] [Indexed: 02/07/2023] Open
Abstract
Pulmonary hypertension is a fatal rare disease that causes right heart failure by elevated pulmonary arterial resistance. There is an unmet medical need for the development of therapeutics focusing on the pulmonary vascular remodeling. Bioactive lipids produced by perivascular inflammatory cells might modulate the vascular remodeling. Here, we show that ω-3 fatty acid-derived epoxides (ω-3 epoxides) released from mast cells by PAF-AH2, an oxidized phospholipid-selective phospholipase A2, negatively regulate pulmonary hypertension. Genetic deletion of Pafah2 in mice accelerate vascular remodeling, resulting in exacerbation of hypoxic pulmonary hypertension. Treatment with ω-3 epoxides suppresses the lung fibroblast activation by inhibiting TGF-β signaling. In vivo ω-3 epoxides supplementation attenuates the progression of pulmonary hypertension in several animal models. Furthermore, whole-exome sequencing for patients with pulmonary arterial hypertension identifies two candidate pathogenic variants of Pafah2. Our findings support that the PAF-AH2-ω-3 epoxide production axis could be a promising therapeutic target for pulmonary hypertension. Pulmonary hypertension is a fatal disease that causes right heart failure due to pulmonary artery stenosis. Here, the authors find that ω-3 epoxides produced by the phospholipase PAF-AH2 in mast cells regulate pulmonary vascular remodeling.
Collapse
|
9
|
Huang J, Zhang J, Wang X, Jin Z, Zhang P, Su H, Sun X. Effect of Probiotics on Respiratory Tract Allergic Disease and Gut Microbiota. Front Nutr 2022; 9:821900. [PMID: 35295917 PMCID: PMC8920559 DOI: 10.3389/fnut.2022.821900] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
Allergy is a hypersensitivity reaction triggered by specific cell or antibody-mediated immune mechanisms. Allergies have increased in industrialized countries in recent decades. The rise in allergic respiratory diseases such as allergic rhinitis (AR) and allergic asthma (AA) is a potential threat to public health. Searches were conducted using PubMed, Google Scholar and Medline using the following key terms: allergic rhinitis OR asthma AND probiotics, allergic airway inflammation AND immune disorders, probiotics OR gut microbiota AND allergic disease, probiotics AND inflammatory. Studies from all years were included, specifically those published within the last 10 years. Some review articles and their reference lists were searched to identify related articles. The role of microbiota in respiratory allergic diseases has attracted more and more attention. Pieces of evidence suggested that the development of allergic diseases causes a possible imbalance in the composition of the gut microbiota. Compared to colonized mice, germ-free mice exhibit exaggerated allergic airway responses, suggesting that microbial host interactions play an important role in the development of allergic diseases. Probiotics modulate both the innate and adaptive inflammatory immune responses, often used as dietary supplements to provide health benefits in gastrointestinal disorders. Probiotics may serve as immunomodulators and activators of host defense pathways. Besides, oral probiotics can modulate the immune response in the respiratory system. Recently, studies in humans and animals have demonstrated the role of probiotic in RA and AA. To understand the characterization, microbiota, and the potential role of probiotics intervention of AA/AR, this review provides an overview of clinical features of AA and AR, probiotics for the prevention and treatment of AR, AA, changes in gut microbiota, and their mechanisms of action.
Collapse
Affiliation(s)
- Jinli Huang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Juan Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xingzhi Wang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zenghui Jin
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Panpan Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hui Su
- Department of Geratology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xin Sun
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
10
|
Rönnberg E, Boey DZH, Ravindran A, Säfholm J, Orre AC, Al-Ameri M, Adner M, Dahlén SE, Dahlin JS, Nilsson G. Immunoprofiling Reveals Novel Mast Cell Receptors and the Continuous Nature of Human Lung Mast Cell Heterogeneity. Front Immunol 2022; 12:804812. [PMID: 35058936 PMCID: PMC8764255 DOI: 10.3389/fimmu.2021.804812] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/06/2021] [Indexed: 12/12/2022] Open
Abstract
Background Immunohistochemical analysis of granule-associated proteases has revealed that human lung mast cells constitute a heterogeneous population of cells, with distinct subpopulations identified. However, a systematic and comprehensive analysis of cell-surface markers to study human lung mast cell heterogeneity has yet to be performed. Methods Human lung mast cells were obtained from lung lobectomies, and the expression of 332 cell-surface markers was analyzed using flow cytometry and the LEGENDScreen™ kit. Markers that exhibited high variance were selected for additional analyses to reveal whether they were correlated and whether discrete mast cell subpopulations were discernable. Results We identified the expression of 102 surface markers on human lung mast cells, 23 previously not described on mast cells, of which several showed high continuous variation in their expression. Six of these markers were correlated: SUSD2, CD49a, CD326, CD34, CD66 and HLA-DR. The expression of these markers was also correlated with the size and granularity of mast cells. However, no marker produced an expression profile consistent with a bi- or multimodal distribution. Conclusions LEGENDScreen analysis identified more than 100 cell-surface markers on mast cells, including 23 that, to the best of our knowledge, have not been previously described on human mast cells. The comprehensive expression profiling of the 332 surface markers did not identify distinct mast cell subpopulations. Instead, we demonstrate the continuous nature of human lung mast cell heterogeneity.
Collapse
Affiliation(s)
- Elin Rönnberg
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Clinical Immunology and Transfusion Medicine, Stockholm, Sweden.,Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Daryl Zhong Hao Boey
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Clinical Immunology and Transfusion Medicine, Stockholm, Sweden.,Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Avinash Ravindran
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Clinical Immunology and Transfusion Medicine, Stockholm, Sweden.,Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Jesper Säfholm
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden.,Unit for Experimental Asthma and Allergy Research, The Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ann-Charlotte Orre
- Thoracic Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Mamdoh Al-Ameri
- Thoracic Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Mikael Adner
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden.,Unit for Experimental Asthma and Allergy Research, The Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sven-Erik Dahlén
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden.,Unit for Experimental Asthma and Allergy Research, The Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Joakim S Dahlin
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Clinical Immunology and Transfusion Medicine, Stockholm, Sweden.,Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Gunnar Nilsson
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Clinical Immunology and Transfusion Medicine, Stockholm, Sweden.,Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden.,Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
11
|
Tiotiu A, Badi Y, Kermani NZ, Sanak M, Kolmert J, Wheelock CE, Hansbro PM, Dahlén SE, Sterk PJ, Djukanovic R, Guo Y, Mumby S, Adcock IM, Chung KF. Association of Differential Mast Cell Activation to Granulocytic Inflammation in Severe Asthma. Am J Respir Crit Care Med 2021; 205:397-411. [PMID: 34813381 DOI: 10.1164/rccm.202102-0355oc] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Mast cells (MC) play a role in inflammation and both innate and adaptive immunity but their involvement in severe asthma (SA) remains undefined. OBJECTIVE We investigated the phenotypic characteristics of the U-BIOPRED asthma cohort by applying published MC activation signatures to the sputum cell transcriptome. METHODS 84 SA, 20 mild/moderate (MMA) asthma, and 16 non-asthmatic healthy participants were studied. We calculated enrichment scores (ES) for nine MC activation signatures by asthma severity, sputum granulocyte status and three previously-defined sputum molecular phenotypes or transcriptome-associated clusters (TAC1, 2, 3) using gene-set variation analysis. RESULTS MC signatures except unstimulated, repeated FcεR1-stimulated and IFNγ-stimulated were enriched in SA. A FcεR1-IgE-stimulated and a single cell signature from asthmatic bronchial biopsies were highly enriched in eosinophilic asthma and in the TAC1 molecular phenotype. Subjects with a high ES for these signatures had elevated sputum levels of similar genes and pathways. IL-33- and LPS-stimulated MC signatures had greater ES in neutrophilic and mixed granulocytic asthma and in the TAC2 molecular phenotype. These subjects exhibited neutrophil, NF-κB, and IL-1β/TNFα pathway activation. The IFNγ-stimulated signature had the greatest ES in TAC2 and TAC3 that was associated with responses to viral infection. Similar results were obtained in an independent ADEPT asthma cohort. CONCLUSIONS Gene signatures of MC activation allow the detection of SA phenotypes and indicate that MC can be induced to take on distinct transcriptional phenotypes associated with specific clinical phenotypes. IL-33-stimulated MCs signature was associated with severe neutrophilic asthma while IgE-activated MC with an eosinophilic phenotype.
Collapse
Affiliation(s)
- Angelica Tiotiu
- National Heart and Lung Institute Division of Respiratory Science, 228067, London, United Kingdom of Great Britain and Northern Ireland.,University Hospital Centre Nancy, 26920, Nancy, France
| | - Yusef Badi
- National Heart and Lung Institute Division of Respiratory Science, 228067, London, United Kingdom of Great Britain and Northern Ireland
| | | | - Marek Sanak
- Jagiellonian University School of Medicine, Department of Medicine, Kraków, Poland
| | - Johan Kolmert
- Karolinska Institutet, Institute of Environmental Medicine, Stockholm, Sweden
| | - Craig E Wheelock
- Karolinska Institutet, 27106, Medical Biochemistry and Biophysics, Stockholm, Sweden
| | - Philip M Hansbro
- University of Technology Sydney, 1994, Sydney, New South Wales, Australia
| | - Sven-Erik Dahlén
- Karolinska Intitutet, Centre for Allergy Research, Stockholm, Sweden
| | - Peter J Sterk
- University of Amsterdam, 1234, Amsterdam, Netherlands
| | - Ratko Djukanovic
- Southampton University, Clinical and Experimental Sciences and Southampton NIHR Respiratory Biomedical Research Unit, Southampton, United Kingdom of Great Britain and Northern Ireland
| | - Yike Guo
- Imperial College London, 4615, London, United Kingdom of Great Britain and Northern Ireland
| | - Sharon Mumby
- Imperial College London, 4615, London, United Kingdom of Great Britain and Northern Ireland
| | - Ian M Adcock
- NHLI, Imperial College London, Airways Disease, London, United Kingdom of Great Britain and Northern Ireland
| | - Kian Fan Chung
- National Heart and Lung Institute, London, United Kingdom of Great Britain and Northern Ireland;
| | | |
Collapse
|
12
|
Reyes-García J, Montaño LM, Carbajal-García A, Wang YX. Sex Hormones and Lung Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:259-321. [PMID: 34019274 DOI: 10.1007/978-3-030-68748-9_15] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammation is a characteristic marker in numerous lung disorders. Several immune cells, such as macrophages, dendritic cells, eosinophils, as well as T and B lymphocytes, synthetize and release cytokines involved in the inflammatory process. Gender differences in the incidence and severity of inflammatory lung ailments including asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (PF), lung cancer (LC), and infectious related illnesses have been reported. Moreover, the effects of sex hormones on both androgens and estrogens, such as testosterone (TES) and 17β-estradiol (E2), driving characteristic inflammatory patterns in those lung inflammatory diseases have been investigated. In general, androgens seem to display anti-inflammatory actions, whereas estrogens produce pro-inflammatory effects. For instance, androgens regulate negatively inflammation in asthma by targeting type 2 innate lymphoid cells (ILC2s) and T-helper (Th)-2 cells to attenuate interleukin (IL)-17A-mediated responses and leukotriene (LT) biosynthesis pathway. Estrogens may promote neutrophilic inflammation in subjects with asthma and COPD. Moreover, the activation of estrogen receptors might induce tumorigenesis. In this chapter, we summarize the most recent advances in the functional roles and associated signaling pathways of inflammatory cellular responses in asthma, COPD, PF, LC, and newly occurring COVID-19 disease. We also meticulously deliberate the influence of sex steroids on the development and progress of these common and severe lung diseases.
Collapse
Affiliation(s)
- Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico.,Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Luis M Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
13
|
Inoue Y, Okamoto T, Honda T, Nukui Y, Akashi T, Takemura T, Tozuka M, Miyazaki Y. Disruption in the balance between apolipoprotein A-I and mast cell chymase in chronic hypersensitivity pneumonitis. IMMUNITY INFLAMMATION AND DISEASE 2020; 8:659-671. [PMID: 33016012 PMCID: PMC7654418 DOI: 10.1002/iid3.355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023]
Abstract
Background Apolipoprotein A‐I (apoA‐I) has an antifibrotic effect in idiopathic pulmonary fibrosis. Although pulmonary fibrosis is associated with poor prognosis of patients with hypersensitivity pneumonitis (HP), little is known regarding the role of apoA‐I in the pathogenesis of HP. Methods Two‐dimensional electrophoresis, immunoblotting, and enzyme‐linked immunosorbent assays were performed for the identification and quantification of apoA‐I in bronchoalveolar lavage fluid (BALF) from patients with acute and chronic HP. To investigate the degradation of apoA‐I, apoA‐I was incubated with BALF. Moreover, the role of apoA‐I in TGF‐β1‐induced epithelial–mesenchymal transition of A549 cells was examined. Results The concentration of apoA‐I in the BALF was significantly lower in chronic HP (n = 56) compared with acute HP (n = 31). The expression level of apoA‐I was also low in the lung tissues of chronic HP. ApoA‐I was degraded by BALF from HP patients. The number of chymase‐positive mast cells in the alveolar parenchyma was inversely correlated with apoA‐I levels in the BALF of chronic HP patients. In vitro experiment using A549 cells, untreated apoA‐I inhibited TGF‐β1‐induced epithelial–mesenchymal transition, although this trend was not observed in the chymase‐treated apoA‐I. Conclusions A decrease of apoA‐I was associated with the pathogenesis of chronic HP in terms of pulmonary fibrosis and mast cell chymase attenuated the protective effect of apoA‐I against pulmonary fibrosis. Furthermore, apoA‐I could be a crucial molecule associated with lung fibrogenesis of HP.
Collapse
Affiliation(s)
- Yukihisa Inoue
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tsukasa Okamoto
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takayuki Honda
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshihisa Nukui
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takumi Akashi
- Department of Pathology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tamiko Takemura
- Department of Pathology, Japan Red Cross Centre, Tokyo, Japan
| | - Minoru Tozuka
- Department of Analytical Laboratory Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasunari Miyazaki
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
14
|
Zhang L, Jin H, Song Y, Chen SY, Wang Y, Sun Y, Tang C, Du J, Huang Y. Endogenous sulfur dioxide is a novel inhibitor of hypoxia-induced mast cell degranulation. J Adv Res 2020; 29:55-65. [PMID: 33842005 PMCID: PMC8020161 DOI: 10.1016/j.jare.2020.08.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 08/25/2020] [Accepted: 08/28/2020] [Indexed: 01/25/2023] Open
Abstract
Endogenous SO2/AAT pathway exists in mast cells (MCs). Endogenous SO2 is a novel MC membrane stabilizer under hypoxic circumstance. MC-derived SO2 upregulates cAMP level, thereby suppressing MC degranulation.
Introduction Mast cell (MC) degranulation is an important step in the pathogenesis of inflammatory reactions and allergies; however, the mechanism of stabilizing MC membranes to reduce their degranulation is unclear. Methods SO2 content in MC culture supernatant was measured by HPLC-FD. The protein and mRNA expressions of the key enzymes aspartate aminotransferase 1 (AAT1) and AAT2 and intracellular AAT activity were detected. The cAMP level in MCs was detected by immunofluorescence and ELISA. The release rate of MC degranulation marker β-hexosaminidase was measured. The expression of AAT1 and cAMP, the MC accumulation and degranulation in lung tissues were detected. Objectives To exam whether an endogenous sulfur dioxide (SO2) pathway exists in MCs and if it serves as a novel endogenous MC stabilizer. Results We firstly show the existence of the endogenous SO2/AAT pathway in MCs. Moreover, when AAT1 was knocked down in MCs, MC degranulation was significantly increased, and could be rescued by a SO2 donor. Mechanistically, AAT1 knockdown decreased the cyclic adenosine monophosphate (cAMP) content in MCs, while SO2 prevented this reduction in a dose-independent manner. Pretreatment with the cAMP-synthesizing agonist forskolin or the cAMP degradation inhibitor IBMX significantly blocked the increase in AAT1 knockdown-induced MC degranulation. Furthermore, in hypoxia-stimulated MCs, AAT1 protein expression and SO2 production were markedly down regulated, and MC degranulation was activated, which were blunted by AAT1 overexpression. The cAMP synthesis inhibitor SQ22536 disrupted the suppressive effect of AAT1 overexpression on hypoxia-induced MC degranulation. In a hypoxic environment, mRNA and protein expression of AAT1 was significantly reduced in lung tissues of rats. Supplementation of SO2 elevated the cAMP level and reduced perivascular MC accumulation and degranulation in lung tissues of rats exposed to a hypoxic environment in vivo. Conclusion SO2 serves as an endogenous MC stabilizer via upregulating the cAMP pathway under hypoxic circumstance.
Collapse
Affiliation(s)
- Lulu Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Research Unit of Clinical Diagnosis and Treatment of Pediatric Syncope and Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Hongfang Jin
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yunjia Song
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Selena Ying Chen
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Yi Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yan Sun
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Chaoshu Tang
- Department of Physiology and Pathophysiology, Peking University Health Science Centre, Beijing, China
- Key Laboratory of Molecular Cardiology, Ministry of Education, Beijing, China
| | - Junbao Du
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Key Laboratory of Molecular Cardiology, Ministry of Education, Beijing, China
| | - Yaqian Huang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Corresponding author at: Department of Pediatrics, Peking University First Hospital, Beijing 100034, China.
| |
Collapse
|
15
|
Tavares LP, Peh HY, Tan WSD, Pahima H, Maffia P, Tiligada E, Levi-Schaffer F. Granulocyte-targeted therapies for airway diseases. Pharmacol Res 2020; 157:104881. [PMID: 32380052 PMCID: PMC7198161 DOI: 10.1016/j.phrs.2020.104881] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 12/24/2022]
Abstract
The average respiration rate for an adult is 12-20 breaths per minute, which constantly exposes the lungs to allergens and harmful particles. As a result, respiratory diseases, which includes asthma, chronic obstructive pulmonary disease (COPD) and acute lower respiratory tract infections (LTRI), are a major cause of death worldwide. Although asthma, COPD and LTRI are distinctly different diseases with separate mechanisms of disease progression, they do share a common feature - airway inflammation with intense recruitment and activation of granulocytes and mast cells. Neutrophils, eosinophils, basophils, and mast cells are crucial players in host defense against pathogens and maintenance of lung homeostasis. Upon contact with harmful particles, part of the pulmonary defense mechanism is to recruit these cells into the airways. Despite their protective nature, overactivation or accumulation of granulocytes and mast cells in the lungs results in unwanted chronic airway inflammation and damage. As such, understanding the bright and the dark side of these leukocytes in lung physiology paves the way for the development of therapies targeting this important mechanism of disease. Here we discuss the role of granulocytes in respiratory diseases and summarize therapeutic strategies focused on granulocyte recruitment and activation in the lungs.
Collapse
Affiliation(s)
- Luciana P Tavares
- ImmuPhar - Immunopharmacology Section Committee of International Union of Basic and Clinical Pharmacology (IUPHAR); Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Hong Yong Peh
- ImmuPhar - Immunopharmacology Section Committee of International Union of Basic and Clinical Pharmacology (IUPHAR); Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, 16 Medical Drive, 117600, Singapore
| | - Wan Shun Daniel Tan
- ImmuPhar - Immunopharmacology Section Committee of International Union of Basic and Clinical Pharmacology (IUPHAR); Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, 16 Medical Drive, 117600, Singapore
| | - Hadas Pahima
- ImmuPhar - Immunopharmacology Section Committee of International Union of Basic and Clinical Pharmacology (IUPHAR); Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Pasquale Maffia
- ImmuPhar - Immunopharmacology Section Committee of International Union of Basic and Clinical Pharmacology (IUPHAR); Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Ekaterini Tiligada
- ImmuPhar - Immunopharmacology Section Committee of International Union of Basic and Clinical Pharmacology (IUPHAR); Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Francesca Levi-Schaffer
- ImmuPhar - Immunopharmacology Section Committee of International Union of Basic and Clinical Pharmacology (IUPHAR); Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
16
|
Wang Y, Ma H, Tao X, Luo Y, Wang H, He J, Fang Q, Guo S, Song C. SCF promotes the production of IL-13 via the MEK-ERK-CREB signaling pathway in mast cells. Exp Ther Med 2019; 18:2491-2496. [PMID: 31555361 PMCID: PMC6755428 DOI: 10.3892/etm.2019.7866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 07/12/2018] [Indexed: 12/26/2022] Open
Abstract
Mast cells serve a key role in the occurrence and development of allergy. As an important growth factor of mast cells, stem cell factor (SCF) has an effect on the apoptosis, chemotaxis, adhesion, degranulation and other biological characteristics of mast cells. However, there are few studies regarding the effect of SCF signal on the production of cytokines from mast cells, particularly Th2 type cytokines. In the present study, the expression and secretion of IL-13 in P815 cells stimulated by SCF were detected by fluorescence quantitative PCR and ELISA, and western blotting and EMSA were used to detect ERK phosphorylation and activation of CREB in stimulated P815 cells. The results demonstrated that the production of IL-13 was significantly increased in P815 cells stimulated by SCF (1–100 ng/ml; P<0.01). There was an obvious phosphorylation of ERK and CREB activation in P815 cells stimulated by SCF (50 ng/ml). Compared with the SCF single stimulation group, the production of IL-13 was significantly reduced in P815 cells stimulated with U0126 (ERK-MEK/pathway inhibitor) or H-89 (CREB inhibitor) combined with SCF stimulation group (P<0.01). However, JSI-124 (JAK/STAT3 pathway inhibitor), Wortmannin (PI3K/Akt pathway inhibitor) and PDTC (NF-κB inhibitor) had no effect on the role of SCF promoting the P815 cells producing IL-13. Therefore, SCF signaling promotes mast cell P815 to produce IL-13, and this effect is associated with the MEK-ERK-CREB signaling pathway.
Collapse
Affiliation(s)
- Yimeng Wang
- Department of Immunology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Hua Ma
- Department of Immunology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Xiangnan Tao
- Department of Immunology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Yulan Luo
- Department of Immunology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Helong Wang
- Department of Immunology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Jing He
- Department of Immunology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Qiang Fang
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Shujun Guo
- Department of Immunology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Chuanwang Song
- Department of Immunology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| |
Collapse
|
17
|
Elieh Ali Komi D, Bjermer L. Mast Cell-Mediated Orchestration of the Immune Responses in Human Allergic Asthma: Current Insights. Clin Rev Allergy Immunol 2019; 56:234-247. [PMID: 30506113 DOI: 10.1007/s12016-018-8720-1] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Improving the lung function after experimental allergen challenge by blocking of mast cell (MC) mediators and the capability of MC mediators (including histamine, prostaglandin (PG) D2, and leukotriene (LT) C4) in induction of mucosal edema, bronchoconstriction, and mucus secretion provide evidence that MCs play a key role in pathophysiology of asthma. In asthma, the number of MCs increases in the airways and infiltration of MCs in a variety of anatomical sites including the epithelium, the submucosal glands, and the smooth muscle bundles occurs. MC localization within the ASM is accompanied with the hypertrophy and hyperplasia of the layer, and smooth muscle dysfunction that is mainly observed in forms of bronchial hyperresponsiveness, and variable airflow obstruction. Owing to the expression of a wide range of surface receptors and releasing various cytoplasmic mediators, MCs orchestrate the pathologic events of the disease. MC-released preformed mediators including chymase, tryptase, and histamine and de novo synthesized mediators such as PGD2, LTC4, and LTE4 in addition of cytokines mainly TGFβ1, TSLP, IL-33, IL-4, and IL-13 participate in pathogenesis of asthma. The release of MC mediators and MC/airway cell interactions during remodeling phase of asthma results in persistent cellular and structural changes in the airway wall mainly epithelial cell shedding, goblet cell hyperplasia, hypertrophy of ASM bundles, fibrosis in subepithelial region, abnormal deposition of extracellular matrix (ECM), increased tissue vascularity, and basement membrane thickening. We will review the current knowledge regarding the participation of MCs in each stage of asthma pathophysiology including the releasing mediators and their mechanism of action, expression of receptors by which they respond to stimuli, and finally the pharmaceutical products designed based on the strategy of blocking MC activation and mediator release.
Collapse
Affiliation(s)
- Daniel Elieh Ali Komi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leif Bjermer
- Department of Respiratory Medicine & Allergology, Inst for Clinical Science, Lund University, Lund, Sweden.
- Lung and Allergy Research, Skane University Hospital, Lasarettsgatan 7, 22185, Lund, Sweden.
| |
Collapse
|
18
|
Mathew G, Sharma A, Pickering RJ, Rosado CJ, Lemarie J, Mudgal J, Thambi M, Sebastian S, Jandeleit-Dahm KA, de Haan JB, Unnikrishnan MK. A novel synthetic small molecule DMFO targets Nrf2 in modulating proinflammatory/antioxidant mediators to ameliorate inflammation. Free Radic Res 2018; 52:1140-1157. [PMID: 30422019 DOI: 10.1080/10715762.2018.1533636] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Inflammation is a protective immune response against invading pathogens, however, dysregulated inflammation is detrimental. As the complex inflammatory response involves multiple mediators, including the involvement of reactive oxygen species, concomitantly targeting proinflammatory and antioxidant check-points may be a more rational strategy. We report the synthesis and anti-inflammatory/antioxidant activity of a novel indanedione derivative DMFO. DMFO scavenged reactive oxygen species (ROS) in in-vitro radical scavenging assays and in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. In acute models of inflammation (carrageenan-induced inflammation in rat paw and air pouch), DMFO effectively reduced paw oedema and leucocyte infiltration with an activity comparable to diclofenac. DMFO stabilised mast cells (MCs) in in-vitro A23187 and compound 48/80-induced assays. Additionally, DMFO stabilised MCs in an antigen (ovalbumin)-induced MC degranulation model in-vivo, without affecting serum IgE levels. In a model of chronic immune-mediated inflammation, Freund's adjuvant-induced arthritis, DMFO reduced arthritic score and contralateral paw oedema, and increased the pain threshold with an efficacy comparable to diclofenac but without being ulcerogenic. Additionally, DMFO significantly reduced serum TNFα levels. Mechanistic studies revealed that DMFO reduced proinflammatory genes (IL1β, TNFα, IL6) and protein levels (COX2, MCP1), with a concurrent increase in antioxidant genes (NQO1, haem oxygenase 1 (HO-1), Glo1, Nrf2) and protein (HO-1) in LPS-stimulated macrophages. Importantly, the anti-inflammatory/antioxidant effect on gene expression was absent in primary macrophages isolated from Nrf2 KO mice suggesting an Nrf2-targeted activity, which was subsequently confirmed using siRNA transfection studies in RAW macrophages. Therefore, DMFO is a novel, orally-active, safe (even at 2 g/kg p.o.), a small molecule which targets Nrf2 in ameliorating inflammation.
Collapse
Affiliation(s)
- Geetha Mathew
- a Department of Pharmacology, Manipal College of Pharmaceutical Sciences , Manipal Academy of Higher Education , Manipal , India.,b Oxidative Stress Laboratory, Basic Science Domain , Baker Heart and Diabetes Institute , Melbourne , Australia.,c Department of Diabetes, the Alfred Centre , Monash University , Melbourne , Australia
| | - Arpeeta Sharma
- b Oxidative Stress Laboratory, Basic Science Domain , Baker Heart and Diabetes Institute , Melbourne , Australia
| | - Raelene J Pickering
- c Department of Diabetes, the Alfred Centre , Monash University , Melbourne , Australia
| | - Carlos J Rosado
- c Department of Diabetes, the Alfred Centre , Monash University , Melbourne , Australia
| | - Jeremie Lemarie
- b Oxidative Stress Laboratory, Basic Science Domain , Baker Heart and Diabetes Institute , Melbourne , Australia
| | - Jayesh Mudgal
- a Department of Pharmacology, Manipal College of Pharmaceutical Sciences , Manipal Academy of Higher Education , Manipal , India
| | - Magith Thambi
- a Department of Pharmacology, Manipal College of Pharmaceutical Sciences , Manipal Academy of Higher Education , Manipal , India
| | - Sarine Sebastian
- a Department of Pharmacology, Manipal College of Pharmaceutical Sciences , Manipal Academy of Higher Education , Manipal , India
| | - Karin A Jandeleit-Dahm
- b Oxidative Stress Laboratory, Basic Science Domain , Baker Heart and Diabetes Institute , Melbourne , Australia.,c Department of Diabetes, the Alfred Centre , Monash University , Melbourne , Australia
| | - Judy B de Haan
- b Oxidative Stress Laboratory, Basic Science Domain , Baker Heart and Diabetes Institute , Melbourne , Australia
| | - Mazhuvancherry K Unnikrishnan
- d Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences , Manipal Academy of Higher Education , Manipal , India
| |
Collapse
|
19
|
Kempuraj D, Thangavel R, Selvakumar GP, Ahmed ME, Zaheer S, Raikwar SP, Zahoor H, Saeed D, Dubova I, Giler G, Herr S, Iyer SS, Zaheer A. Mast Cell Proteases Activate Astrocytes and Glia-Neurons and Release Interleukin-33 by Activating p38 and ERK1/2 MAPKs and NF-κB. Mol Neurobiol 2018; 56:1681-1693. [PMID: 29916143 DOI: 10.1007/s12035-018-1177-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/04/2018] [Indexed: 12/13/2022]
Abstract
Inflammatory mediators released from activated microglia, astrocytes, neurons, and mast cells mediate neuroinflammation. Parkinson's disease (PD) is characterized by inflammation-dependent dopaminergic neurodegeneration in substantia nigra. 1-Methyl-4-phenylpyridinium (MPP+), a metabolite of parkinsonian neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), induces inflammatory mediators' release from brain cells and mast cells. Brain cells' interaction with mast cells is implicated in neuroinflammation. However, the exact mechanisms involved are not yet clearly understood. Mouse fetal brain-derived cultured primary astrocytes and glia-neurons were incubated with mouse mast cell protease-6 (MMCP-6) and MMCP-7, and mouse bone marrow-derived mast cells (BMMCs) were incubated with MPP+ and brain protein glia maturation factor (GMF). Interleukin-33 (IL-33) released from these cells was quantitated by enzyme-linked immunosorbent assay. Both MMCP-6 and MMCP-7 induced IL-33 release from astrocytes and glia-neurons. MPP+ and GMF were used as a positive control-induced IL-33 and reactive oxygen species expression in mast cells. Mast cell proteases and MPP+ activate p38 and extracellular signal-regulated kinases 1/2 (ERK1/2), mitogen-activated protein kinases (MAPKs), and transcription factor nuclear factor-kappa B (NF-κB) in astrocytes, glia-neurons, or mast cells. Addition of BMMCs from wt mice and transduction with adeno-GMF show higher chemokine (C-C motif) ligand 2 (CCL2) release. MPP+ activated glial cells and reduced microtubule-associated protein 2 (MAP-2) expression indicating neurodegeneration. IL-33 expression increased in the midbrain and striatum of PD brains as compared with age- and sex-matched control subjects. Glial cells and neurons interact with mast cells and accelerate neuroinflammation and these interactions can be explored as a new therapeutic target to treat PD.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA.
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA.
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Gvindhasamy Pushpavathi Selvakumar
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Mohammad Ejaz Ahmed
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Smita Zaheer
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Haris Zahoor
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Daniyal Saeed
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Iuliia Dubova
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Gema Giler
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Shelby Herr
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Shankar S Iyer
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Asgar Zaheer
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA.
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA.
| |
Collapse
|
20
|
Ng CW, Tam IYS, Sam SW, Yu Y, Lau HYA. Immobilized Osteopontin Enhances Adhesion but Suppresses Cytokine Release of Anti-IgE Activated Human Mast Cells. Front Immunol 2018; 9:1109. [PMID: 29872439 PMCID: PMC5972195 DOI: 10.3389/fimmu.2018.01109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/02/2018] [Indexed: 01/27/2023] Open
Abstract
Osteopontin (OPN) is an Arg-Gly-Asp (RGD)-containing extracellular matrix protein which is upregulated in inflamed tissues and has been reported to modulate mast cell activities in mice. Due to the known heterogeneity among mast cells of different species and the important roles of mast cells in allergic reactions, we investigated the effects of human OPN (hOPN) on human mast cell activities. Mature primary human cultured mast cells (HCMC) were derived from peripheral blood CD34+ progenitors and the modulation of their activation by soluble and plate-bound immobilized hOPN were examined by studying their release of inflammatory mediators (histamine, IL-5, IL-8, TNF-α, and VEGF) and matrix adhesion following stimulation by anti-IgE. Immobilized hOPN enhanced the adhesion, but suppressed the release of IL-5, IL-8, and TNF-α of anti-IgE-activated HCMC while soluble hOPN failed to demonstrate any significant effects. By employing cyclic RGD peptide and neutralizing antibodies against different classes of integrin and CD44, we demonstrated that the interaction of immobilized hOPN and HCMC was mediated by the RGD domain of hOPN and integrin but not CD44 on HCMC. Our results suggest that immobilized hOPN anchored to extracellular matrix can regulate adaptive immunity in humans by retaining mast cells at the site of inflammation and suppressing anti-IgE-induced cytokine release from HCMC.
Collapse
Affiliation(s)
- Chun Wai Ng
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Issan Yee San Tam
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Sze Wing Sam
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Yangyang Yu
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong.,Department of Physiology, Shenzhen University Health Science Center, Shenzhen, China
| | - Hang Yung Alaster Lau
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
21
|
Xue L, Geng Y, Li M, Jin YF, Ren HX, Li X, Wu F, Wang B, Cheng WY, Chen T, Chen YJ. Inhibitory effects of methamphetamine on mast cell activation and cytokine/chemokine production stimulated by lipopolysaccharide in C57BL/6J mice. Exp Ther Med 2018; 15:3544-3550. [PMID: 29545881 PMCID: PMC5841010 DOI: 10.3892/etm.2018.5837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/13/2017] [Indexed: 01/09/2023] Open
Abstract
Previous studies have demonstrated that methamphetamine (MA) influences host immunity; however, the effect of MA on lipopolysaccharide (LPS)-induced immune responses remains unknown. Mast cells (MCs) are considered to serve an important role in the innate and acquired immune response, but it remains unknown whether MA modulates MC activation and LPS-stimulated cytokine production. The present study aimed to investigate the effect of MA on LPS-induced MC activation and the production of MC-derived cytokines in mice. Markers for MC activation, including cluster of differentiation 117 and the type I high affinity immunoglobulin E receptor, were assessed in mouse intestines. Levels of MC-derived cytokines in the lungs and thymus were also examined. The results demonstrated that cytokines were produced in the bone marrow-derived mast cells (BMMCs) of mice. The present study demonstrated that MA suppressed the LPS-mediated MC activation in mouse intestines. MA also altered the release of MC cytokines in the lung and thymus following LPS stimulation. In addition, LPS-stimulated cytokines were decreased in the BMMCs of mice following treatment with MA. The present study demonstrated that MA may regulate LPS-stimulated MC activation and cytokine production.
Collapse
Affiliation(s)
- Li Xue
- Department of Immunology and Pathogenic Biology, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, Shaanxi 710061, P.R. China
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yan Geng
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Ming Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yao-Feng Jin
- Department of Pathology, The Second Affiliated Hospital Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Hui-Xun Ren
- Department of Immunology and Pathogenic Biology, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, Shaanxi 710061, P.R. China
| | - Xia Li
- VIP Internal Medicine Department, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Feng Wu
- Graduate Teaching and Experiment Centre, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, Shaanxi 710061, P.R. China
| | - Biao Wang
- Department of Immunology and Pathogenic Biology, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, Shaanxi 710061, P.R. China
| | - Wei-Ying Cheng
- Department of Immunology and Pathogenic Biology, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, Shaanxi 710061, P.R. China
| | - Teng Chen
- Forensic Medicine College of Xi'an Jiaotong University, Key Laboratory of The Health Ministry for Forensic Medicine, Key Laboratory of The Ministry of Education for Environment and Genes Related to Diseases, Xi'an, Shaanxi 710061, P.R. China
| | - Yan-Jiong Chen
- Department of Immunology and Pathogenic Biology, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
22
|
Fang H, Zhang Y, Li N, Wang G, Liu Z. The Autoimmune Skin Disease Bullous Pemphigoid: The Role of Mast Cells in Autoantibody-Induced Tissue Injury. Front Immunol 2018; 9:407. [PMID: 29545809 PMCID: PMC5837973 DOI: 10.3389/fimmu.2018.00407] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/14/2018] [Indexed: 01/09/2023] Open
Abstract
Bullous pemphigoid (BP) is an autoimmune and inflammatory skin disease associated with subepidermal blistering and autoantibodies directed against the hemidesmosomal components BP180 and BP230. Animal models of BP were developed by passively transferring anti-BP180 IgG into mice, which recapitulates the key features of human BP. By using these in vivo model systems, key cellular and molecular events leading to the BP disease phenotype are identified, including binding of pathogenic IgG to its target, complement activation of the classical pathway, mast cell degranulation, and infiltration and activation of neutrophils. Proteinases released by infiltrating neutrophils cleave BP180 and other hemidesmosome-associated proteins, causing DEJ separation. Mast cells and mast cell-derived mediators including inflammatory cytokines and proteases are increased in lesional skin and blister fluids of BP. BP animal model evidence also implicates mast cells in the pathogenesis of BP. However, recent studies questioned the pathogenic role of mast cells in autoimmune diseases such as multiple sclerosis, rheumatoid arthritis, and epidermolysis bullosa acquisita. This review highlights the current knowledge on BP pathophysiology with a focus on a potential role for mast cells in BP and mast cell-related critical issues needing to be addressed in the future.
Collapse
Affiliation(s)
- Hui Fang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yang Zhang
- Department of Dermatology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Dermatology, The Second Hospital, School of Medicine, Xi’an Jiaotong University, Xi’an, China
| | - Ning Li
- Department of Dermatology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhi Liu
- Department of Dermatology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
23
|
Bird SD. Calcium mediates cell shape change in human peritoneal mesothelial cells. Cell Calcium 2018; 72:116-126. [PMID: 29730478 DOI: 10.1016/j.ceca.2018.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/15/2018] [Accepted: 02/15/2018] [Indexed: 12/13/2022]
Abstract
Mast cells in the peritoneal membrane (PM) may degranulate to release preformed inflammatory mediators including histamine which is capable of diffusing into the surrounding interstitium, modulating cells in their vicinity including, human peritoneal mesothelial cells (hPMC). The present study aimed to investigate the quorum intracellular calcium ([Ca2+i]) response to histamine compared to the membrane soluble ionophore, A23187, in adherent cultured hPMC. To examine [Ca2+i] handling, Fura - 2 loaded cells were exposed to histamine and A23187. Agonist induced transient [Ca2+i] event(s) (TCE) were defined and compared including, resting calcium, peak height, recovery and transient kinetics. Changes in cell shape were examined with immunocytochemistry of the cortical actin (CA) and microtubule (MT) cytoskeleton. To investigate whether histamine induced changes in cell shape were mediated by [Ca2+i], mobilization of [Ca2+i] was prevented with 20 μmol/l of the calcium chelator 1,2-bis-(2-aminophenoxy) ethane-N,N,N',N'-tetraacetic acid (BAPTA-AM). Histamine produced a dose dependent increase of [Ca2+i], maximal at 1.0 mmol/l which recovered to the pre-challenge resting value. Transient multiplicity with repeated challenge was evident below a histamine threshold of 10-2 mmol/l. Morphometric analysis of MTs and CA showed significant cell elongation plus histamine and A23187. The histamine induced cell elongation was eliminated with [Ca2+i] clamping. This data indicated that increased [Ca2+i] was essential for cell elongation and the formation of stress fibres and therefore has a pivotal role in the regulation of the PM barrier.
Collapse
Affiliation(s)
- Stephen D Bird
- Department of Obstetrics and Gynaecology, The University of Melbourne, Victoria, Australia; Department of Medicine, Dunedin School of Medicine, The University of Otago, Dunedin, New Zealand.
| |
Collapse
|
24
|
Kempuraj D, Selvakumar GP, Thangavel R, Ahmed ME, Zaheer S, Raikwar SP, Iyer SS, Bhagavan SM, Beladakere-Ramaswamy S, Zaheer A. Mast Cell Activation in Brain Injury, Stress, and Post-traumatic Stress Disorder and Alzheimer's Disease Pathogenesis. Front Neurosci 2017; 11:703. [PMID: 29302258 PMCID: PMC5733004 DOI: 10.3389/fnins.2017.00703] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 11/30/2017] [Indexed: 12/30/2022] Open
Abstract
Mast cells are localized throughout the body and mediate allergic, immune, and inflammatory reactions. They are heterogeneous, tissue-resident, long-lived, and granulated cells. Mast cells increase their numbers in specific site in the body by proliferation, increased recruitment, increased survival, and increased rate of maturation from its progenitors. Mast cells are implicated in brain injuries, neuropsychiatric disorders, stress, neuroinflammation, and neurodegeneration. Brain mast cells are the first responders before microglia in the brain injuries since mast cells can release prestored mediators. Mast cells also can detect amyloid plaque formation during Alzheimer's disease (AD) pathogenesis. Stress conditions activate mast cells to release prestored and newly synthesized inflammatory mediators and induce increased blood-brain barrier permeability, recruitment of immune and inflammatory cells into the brain and neuroinflammation. Stress induces the release of corticotropin-releasing hormone (CRH) from paraventricular nucleus of hypothalamus and mast cells. CRH activates glial cells and mast cells through CRH receptors and releases neuroinflammatory mediators. Stress also increases proinflammatory mediator release in the peripheral systems that can induce and augment neuroinflammation. Post-traumatic stress disorder (PTSD) is a traumatic-chronic stress related mental dysfunction. Currently there is no specific therapy to treat PTSD since its disease mechanisms are not yet clearly understood. Moreover, recent reports indicate that PTSD could induce and augment neuroinflammation and neurodegeneration in the pathogenesis of neurodegenerative diseases. Mast cells play a crucial role in the peripheral inflammation as well as in neuroinflammation due to brain injuries, stress, depression, and PTSD. Therefore, mast cells activation in brain injury, stress, and PTSD may accelerate the pathogenesis of neuroinflammatory and neurodegenerative diseases including AD. This review focusses on how mast cells in brain injuries, stress, and PTSD may promote the pathogenesis of AD. We suggest that inhibition of mast cells activation and brain cells associated inflammatory pathways in the brain injuries, stress, and PTSD can be explored as a new therapeutic target to delay or prevent the pathogenesis and severity of AD.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veteran's Hospital, United States Department of Veterans Affairs, Columbia, MO, United States
| | - Govindhasamy P. Selvakumar
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veteran's Hospital, United States Department of Veterans Affairs, Columbia, MO, United States
| | - Ramasamy Thangavel
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veteran's Hospital, United States Department of Veterans Affairs, Columbia, MO, United States
| | - Mohammad E. Ahmed
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veteran's Hospital, United States Department of Veterans Affairs, Columbia, MO, United States
| | - Smita Zaheer
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Sudhanshu P. Raikwar
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veteran's Hospital, United States Department of Veterans Affairs, Columbia, MO, United States
| | - Shankar S. Iyer
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veteran's Hospital, United States Department of Veterans Affairs, Columbia, MO, United States
| | - Sachin M. Bhagavan
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Swathi Beladakere-Ramaswamy
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Asgar Zaheer
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veteran's Hospital, United States Department of Veterans Affairs, Columbia, MO, United States
| |
Collapse
|
25
|
Felix-Patrício B, Miranda AF, Medeiros JL, Gallo CBM, Gregório BM, Souza DB, Costa WS, Sampaio FJB. The prostate after castration and hormone replacement in a rat model: structural and ultrastructural analysis. Int Braz J Urol 2017; 43:957-965. [PMID: 28379662 PMCID: PMC5678530 DOI: 10.1590/s1677-5538.ibju.2016.0484] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 01/31/2017] [Indexed: 12/13/2022] Open
Abstract
Purpose: To evaluate if late hormonal replacement is able to recover the prostatic tissue modified by androgenic deprivation. Materials and Methods: 24 rats were assigned into a Sham group; an androgen deficient group, submitted to bilateral orchiectomy (Orch); and a group submitted to bilateral orchiectomy followed by testosterone replacement therapy (Orch+T). After 60 days from surgery blood was collected for determination of testosterone levels and the ventral prostate was collected for quantitative and qualitative microscopic analysis. The acinar epithelium height, the number of mast cells per field, and the densities of collagen fibers and acinar lumen were analyzed by stereological methods under light microscopy. The muscle fibers and types of collagen fibers were qualitatively assessed by scanning electron microscopy and polarization microscopy. Results: Hormone depletion (in group Orch) and return to normal levels (in group Orch+T) were effective as verified by serum testosterone analysis. The androgen deprivation promoted several alterations in the prostate: the acinar epithelium height diminished from 16.58±0.47 to 11.48±0.29μm; the number of mast cells per field presented increased from 0.45±0.07 to 2.83±0.25; collagen fibers density increased from 5.83±0.92 to 24.70±1.56%; and acinar lumen density decreased from 36.78±2.14 to 16.47±1.31%. Smooth muscle was also increased in Orch animals, and type I collagen fibers became more predominant in these animals. With the exception of the densities of collagen fibers and acinar lumen, in animals receiving testosterone replacement therapy all parameters became statistically similar to Sham. Collagen fibers density became lower and acinar lumen density became higher in Orch+T animals, when compared to Sham. This is the first study to demonstrate a relation between mast cells and testosterone levels in the prostate. This cells have been implicated in prostatic cancer and benign hyperplasia, although its specific role is not understood. Conclusion: Testosterone deprivation promotes major changes in the prostate of rats. The hormonal replacement therapy was effective in reversing these alterations.
Collapse
Affiliation(s)
- Bruno Felix-Patrício
- Instituto de Ciências Humanas e da Saúde, Universidade Federal Fluminense, Rio das Ostras, RJ, Brasil
| | - Alexandre F Miranda
- Urogenital Research Unit, Universidade Estadual do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | | | - Carla B M Gallo
- Urogenital Research Unit, Universidade Estadual do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Bianca M Gregório
- Urogenital Research Unit, Universidade Estadual do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Diogo B Souza
- Urogenital Research Unit, Universidade Estadual do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Waldemar S Costa
- Urogenital Research Unit, Universidade Estadual do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Francisco J B Sampaio
- Urogenital Research Unit, Universidade Estadual do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
26
|
Cellular and molecular mechanisms of asthma and COPD. Clin Sci (Lond) 2017; 131:1541-1558. [PMID: 28659395 DOI: 10.1042/cs20160487] [Citation(s) in RCA: 319] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/28/2017] [Accepted: 03/01/2017] [Indexed: 12/19/2022]
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) both cause airway obstruction and are associated with chronic inflammation of the airways. However, the nature and sites of the inflammation differ between these diseases, resulting in different pathology, clinical manifestations and response to therapy. In this review, the inflammatory and cellular mechanisms of asthma and COPD are compared and the differences in inflammatory cells and profile of inflammatory mediators are highlighted. These differences account for the differences in clinical manifestations of asthma and COPD and their response to therapy. Although asthma and COPD are usually distinct, there are some patients who show an overlap of features, which may be explained by the coincidence of two common diseases or distinct phenotypes of each disease. It is important to better understand the underlying cellular and molecular mechanisms of asthma and COPD in order to develop new treatments in areas of unmet need, such as severe asthma, curative therapy for asthma and effective anti-inflammatory treatments for COPD.
Collapse
|
27
|
Srivastava S, Li Z, Skolnik EY. Phosphatidlyinositol-3-kinase C2 beta (PI3KC2β) is a potential new target to treat IgE mediated disease. PLoS One 2017; 12:e0183474. [PMID: 28820911 PMCID: PMC5562315 DOI: 10.1371/journal.pone.0183474] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 08/04/2017] [Indexed: 12/14/2022] Open
Abstract
Cross linking of the IgE receptor (FcεRI) on mast cells plays a critical role in IgE-dependent allergy including allergic rhinitis, asthma, anaphylaxis, and delayed type hypersensitivity reactions. The Ca2+ activated K+ channel, KCa3.1, plays a critical role in IgE-stimulated Ca2+ entry and degranulation in mast cells by helping to maintain a negative membrane potential, which provides an electrochemical gradient to drive Ca2+ influx. Of the 3 classes of PI3K, the class II PI3Ks are the least studied and little is known about the roles for class II PI3Ks in vivo in the context of the whole organism under normal and pathological conditions. Studying bone marrow derived mast cells (BMMC) isolated from PI3KC2β-/- mice, we now show that the class II PI3KC2β is critical for FcεRI stimulated KCa3.1 channel activation and the subsequent activation of mast cells. We found FcεRI-stimulated Ca2+ entry, cytokine production, and degranulation are decreased in BMMC isolated from PI3KC2β-/- mice. In addition, PI3KC2β-/- mice are markedly resistant to both passive cutaneous and passive systemic anaphylaxis. These findings identify PI3KC2β as a new pharmacologic target to treat IgE-mediated disease.
Collapse
Affiliation(s)
- Shekhar Srivastava
- Division of Nephrology, New York University Langone Medical Center, New York, New York, United States of America
- Department of Molecular Pathogenesis, Skirball Institute for Biomolecular Medicine, New York University Langone Medical Center, New York, New York, United States of America
- The Helen L. and Martin S. Kimmel Center for Biology and Medicine, Skirball Institute for Biomolecular Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Zhai Li
- Division of Nephrology, New York University Langone Medical Center, New York, New York, United States of America
- Department of Molecular Pathogenesis, Skirball Institute for Biomolecular Medicine, New York University Langone Medical Center, New York, New York, United States of America
- The Helen L. and Martin S. Kimmel Center for Biology and Medicine, Skirball Institute for Biomolecular Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Edward Y. Skolnik
- Division of Nephrology, New York University Langone Medical Center, New York, New York, United States of America
- Department of Molecular Pathogenesis, Skirball Institute for Biomolecular Medicine, New York University Langone Medical Center, New York, New York, United States of America
- The Helen L. and Martin S. Kimmel Center for Biology and Medicine, Skirball Institute for Biomolecular Medicine, New York University Langone Medical Center, New York, New York, United States of America
- Department of Pharmacology, Skirball Institute for Biomolecular Medicine, New York University Langone Medical Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
28
|
Abstract
β2-adrenoceptor agonists, often used in combination with corticosteroids, have been extensively used for the treatment of asthma. However, concerns have been raised regarding their adverse effects and safety including poor asthma control, life-threatening exacerbations, exacerbations that often require hospitalization, and asthma-related deaths. The question as to whether these adverse effects relate to the loss of their bronchoprotective action remains an interesting possibility. In the chapter, we will review the experimental evidence that describes the different potential factors and associated mechanisms that can blunt the therapeutic action of β2-adrenoceptor agonists in asthma. We show here evidence that various key inflammatory cytokines, growth factors, some respiratory viruses, certain allergens, unknown factors present in serum from atopic asthmatics have the capacity to impair β2-adrenoceptor function in airway smooth muscle, the main target of these drugs. More importantly, we present our latest research describing the role played by mast cells in impairing β2-adrenoceptor function. Although no definitive conclusion could be made regarding the implication of one single mechanism, receptor uncoupling, or receptor desensitization due to phosphorylation represents the main inhibitory pathways associated with a loss of β2-adrenoceptor function in airway smooth muscle. Targeting the pathways leading to β2-adrenoceptor dysfunction will likely provide novel therapies to improve the efficacy of β2-agonists in asthma.
Collapse
|
29
|
Galdiero MR, Varricchi G, Seaf M, Marone G, Levi-Schaffer F, Marone G. Bidirectional Mast Cell-Eosinophil Interactions in Inflammatory Disorders and Cancer. Front Med (Lausanne) 2017; 4:103. [PMID: 28791287 PMCID: PMC5523083 DOI: 10.3389/fmed.2017.00103] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 06/26/2017] [Indexed: 12/19/2022] Open
Abstract
Human mast cells (MCs) and eosinophils were first described and named by Paul Ehrlich. These cells have distinct myeloid progenitors and differ morphologically, ultrastructurally, immunologically, biochemically, and pharmacologically. However, MCs and eosinophils play a pivotal role in several allergic disorders. In addition, these cells are involved in autoimmune disorders, cardiovascular diseases, and cancer. MCs are distributed throughout all normal human tissues, whereas eosinophils are present only in gastrointestinal tract, secondary lymphoid tissues, and adipose tissue, thymus, mammary gland, and uterus. However, in allergic disorders, MCs and eosinophils can form the "allergic effector unit." Moreover, in several tumors, MCs and eosinophils can be found in close proximity. Therefore, it is likely that MCs have the capacity to modulate eosinophil functions and vice versa. For example, interleukin 5, stem cell factor, histamine, platelet-activating factor (PAF), prostaglandin D2 (PGD2), cysteinyl leukotrienes, and vascular endothelial growth factors (VEGFs), produced by activated MCs, can modulate eosinophil functions through the engagement of specific receptors. In contrast, eosinophil cationic proteins such as eosinophil cationic protein and major basic protein (MBP), nerve growth factor, and VEGFs released by activated eosinophils can modulate MC functions. These bidirectional interactions between MCs and eosinophils might be relevant not only in allergic diseases but also in several inflammatory and neoplastic disorders.
Collapse
Affiliation(s)
- Maria Rosaria Galdiero
- Department of Translational Medical Sciences (DiSMeT), Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences (DiSMeT), Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Mansour Seaf
- Pharmacology and Experimental Therapeutics Unit, Faculty of Medicine, School of Pharmacy, Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Giancarlo Marone
- Department of Public Health, University of Naples Federico II, Monaldi Hospital Pharmacy, Naples, Italy
| | - Francesca Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit, Faculty of Medicine, School of Pharmacy, Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gianni Marone
- Department of Translational Medical Sciences (DiSMeT), Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore” (IEOS), National Research Council (CNR), Naples, Italy
| |
Collapse
|
30
|
Lu F, Huang S. The Roles of Mast Cells in Parasitic Protozoan Infections. Front Immunol 2017; 8:363. [PMID: 28428784 PMCID: PMC5382204 DOI: 10.3389/fimmu.2017.00363] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 03/14/2017] [Indexed: 12/26/2022] Open
Abstract
Protozoan parasites such as Plasmodium spp., Leishmania spp., Trypanosoma spp., and Toxoplasma gondii are major causes of parasitic diseases in both humans and animals. The immune system plays a critical role against protozoa, but their immune mechanism remains poorly understood. This highlights the need to investigate the function of immune cells involved in the process of parasite infections and the responses of host immune system to parasite infections. Mast cells (MCs) are known to be central players in allergy and anaphylaxis, and it has been demonstrated that MCs have crucial roles in host defense against a number of different pathogens, including parasites. To date, there are many studies that have examined the interaction of helminth-derived antigens and MCs. As one of the major effector cells, MCs also play an important role in the immune response against some parasitic protozoa, but their role in protozoan infections is, however, less well characterized. Herein, we review the current knowledge about the roles of MCs and their mediators during infections involving highly pathogenic protozoa including Plasmodium spp., Leishmania spp., Trypanosoma spp., and T. gondii. We offer a general review of the data from patients and experimental animal models infected with the aforementioned protozoa, which correlate MCs and MC-derived mediators with exacerbated inflammation and disease progression as well as protection against the parasitic infections in different circumstances. This review updates our current understanding of the roles of MCs during parasitic protozoan infections, and the participation of MCs in parasitic protozoan infections could be of a potential therapeutic target.
Collapse
Affiliation(s)
- Fangli Lu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Shiguang Huang
- School of Stomatology, Jinan University, Guangzhou, China
| |
Collapse
|
31
|
Gangwar RS, Landolina N, Arpinati L, Levi-Schaffer F. Mast cell and eosinophil surface receptors as targets for anti-allergic therapy. Pharmacol Ther 2016; 170:37-63. [PMID: 27773785 DOI: 10.1016/j.pharmthera.2016.10.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Roopesh Singh Gangwar
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Nadine Landolina
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Ludovica Arpinati
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Francesca Levi-Schaffer
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel.
| |
Collapse
|