1
|
Newton HS, Cedrone E, Grunberger J, Xie S, Zhao Y, Tran B, Toms BS, Xu W, Plant-Hately A, Liptrott NJ, Dobrovolskaia MA. Nanoparticles may influence mast cells gene expression profiles without affecting their degranulation function. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2025; 66:102818. [PMID: 40185352 DOI: 10.1016/j.nano.2025.102818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/09/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
An in vitro method for monitoring nanoparticle effects on IgE-dependent mast cell degranulation was developed and validated. The assayed nanoparticles included four clinical-grade nanomedicines (Abraxane, Doxil, AmBisome, and Feraheme) and three commercial research-grade nanomaterials (generation 5 PAMAM dendrimers with carboxy-, hydroxy-, or amine- surface functionalities). Most of the tested materials did not alter IgE-dependent mast cell degranulation, suggesting that nanoparticles and nanomedicines are unlikely to worsen pre-existing allergies to other antigens. Two clinical-grade formulations containing cytotoxic oncology drugs-Abraxane and Doxil-decreased degranulation. Abraxane but not Doxil decreased FcεR expression on the cell surface. Single-cell sequencing revealed the most differentially expressed genes (DEG) in Abraxane and Doxil-treated cultures. Interestingly, Feraheme and amine-terminated dendrimers induced DEG without affecting degranulation. These data demonstrate that some nanomaterials have more effects on immune cells than can be detected by a functional immunoassay.
Collapse
Affiliation(s)
- Hannah S Newton
- Nanotechnology Characterization Lab., Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702, USA
| | - Edward Cedrone
- Nanotechnology Characterization Lab., Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702, USA
| | - Jason Grunberger
- Nanotechnology Characterization Lab., Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702, USA; Utah Center for Nanomedicine, Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, USA
| | - Shaojun Xie
- Sequencing Facility Bioinformatics Group, Biomedical Informatics and Data Science Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702, USA
| | - Yongmei Zhao
- Sequencing Facility Bioinformatics Group, Biomedical Informatics and Data Science Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702, USA
| | - Bao Tran
- CCR Sequencing Facility, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702, USA
| | | | - Weining Xu
- 10x Genomics, Inc., Pleasanton, CA 94588-3260, USA
| | - Alexander Plant-Hately
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L7 3NY, UK; Centre of Excellence for Long-Acting Therapeutics (CELT), Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L7 8TX, UK
| | - Neill J Liptrott
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L7 3NY, UK; Centre of Excellence for Long-Acting Therapeutics (CELT), Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L7 8TX, UK
| | - Marina A Dobrovolskaia
- Nanotechnology Characterization Lab., Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702, USA.
| |
Collapse
|
2
|
Elieh-Ali-Komi D, Shafaghat F, Alipoor SD, Kazemi T, Atiakshin D, Pyatilova P, Maurer M. Immunomodulatory Significance of Mast Cell Exosomes (MC-EXOs) in Immune Response Coordination. Clin Rev Allergy Immunol 2025; 68:20. [PMID: 39976807 PMCID: PMC11842441 DOI: 10.1007/s12016-025-09033-6] [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] [Accepted: 02/05/2025] [Indexed: 02/23/2025]
Abstract
Mast cells (MCs) communicate with other cells by direct cell-to-cell interaction, secreting mediators, and releasing exosomes (EXOs). MC-exosomes (MC-EXOs) contain proteins, lipids, mRNAs, and noncoding RNAs (ncRNAs), exhibit typical EXO markers such as heat shock proteins, tetraspanins, tumor susceptibility gene 101 protein (TSG101), and ALG-2-interacting protein X (ALIX), and are released constitutively or following MC degranulation. MC-EXOs also have signature MC markers like FcεRI and KIT (CD117), which allows for their identification and comparison with other EXO populations. Following their release, MC-EXOs may interact with the recipient cell(s) directly or be internalized and then release their protein and nucleic acid content. This may contribute to the regulation of immune responses and other biological processes and reprogramming of recipient cells. MC-EXO proteins may integrate and become a functional part of the recipient cell membrane. The mRNA transferred by MC-EXOs is functional and the transfer of exosomal RNA to other MCs results in the expression of donor MC proteins in the recipient MCs. Moreover, MCs may function as the recipients of EXOs that are released by other non-immune and immune cells, altering the secretome of MCs. In this review, we focus on how MC-EXOs modulate the biology of other cells and vice versa; and we highlight the role of MC-EXOs in the pathogenesis of allergic and non-allergic diseases.
Collapse
Affiliation(s)
- Daniel Elieh-Ali-Komi
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology, Berlin, Germany.
| | - Farzaneh Shafaghat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shamila D Alipoor
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dmitrii Atiakshin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultra-Structural Analysis Innovative Technologies, Peoples' Friendship University of Russia, 6 Miklukho-Maklaya St, 117198, Moscow, Russia
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036, Voronezh, Russia
| | - Polina Pyatilova
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology, Berlin, Germany
| | - Marcus Maurer
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology, Berlin, Germany
| |
Collapse
|
3
|
Pfeiffer A, Bandara G, Petersen JD, Falduto GH, Zimmerberg J, Metcalfe DD, Olivera A. Activation of the receptor KIT induces the secretion of exosome-like small extracellular vesicles. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e139. [PMID: 38938682 PMCID: PMC11080788 DOI: 10.1002/jex2.139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/01/2023] [Accepted: 01/01/2024] [Indexed: 06/29/2024]
Abstract
The receptor tyrosine kinase (RTK) KIT and its ligand stem cell factor (SCF) are essential for human mast cell (huMC) survival and proliferation. HuMCs expressing oncogenic KIT variants secrete large numbers of extracellular vesicles (EVs). The role KIT plays in regulating EV secretion has not been examined. Here, we investigated the effects of stimulation or inhibition of KIT activity on the secretion of small EVs (sEVs). In huMCs expressing constitutively active KIT, the quantity and quality of secreted sEVs positively correlated with the activity status of KIT. SCF-mediated stimulation of KIT in huMCs or murine MCs, or of transiently expressed KIT in HeLa cells, enhanced the release of sEVs expressing exosome markers. In contrast, ligand-mediated stimulation of the RTK EGFR in HeLa cells did not affect sEV secretion. The release of sEVs induced by either constitutively active or ligand-activated KIT was remarkably decreased when cells were treated with KIT inhibitors, concomitant with reduced exosome markers in sEVs. Similarly, inhibition of oncogenic KIT signalling kinases like PI3K, and MAPK significantly reduced the secretion of sEVs. Thus, activation of KIT and its early signalling cascades stimulate the secretion of exosome-like sEVs in a regulated fashion, which may have implications for KIT-driven functions.
Collapse
Affiliation(s)
- Annika Pfeiffer
- Mast Cell Biology SectionLaboratory of Allergic DiseasesNational Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMarylandUSA
| | - Geethani Bandara
- Mast Cell Biology SectionLaboratory of Allergic DiseasesNational Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMarylandUSA
| | - Jennifer D. Petersen
- Section on Integrative BiophysicsDivision of Basic and Translational BiophysicsEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthBethesdaMarylandUSA
- Inherited Movement Disorders UnitNeurogenetics BranchNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
| | - Guido H. Falduto
- Mast Cell Biology SectionLaboratory of Allergic DiseasesNational Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMarylandUSA
| | - Joshua Zimmerberg
- Section on Integrative BiophysicsDivision of Basic and Translational BiophysicsEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthBethesdaMarylandUSA
| | - Dean D. Metcalfe
- Mast Cell Biology SectionLaboratory of Allergic DiseasesNational Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMarylandUSA
| | - Ana Olivera
- Mast Cell Biology SectionLaboratory of Allergic DiseasesNational Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
4
|
Wu T, He J, Yan S, Li J, Chen K, Zhang D, Cheng M, Xiang Z, Fang Y. Human placental extract suppresses mast cell activation and induces mast cell apoptosis. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2023; 19:98. [PMID: 38012745 PMCID: PMC10683163 DOI: 10.1186/s13223-023-00850-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/18/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Human placental extract (HPE) has been documented to facilitate the healing of certain disorders including allergy. However, the effects of HPE on the functionality of mast cells, a critical cell type in allergic diseases, have not been reported. METHODS To investigate the effects of HPE on the regulation of allergy with respect to the biological functions of mast cells, the mast cell line C57 or HMC-1 cells were treated with HPE followed by the assessment of cell proliferation, apoptosis, activation, chemotaxis and phagocytosis. Mouse peritoneal mast cells were also investigated for their responses to induction of apoptosis by HPE in vivo. Furthermore, the effect of HPE on mast cell degranulation was confirmed using the passive cutaneous anaphylaxis (PCA) assay, an acute allergy model. RESULTS HPE was capable of suppressing mast cell proliferation and inducing mast cell apoptosis. Mast cell degranulation in response to compound 48/80- or anti-DNP IgE and DNP-mediated activation was suppressed. In addition, treatment with HPE compromised the production of cytokines by mast cells and cell chemotaxis. These observations were consistent with the dampened passive cutaneous anaphylaxis (PCA) assay following treatment with HPE. CONCLUSION This study revealed a suppressive effect of HPE on overall mast cell activities, suggesting a potential regulatory role of HPE on the alleviation of allergic diseases through mast cells.
Collapse
Affiliation(s)
- Tongqian Wu
- Center for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyi Street 28, Guiyang, 550004, Guizhou, China
- School of Laboratory Science, Guizhou Medical University, Guiyang, China
- Clinical Research Center, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jingjing He
- School of Laboratory Science, Guizhou Medical University, Guiyang, China
| | - Shirong Yan
- Center for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyi Street 28, Guiyang, 550004, Guizhou, China
- School of Laboratory Science, Guizhou Medical University, Guiyang, China
| | - Jing Li
- Center for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyi Street 28, Guiyang, 550004, Guizhou, China
- School of Laboratory Science, Guizhou Medical University, Guiyang, China
| | - Ke Chen
- School of Laboratory Science, Guizhou Medical University, Guiyang, China
| | - Dingshan Zhang
- School of Laboratory Science, Guizhou Medical University, Guiyang, China
| | - Mingliang Cheng
- Department of Infectious Disease, Affiliated Hospital of Guizhou Medical University, Guiyi Street 28, Guiyang, 550004, Guizhou, China.
| | - Zou Xiang
- Department of Health Technology and Informatics, Faculty of Health and Social Science, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Yu Fang
- Center for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyi Street 28, Guiyang, 550004, Guizhou, China.
- School of Laboratory Science, Guizhou Medical University, Guiyang, China.
| |
Collapse
|
5
|
Yong PYA, Yip AJW, Islam F, Hong HJ, Teh YE, Tham CL, Tan JW. The anti-allergic potential of stingless bee honey from different botanical sources via modulation of mast cell degranulation. BMC Complement Med Ther 2023; 23:307. [PMID: 37667314 PMCID: PMC10476411 DOI: 10.1186/s12906-023-04129-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 08/16/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Allergy is an inflammatory disorder affecting around 20% of the global population. The adverse effects of current conventional treatments give rise to the increased popularity of using natural food products as complementary and alternative medicine against allergic diseases. Stingless bee honey, commonly known as Kelulut honey (KH) in Malaysia, has been used locally as a traditional remedy to relieve cough and asthma. This study evaluated the anti-allergic potential of KH collected from four different botanical sources on phorbol ester 12-myristate-3-acetate and calcium ionophore-activated human mast cells. METHODS The present study examined the inhibitory effects of all collected honey on the release of selected inflammatory mediators, such as tumor necrosis factor-α (TNF-α), interleukin (IL)-4, IL-6, IL-8, histamine, and β-hexosaminidase in an activated HMC. Besides that, all honey's total phenolic content (TPC) was also examined, followed by using liquid chromatography with tandem mass spectrometry (LC-MS/MS) to identify the phytochemicals in the honey. Further examination of the identified phytochemicals on their potential interaction with selected signaling molecules in an activated mast cell was conducted using computational methods. RESULTS The results indicated that there were significant inhibitory effects on all selected inflammatory mediators' release by KH sourced from bamboo (BH) and rubber tree (RH) at 0.5% and 1%, but not KH sourced from mango (AH) and noni (EH). BH and RH were found to have higher TPC values and were rich in their phytochemical profiles based on the LC-MS/MS results. Computational studies were employed to determine the possible molecular target of KH through molecular docking using HADDOCK and PRODIGY web servers. CONCLUSIONS In short, the results indicated that KH possesses anti-allergic effects towards an activated HMC, possibly by targeting downstream MAPKs. However, their anti-allergic effects may vary according to their botanical sources. Nevertheless, the present study has provided insight into the potential application of stingless bee honey as a complementary and alternative medicine to treat various allergic diseases.
Collapse
Affiliation(s)
- Poi Yi Aw Yong
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, 47500, Selangor, Malaysia
| | - Ashley Jia Wen Yip
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, 47500, Selangor, Malaysia
| | - Fahmida Islam
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, 47500, Selangor, Malaysia
- Australian Regenerative Medicine Institute, Monash University, Clayton, 3800, VIC, Australia
| | - Hui Jing Hong
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, 47500, Selangor, Malaysia
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Pulau Pinang, 11800, Malaysia
| | - Yi En Teh
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, 47500, Selangor, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia.
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia.
| | - Ji Wei Tan
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, 47500, Selangor, Malaysia.
| |
Collapse
|
6
|
Proteins Derived from Cnidium officinale Makino React with Serum IgE of Allergic Patients and Stimulate ERK/NF-kB Activation in Human Mast Cell Line HMC-1 Cells. BIOCHIP JOURNAL 2021. [DOI: 10.1007/s13206-021-00008-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
7
|
Folkerts J, Gaudenzio N, Maurer M, Hendriks RW, Stadhouders R, Tam SY, Galli SJ. Rapid identification of human mast cell degranulation regulators using functional genomics coupled to high-resolution confocal microscopy. Nat Protoc 2020; 15:1285-1310. [PMID: 32060492 PMCID: PMC7197894 DOI: 10.1038/s41596-019-0288-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 12/18/2019] [Indexed: 12/26/2022]
Abstract
Targeted functional genomics represents a powerful approach for studying gene function in vivo and in vitro. However, its application to gene expression studies in human mast cells has been hampered by low yields of human mast cell cultures and their poor transfection efficiency. We developed an imaging system in which mast cell degranulation can be visualized in single cells subjected to shRNA knockdown or CRISPR-Cas 9 gene editing. By using high resolution confocal microscopy and a fluorochrome-labeled avidin probe, one can directly assess the suppression of functional responses, i.e. degranulation, in single human mast cells. The elimination of a drug or marker selection step avoids the use of potentially toxic treatment procedures and the short hands-on time of the functional analysis step enables the high-throughput screening of shRNA or CRISPR-Cas9 constructs to identify genes that regulate human mast cell degranulation. The ability to analyse single cells significantly reduces the total number of cells required, and allows for the parallel visualization of the degranulation profile of both edited and non-edited mast cells, offering a consistent internal control not found in other protocols. Moreover, our protocol offers a flexible choice between RNA interference and CRISPR-Cas9 genome editing for perturbation of gene expression using our human mast cell single-cell imaging system. Perturbation of gene expression, acquisition of microscopy data, and image analysis can be completed within 5 days, requiring only standard laboratory equipment and expertise. This protocol presents an an imaging system in which mast cell degranulation can be visualized in single cells subjected to shRNA knockdown or CRISPR-Cas 9 gene editing using high resolution confocal microscopy with a fluorochrome-labeled avidin probe.
Collapse
Affiliation(s)
- Jelle Folkerts
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA.,Department of Dermatology and Allergy, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, the Netherlands
| | - Nicolas Gaudenzio
- Unité de Différenciation Epithéliale et Autoimmunité Rhumatoïde (UDEAR), UMR 1056, INSERM, Université de Toulouse, Toulouse, France
| | - Marcus Maurer
- Department of Dermatology and Allergy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, the Netherlands
| | - Ralph Stadhouders
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, the Netherlands.,Department of Cell Biology, Erasmus MC Rotterdam, Rotterdam, the Netherlands
| | - See-Ying Tam
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA.
| | - Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA. .,Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, California, USA.
| |
Collapse
|
8
|
Abstract
Mast cells and basophils play a crucial role during type I hypersensitivity reactions. However, despite efforts to elucidate their role in the pathogenesis of allergy and inflammation, our understanding of MC and basophil biology is still relatively scarce. The practical difficulty in obtaining a sufficient number of purified primary cells from biological samples has slowed down the process of reaching a full understanding of the physiological role of these functionally similar cell types. The establishment of several immortalized cell lines has been a useful tool to establish and perform sophisticated laboratory protocols that are impractical using primary cells. Continuous cell lines have been extensively used to investigate allergen/IgE-mediated cell activation, to elucidate the degranulation dynamics, to investigate structural and functional properties of the high-affinity receptor (FcεRI), and to test cell-stabilizing compounds. In this chapter, we review the most widely used and better-characterized MC and basophil cell lines, highlighting their advantages and drawbacks. It must be pointed out, however, that while cell lines represent a useful in vitro tool due to their easy manipulability and reduced culture costs, they often show aberrant characteristics which are not fully representative of primary cell physiology; results obtained with such cells therefore must be interpreted with due care.
Collapse
|
9
|
A review on anti-inflammatory activity of green synthesized zinc oxide nanoparticle: Mechanism-based approach. Bioorg Chem 2019; 94:103423. [PMID: 31776035 DOI: 10.1016/j.bioorg.2019.103423] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/18/2019] [Accepted: 11/04/2019] [Indexed: 12/26/2022]
Abstract
Inflammation plays a very important role in the pathogenesis of various diseases like atherosclerosis, rheumatoid arthritis, asthma, and cancer. Lack of anti-inflammatory drugs and vectors provokes the need for developing new molecules for the management of inflammatory disorders. Nanotechnology has emerged as a wonderful research area in the past decade owing to its enhanced properties than bulk counterparts. This paper discusses the green synthesis of zinc oxide nanoparticle (ZnO NPs) and various characterization tools employed to comprehend the physiochemical properties of nanoparticles. ZnO NPs interaction with cells and its pharmacokinetic behavior inside the cells has also been discussed. The anti-inflammatory activity of ZnO NPs has been elucidated with the mechanism-based approach. A concise literature review has been included which summarizes the size, shape of ZnO NPs and the inflammatory model used for analyzing the anti-inflammatory activity of ZnO NPs. ZnO NPs potential offering towards anti-inflammatory activity like stable nature, selective targeting has been discussed briefly. The present study highlights the potential of ZnO NPs as an anti-inflammatory drug molecule or a vector for drug delivery.
Collapse
|
10
|
Yang DW, Kang OH, Lee YS, Han SH, Lee SW, Cha SW, Seo YS, Mun SH, Gong R, Shin DW, Kwon DY. Anti-inflammatory effect of salidroside on phorbol-12-myristate-13-acetate plus A23187-mediated inflammation in HMC-1 cells. Int J Mol Med 2016; 38:1864-1870. [PMID: 27779653 DOI: 10.3892/ijmm.2016.2781] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 09/27/2016] [Indexed: 11/05/2022] Open
Abstract
Salidroside [2-(4-hydroxyphenyl)ethyl β-D-gluco-pyranoside (SAS)] has been identified as the most potent ingredient of the plant Rhodiola rosea L. Previous studies have demonstrated that it possesses a number of pharmacological properties, including anti-aging, anti-fatigue, antioxidant, anticancer and anti-inflammatory properties. In this study, to ascertain the molecular mechanisms responsible for the anti-inflammatory activity of SAS, we used phorbol-12-myristate-13-acetate (PMA) plus A23187 to induce inflammation in human mast cell line-1 (HMC-1). The HMC-1 cells were treated with SAS prior to being stimulated with PMA plus A23187. Pro-inflammatory cytokine production was measured by enzyme-linked immunosorbent assay (ELISA) and reverse transcription-polymerase chain reaction (RT-PCR). Western blot analysis was used to examine the activation of mitogen-activated protein kinases (MAPKs) and nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB). SAS inhibited the mRNA expression and production of interleukin (IL)-6, IL-8 and tumor necrosis factor (TNF). In cells stimulated with PMA plus A23187, SAS suppressed the phosphorylation of extracellular signal-regulated kinase (ERK)1/2 and c-jun N-terminal kinase 1/2 (JNK1/2), but not that of p38 MAPK. SAS suppressed the expression of NF-κB in the nucleus. On the whole, our results suggest that SAS exerts an anti-inflammatory effect by inhibiting the production of pro-inflammatory cytokines through the blocking of the NF-κB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Da-Wun Yang
- BK21 Plus Team, Professional Graduate School of Oriental Medicine,Wonkwang University, Iksan, Jeonbuk 570‑749, Republic of Korea
| | - Ok-Hwa Kang
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang-Oriental Medicines Research Institute, Institute of Biotechnology, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| | - Young-Seob Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, Chungbuk 369-873, Republic of Korea
| | - Sin-Hee Han
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, Chungbuk 369-873, Republic of Korea
| | - Sang-Won Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, Chungbuk 369-873, Republic of Korea
| | - Seon-Woo Cha
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, Chungbuk 369-873, Republic of Korea
| | - Yun-Soo Seo
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang-Oriental Medicines Research Institute, Institute of Biotechnology, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| | - Su-Hyun Mun
- BK21 Plus Team, Professional Graduate School of Oriental Medicine,Wonkwang University, Iksan, Jeonbuk 570‑749, Republic of Korea
| | - Ryong Gong
- BK21 Plus Team, Professional Graduate School of Oriental Medicine,Wonkwang University, Iksan, Jeonbuk 570‑749, Republic of Korea
| | - Dong-Won Shin
- Department of Oriental Medicine Resources, Sunchon National University, Jeonnam 540-742, Republic of Korea
| | - Dong-Yeul Kwon
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang-Oriental Medicines Research Institute, Institute of Biotechnology, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| |
Collapse
|
11
|
Feltis B, Elbaz A, Wright P, Mackay G, Turney T, Lopata A. Characterizing the inhibitory action of zinc oxide nanoparticles on allergic-type mast cell activation. Mol Immunol 2015; 66:139-46. [DOI: 10.1016/j.molimm.2015.02.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 02/11/2015] [Accepted: 02/19/2015] [Indexed: 12/19/2022]
|
12
|
Balletta A, Lorenz D, Rummel A, Gerhard R, Bigalke H, Wegner F. Clostridium difficile toxin B inhibits the secretory response of human mast cell line-1 (HMC-1) cells stimulated with high free-Ca²⁺ and GTPγS. Toxicology 2014; 328:48-56. [PMID: 25497110 DOI: 10.1016/j.tox.2014.12.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 11/19/2014] [Accepted: 12/09/2014] [Indexed: 01/06/2023]
Abstract
Clostridium difficile toxins A and B (TcdA and TcdB) belong to the class of large clostridial cytotoxins and inactivate by glucosylation some low molecular mass GTPases of the Rho-family (predominantly Rho, Rac and Cdc42), known as regulators of the actin cytoskeleton. TcdA and B also represent the main virulence factors of the anaerobic gram-positive bacterium that is the causal agent of pseudomembranous colitis. In our study, TcdB was chosen instead of TcdA for the well-known higher cytotoxic potency. Inactivation of Rho-family GTPases by this toxin in our experimental conditions induced morphological changes and reduction of electron-dense mast cell-specific granules in human mast cell line-1 (HMC-1) cells, but not cell death or permeabilisation of plasma-membranes. Previously reported patch-clamp dialysis experiments revealed that high intracellular free-Ca(2+) and GTPγS concentrations are capable of inducing exocytosis as indicated by significant membrane capacitance (Cm) increases in HMC-1 cells. In this study, we investigated the direct effects of TcdB upon HMC-1 cell "stimulated" Cm increase, as well as on "constitutive" secretion of hexosaminidase and interleukin-16 (IL-16). Compared to untreated control cells, HMC-1 cells incubated with TcdB for 3-24h exhibited a significant reduction of the mean absolute and relative Cm increase in response to free-Ca(2+) and GTPγS suggesting an inhibition of secretory processes by TcdB. In conclusion, the HMC-1 cell line represents a suitable model for the study of direct effects of C. difficile toxins on human mast cell secretory activity.
Collapse
Affiliation(s)
- Andrea Balletta
- Department of Toxicology, Hannover Medical School, Carl Neuberg Str. 1, 30625 Hannover, Germany; Department of Neurology, Hannover Medical School, Carl Neuberg Str. 1, 30625 Hannover, Germany.
| | - Dorothea Lorenz
- Department of Cellular Imaging and Electron Microscopy, Leibniz Institute of Molecular Pharmacology, Robert Rössle Str. 10, 13125 Berlin, Germany.
| | - Andreas Rummel
- Department of Toxicology, Hannover Medical School, Carl Neuberg Str. 1, 30625 Hannover, Germany.
| | - Ralf Gerhard
- Department of Toxicology, Hannover Medical School, Carl Neuberg Str. 1, 30625 Hannover, Germany.
| | - Hans Bigalke
- Department of Toxicology, Hannover Medical School, Carl Neuberg Str. 1, 30625 Hannover, Germany.
| | - Florian Wegner
- Department of Neurology, Hannover Medical School, Carl Neuberg Str. 1, 30625 Hannover, Germany.
| |
Collapse
|
13
|
Balletta A, Lorenz D, Rummel A, Gerhard R, Bigalke H, Wegner F. Human mast cell line-1 (HMC-1) cells exhibit a membrane capacitance increase when dialysed with high free-Ca(2+) and GTPγS containing intracellular solution. Eur J Pharmacol 2013; 720:227-36. [PMID: 24406507 DOI: 10.1016/j.ejphar.2013.10.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 10/04/2013] [Accepted: 10/16/2013] [Indexed: 01/17/2023]
Abstract
An increase in cytosolic free calcium concentration [Ca(2+)]i initiates the exocytotic activity in various types of secretory cells. The guanosine 5'-O-[3-thio]triphosphate (GTPγS), a non-hydrolysable analogue of GTP (guanosine 5'-triphosphate), is an effective secretagogue for different cell types of different species, like mast cells, neutrophils or eosinophils. Consequently, the internal administration of GTPγS causes degranulation of mouse and rat mast cells. Regarding rat mast cells, it is proved that Ca(2+) can cooperate with GTP or GTPγS in accelerating and increasing amplitude of the secretory response. All the previous studies with respect to capacitance recordings and mast cells were performed using mouse or rat mast cells, usually derived from peritoneum or the rat basophilic leukaemia cell line RBL. In this study, we applied the capacitance measurement technique to the human mast cell line-1 (HMC-1) cells, an immature cell line established from a patient with mast cell leukaemia. Patch-clamp dialysis experiments revealed that high intracellular free Ca(2+) and GTPγS concentrations are both required for considerable capacitance increases in HMC-1 cells. During degranulation of HMC-1 cells, the total membrane capacitance (Cm) increase appeared continuously and, in some cases, as a discrete capacitance change, developing in a stepwise manner. Then, we tested the effect of latrunculin B upon HMC-1 cell capacitance increase as well as of some classic mast cell stimulators like PMA, A23187 and IL-1β in hexosaminidase release. Finally, we could conclude that the HMC-1 cell line represents a suitable model for the study of human mast cell degranulation.
Collapse
Affiliation(s)
- Andrea Balletta
- Department of Toxicology, Hannover Medical School, Carl Neuberg Str. 1, 30625 Hannover, Germany; Department of Neurology, Hannover Medical School, Carl Neuberg Str. 1, 30625 Hannover, Germany.
| | - Dorothea Lorenz
- Department of Cellular Imaging and Electron Microscopy, Leibniz Institute of Molecular Pharmacology, Robert Rössle Str. 10, 13125 Berlin, Germany.
| | - Andreas Rummel
- Department of Toxicology, Hannover Medical School, Carl Neuberg Str. 1, 30625 Hannover, Germany.
| | - Ralf Gerhard
- Department of Toxicology, Hannover Medical School, Carl Neuberg Str. 1, 30625 Hannover, Germany.
| | - Hans Bigalke
- Department of Toxicology, Hannover Medical School, Carl Neuberg Str. 1, 30625 Hannover, Germany.
| | - Florian Wegner
- Department of Neurology, Hannover Medical School, Carl Neuberg Str. 1, 30625 Hannover, Germany.
| |
Collapse
|
14
|
Xia YC, Redhu NS, Moir LM, Koziol-White C, Ammit AJ, Al-Alwan L, Camoretti-Mercado B, Clifford RL. Pro-inflammatory and immunomodulatory functions of airway smooth muscle: Emerging concepts. Pulm Pharmacol Ther 2013; 26:64-74. [DOI: 10.1016/j.pupt.2012.05.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 05/08/2012] [Accepted: 05/09/2012] [Indexed: 12/22/2022]
|
15
|
Xia YC, Harris T, Stewart AG, Mackay GA. Secreted factors from human mast cells trigger inflammatory cytokine production by human airway smooth muscle cells. Int Arch Allergy Immunol 2012; 160:75-85. [PMID: 22948287 DOI: 10.1159/000339697] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 05/21/2012] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND A notable feature of allergic asthma is the infiltration of mast cells into smooth muscle in the human airway. Thus, mast cells and human airway smooth muscle (hASM) cells are likely to exhibit mutual functional modulation via direct cell-cell contact or through released factors. This study examined mast cell modulation of hASM cell cytokine release. METHODS The mast cell line HMCα was used to model mast cell function. hASM cells were either co-cultured directly with resting or IgE/antigen-stimulated HMCα cells or treated with HMCα-conditioned media to examine the impact on cytokine release. The activation pathways triggered in hASM cells by the mast cell-derived factors were examined through the use of selective inhibitors and by Western blotting. RESULTS HMCα cells, or their conditioned media, induced the expression of cytokines (IL-8 and IL-6) by hASM cells at both the mRNA and the protein level. Cytokine expression in hASM cells was greatly amplified when HMCα cells were IgE/antigen-activated. The effects of the conditioned media were not mediated by the chemokines MCP-1 and MIP-1α or by exosomes. While the mast cell-derived factor(s) increased p38(MAPK) phosphorylation in hASM cells, cytokine production was not inhibited by the p38(MAPK) inhibitor SB203580. hASM cell production of IL-8 induced by HMCα condition media but not IL-6 was, however, attenuated by the Src tyrosine kinase inhibitor PP2. CONCLUSIONS Our study shows that the release of soluble mediators by activated mast cells can stimulate hASM cells to elicit production of proinflammatory cytokines that may then exacerbate airway inflammation in asthma.
Collapse
Affiliation(s)
- Y C Xia
- Department of Pharmacology, University of Melbourne, Parkville, Vic., Australia
| | | | | | | |
Collapse
|
16
|
The role of P2Y(14) and other P2Y receptors in degranulation of human LAD2 mast cells. Purinergic Signal 2012; 9:31-40. [PMID: 22825617 DOI: 10.1007/s11302-012-9325-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 06/21/2012] [Indexed: 10/28/2022] Open
Abstract
Mast cell degranulation affects many conditions, e.g., asthma and urticaria. We explored the potential role of the P2Y(14) receptor (P2Y(14)R) and other P2Y subtypes in degranulation of human LAD2 mast cells. All eight P2YRs were expressed at variable levels in LAD2 cells (quantitative real-time RT-PCR). Gene expression levels of ADP receptors, P2Y(1)R, P2Y(12)R, and P2Y(13)R, were similar, and P2Y(11)R and P2Y(4)R were highly expressed at 5.8- and 3.8-fold of P2Y(1)R, respectively. Least expressed P2Y(2)R was 40-fold lower than P2Y(1)R, and P2Y(6)R and P2Y(14)R were ≤50 % of P2Y(1)R. None of the native P2YR agonists alone induced β-hexosaminidase (β-Hex) release, but some nucleotides significantly enhanced β-Hex release induced by C3a or antigen, with a rank efficacy order of ATP > UDPG ≥ ADP >> UDP, UTP. Although P2Y(11)R and P2Y(4)R are highly expressed, they did not seem to play a major role in degranulation as neither P2Y(4)R agonist UTP nor P2Y(11)R agonists ATPγS and NF546 had a substantial effect. P2Y(1)R-selective agonist MRS2365 enhanced degranulation, but ~1,000-fold weaker compared to its P2Y(1)R potency, and the effect of P2Y(6)R agonist 3-phenacyl-UDP was negligible. The enhancement by ADP and ATP appears mediated via multiple receptors. Both UDPG and a synthetic agonist of the P2Y(14)R, MRS2690, enhanced C3a-induced β-Hex release, which was inhibited by a P2Y(14)R antagonist, specific P2Y(14)R siRNA and pertussis toxin, suggesting a role of P2Y(14)R activation in promoting human mast cell degranulation.
Collapse
|
17
|
Yeganeh B, Xia C, Movassagh H, Koziol-White C, Chang Y, Al-Alwan L, Bourke JE, Oliver BGG. Emerging mediators of airway smooth muscle dysfunction in asthma. Pulm Pharmacol Ther 2012; 26:105-11. [PMID: 22776693 DOI: 10.1016/j.pupt.2012.06.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 06/27/2012] [Accepted: 06/27/2012] [Indexed: 12/26/2022]
Abstract
Phenotypic changes in airway smooth muscle are integral to the pathophysiological changes that constitute asthma - namely inflammation, airway wall remodelling and bronchial hyperresponsiveness. In vitro and in vivo studies have shown that the proliferative, secretory and contractile functions of airway smooth muscle are dysfunctional in asthma. These functions can be modulated by various mediators whose levels are altered in asthma, derived from inflammatory cells or produced by airway smooth muscle itself. In this review, we describe the emerging roles of the CXC chemokines (GROs, IP-10), Th17-derived cytokines (IL-17, IL-22) and semaphorins, as well as the influence of viral infection on airway smooth muscle function, with a view to identifying new opportunities for therapeutic intervention in asthma.
Collapse
Affiliation(s)
- Behzad Yeganeh
- Department of Physiology, Manitoba Institute of Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Antibody repertoire complexity and effector cell biology determined by assays for IgE-mediated basophil and T-cell activation. J Immunol Methods 2012; 383:4-20. [PMID: 22683539 DOI: 10.1016/j.jim.2012.05.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 05/23/2012] [Accepted: 05/29/2012] [Indexed: 11/23/2022]
Abstract
Effector cell activation and T-cell activation, the latter mediated by facilitated antigen presentation, are immunological mechanisms that play crucial roles in the manifestation and maintenance of allergic disease. In addition to their relevance for the pathogenesis of allergy in-vivo, in-vitro assays based on these immunological mechanisms have been established and used for diagnostics, for monitoring the progression of disease and for the effect of specific immunotherapy as well as for basic research purposes. Here we review different parameters that affect effector cell activation and facilitated antigen uptake and presentation, including assay designs, readout parameters and critical experimental conditions. Central to the two immunological mechanisms is complex formation between allergen-specific IgE, allergen, and cell surface-anchored immunoglobulin receptor; the high affinity IgE-receptor FcεRI on basophils and mast cells, and the low affinity IgE-receptor FcεRII (CD23) on B-cells. Accordingly, the effect of IgE repertoire complexity and allergen diversity on effector cell and facilitated antigen presentation is discussed in detail.
Collapse
|