1
|
Mihlan M, Wissmann S, Gavrilov A, Kaltenbach L, Britz M, Franke K, Hummel B, Imle A, Suzuki R, Stecher M, Glaser KM, Lorentz A, Carmeliet P, Yokomizo T, Hilgendorf I, Sawarkar R, Diz-Muñoz A, Buescher JM, Mittler G, Maurer M, Krause K, Babina M, Erpenbeck L, Frank M, Rambold AS, Lämmermann T. Neutrophil trapping and nexocytosis, mast cell-mediated processes for inflammatory signal relay. Cell 2024; 187:5316-5335.e28. [PMID: 39096902 DOI: 10.1016/j.cell.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 04/10/2024] [Accepted: 07/08/2024] [Indexed: 08/05/2024]
Abstract
Neutrophils are sentinel immune cells with essential roles for antimicrobial defense. Most of our knowledge on neutrophil tissue navigation derived from wounding and infection models, whereas allergic conditions remained largely neglected. Here, we analyzed allergen-challenged mouse tissues and discovered that degranulating mast cells (MCs) trap living neutrophils inside them. MCs release the attractant leukotriene B4 to re-route neutrophils toward them, thus exploiting a chemotactic system that neutrophils normally use for intercellular communication. After MC intracellular trap (MIT) formation, neutrophils die, but their undigested material remains inside MC vacuoles over days. MCs benefit from MIT formation, increasing their functional and metabolic fitness. Additionally, they are more pro-inflammatory and can exocytose active neutrophilic compounds with a time delay (nexocytosis), eliciting a type 1 interferon response in surrounding macrophages. Together, our study highlights neutrophil trapping and nexocytosis as MC-mediated processes, which may relay neutrophilic features over the course of chronic allergic inflammation.
Collapse
Affiliation(s)
- Michael Mihlan
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Münster 48149, Germany.
| | - Stefanie Wissmann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Institute for Biomechanics, ETH Zürich, Zürich 8092, Switzerland
| | - Alina Gavrilov
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Roche Pharma Research and Early Development (pRED), Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology (CMI2O), Roche Innovation Center, Basel 4070, Switzerland
| | - Lukas Kaltenbach
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Marie Britz
- Department of Dermatology, Universitätsklinikum Münster, Münster 48149, Germany
| | - Kristin Franke
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Berlin 12203, Germany; Charité-Universitätsmedizin Berlin, Institute of Allergology, Berlin 12203, Germany
| | - Barbara Hummel
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Andrea Imle
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg 69117, Germany
| | - Ryo Suzuki
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Manuel Stecher
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Münster 48149, Germany
| | - Katharina M Glaser
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Institut Curie, PSL Research University, INSERM U932, Paris 75005, France
| | - Axel Lorentz
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart 70593, Germany
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium; Center for Biotechnology, Khalifa University of Science and Technology, PO Box 127788, Abu Dhabi, United Arab Emirates
| | - Takehiko Yokomizo
- Department of Biochemistry, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center and Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Ritwick Sawarkar
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Medical Research Council (MRC) Toxicology Unit and Department of Genetics, University of Cambridge, Cambridge CB21QR, UK
| | - Alba Diz-Muñoz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg 69117, Germany
| | - Joerg M Buescher
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Gerhard Mittler
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Marcus Maurer
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Berlin 12203, Germany; Charité-Universitätsmedizin Berlin, Institute of Allergology, Berlin 12203, Germany
| | - Karoline Krause
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Berlin 12203, Germany; Charité-Universitätsmedizin Berlin, Institute of Allergology, Berlin 12203, Germany
| | - Magda Babina
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Berlin 12203, Germany; Charité-Universitätsmedizin Berlin, Institute of Allergology, Berlin 12203, Germany
| | - Luise Erpenbeck
- Department of Dermatology, Universitätsklinikum Münster, Münster 48149, Germany
| | - Marcus Frank
- Medical Biology and Electron Microscopy Center, Rostock University Medical Center, Rostock 18057, Germany; Department Life, Light and Matter, Rostock University, Rostock 18051, Germany
| | - Angelika S Rambold
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Tim Lämmermann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Münster 48149, Germany.
| |
Collapse
|
2
|
Sharma A, Deshmukh P, Jain S, Gaurkar S, Sharma A. Unraveling the Molecular Threads: A Comprehensive Review of the Pathogenesis and Therapeutic Insights Into Allergic Rhinitis. Cureus 2024; 16:e64410. [PMID: 39131035 PMCID: PMC11317112 DOI: 10.7759/cureus.64410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 07/12/2024] [Indexed: 08/13/2024] Open
Abstract
Allergic rhinitis (AR) is a prevalent inflammatory disorder of the nasal mucosa, triggered by allergen exposure and characterized by symptoms such as sneezing, nasal congestion, itching, and rhinorrhea. This comprehensive review aims to unravel the molecular mechanisms underpinning AR, exploring the pathogenesis from allergen recognition to chronic inflammation and tissue remodelling. Central to the disease are immunoglobulin E (IgE)-mediated hypersensitivity reactions, involving key inflammatory mediators and cellular players such as mast cells, eosinophils, and T cells. Genetic predisposition and environmental factors also play significant roles in susceptibility and disease progression. Therapeutic strategies for AR are varied, ranging from symptomatic relief through antihistamines and nasal corticosteroids to more targeted approaches like allergen-specific immunotherapy. Emerging treatments focus on novel molecular pathways, with a growing emphasis on personalized medicine to optimize patient outcomes. Despite advancements, challenges remain in fully understanding the heterogeneity of AR and developing universally effective treatments. This review synthesizes current knowledge, highlighting critical insights into the molecular basis of AR and their implications for clinical practice. It underscores the need for integrated, multidisciplinary approaches to enhance therapeutic efficacy and calls for ongoing research to address unresolved questions and explore new frontiers in AR management. Through this comprehensive synthesis, the review aims to inform and inspire future research and clinical strategies, ultimately improving the quality of life for individuals affected by AR.
Collapse
Affiliation(s)
- Abhijeet Sharma
- Otorhinolaryngology and Head and Neck Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Prasad Deshmukh
- Otorhinolaryngology and Head and Neck Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Shraddha Jain
- Otorhinolaryngology and Head and Neck Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sagar Gaurkar
- Otorhinolaryngology and Head and Neck Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Ayushi Sharma
- Dental Sciences, People's College of Dental Science and Research Centre, Bhopal, IND
| |
Collapse
|
3
|
Yilmaz D, Sahin E. All-cause mortality and related factors in patients with varying degrees of peripheral blood eosinophilia. Medicine (Baltimore) 2024; 103:e38359. [PMID: 39259127 PMCID: PMC11142823 DOI: 10.1097/md.0000000000038359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/26/2024] [Accepted: 05/03/2024] [Indexed: 09/12/2024] Open
Abstract
The aim of this study was to investigate all-cause mortality rates and related factors in patients with different levels of eosinophilia. This retrospective cohort study was conducted between January 2020 and December 2022 in the Internal Medicine Department of Dr Sadi Konuk Training and Research Hospital, Istanbul, Turkiye. A total of 161 patients with eosinophilia (at least 3 times) were included and divided into groups with absolute eosinophil counts of 500-999/µL (mild), 1000-1500/µL (moderate), and >1500/µL (severe). The mean age of patients was 65.67 ± 16.64 years at the time of admission, and 45 patients (57.8%) were male. The rates of mortality, oncological disease, and organ involvement were significantly higher in the severe group (P < .05). Increased serum total immunoglobulin E and vitamin B12, hematocrit value, eosinophil-to-lymphocyte ratio, and leukocyte were observed in eosinophilic patients. Decreased lymphocyte count, hemoglobin and hematocrit values were higher in deceased patients than in survivors (P < .05). Increased eosinophil-to-lymphocyte ratio, C-reactive protein, vitamin B12, and lactate dehydrogenase (LDH) activity were observed in participants who died compared to those who survived (P < .05). Multivariable logistic regression revealed that advanced age and higher LDH activity were independently associated with greater mortality risk while receiving non-steroid anti-inflammatory drugs or proton-pump inhibitors were associated with reduced mortality risk (P < .05). Advanced age and increased LDH activity were independently associated with greater risk for mortality, whereas absolute eosinophil counts was not. Considering the literature on this topic, our results show the need for further clinical and fundamental research to understand the role of eosinophils in human disease.
Collapse
Affiliation(s)
- Deniz Yilmaz
- Department of Internal Diseases, University of Health Sciences Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Ezgi Sahin
- Department of Rheumatology, Istanbul University Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
4
|
Volarić M, Šojat D, Majnarić LT, Vučić D. The Association between Functional Dyspepsia and Metabolic Syndrome-The State of the Art. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:237. [PMID: 38397726 PMCID: PMC10888556 DOI: 10.3390/ijerph21020237] [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: 01/10/2024] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024]
Abstract
Functional dyspepsia is a common functional disorder of the gastrointestinal tract that is responsible for many primary care visits. No organic changes have been found to explain its symptoms. We hypothesize that modern lifestyles and environmental factors, especially psychological stress, play a crucial role in the high prevalence of functional dyspepsia and metabolic syndrome. While gastrointestinal tract diseases are rarely linked to metabolic disorders, chronic stress, obesity-related metabolic syndrome, chronic inflammation, intestinal dysbiosis, and functional dyspepsia have significant pathophysiological associations. Functional dyspepsia, often associated with anxiety and chronic psychological stress, can activate the neuroendocrine stress axis and immune system, leading to unhealthy habits that contribute to obesity. Additionally, intestinal dysbiosis, which is commonly present in functional dyspepsia, can exacerbate systemic inflammation and obesity, further promoting metabolic syndrome-related disorders. It is worth noting that the reverse is also true: obesity-related metabolic syndrome can worsen functional dyspepsia and its associated symptoms by triggering systemic inflammation and intestinal dysbiosis, as well as negative emotions (depression) through the brain-gut axis. To understand the pathophysiology and deliver an effective treatment strategy for these two difficult-to-cure disorders, which are challenging for both caregivers and patients, a psychosocial paradigm is essential.
Collapse
Affiliation(s)
- Mile Volarić
- Department of Family Medicine, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, 31000 Osijek, Croatia; (M.V.); (L.T.M.)
- Department of Gastroenterology and Hepatology, School of Medicine, University of Mostar Clinical Hospital, University of Mostar, Bijeli Brijeg bb, 88000 Mostar, Bosnia and Herzegovina
| | - Dunja Šojat
- Department of Family Medicine, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, 31000 Osijek, Croatia; (M.V.); (L.T.M.)
| | - Ljiljana Trtica Majnarić
- Department of Family Medicine, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, 31000 Osijek, Croatia; (M.V.); (L.T.M.)
| | - Domagoj Vučić
- Department of Cardiology, General Hospital “Dr. Josip Benčević”, A. Štampara, 35105 Slavonski Brod, Croatia;
| |
Collapse
|
5
|
Tiligada E, Gafarov D, Zaimi M, Vitte J, Levi-Schaffer F. Novel Immunopharmacological Drugs for the Treatment of Allergic Diseases. Annu Rev Pharmacol Toxicol 2024; 64:481-506. [PMID: 37722722 DOI: 10.1146/annurev-pharmtox-051623-091038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2023]
Abstract
The exponential rise in the prevalence of allergic diseases since the mid-twentieth century has led to a genuine public health emergency and has also fostered major progress in research on the underlying mechanisms and potential treatments. The management of allergic diseases benefits from the biological revolution, with an array of novel immunomodulatory therapeutic and investigational tools targeting players of allergic inflammation at distinct pathophysiological steps. Prominent examples include therapeutic monoclonal antibodies against cytokines, alarmins, and their receptors, as well as small-molecule modifiers of signal transduction mainly mediated by Janus kinases and Bruton's tyrosine kinases. However, the first-line therapeutic options have yet to switch from symptomatic to disease-modifying interventions. Here we present an overview of available drugs in the context of our current understanding of allergy pathophysiology, identify potential therapeutic targets, and conclude by providing a selection of candidate immunopharmacological molecules under investigation for potential future use in allergic diseases.
Collapse
Affiliation(s)
- Ekaterini Tiligada
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel;
| | - Daria Gafarov
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel;
| | - Maria Zaimi
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Joana Vitte
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel;
- Desbrest Institute of Epidemiology and Public Health, University of Montpellier, INSERM
- Montpellier, France
| | - Francesca Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel;
| |
Collapse
|
6
|
Vraila M, Asp E, Melo FR, Grujic M, Rollman O, Pejler G, Lampinen M. Monensin induces secretory granule-mediated cell death in eosinophils. J Allergy Clin Immunol 2023; 152:1312-1320.e3. [PMID: 37536509 DOI: 10.1016/j.jaci.2023.07.012] [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: 08/24/2022] [Revised: 07/08/2023] [Accepted: 07/19/2023] [Indexed: 08/05/2023]
Abstract
BACKGROUND Eosinophils contribute to the pathology of several types of disorders, in particular of allergic nature, and strategies to limit their actions are therefore warranted. OBJECTIVE We sought to evaluate the possibility of targeting the acidic, lysosome-like eosinophil granules as a potential means of inducing eosinophil cell death. METHODS To this end, we used monensin, an ionophoric drug that has previously been shown to permeabilize the secretory granules of mast cells, thereby inducing cell death. RESULTS Our findings reveal that monensin induces cell death in human eosinophils, whereas neutrophils were less affected. Blockade of granule acidification reduced the effect of monensin on the eosinophils, demonstrating that granule acidity is an important factor in the mechanism of cell death. Furthermore, monensin caused an elevation of the granule pH, which was accompanied by a decrease of the cytosolic pH, hence indicating that monensin caused leakage of acidic contents from the granules into the cytosol. In agreement with a granule-targeting mechanism, transmission electron microscopy analysis revealed that monensin caused extensive morphological alterations of the eosinophil granules, as manifested by a marked loss of electron density. Eosinophil cell death in response to monensin was caspase-independent, but dependent on granzyme B, a pro-apoptotic serine protease known to be expressed by eosinophils. CONCLUSIONS We conclude that monensin causes cell death of human eosinophils through a granule-mediated mechanism dependent on granzyme B.
Collapse
Affiliation(s)
- Marianthi Vraila
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Elin Asp
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Fabio Rabelo Melo
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Mirjana Grujic
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Ola Rollman
- Department of Medical Sciences, Dermatology and Venereology, Uppsala University, Uppsala, Sweden
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Maria Lampinen
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Dermatology and Venereology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
7
|
Cao Y, Rische CH, Bochner BS, O’Sullivan JA. Interactions between Siglec-8 and endogenous sialylated cis ligands restrain cell death induction in human eosinophils and mast cells. Front Immunol 2023; 14:1283370. [PMID: 37928558 PMCID: PMC10623328 DOI: 10.3389/fimmu.2023.1283370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
Sialic acid-binding immunoglobulin-like lectin (Siglec)-8 is a sialoside-binding receptor expressed by eosinophils and mast cells that exhibits priming status- and cell type-dependent inhibitory activity. On eosinophils that have been primed with IL-5, GM-CSF, or IL-33, antibody ligation of Siglec-8 induces cell death through a pathway involving the β2 integrin-dependent generation of reactive oxygen species (ROS) via NADPH oxidase. In contrast, Siglec-8 engagement on mast cells inhibits cellular activation and mediator release but reportedly does not impact cell viability. The differences in responses between cytokine-primed and unprimed eosinophils, and between eosinophils and mast cells, to Siglec-8 ligation are not understood. We previously found that Siglec-8 binds to sialylated ligands present on the surface of the same cell (so-called cis ligands), preventing Siglec-8 ligand binding in trans. However, the functional relevance of these cis ligands has not been elucidated. We therefore explored the potential influence of cis ligands of Siglec-8 on both eosinophils and mast cells. De-sialylation using exogenous sialidase profoundly altered the consequences of Siglec-8 antibody engagement on both cell types, eliminating the need for cytokine priming of eosinophils to facilitate cell death and enabling Siglec-8-dependent mast cell death without impacting anti-Siglec-8 antibody binding. The cell death process licensed by de-sialylation resembled that characterized in IL-5-primed eosinophils, including CD11b upregulation, ROS production, and the activities of Syk, PI3K, and PLC. These results implicate cis ligands in restraining Siglec-8 function on eosinophils and mast cells and reveal a promising approach to the selective depletion of mast cells in patients with mast cell-mediated diseases.
Collapse
Affiliation(s)
- Yun Cao
- Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Clayton H. Rische
- Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Biomedical Engineering, Northwestern University McCormick School of Engineering, Evanston, IL, United States
| | - Bruce S. Bochner
- Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jeremy A. O’Sullivan
- Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
8
|
O'Sullivan JA, Youngblood BA, Schleimer RP, Bochner BS. Siglecs as potential targets of therapy in human mast cell- and/or eosinophil-associated diseases. Semin Immunol 2023; 69:101799. [PMID: 37413923 PMCID: PMC10528103 DOI: 10.1016/j.smim.2023.101799] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Siglecs (sialic acid-binding immunoglobulin-like lectins) are a family of vertebrate glycan-binding cell-surface proteins. The majority mediate cellular inhibitory activity once engaged by specific ligands or ligand-mimicking molecules. As a result, Siglec engagement is now of interest as a strategy to therapeutically dampen unwanted cellular responses. When considering allergic inflammation, human eosinophils and mast cells express overlapping but distinct patterns of Siglecs. For example, Siglec-6 is selectively and prominently expressed on mast cells while Siglec-8 is highly specific for both eosinophils and mast cells. This review will focus on a subset of Siglecs and their various endogenous or synthetic sialoside ligands that regulate eosinophil and mast cell function and survival. It will also summarize how certain Siglecs have become the focus of novel therapies for allergic and other eosinophil- and mast cell-related diseases.
Collapse
Affiliation(s)
- Jeremy A O'Sullivan
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Robert P Schleimer
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bruce S Bochner
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
9
|
Atiakshin D, Kostin A, Volodkin A, Nazarova A, Shishkina V, Esaulenko D, Buchwalow I, Tiemann M, Noda M. Mast Cells as a Potential Target of Molecular Hydrogen in Regulating the Local Tissue Microenvironment. Pharmaceuticals (Basel) 2023; 16:817. [PMID: 37375765 DOI: 10.3390/ph16060817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/18/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Knowledge of the biological effects of molecular hydrogen (H2), hydrogen gas, is constantly advancing, giving a reason for the optimism in several healthcare practitioners regarding the management of multiple diseases, including socially significant ones (malignant neoplasms, diabetes mellitus, viral hepatitis, mental and behavioral disorders). However, mechanisms underlying the biological effects of H2 are still being actively debated. In this review, we focus on mast cells as a potential target for H2 at the specific tissue microenvironment level. H2 regulates the processing of pro-inflammatory components of the mast cell secretome and their entry into the extracellular matrix; this can significantly affect the capacity of the integrated-buffer metabolism and the structure of the immune landscape of the local tissue microenvironment. The analysis performed highlights several potential mechanisms for developing the biological effects of H2 and offers great opportunities for translating the obtained findings into clinical practice.
Collapse
Affiliation(s)
- Dmitri Atiakshin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia Named after Patrice Lumumba, 117198 Moscow, Russia
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Andrey Kostin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia Named after Patrice Lumumba, 117198 Moscow, Russia
| | - Artem Volodkin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia Named after Patrice Lumumba, 117198 Moscow, Russia
| | - Anna Nazarova
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia Named after Patrice Lumumba, 117198 Moscow, Russia
| | - Viktoriya Shishkina
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Dmitry Esaulenko
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Igor Buchwalow
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia Named after Patrice Lumumba, 117198 Moscow, Russia
- Institute for Hematopathology, Fangdieckstr. 75a, 22547 Hamburg, Germany
| | - Markus Tiemann
- Institute for Hematopathology, Fangdieckstr. 75a, 22547 Hamburg, Germany
| | - Mami Noda
- Laboratory of Pathophysiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 816-0811, Japan
| |
Collapse
|
10
|
Puzzovio PG, Pahima H, George T, Mankuta D, Eliashar R, Tiligada E, Levy BD, Levi-Schaffer F. Mast cells contribute to the resolution of allergic inflammation by releasing resolvin D1. Pharmacol Res 2023; 189:106691. [PMID: 36773709 PMCID: PMC10285510 DOI: 10.1016/j.phrs.2023.106691] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023]
Abstract
BACKGROUND Mast cells are initiators and main effectors of allergic inflammation, together with eosinophils, with whom they can interact in a physical and soluble cross-talk with marked pro-inflammatory features, the Allergic Effector Unit. The pro-resolution role of mast cells, alone or in co-culture with eosinophils, has not been characterized yet. OBJECTIVES We aimed to investigate select pro-resolution pathways in mast cells in vitro and in vivo in allergic inflammation. METHODS In vitro, we employed human and murine mast cells and analyzed release of resolvin D1 and expression of 15-lipoxygenase after IgE-mediated activation. We performed co-culture of IgE-activated mast cells with peripheral blood eosinophils and investigated 15-lipoxygenase expression and Resolvin D1 release. In vivo, we performed Ovalbumin/Alum and Ovalbumin/S. aureus enterotoxin B allergic peritonitis model in Wild Type mice following a MC "overshoot" protocol. RESULTS We found that IgE-activated mast cells release significant amounts of resolvin D1 30 min after activation, while 15-lipoxygenase expression remained unchanged. Resolvin D1 release was found to be decreased in IgE-activated mast cells co-cultured with peripheral blood eosinophils for 30 min In vivo, mast cell-overshoot mice exhibited a trend of reduced inflammation, together with increased peritoneal resolvin D1 release. CONCLUSIONS Mast cells can actively contribute to resolution of allergic inflammation by releasing resolvin D1.
Collapse
Affiliation(s)
- Pier Giorgio Puzzovio
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hadas Pahima
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tresa George
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - David Mankuta
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Ron Eliashar
- Department of Otolaryngology / Head and Neck Surgery, Hadassah Hebrew University Medical Center and the Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ekaterini Tiligada
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel; Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Bruce D Levy
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Francesca Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
11
|
Mast Cells in Regeneration of the Skin in Burn Wound with Special Emphasis on Molecular Hydrogen Effect. Pharmaceuticals (Basel) 2023; 16:ph16030348. [PMID: 36986447 PMCID: PMC10059032 DOI: 10.3390/ph16030348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
The mechanisms of regeneration for the fibrous component of the connective tissue of the dermis are still insufficiently studied. The aim of this study was to evaluate the effectiveness of the use of molecular hydrogen on the local therapy of a II degree burn wound with the intensification of collagen fibrillogenesis in the skin. We analyzed the involvement of mast cells (MCs) in the regeneration of the collagen fibers of the connective tissue using water with a high content of molecular hydrogen and in a therapeutic ointment for the cell wounds. Thermal burns led to an increase in the skin MC population, accompanied by a systemic rearrangement of the extracellular matrix. The use of molecular hydrogen for the treatment of burn wounds stimulated the regeneration processes by activating the formation of the fibrous component of the dermis, accelerating wound healing. Thus, the intensification of collagen fibrillogenesis was comparable to the effects of a therapeutic ointment. The remodeling of the extracellular matrix correlated with a decrease in the area of damaged skin. Skin regeneration induced by the activation of the secretory activity of MCs may be one of the possible points of implementation of the biological effects of molecular hydrogen in the treatment of burn wounds. Thus, the positive effects of molecular hydrogen on skin repair can be used in clinical practice to increase the effectiveness of therapy after thermal exposure.
Collapse
|
12
|
Zhou B, Li J, Liu R, Zhu L, Peng C. The Role of Crosstalk of Immune Cells in Pathogenesis of Chronic Spontaneous Urticaria. Front Immunol 2022; 13:879754. [PMID: 35711438 PMCID: PMC9193815 DOI: 10.3389/fimmu.2022.879754] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 05/02/2022] [Indexed: 12/15/2022] Open
Abstract
Chronic spontaneous urticaria (CSU) is defined as recurrent episodes of spontaneous wheal development and/or angioedema for more than six weeks and at least twice a week. The core link in the pathogenesis of CSU is the activation of mast cells, T cells, eosinophils, and other immune cells infiltrating around the small venules of the lesion. Increased vascular permeability, vasodilatation, and recruitment of inflammatory cells directly depend on mast cell mediators’ release. Complex regulatory systems tightly influence the critical roles of mast cells in the local microenvironment. The bias toward Th2 inflammation and autoantibodies derived from B cells, histamine expressed by basophils, and initiation of the extrinsic coagulation pathway by eosinophils or monocytes exerts powerful modulatory influences on mast cells. Cell-to-cell interactions between mast cells and eosinophils/T cells also are regulators of their function and may involve CSU’s pathomechanism. This review summarizes up-to-date knowledge regarding the crosstalk between mast cells and other immune cells, providing the impetus to develop new research concepts and treatment strategies for CSU.
Collapse
Affiliation(s)
- Bingjing Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Runqiu Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Zhu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Cong Peng,
| |
Collapse
|
13
|
Ceulemans M, Jacobs I, Wauters L, Vanuytsel T. Immune Activation in Functional Dyspepsia: Bystander Becoming the Suspect. Front Neurosci 2022; 16:831761. [PMID: 35557605 PMCID: PMC9087267 DOI: 10.3389/fnins.2022.831761] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Abstract
Disorders of gut-brain interaction (DGBI), formerly termed functional gastrointestinal disorders (FGID), are highly prevalent although exact pathophysiological mechanisms remain unclear. Intestinal immune activation has been recognized, but increasing evidence supports a pivotal role for an active inflammatory state in these disorders. In functional dyspepsia (FD), marked eosinophil and mast cell infiltration has been repeatedly demonstrated and associations with symptoms emphasize the relevance of an eosinophil-mast cell axis in FD pathophysiology. In this Review, we highlight the importance of immune activation in DGBI with a focus on FD. We summarize eosinophil biology in both homeostasis and inflammatory processes. The evidence for immune activation in FD is outlined with attention to alterations on both cellular and molecular level, and how these may contribute to FD symptomatology. As DGBI are complex and multifactorial conditions, we shed light on factors associated to, and potentially influencing immune activation, including bidirectional gut-brain interaction, allergy and the microbiota. Crucial studies reveal a therapeutic benefit of treatments targeting immune activation, suggesting that specific anti-inflammatory therapies could offer renewed hope for at least a subset of DGBI patients. Lastly, we explore the future directions for DGBI research that could advance the field. Taken together, emerging evidence supports the recognition of FD as an immune-mediated organic-based disorder, challenging the paradigm of a strictly functional nature.
Collapse
Affiliation(s)
- Matthias Ceulemans
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Inge Jacobs
- Allergy and Clinical Immunology Research Group, Department of Microbiology, Immunology and Transplantation, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Lucas Wauters
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Tim Vanuytsel
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
14
|
Atiakshin D, Kostin A, Trotsenko I, Samoilova V, Buchwalow I, Tiemann M. Carboxypeptidase A3—A Key Component of the Protease Phenotype of Mast Cells. Cells 2022; 11:cells11030570. [PMID: 35159379 PMCID: PMC8834431 DOI: 10.3390/cells11030570] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 11/16/2022] Open
Abstract
Carboxypeptidase A3 (CPA3) is a specific mast cell (MC) protease with variable expression. This protease is one of the preformed components of the secretome. During maturation of granules, CPA3 becomes an active enzyme with a characteristic localization determining the features of the cytological and ultrastructural phenotype of MC. CPA3 takes part in the regulation of a specific tissue microenvironment, affecting the implementation of innate immunity, the mechanisms of angiogenesis, the processes of remodeling of the extracellular matrix, etc. Characterization of CPA3 expression in MC can be used to refine the MC classification, help in a prognosis, and increase the effectiveness of targeted therapy.
Collapse
Affiliation(s)
- Dmitri Atiakshin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples’ Friendship University of Russia, Miklukho-Maklaya Str. 6, 117198 Moscow, Russia; (D.A.); (A.K.); (I.T.)
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, Studencheskaya Str. 10, 394036 Voronezh, Russia
| | - Andrey Kostin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples’ Friendship University of Russia, Miklukho-Maklaya Str. 6, 117198 Moscow, Russia; (D.A.); (A.K.); (I.T.)
| | - Ivan Trotsenko
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples’ Friendship University of Russia, Miklukho-Maklaya Str. 6, 117198 Moscow, Russia; (D.A.); (A.K.); (I.T.)
| | - Vera Samoilova
- Institute for Hematopathology, Fangdieckstr. 75a, 22547 Hamburg, Germany; (V.S.); (M.T.)
| | - Igor Buchwalow
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples’ Friendship University of Russia, Miklukho-Maklaya Str. 6, 117198 Moscow, Russia; (D.A.); (A.K.); (I.T.)
- Institute for Hematopathology, Fangdieckstr. 75a, 22547 Hamburg, Germany; (V.S.); (M.T.)
- Correspondence: ; Tel.: +49-(040)-7070-85317; Fax: +49-(040)-7070-85110
| | - Markus Tiemann
- Institute for Hematopathology, Fangdieckstr. 75a, 22547 Hamburg, Germany; (V.S.); (M.T.)
| |
Collapse
|
15
|
Puzzovio PG, Levi-Schaffer F. The allergic effector unit: From basic science to drug-targetable mast cell-eosinophil interactions in patients. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 9:3845-3846. [PMID: 34627540 DOI: 10.1016/j.jaip.2021.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 07/02/2021] [Indexed: 12/29/2022]
Affiliation(s)
- Pier Giorgio Puzzovio
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Francesca Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, the Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
16
|
Tontini C, Bulfone-Paus S. Novel Approaches in the Inhibition of IgE-Induced Mast Cell Reactivity in Food Allergy. Front Immunol 2021; 12:613461. [PMID: 34456900 PMCID: PMC8387944 DOI: 10.3389/fimmu.2021.613461] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 06/23/2021] [Indexed: 01/21/2023] Open
Abstract
Allergy is an IgE-dependent type-I hypersensitivity reaction that can lead to life-threatening systemic symptoms such as anaphylaxis. In the pathogenesis of the allergic response, the common upstream event is the binding of allergens to specific IgE, inducing cross-linking of the high-affinity FcεRI on mast cells, triggering cellular degranulation and the release of histamine, proteases, lipids mediators, cytokines and chemokines with inflammatory activity. A number of novel therapeutic options to curb mast cell activation are in the pipeline for the treatment of severe allergies. In addition to anti-IgE therapy and allergen-specific immunotherapy, monoclonal antibodies targeted against several key Th2/alarmin cytokines (i.e. IL-4Rα, IL-33, TSLP), active modification of allergen-specific IgE (i.e. inhibitory compounds, monoclonal antibodies, de-sialylation), engagement of inhibitory receptors on mast cells and allergen-specific adjuvant vaccines, are new promising options to inhibit the uncontrolled release of mast cell mediators upon allergen exposure. In this review, we critically discuss the novel approaches targeting mast cells limiting allergic responses and the immunological mechanisms involved, with special interest on food allergy treatment.
Collapse
Affiliation(s)
- Chiara Tontini
- Lydia Becker Institute for Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Silvia Bulfone-Paus
- Lydia Becker Institute for Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
17
|
Zheng WB, Zou Y, Liu Q, Hu MH, Elsheikha HM, Zhu XQ. Toxocara canis Infection Alters lncRNA and mRNA Expression Profiles of Dog Bone Marrow. Front Cell Dev Biol 2021; 9:688128. [PMID: 34277631 PMCID: PMC8277978 DOI: 10.3389/fcell.2021.688128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/31/2021] [Indexed: 01/05/2023] Open
Abstract
Bone marrow is the main hematopoietic organ that produces red blood cells, granulocytes, monocyte/macrophages, megakaryocytes, lymphocytes, and myeloid dendritic cells. Many of these cells play roles in the pathogenesis of Toxocara canis infection, and understanding how infection alters the dynamics of transcription regulation in bone marrow is therefore critical for deciphering the global changes in the dog transcriptional signatures during T. canis infection. In this study, long non-coding RNA (lncRNA) and messenger RNA (mRNA) expression profiles in the bone marrow of Beagle dogs infected with T. canis were determined at 12 h post-infection (hpi), 24 hpi, 96 hpi, and 36 days post-infection (dpi). RNA-sequencing and bioinformatics analysis identified 1,098, 984, 1,120, and 1,305 differentially expressed lncRNAs (DElncRNAs), and 196, 253, 223, and 328 differentially expressed mRNAs (DEmRNAs) at 12 h, 24 h, 96 h, and 36 days after infection, respectively. We also identified 29, 36, 38, and 68 DEmRNAs potentially cis-regulated by 44, 44, 51, and 80 DElncRNAs at 12 hpi, 24 hpi, 96 hpi, and 36 dpi, respectively. To validate the sequencing findings, qRT-PCR was performed on 10 randomly selected transcripts. Many altered genes were involved in the differentiation of bone marrow cells. GO of DElncRNAs and GO and KEGG pathway analyses of DEmRNAs revealed alterations in several signaling pathways, including pathways involved in energy metabolism, amino acid biosynthesis and metabolism, Wnt signaling pathway, Huntington's disease, HIF-1 signaling pathway, cGMP–PKG signaling pathway, dilated cardiomyopathy, and adrenergic signaling in cardiomyocytes. These findings revealed that bone marrow of T. canis-infected dogs exhibits distinct lncRNA and mRNA expression patterns compared to healthy control dogs. Our data provide novel insights into T. canis interaction with the definitive host and shed light on the significance of the non-coding portion of the dog genome in the pathogenesis of toxocariasis.
Collapse
Affiliation(s)
- Wen-Bin Zheng
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yang Zou
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Qing Liu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Min-Hua Hu
- National Canine Laboratory Animal Resource Center, Guangzhou General Pharmaceutical Research Institute Co., Ltd, Guangzhou, China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Xing-Quan Zhu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Veterinary Public Health of Higher Education of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
18
|
Pahima H, Puzzovio PG, Levi-Schaffer F. A novel mast cell-dependent allergic peritonitis model. Clin Exp Immunol 2021; 205:306-315. [PMID: 33999404 DOI: 10.1111/cei.13619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/04/2021] [Accepted: 05/03/2021] [Indexed: 11/29/2022] Open
Abstract
Typical murine models of allergic inflammation are induced by the combination of ovalbumin and aluminum hydroxide. However, accumulating evidence indicates that, in models of asthma and atopic dermatitis, allergic inflammation can be generated in the absence of aluminum hydroxide. Moreover, co-administration of Staphylococcus aureus enterotoxin B with ovalbumin can enhance inflammation. The objective of this study was to establish a rapid and mast cell-dependent murine model of allergic inflammation by inducing allergic peritonitis using ovalbumin and S. aureus enterotoxin B. Allergic peritonitis was induced in C57BL/6 mice by subcutaneous sensitization and intraperitoneal challenge with ovalbumin and S. aureus enterotoxin B. Disease characteristics were assessed by flow cytometry, enzyme-linked immunosorbent assay (ELISA), trypan blue exclusion and colorimetric assays. The time-course of the allergic peritonitis revealed a peak of peritoneal inflammation 48 h after challenge, as assessed by total cells and eosinophil counts. The decrease of cell numbers started 96 h post-challenge, with complete clearance within 168 h. Moreover, significantly higher levels of tryptase and increased vascular permeability were found 30 min following challenge. Allergic inflammation induction by ovalbumin and S. aureus enterotoxin B was impaired in mast cell-deficient mice and partially restored by mice reconstitution with bone marrow-derived mast cells, indicating the mast cell role in this model. We present a novel model of allergic peritonitis that is mast cell-dependent, simple and robust. Moreover, the use of S. aureus enterotoxin B better resembles human allergic inflammation, which is known to be characterized by the colonization of S. aureus.
Collapse
Affiliation(s)
- Hadas Pahima
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Pier Giorgio Puzzovio
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Francesca Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
19
|
Cui K, Hu S, Mei X, Cheng M. Innate Immune Cells in the Esophageal Tumor Microenvironment. Front Immunol 2021; 12:654731. [PMID: 33995371 PMCID: PMC8113860 DOI: 10.3389/fimmu.2021.654731] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/13/2021] [Indexed: 01/10/2023] Open
Abstract
Esophageal cancer (EC) is one of the most common mucosa-associated tumors, and is characterized by aggressiveness, poor prognosis, and unfavorable patient survival rates. As an organ directly exposed to the risk of foodborne infection, the esophageal mucosa harbors distinct populations of innate immune cells, which play vital roles in both maintenance of esophageal homeostasis and immune defense and surveillance during mucosal anti-infection and anti-tumor responses. In this review, we highlight recent progress in research into innate immune cells in the microenvironment of EC, including lymphatic lineages, such as natural killer and γδT cells, and myeloid lineages, including macrophages, dendritic cells, neutrophils, myeloid-derived suppressor cells, mast cells and eosinophils. Further, putative innate immune cellular and molecular mechanisms involved in tumor occurrence and progression are discussed, to highlight potential directions for the development of new biomarkers and effective intervention targets, which can hopefully be applied in long-term multilevel clinical EC treatment. Fully understanding the innate immunological mechanisms involved in esophageal mucosa carcinogenesis is of great significance for clinical immunotherapy and prognosis prediction for patients with EC.
Collapse
Affiliation(s)
- Kele Cui
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China
- Cancer Immunotherapy Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shouxin Hu
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China
- Cancer Immunotherapy Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xinyu Mei
- Department of Thoracic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Min Cheng
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China
- Cancer Immunotherapy Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
20
|
Cheng L, Luo QQ, Chen SL. The role of intestinal mast cell infiltration in irritable bowel syndrome. J Dig Dis 2021; 22:143-151. [PMID: 33511763 DOI: 10.1111/1751-2980.12971] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 01/17/2021] [Accepted: 01/25/2021] [Indexed: 12/11/2022]
Abstract
As an essential part of the immune system, mast cells (MCs) play an important role in the pathogenesis of irritable bowel syndrome (IBS). Accumulating evidence has identified altered MC count and density in intestinal mucosa of patients with IBS; however, conflicting findings yield inconsistent conclusions. Currently, most studies have suggested intestinal MC infiltration in IBS patients. Considering the pivotal role of MCs in IBS, it is necessary to achieve a better understanding about the pathological changes in the intestine. The risk factors for IBS, including dietary habits, psychological factors, infection, and dysbiosis, are implicated to induce intestinal MC infiltration. Mechanistically, food may trigger immune-related allergic reactions and affect the intestinal microbiota activity. Some exogenous pathogens and altered profile of commensal bacteria promote intestinal MC recruitment through promoted release of chemokines from epithelial cells or direct activation of the immune system. In addition, psychological factors may affect the microenvironment where MCs live. MCs have been proven to interact with the enteric neurons and other immunocytes, evidenced by the close proximity of MCs to neurons and regional altered immune system components. A variety of mediators released by the enteric neurons, immunocytes, and MCs per se, such as neurotrophins, neuropeptides, cytokines, and chemokines, may have stimulant effects on MCs by modulating the survival, proliferation, and recruitment process of MCs in the intestine. In this review, the associations between IBS and intestinal MC density and the underlying mechanisms are discussed.
Collapse
Affiliation(s)
- Li Cheng
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Qing Qing Luo
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Sheng Liang Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| |
Collapse
|
21
|
First Evidence for a Role of Siglec-8 in Breast Cancer. Int J Mol Sci 2021; 22:ijms22042000. [PMID: 33670444 PMCID: PMC7922794 DOI: 10.3390/ijms22042000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 12/20/2022] Open
Abstract
Sialic acid-binding immunoglobulin-like lectins (Siglecs) are involved in various immune cell-mediated diseases. Their role in cancer is poorly investigated, and research focusses on Siglec-expression on immune cells interacting with tumor cells. This study evaluates the role of Siglec-8 in breast cancer (BC). Siglec-8 expression was analyzed immunohistochemically on 235 primary BC cases and was correlated with clinical and pathological parameters and outcome. Cell culture experiments were performed with various BC cell lines. Siglec-8 was expressed in 215 BC cases and expression was lowest in triple-negative BC. It correlated with estrogen receptor-status, grading and the prognostic factors galectin (Gal)-7 and tumor-associated mucin-1 (TA-MUC1). However, Gal-7 and TA-MUC1 were only prognosticators for clinical outcome in the cohort expressing high (Immunoreactivity score IRS > 3) Siglec-8 levels but not in the low-expressing cohort. Siglec-8 knockdown led to a significantly reduced Gal-7 expression in MCF7 cells. All BC cell lines expressed low Siglec-8-levels, that could be elevated in MCF7 by Peroxisome proliferator-activated receptor (PPARγ)-stimulation. This study demonstrates that Siglec-8 is expressed in BC cells and correlates with known clinical and prognostic parameters. It is probably associated with Gal-7 and TA-MUC1 and might be regulated via PPARγ. Further analyses focusing on functional associations will clarify Siglec-8’s eligibility as a possible therapeutic target.
Collapse
|
22
|
Abstract
Mast cells and eosinophils are the key effector cells of allergy [1]. In general, allergic reactions are composed of two phases, namely an early phase and a late phase, and after that resolution occurs. If the allergic reactions fail to resolve after the late phase, allergic inflammation (AI) can evolve into a chronic phase mainly involving mast cells and eosinophils that abundantly coexist in the inflamed tissue in the late and chronic phases and cross-talk in a bidirectional manner. We defined these bidirectional interactions between MCs and Eos, as the "allergic effector unit." This cross talk is mediated by both physical cell-cell contacts through cell surface receptors such as CD48, 2B4, and respective ligands and through released mediators such as various specific granular mediators, arachidonic acid metabolites, cytokines, and chemokines [2, 3]. The allergic effector unit can be studied in vitro in a customized co-culture system using mast cells and eosinophils derived from either mouse or human sources.
Collapse
|
23
|
Mattei F, Andreone S, Marone G, Gambardella AR, Loffredo S, Varricchi G, Schiavoni G. Eosinophils in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1273:1-28. [PMID: 33119873 DOI: 10.1007/978-3-030-49270-0_1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Eosinophils are rare blood-circulating and tissue-infiltrating immune cells studied for decades in the context of allergic diseases and parasitic infections. Eosinophils can secrete a wide array of soluble mediators and effector molecules, with potential immunoregulatory activities in the tumor microenvironment (TME). These findings imply that these cells may play a role in cancer immunity. Despite these cells were known to infiltrate tumors since many years ago, their role in TME is gaining attention only recently. In this chapter, we will review the main biological functions of eosinophils that can be relevant within the TME. We will discuss how these cells may undergo phenotypic changes acquiring pro- or antitumoricidal properties according to the surrounding stimuli. Moreover, we will analyze canonical (i.e., degranulation) and unconventional mechanisms (i.e., DNA traps, exosome secretion) employed by eosinophils in inflammatory contexts, which can be relevant for tumor immune responses. Finally, we will review the available preclinical models that could be employed for the study of the role in vivo of eosinophils in cancer.
Collapse
Affiliation(s)
- Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Sara Andreone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Giancarlo Marone
- Department of Public Health, University of Naples Federico II, Naples, Italy.,Azienda Ospedaliera Ospedali dei Colli - Monaldi Hospital Pharmacy, Naples, Italy
| | | | - Stefania Loffredo
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy. .,WAO Center of Excellence, Naples, Italy. .,Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy.
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
24
|
Discovery, Function, and Therapeutic Targeting of Siglec-8. Cells 2020; 10:cells10010019. [PMID: 33374255 PMCID: PMC7823959 DOI: 10.3390/cells10010019] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/18/2020] [Accepted: 12/20/2020] [Indexed: 12/11/2022] Open
Abstract
Siglecs (sialic acid-binding immunoglobulin-like lectins) are single-pass cell surface receptors that have inhibitory activities on immune cells. Among these, Siglec-8 is a CD33-related family member selectively expressed on human mast cells and eosinophils, and at low levels on basophils. These cells can participate in inflammatory responses by releasing mediators that attract or activate other cells, contributing to the pathogenesis of allergic and non-allergic diseases. Since its discovery in 2000, initial in vitro studies have found that the engagement of Siglec-8 with a monoclonal antibody or with selective polyvalent sialoglycan ligands induced the cell death of eosinophils and inhibited mast cell degranulation. Anti-Siglec-8 antibody administration in vivo to humanized and transgenic mice selectively expressing Siglec-8 on mouse eosinophils and mast cells confirmed the in vitro findings, and identified additional anti-inflammatory effects. AK002 (lirentelimab) is a humanized non-fucosylated IgG1 antibody against Siglec-8 in clinical development for mast cell- and eosinophil-mediated diseases. AK002 administration has safely demonstrated the inhibition of mast cell activity and the depletion of eosinophils in several phase 1 and phase 2 trials. This article reviews the discovery and functions of Siglec-8, and strategies for its therapeutic targeting for the treatment of eosinophil- and mast cell-associated diseases.
Collapse
|
25
|
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: 3.0] [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
|
26
|
The emerging roles of eosinophils in mucosal homeostasis. Mucosal Immunol 2020; 13:574-583. [PMID: 32157190 DOI: 10.1038/s41385-020-0281-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 02/04/2023]
Abstract
Eosinophils are granulocytes, typically implicated as end-stage effector cells in type-II immune responses. They are capable of producing a wide array of pre-formed molecules which render them with vast potential to influence a wide variety of processes. Nonetheless, eosinophil research has traditionally focused on their role in anti-helminthic responses and pathophysiological processes in type-II immune disorders, such as allergy and asthma, where eosinophilia is a hallmark phenotype. However, a number of key studies over the past decade have placed this restricted view of eosinophil function into question, presenting additional evidence for eosinophils as critical regulators of various homeostatic processes including immune maintenance, organ development, and tissue regeneration.
Collapse
|
27
|
O'Sullivan JA, Chang AT, Youngblood BA, Bochner BS. Eosinophil and mast cell Siglecs: From biology to drug target. J Leukoc Biol 2020; 108:73-81. [PMID: 31965606 PMCID: PMC7531194 DOI: 10.1002/jlb.2mr0120-352rr] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/04/2020] [Accepted: 01/06/2020] [Indexed: 12/26/2022] Open
Abstract
Mast cells and eosinophils are innate immune cells involved in both acute and chronic inflammatory responses. Siglecs are a family of cell surface receptors that share sialic acid binding activity. Over the past 20 years, our knowledge of the expression and function of Siglecs on cells of the immune system and others has greatly expanded, as has our understanding of their signaling, ligands, and possible roles in disease pathophysiology. Because of this, Siglecs have garnered interest as potential drug targets using strategies ranging from biologics to ligand-directed nanoparticles. This mini-review will highlight the state of our knowledge regarding human eosinophil and mast cell Siglecs, their biology, what they recognize, tools developed for in vitro and preclinical experimentation, and the status of ongoing efforts to develop drugs that engage eosinophil and mast cell Siglecs for potential therapeutic benefit.
Collapse
Affiliation(s)
- Jeremy A O'Sullivan
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | | | | | - Bruce S Bochner
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
28
|
Andrade CA, Pacheco GA, Gálvez NMS, Soto JA, Bueno SM, Kalergis AM. Innate Immune Components that Regulate the Pathogenesis and Resolution of hRSV and hMPV Infections. Viruses 2020; 12:E637. [PMID: 32545470 PMCID: PMC7354512 DOI: 10.3390/v12060637] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023] Open
Abstract
The human respiratory syncytial virus (hRSV) and human Metapneumovirus (hMPV) are two of the leading etiological agents of acute lower respiratory tract infections, which constitute the main cause of mortality in infants. However, there are currently approved vaccines for neither hRSV nor hMPV. Moreover, despite the similarity between the pathology caused by both viruses, the immune response elicited by the host is different in each case. In this review, we discuss how dendritic cells, alveolar macrophages, neutrophils, eosinophils, natural killer cells, innate lymphoid cells, and the complement system regulate both pathogenesis and the resolution of hRSV and hMPV infections. The roles that these cells play during infections by either of these viruses will help us to better understand the illnesses they cause. We also discuss several controversial findings, relative to some of these innate immune components. To better understand the inflammation in the lungs, the role of the respiratory epithelium in the recruitment of innate immune cells is briefly discussed. Finally, we review the main prophylactic strategies and current vaccine candidates against both hRSV and hMPV.
Collapse
Affiliation(s)
- Catalina A. Andrade
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Gaspar A. Pacheco
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Nicolas M. S. Gálvez
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Jorge A. Soto
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Susan M. Bueno
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Alexis M. Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| |
Collapse
|
29
|
Altrichter S, Frischbutter S, Fok JS, Kolkhir P, Jiao Q, Skov PS, Metz M, Church MK, Maurer M. The role of eosinophils in chronic spontaneous urticaria. J Allergy Clin Immunol 2020; 145:1510-1516. [DOI: 10.1016/j.jaci.2020.03.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/26/2020] [Accepted: 03/02/2020] [Indexed: 12/21/2022]
|
30
|
Hughes MR, Canals Hernaez D, Cait J, Refaeli I, Lo BC, Roskelley CD, McNagny KM. A sticky wicket: Defining molecular functions for CD34 in hematopoietic cells. Exp Hematol 2020; 86:1-14. [PMID: 32422232 DOI: 10.1016/j.exphem.2020.05.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/05/2020] [Accepted: 05/09/2020] [Indexed: 02/06/2023]
Abstract
The CD34 cell surface antigen is widely expressed in tissues on cells with progenitor-like properties and on mature vascular endothelia. In adult human bone marrow, CD34 marks hematopoietic stem and progenitor cells (HSPCs) starting from the bulk of hematopoietic stem cells with long-term repopulating potential (LT-HSCs) throughout expansion and differentiation of oligopotent and unipotent progenitors. CD34 protein surface expression is typically lost as cells mature into terminal effectors. Because of this expression pattern of HSPCs, CD34 has had a central role in the evaluation or selection of donor graft tissue in HSC transplant (HSCT). Given its clinical importance, it is surprising that the biological functions of CD34 are still poorly understood. This enigma is due, in part, to CD34's context-specific role as both a pro-adhesive and anti-adhesive molecule and its potential functional redundancy with other sialomucins. Moreover, there are also critical differences in the regulation of CD34 expression on HSPCs in humans and experimental mice. In this review, we highlight some of the more well-defined functions of CD34 in HSPCs with a focus on proposed functions most relevant to HSCT biology.
Collapse
Affiliation(s)
- Michael R Hughes
- The Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Diana Canals Hernaez
- The Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Jessica Cait
- The Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Ido Refaeli
- The Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Bernard C Lo
- The Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Calvin D Roskelley
- Life Sciences Institute, Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Kelly M McNagny
- The Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
31
|
Atiakshin D, Buchwalow I, Tiemann M. Mast cells and collagen fibrillogenesis. Histochem Cell Biol 2020; 154:21-40. [PMID: 32222902 DOI: 10.1007/s00418-020-01875-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2020] [Indexed: 02/07/2023]
Abstract
This article presents 20 combinations of histochemical stainings for the determination of mast cell co-localization with the fibrous component of the connective tissue in the fibrillogenesis course. Best results were obtained using metachromatic detection of mast cells in combination with silver or picro-fuchsin impregnation, staining with brilliant green using van Gieson staining, and a combination of aniline blue staining with neutral red. Proposed variants of histochemical protocols open up new opportunities to analyze the participation of mast cells in extracellular matrix remodeling of the tissue microenvironment in the course of adaptive and pathological processes. Results obtained expand the current theoretical views of the process of fibrillogenesis in the extracellular matrix. They also shed new light on the participation of mast cell secretion components in the molecular mechanisms of fiber formation.
Collapse
Affiliation(s)
- Dmitri Atiakshin
- Research Institute of Experimental Biology and Medicine, Voronezh N. N. Burdenko State Medical University, Voronezh, Russia
| | - Igor Buchwalow
- Institute of Hematopathology, Fangdieckstr. 75a, 22547, Hamburg, Germany.
| | - Markus Tiemann
- Institute of Hematopathology, Fangdieckstr. 75a, 22547, Hamburg, Germany
| |
Collapse
|
32
|
Marone G, Gambardella AR, Mattei F, Mancini J, Schiavoni G, Varricchi G. Basophils in Tumor Microenvironment and Surroundings. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1224:21-34. [PMID: 32036602 DOI: 10.1007/978-3-030-35723-8_2] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Basophils represent approximately 1% of human peripheral blood leukocytes. Their effector functions were initially appreciated in the 1970s when basophils were shown to express the high-affinity receptor (FcεRI) for IgE and to release proinflammatory mediators (histamine and cysteinyl leukotriene C4) and immunoregulatory cytokines (i.e., IL-4 and IL-13). Basophils in the mouse were subsequently identified and immunologically characterized. There are many similarities but also several differences between human and mouse basophils. Basophil-deficient mice have enabled to examine the in vivo roles of basophils in several immune disorders and, more recently, in tumor immunity. Activated human basophils release several proangiogenic molecules such as vascular endothelial growth factor-A (VEGF-A), vascular endothelial growth factor-B (VEGF-B), CXCL8, angiopoietin 1 (ANGPT1), and hepatocyte growth factor (HGF). On the other side, basophils can exert anti-tumorigenic effects by releasing granzyme B, TNF-α, and histamine. Circulating basophils have been associated with certain human hematologic (i.e., chronic myeloid leukemia) and solid tumors. Basophils have been found in tumor microenvironment (TME) of human lung adenocarcinoma and pancreatic cancer. Basophils played a role in melanoma rejection in basophil-deficient mouse model. By contrast, basophils appear to play a pro-tumorigenic role in experimental and human pancreatic cancer. In conclusion, the roles of basophils in experimental and human cancers have been little investigated and remain largely unknown. The elucidation of the roles of basophils in tumor immunity will demand studies on increasing complexity beyond those assessing basophil density and their microlocalization in TME. There are several fundamental questions to be addressed in experimental models and clinical studies before we understand whether basophils are an ally, adversary, or even innocent bystanders in cancers.
Collapse
Affiliation(s)
- Giancarlo Marone
- Department of Public Health, University of Naples Federico II, Naples, Italy
- Azienda Ospedaliera dei Colli-Monaldi Hospital Pharmacy, Naples, Italy
| | | | - Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Jacopo Mancini
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Gilda Varricchi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.
- WAO Center of Excellence, Naples, Italy.
- Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy.
| |
Collapse
|
33
|
Amorim NRT, Souza-Almeida G, Luna-Gomes T, Bozza PT, Canetti C, Diaz BL, Maya-Monteiro CM, Bandeira-Melo C. Leptin Elicits In Vivo Eosinophil Migration and Activation: Key Role of Mast Cell-Derived PGD 2. Front Endocrinol (Lausanne) 2020; 11:572113. [PMID: 33117286 PMCID: PMC7551309 DOI: 10.3389/fendo.2020.572113] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/09/2020] [Indexed: 12/16/2022] Open
Abstract
Eosinophils are key regulators of adipose tissue homeostasis, thus characterization of adipose tissue-related molecular factors capable of regulating eosinophil activity is of great interest. Leptin is known to directly activate eosinophils in vitro, but leptin ability of inducing in vivo eosinophilic inflammatory response remains elusive. Here, we show that leptin elicits eosinophil influx as well as its activation, characterized by increased lipid body biogenesis and LTC4 synthesis. Such leptin-triggered eosinophilic inflammatory response was shown to be dependent on activation of the mTOR signaling pathway, since it was (i) inhibited by rapamycin pre-treatment and (ii) reduced in PI3K-deficient mice. Local infiltration of activated eosinophils within leptin-driven inflammatory site was preceded by increased levels of classical mast cell-derived molecules, including TNFα, CCL5 (RANTES), and PGD2. Thus, mice were pre-treated with a mast cell degranulating agent compound 48/80 which was capable to impair leptin-induced PGD2 release, as well as eosinophil recruitment and activation. In agreement with an indirect mast cell-driven phenomenon, eosinophil accumulation induced by leptin was abolished in TNFR-1 deficient and also in HQL-79-pretreated mice, but not in mice pretreated with neutralizing antibodies against CCL5, indicating that both typical mast cell-driven signals TNFα and PGD2, but not CCL5, contribute to leptin-induced eosinophil influx. Distinctly, leptin-induced eosinophil lipid body (lipid droplet) assembly and LTC4 synthesis appears to depend on both PGD2 and CCL5, since both HQL-79 and anti-CCL5 treatments were able to inhibit these eosinophil activation markers. Altogether, our data show that leptin triggers eosinophilic inflammation in vivo via an indirect mechanism dependent on activation of resident mast cell secretory activity and mediation by TNFα, CCL5, and specially PGD2.
Collapse
Affiliation(s)
- Natália R. T. Amorim
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Glaucia Souza-Almeida
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz - IOC, FIOCRUZ, Rio de Janeiro, Brazil
- Laboratório de Imunoinflamação, Instituto de Biologia, Universidade de Campinas, Campinas, Brazil
| | - Tatiana Luna-Gomes
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Ciências da Natureza, Instituto de Aplicação Fernando Rodrigues da Silveira, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia T. Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz - IOC, FIOCRUZ, Rio de Janeiro, Brazil
| | - Claudio Canetti
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno L. Diaz
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clarissa M. Maya-Monteiro
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz - IOC, FIOCRUZ, Rio de Janeiro, Brazil
- *Correspondence: Christianne Bandeira-Melo, ; Clarissa M. Maya-Monteiro,
| | - Christianne Bandeira-Melo
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- *Correspondence: Christianne Bandeira-Melo, ; Clarissa M. Maya-Monteiro,
| |
Collapse
|
34
|
Zheng M, Gao S, Yu Y, Xu J, Huang Z, Li J, Xie S, Lin J. Surface-enhanced Raman spectroscopy analysis of mast cell degranulation induced by low-intensity laser. IET Nanobiotechnol 2019; 13:983-988. [PMID: 31811770 DOI: 10.1049/iet-nbt.2019.0145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Mast cell (MC) degranulation is an important step in the healing process. In this study, silver-nanoparticles-based surface-enhanced Raman spectroscopy (SERS) was used to investigate the spectral characteristics of degranulation of MCs activated by low-intensity laser. The significant spectral changes, such as Raman peak intensities, suggested the concentration variation of some degranulated substances. The Raman intensity ratio of 799-554 cm-1 could be used as a potential internal indicator for the degranulation degree of MCs. Principal component analysis (PCA) was employed to reduce the high dimension of spectra into a few principal components (PCs) while retaining the most diagnostically significant information for sample differentiation. Using the diagnostically significant PC scores (P < 0.05), linear discriminate analysis (LDA) was applied to identify different cell degranulation groups with high sensitivity, specificity and accuracy. This exploratory work demonstrates that SERS technique combined with a PCA-LDA algorithm possesses great potential for developing a label-free, comprehensive, non-invasive and accurate method for measuring MC degranulation.
Collapse
Affiliation(s)
- Mengmeng Zheng
- Key Laboratory of OptoElectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou, Fujian, People's Republic of China
| | - Siqi Gao
- Key Laboratory of OptoElectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou, Fujian, People's Republic of China
| | - Yun Yu
- College of Integrated Traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, People's Republic of China
| | - Jianshu Xu
- Key Laboratory of OptoElectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou, Fujian, People's Republic of China
| | - Zufang Huang
- Key Laboratory of OptoElectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou, Fujian, People's Republic of China
| | - Juan Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou, Fujian, People's Republic of China
| | - Shusen Xie
- Key Laboratory of OptoElectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou, Fujian, People's Republic of China
| | - Juqiang Lin
- Key Laboratory of OptoElectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou, Fujian, People's Republic of China.
| |
Collapse
|
35
|
Magrone T, Magrone M, Jirillo E. Mast Cells as a Double-Edged Sword in Immunity: Their Function in Health and Disease. First of Two Parts. Endocr Metab Immune Disord Drug Targets 2019; 20:654-669. [PMID: 31789135 DOI: 10.2174/1871530319666191202120301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/08/2019] [Accepted: 11/21/2019] [Indexed: 11/22/2022]
Abstract
Mast cells (MCs) have recently been re-interpreted in the context of the immune scenario in the sense that their pro-allergic role is no longer exclusive. In fact, MCs even in steady state conditions maintain homeostatic functions, producing mediators and intensively cross-talking with other immune cells. Here, emphasis will be placed on the array of receptors expressed by MCs and the variety of cytokines they produce. Then, the bulk of data discussed will provide readers with a wealth of information on the dual ability of MCs not only to defend but also to offend the host. This double attitude of MCs relies on many variables, such as their subsets, tissues of residency and type of stimuli ranging from microbes to allergens and food antigens. Finally, the relationship between MCs with basophils and eosinophils will be discussed.
Collapse
Affiliation(s)
- Thea Magrone
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, School of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Manrico Magrone
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, School of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Emilio Jirillo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, School of Medicine, University of Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
36
|
Youngblood BA, Brock EC, Leung J, Falahati R, Bryce PJ, Bright J, Williams J, Shultz LD, Greiner DL, Brehm MA, Bebbington C, Tomasevic N. AK002, a Humanized Sialic Acid-Binding Immunoglobulin-Like Lectin-8 Antibody that Induces Antibody-Dependent Cell-Mediated Cytotoxicity against Human Eosinophils and Inhibits Mast Cell-Mediated Anaphylaxis in Mice. Int Arch Allergy Immunol 2019; 180:91-102. [PMID: 31401630 DOI: 10.1159/000501637] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 06/19/2019] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Pathologic accumulation and activation of mast cells and eosinophils are implicated in allergic and inflammatory diseases. Sialic acid-binding immunoglobulin-like lectin (Siglec)-8 is an inhibitory receptor selectively expressed on mast cells, eosinophils and, at a lower extent, basophils. When engaged with an antibody, Siglec-8 can induce apoptosis of activated eosinophils and inhibit mast cell activation. AK002 is a humanized, non-fucosylated IgG1 anti-Siglec-8 antibody undergoing clinical investigation for treatment of allergic, inflammatory, and proliferative diseases. Here we examine the human tissue selectivity of AK002 and evaluate the in vitro, ex vivo, and in vivo activity of AK002 on eosinophils and mast cells. METHODS The affinity of AK002 for Siglec-8 and CD16 was determined by biolayer interferometry. Ex vivo activity of AK002 on human eosinophils from blood and dissociated human tissue was tested in apoptosis and antibody-dependent cell-mediated cytotoxicity (ADCC) assays. The in vivo activity of a murine precursor of AK002 (mAK002) was tested in a passive systemic anaphylaxis (PSA) humanized mouse model. RESULTS AK002 bound selectively to mast cells, eosinophils and, at a lower level, to basophils in human blood and tissue and not to other cell types examined. AK002 induced apoptosis of interleukin-5-activated blood eosinophils and demonstrated potent ADCC activity against blood eosinophils in the presence of natural killer cells. AK002 also significantly reduced eosinophils in dissociated human lung tissue. Furthermore, mAK002 prevented PSA in humanized mice through mast cell inhibition. CONCLUSION AK002 selectively evokes potent apoptotic and ADCC activity against eosinophils and prevents systemic anaphylaxis through mast cell inhibition.
Collapse
Affiliation(s)
| | | | - John Leung
- Allakos, Inc., Redwood City, California, USA
| | | | - Paul J Bryce
- Division of Allergy-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | | | | | | | - Dale L Greiner
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Michael A Brehm
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | | | |
Collapse
|
37
|
Abstract
During degranulation, mast cells secrete a specific set of mediators defined as "secretome" including the preformed mediators that have already been synthesized by a cell and contained in the cytoplasmic granules. This group includes serine proteases, in particular, chymase and tryptase. Biological significance of chymase depends on the mechanisms of degranulation and is characterized by selective effects on the cellular and non-cellular components of the specific tissue microenvironment. Chymase is known to be closely involved in the mechanisms of inflammation and allergy, angiogenesis, and oncogenesis, remodeling of the extracellular matrix of the connective tissue and changes in organ histoarchitectonics. Number of chymase-positive mast cells in the intra-organ population, and the mechanisms of biogenesis and secretome degranulation appear to be the informative criteria for interpreting the state of the internal organs, characterizing not only the diagnostic efficacy but also the properties of targets of pharmacotherapy. In this review, we discussed the current state of knowledge about mast cell chymase as one of the mast cell secretome proteases. Main issues of the reviewed publications are highlighted with our microscopic images of mast cell chymase visualized using immunohistochemical staining.
Collapse
|
38
|
Loktionov A. Eosinophils in the gastrointestinal tract and their role in the pathogenesis of major colorectal disorders. World J Gastroenterol 2019; 25:3503-3526. [PMID: 31367153 PMCID: PMC6658389 DOI: 10.3748/wjg.v25.i27.3503] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/22/2019] [Accepted: 06/01/2019] [Indexed: 02/06/2023] Open
Abstract
Eosinophils are currently regarded as versatile mobile cells controlling and regulating multiple biological pathways and responses in health and disease. These cells store in their specific granules numerous biologically active substances (cytotoxic cationic proteins, cytokines, growth factors, chemokines, enzymes) ready for rapid release. The human gut is the main destination of eosinophils that are produced and matured in the bone marrow and then transferred to target tissues through the circulation. In health the most important functions of gut-residing eosinophils comprise their participation in the maintenance of the protective mucosal barrier and interactions with other immune cells in providing immunity to microbiota of the gut lumen. Eosinophils are closely involved in the development of inflammatory bowel disease (IBD), when their cytotoxic granule proteins cause damage to host tissues. However, their roles in Crohn’s disease and ulcerative colitis appear to follow different immune response patterns. Eosinophils in IBD are especially important in altering the structure and protective functions of the mucosal barrier and modulating massive neutrophil influx to the lamina propria followed by transepithelial migration to colorectal mucus. IBD-associated inflammatory process involving eosinophils then appears to expand to the mucus overlaying the internal gut surface. The author hypothesises that immune responses within colorectal mucus as well as ETosis exerted by both neutrophils and eosinophils on the both sides of the colonic epithelial barrier act as additional pathogenetic factors in IBD. Literature analysis also shows an association between elevated eosinophil levels and better colorectal cancer (CRC) prognosis, but mechanisms behind this effect remain to be elucidated. In conclusion, the author emphasises the importance of investigating colorectal mucus in IBD and CRC patients as a previously unexplored milieu of disease-related inflammatory responses.
Collapse
|
39
|
van de Veen W, Akdis M. The use of biologics for immune modulation in allergic disease. J Clin Invest 2019; 129:1452-1462. [PMID: 30882368 DOI: 10.1172/jci124607] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The rising prevalence of allergies represents an increasing socioeconomic burden. A detailed understanding of the immunological mechanisms that underlie the development of allergic disease, as well as the processes that drive immune tolerance to allergens, will be instrumental in designing therapeutic strategies to treat and prevent allergic disease. Improved characterization of individual patients through the use of specific biomarkers and improved definitions of disease endotypes are paving the way for the use of targeted therapeutic approaches for personalized treatment. Allergen-specific immunotherapy and biologic therapies that target key molecules driving the Th2 response are already used in the clinic, and a wave of novel drug candidates are under development. In-depth analysis of the cells and tissues of patients treated with such targeted interventions provides a wealth of information on the mechanisms that drive allergies and tolerance to allergens. Here, we aim to deliver an overview of the current state of specific inhibitors used in the treatment of allergy, with a particular focus on asthma and atopic dermatitis, and provide insights into the roles of these molecules in immunological mechanisms of allergic disease.
Collapse
Affiliation(s)
- Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.,Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| |
Collapse
|
40
|
Kappel S, Borgström A, Stokłosa P, Dörr K, Peinelt C. Store-operated calcium entry in disease: Beyond STIM/Orai expression levels. Semin Cell Dev Biol 2019; 94:66-73. [PMID: 30630032 DOI: 10.1016/j.semcdb.2019.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/29/2018] [Accepted: 01/05/2019] [Indexed: 12/19/2022]
Abstract
Precise intracellular calcium signaling is crucial to numerous cellular functions. In non-excitable cells, store-operated calcium entry (SOCE) is a key step in the generation of intracellular calcium signals. Tight regulation of SOCE is important, and dysregulation is involved in several pathophysiological cellular malfunctions. The current underlying SOCE, calcium release-activated calcium current (ICRAC), was first discovered almost three decades ago. Since its discovery, the molecular components of ICRAC, Orai1 and stromal interaction molecule 1 (STIM1), have been extensively investigated. Several regulatory mechanisms and proteins contribute to alterations in SOCE and cellular malfunctions in cancer, immune and neurodegenerative diseases, inflammation, and neuronal disorders. This review summarizes these regulatory mechanisms, including glycosylation, pH sensing, and the regulatory proteins golli, α-SNAP, SARAF, ORMDL3, CRACR2A, and TRPM4 channels.
Collapse
Affiliation(s)
- Sven Kappel
- Institute of Biochemistry and Molecular Medicine, National Center of Competence in Research NCCR TransCure, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland
| | - Anna Borgström
- Institute of Biochemistry and Molecular Medicine, National Center of Competence in Research NCCR TransCure, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland
| | - Paulina Stokłosa
- Institute of Biochemistry and Molecular Medicine, National Center of Competence in Research NCCR TransCure, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland
| | | | - Christine Peinelt
- Institute of Biochemistry and Molecular Medicine, National Center of Competence in Research NCCR TransCure, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland.
| |
Collapse
|
41
|
Maurer M, Pucillo C. What we know (and don't know) about the biology and functions of mast cells and basophils. Immunol Rev 2019; 282:5-7. [PMID: 29431213 DOI: 10.1111/imr.12645] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Marcus Maurer
- Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Carlo Pucillo
- Department of Medicine, University of Udine, Udine, Italy
| |
Collapse
|