1
|
Huyghe P, Ceulemans M, Keita ÅV, Söderholm J, Depoortere I, Tack J, Wauters L, Vanuytsel T. The Duodenal Microenvironment in Functional Dyspepsia. J Neurogastroenterol Motil 2025; 31:186-198. [PMID: 40205896 PMCID: PMC11986653 DOI: 10.5056/jnm24176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 04/11/2025] Open
Abstract
Functional dyspepsia (FD) is a chronic gastrointestinal disorder without a readily identifiable organic cause, resulting in bothersome upper abdominal symptoms. It is a highly prevalent disorder of which the pathophysiology remains mostly elusive, despite intensive research efforts. However, recent studies have found alterations in the microenvironment of the duodenum in patients with FD. In this review we summarize the duodenal microenvironment in homeostatic conditions and the alterations found in patients with FD, highlighting the similarities and discrepancies between different studies. The most consistent findings, being an impaired duodenal barrier and duodenal immune activation, are reviewed. We discuss the potential triggers for these observed alterations, including psychological comorbidities, luminal alterations and food related triggers. In summary, this review presents the evidence of molecular and cellular changes in patients with FD, with an impaired duodenal barrier and activated mucosal eosinophils and mast cells, challenging the notion that FD is purely functional, and offering different targets for potential future treatments.
Collapse
Affiliation(s)
- Pauline Huyghe
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Matthias Ceulemans
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Åsa V Keita
- Department of Biomedical and Clinical Sciences and Department of Surgery, Linköping University, Linköping, Sweden
| | - Johan Söderholm
- Department of Biomedical and Clinical Sciences and Department of Surgery, Linköping University, Linköping, Sweden
| | - Inge Depoortere
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jan Tack
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Lucas Wauters
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Tim Vanuytsel
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
2
|
Cunegundes PS, Wood K, Mao L, Menkes U. Phenolic Preservatives Are Not the Sole Cause of Eosinophilic Infiltration at Infusion Pump Sites. Diabetes Technol Ther 2025. [PMID: 40067457 DOI: 10.1089/dia.2025.0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Background: Skin reactions and discomfort associated with insulin infusion pumps limit user adherence. A recent histopathological study by Kalus et al. (DERMIS study) reported increased eosinophilic infiltration and imputed an inflammatory response to an allergen delivered at the catheter tip. This finding might explain the pruritus reported by pump users. As eosinophils migrate to inflammatory foci, primarily due to IL-5 and CCL11, we aimed to evaluate insulin phenolic preservative (IPP) as a potential allergen in vitro and assess tissue eosinophilic infiltration in vivo. Methods: Histopathological evaluations for eosinophil recruitment were performed over 1 week following IPP infusions in swine tissue. Additional histopathological investigations of eosinophilic infiltration were conducted using three commercial glucose sensors implanted in swine for up to 3 weeks. Results: Eosinophilic infiltration in the dermis and subcutaneous tissue was observed following saline and IPP infusion and at glucose sensor implantation at all time points examined. In vitro studies revealed IPP eosinophil cytotoxicity. However, neither CCL11 nor IL-5 was detected in any of the tested tissue cells after IPP treatment. Conclusion: These findings suggest that IPP is not the only triggering allergen, as IPP did not induce eosinophils in vitro, while glucose sensors also indicated increased eosinophilic infiltration. Therefore, factors other than IPP trigger eosinophil recruitment to insulin infusion pump sets.
Collapse
Affiliation(s)
- Priscila Silva Cunegundes
- Department of Physiology, Integrative Biosciences Center (IBio), Wayne State University, Detroit, MI, United States
| | - Kenneth Wood
- Department of Physiology, Integrative Biosciences Center (IBio), Wayne State University, Detroit, MI, United States
| | - Li Mao
- Department of Physiology, Integrative Biosciences Center (IBio), Wayne State University, Detroit, MI, United States
| | - Ulrike Menkes
- Department of Physiology, Integrative Biosciences Center (IBio), Wayne State University, Detroit, MI, United States
| |
Collapse
|
3
|
Serra J, Alcalá-González LG, Mendive JM, Santander Vaquero C, Serrano Falcón B. Updated document on the management of functional dyspepsia by the Asociación Española de Neurogastroenterologia y Motilidad (ASENEM) and Sociedad Española de Medicina Familiar y Comunitaria (semFYC). REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2025; 117:84-91. [PMID: 39812003 DOI: 10.17235/reed.2025.10572/2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Functional dyspepsia (FD) is a gut-brain axis disorder characterized by postprandial fullness, early satiety, bloating and/or epigastric pain, which are presumed to originate in the gastroduodenal tract. While the international recommendations in the Rome IV consensus require endoscopy to rule out an organic condition before establishing a diagnosis of FD, international guidelines recommend that, in the absence of risk factors, patient management be initiated at the primary care level by establishing Helicobacter pylori infection status, with eradication when positive, followed by empiric therapy with proton pump inhibitors and/or prokinetics, and that endoscopy be reserved for patients refractory to said measures. Second-line therapy includes neuromodulating agents, among which tricyclic antidepressants and atypical antipsychotics such as levosulpiride stand out. The latter has a predominant prokinetic effect, hence it is also used as first-line therapy for patients where early satiety and postprandial fullness predominate. Other therapy alternatives include phytotherapy using STW5 or peppermint/caraway oil, which have shown their superiority over placebo in controlled studies. Concurrently, dietary and lifestyle counseling, as well as psychological interventions such as cognitive-behavioral therapy, when available, may represent a therapeutic alternative worth considering for some patients.
Collapse
Affiliation(s)
- Jordi Serra
- Gastroenterology, Hospital Universitari Vall d'Hebron, Spain
| | | | | | | | | |
Collapse
|
4
|
Peh HY, Chen J. Pro-resolving lipid mediators and therapeutic innovations in resolution of inflammation. Pharmacol Ther 2025; 265:108753. [PMID: 39566561 DOI: 10.1016/j.pharmthera.2024.108753] [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: 05/13/2024] [Revised: 11/07/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024]
Abstract
This review summarizes findings presented at the 19th World Congress of Basic & Clinical Pharmacology 2023 (Glasgow, Scotland, July 3rd to 7th, 2023) from 8 speakers in the field of resolution of inflammation, resolution pharmacology and resolution biology. It is now accepted that the acute inflammatory response is protective to defend the host against infection or tissue injury. Acute inflammation is self-limited and programmed to be limited in space and time: this is achieved through endogenous resolution processes that ensure return to homeostasis. Resolution is brought about by agonist mediators that include specialized pro-resolving lipid mediators (SPMs) and pro-resolving proteins and peptides such as annexin A1 and angiotensin-(1-7), all acting to initiate anti-inflammatory and pro-resolving processes. If the inflammatory reaction remains unchecked through dysfunctional resolution mechanism, it can become chronic and contribute to a plethora of human diseases, including respiratory, cardiovascular, metabolic, allergic diseases, and arthritis. Herein, we discuss how non-resolving inflammation plays a role in the pathogenesis of these diseases. In addition to SPMs, we highlight the discovery, biosynthesis, biofunctions, and latest research updates on innovative therapeutics (including annexin-A1 peptide-mimetic RTP-026, small molecule FPR2 agonist BM-986235/LAR-1219, biased agonist for FPR1/FPR2 Cmpd17b, lipoxin mimetics AT-01-KG and AT-02-CT, melanocortin receptor agonist AP1189, gold nanoparticles, angiotensin-(1-7), and CD300a) that can promote resolution of inflammation directly or through modulation of SPMs production. Drug development strategies based on the biology of the resolution of inflammation can offer novel therapeutic means and/or add-on therapies for the treatment of chronic diseases.
Collapse
Affiliation(s)
- Hong Yong Peh
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Pharmacology, Singapore; Immunology Programme and Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore; Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Jianmin Chen
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom; Centre for inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
5
|
Oboma YI, Ekpenyong BO, Umar MS, Nja GME, Chelimo JJ, Igwe MC, Bunu UO. Histopathological, Cytological and Radiological Correlations in Allergy and Public Health Concerns: A Comprehensive Review. J Asthma Allergy 2024; 17:1333-1354. [PMID: 39749282 PMCID: PMC11693939 DOI: 10.2147/jaa.s498641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
Allergies represent a significant and growing public health concern, affecting millions worldwide and burdening healthcare systems substantially. Accurate diagnosis and understanding of allergy is crucial for effective management and treatment. This review aims to explore the historical evolution, current advances, and prospects of histopathological and cytological techniques in allergy diagnosis, highlighting their crucial role in modern medicine. Major biomedical, public health, and imaging databases such as PubMed, Scopus, Web of Science, and EMBASE were used. The search strategy used include specific keywords and Medical Subject Headings (MeSH) terms related to histopathology, cytology, radiology, allergic diseases, and public health. Histopathological and cytological studies play a pivotal role in elucidating the underlying mechanisms of allergies, offering insights into the cellular and tissue-level changes associated with allergic responses. Histopathology reveals characteristic features such as inflammation, tissue remodeling, and the presence of specific immune cells like eosinophils and mast cells. Cytological analysis can detect cellular changes and abnormalities at a finer scale, providing a complementary perspective to histopathological findings. The correlation between histopathological and cytological findings is critical for achieving accurate and reliable diagnoses. Combined histopathological and cytological studies can reveal the extent of airway inflammation, epithelial damage, and immune cell infiltration, providing a robust basis for clinical decision-making. Recent advancements in diagnostic techniques have further revolutionized the field of allergy diagnosis. These technologies offer increased accuracy, speed, and reproducibility, making them invaluable in both clinical and research settings. Despite these advancements, several challenges and limitations persist. By integrating tissue-level and cellular-level analyses, clinicians can achieve more accurate diagnoses, tailor treatments to individual patients, and ultimately improve the quality of care for those suffering from allergies. In conclusion, histopathological and cytological correlation in allergy diagnosis provides a comprehensive framework for understanding and managing allergic conditions.
Collapse
Affiliation(s)
- Yibala Ibor Oboma
- Department of Medical Laboratory Sciences, School of Allied Health Sciences, Kampala International University Western Campus, Ishaka, Bushenyi, Uganda
| | - Bassey Okon Ekpenyong
- Department of Histopathology, Faculty of Medical Laboratory Science, Rivers State University Nkpolo - Oroworukwo, Port Harcourt, River State, Nigeria
| | - Mohammed Sani Umar
- Department of Radiography, School of Allied Health Sciences, Kampala International University, Western Campus, Ishaka, Bushenyi, Uganda
| | - Glory Mbe Egom Nja
- Department of Public Health, School of Allied Medical Sciences, Kampala International University, Western Campus, Ishaka, Bushenyi, Uganda
| | - Judith Jepkosgei Chelimo
- Department of Public Health, School of Allied Medical Sciences, Kampala International University, Western Campus, Ishaka, Bushenyi, Uganda
| | - Matthew Chibunna Igwe
- Department of Public Health, School of Allied Medical Sciences, Kampala International University, Western Campus, Ishaka, Bushenyi, Uganda
| | - Umi Omar Bunu
- Department of Public Health, School of Allied Medical Sciences, Kampala International University, Western Campus, Ishaka, Bushenyi, Uganda
| |
Collapse
|
6
|
Lefèvre G, Gibier JB, Bongiovanni A, Lhermitte L, Rossignol J, Anglo E, Dendooven A, Dubois R, Terriou L, Launay D, Barete S, Esnault S, Frenzel L, Gourguechon C, Ballul T, Dezoteux F, Staumont-Salle D, Copin MC, Rignault-Bricard R, Maciel TT, Damaj G, Tardivel M, Crinquette-Verhasselt M, Dubreuil P, Maouche-Chrétien L, Bruneau J, Lortholary O, Duployez N, Behal H, Molina TJ, Hermine O. Interactions between eosinophils and IL-5Rα-positive mast cells in nonadvanced systemic mastocytosis. J Allergy Clin Immunol 2024; 154:1523-1533. [PMID: 39151478 DOI: 10.1016/j.jaci.2024.07.025] [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: 12/18/2023] [Revised: 06/27/2024] [Accepted: 07/10/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Bidirectional interactions between eosinophils and mast cells (MCs) have been reported in various allergic diseases. Bone marrow (BM) eosinophilia, and to a lesser extent blood eosinophilia, is common in systemic mastocytosis (SM), but its significance remains unknown. OBJECTIVE We described blood and BM eosinophil characteristics in SM. METHODS A large collection of BM biopsy samples was analyzed using immunohistochemical staining and whole-slide imaging. Eosinophil and extracellular granules were detected by eosinophil peroxidase (EPX) staining and MCs by KIT staining. Complementary analyses were conducted using flow cytometry and immunofluorescence. RESULTS Eosinophil infiltrates and large areas of eosinophil degranulation were observed within or around BM MC infiltrates in SM. EPX staining surface, highlighting intact eosinophils and eosinophil degranulation, was higher in nonadvanced SM (n = 37 BM biopsy samples) compared with both controls (n = 8, P = .0003) and advanced SM (n = 24, P = .014). In nonadvanced SM, positive correlations were observed between serum tryptase levels and percentages of eosinophil counts in BM aspirations (Spearman r coefficient r = 0.38, P = .038), eosinophils count in BM biopsy samples (r = 0.45, P = .007), EPX staining (r = 0.37, P = .035), and eosinophil degranulation (r = 0.39, P = .023). Eosinophil counts in BM biopsy samples also correlated with MC counts (r = 0.47, P = .006) and KIT staining surface (r = 0.49, P = .003). BM MCs expressed IL-5 receptor and other usual eosinophil cytokine/chemokine receptors, and blood eosinophils displayed several increased surface markers compared with controls, suggesting an activated state. CONCLUSION Our data suggest possible cross talk between MCs and eosinophils, supporting MC tryptase release and MC activation-related symptoms. This suggests a rationale for targeting eosinophils in nonadvanced SM not fully controlled by other therapies.
Collapse
Affiliation(s)
- Guillaume Lefèvre
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Lille, and Institute for Translational Research in Inflammation (INFINITE), Lille, France; Institut d'Immunologie, CHU Lille, Lille, France; National Reference Center for Hypereosinophilic Syndromes (CEREO).
| | - Jean-Baptiste Gibier
- University of Lille, Institut de Pathologie, Centre de Biopathologie, CHU Lille, Lille, France
| | - Antonino Bongiovanni
- Centre National de la Recherche Scientifique (CNRS), INSERM, CHU Lille, University of Lille, Institut Pasteur de Lille, Lille, France
| | - Ludovic Lhermitte
- Laboratoire d'Onco-Hématologie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; University of Paris, Institut Imagine, INSERM, Paris, France; French Reference Center for Mastocytosis (CEREMAST), Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Julien Rossignol
- University of Paris, Institut Imagine, INSERM, Paris, France; French Reference Center for Mastocytosis (CEREMAST), Hôpital Necker-Enfants Malades, AP-HP, Paris, France; Department of Hematology, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Emilie Anglo
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Lille, and Institute for Translational Research in Inflammation (INFINITE), Lille, France; Institut d'Immunologie, CHU Lille, Lille, France
| | - Arnaud Dendooven
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Lille, and Institute for Translational Research in Inflammation (INFINITE), Lille, France; Institut d'Immunologie, CHU Lille, Lille, France
| | - Romain Dubois
- University of Lille, Institut de Pathologie, Centre de Biopathologie, CHU Lille, Lille, France
| | - Louis Terriou
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Lille, and Institute for Translational Research in Inflammation (INFINITE), Lille, France; National Reference Center for Hypereosinophilic Syndromes (CEREO); Département de Médecine Interne et Immunologie Clinique, CHU Lille, Lille, France
| | - David Launay
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Lille, and Institute for Translational Research in Inflammation (INFINITE), Lille, France; National Reference Center for Hypereosinophilic Syndromes (CEREO); Département de Médecine Interne et Immunologie Clinique, CHU Lille, Lille, France
| | - Stéphane Barete
- CEREMAST, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Unit de Dermatologie, Sorbonne Université Paris, Paris, France
| | - Stéphane Esnault
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Lille, and Institute for Translational Research in Inflammation (INFINITE), Lille, France; Institut d'Immunologie, CHU Lille, Lille, France; Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wis
| | - Laurent Frenzel
- University of Paris, Institut Imagine, INSERM, Paris, France; French Reference Center for Mastocytosis (CEREMAST), Hôpital Necker-Enfants Malades, AP-HP, Paris, France; Department of Hematology, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | | | - Thomas Ballul
- University of Paris, Institut Imagine, INSERM, Paris, France; French Reference Center for Mastocytosis (CEREMAST), Hôpital Necker-Enfants Malades, AP-HP, Paris, France; Department of Hematology, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Frédéric Dezoteux
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Lille, and Institute for Translational Research in Inflammation (INFINITE), Lille, France; National Reference Center for Hypereosinophilic Syndromes (CEREO); Department of Dermatology, CHU Lille, Lille, France
| | - Delphine Staumont-Salle
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Lille, and Institute for Translational Research in Inflammation (INFINITE), Lille, France; National Reference Center for Hypereosinophilic Syndromes (CEREO); Department of Dermatology, CHU Lille, Lille, France
| | - Marie-Christine Copin
- Department of Pathology, CHU Angers, University of Angers, INSERM, CNRS, CRCI(2)NA, Angers, France
| | | | | | - Gandhi Damaj
- Institut d'Hématologie, University of Caen Normandie, Caen, France
| | - Meryem Tardivel
- Centre National de la Recherche Scientifique (CNRS), INSERM, CHU Lille, University of Lille, Institut Pasteur de Lille, Lille, France
| | | | - Patrice Dubreuil
- Signaling, Hematopoiesis, and Mechanism of Oncogenesis (CRCM), CEREMAST, and Association Française pour les Initiatives de Recherche sur le Mastocyte et les Mastocytose (AFIRMM) studies, INSERM U1068; Institut Paoli-Calmettes; UM105, Aix-Marseille University; and CNRS, UMR7258, Marseille, France
| | | | - Julie Bruneau
- University of Paris, Institut Imagine, INSERM, Paris, France; French Reference Center for Mastocytosis (CEREMAST), Hôpital Necker-Enfants Malades, AP-HP, Paris, France; Department of Pathology, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Olivier Lortholary
- University of Paris, Institut Imagine, INSERM, Paris, France; French Reference Center for Mastocytosis (CEREMAST), Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Nicolas Duployez
- CNRS, INSERM, CHU Lille, Cancer Heterogeneity, Plasticity, and Resistance to Therapies (CANTHER), University of Lille, Institut d'Hématologie, CHU Lille, Lille, France
| | - Hélène Behal
- University of Lille, CHU Lille, Evaluation des Technologies de Santé et des Pratiques Médicales (METRICS), Lille, France
| | - Thierry Jo Molina
- University of Paris, Institut Imagine, INSERM, Paris, France; Signaling, Hematopoiesis, and Mechanism of Oncogenesis (CRCM), CEREMAST, and Association Française pour les Initiatives de Recherche sur le Mastocyte et les Mastocytose (AFIRMM) studies, INSERM U1068; Institut Paoli-Calmettes; UM105, Aix-Marseille University; and CNRS, UMR7258, Marseille, France
| | - Olivier Hermine
- University of Paris, Institut Imagine, INSERM, Paris, France; French Reference Center for Mastocytosis (CEREMAST), Hôpital Necker-Enfants Malades, AP-HP, Paris, France; Department of Hematology, Hôpital Necker-Enfants Malades, AP-HP, Paris, France.
| |
Collapse
|
7
|
Zhang M, Cui Y, Liu P, Mo R, Wang H, Li Y, Wu Y. Oat β-(1 → 3, 1 → 4)-d-glucan alleviates food allergy-induced colonic injury in mice by increasing Lachnospiraceae abundance and butyrate production. Carbohydr Polym 2024; 344:122535. [PMID: 39218555 DOI: 10.1016/j.carbpol.2024.122535] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 07/18/2024] [Accepted: 07/21/2024] [Indexed: 09/04/2024]
Abstract
Oat β-(1 → 3, 1 → 4)-d-glucan (OBG), a linear polysaccharide primarily found in oat bran, has been demonstrated to possess immunomodulatory properties and regulate gut microbiota. This study aimed to investigate the impact of low molecular weight (Mw) OBG (155.2 kDa) on colonic injury and allergic symptoms induced by food allergy (FA), and to explore its potential mechanism. In Experiment 1, results indicated that oral OBG improved colonic inflammation and epithelial barrier, and significantly relieved allergy symptoms. Importantly, the OBG supplement altered the gut microbiota composition, particularly increasing the abundance of Lachnospiraceae and its genera, and promoted the production of short-chain fatty acids, especially butyrate. However, in Experiment 2, the gut microbial depletion eliminated these protective effects of OBG on the colon in allergic mice. Further, in Experiment 3, fecal microbiota transplantation and sterile fecal filtrate transfer directly validated the role of OBG-mediated gut microbiota and its metabolites in relieving FA and its induced colonic injury. Our findings suggest that low Mw OBG can alleviate FA-induced colonic damage by increasing Lachnospiraceae abundance and butyrate production, and provide novel insights into the health benefits and mechanisms of dietary polysaccharide intervention for FA.
Collapse
Affiliation(s)
- Mingrui Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Yingyue Cui
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Pan Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Ruixia Mo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Haotian Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Yingying Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Yi Wu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
8
|
Retzinger AC, Retzinger GS. The Acari Hypothesis, V: deciphering allergenicity. FRONTIERS IN ALLERGY 2024; 5:1454292. [PMID: 39552700 PMCID: PMC11565521 DOI: 10.3389/falgy.2024.1454292] [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: 06/24/2024] [Accepted: 10/18/2024] [Indexed: 11/19/2024] Open
Abstract
The Acari Hypothesis posits that acarians, i.e., mites and ticks, are operative agents of allergy. It derived from observations that allergens are molecular elements of acarians or acarian foodstuffs. A corollary of The Hypothesis provides how acarian dietary elements are selected as allergens; namely, a pattern recognition receptor native to the acarian digestive tract complexes with dietary molecules problematic to the acarian. By virtue of its interspecies operability, the receptor then enables not only removal of the dietary elements by the acarian immune system, but also-should such a complex be inoculated into a human-production of an element-specific IgE. Because pattern recognition receptors bind to molecules problematic to the organism from which the receptors originate, it follows that molecules targeted by adaptive IgE, i.e., allergens, must be problematic to acarians. This claim is supported by evidence that host organisms, when infested by acarians, upregulate representative members of allergenic molecular families. Appreciation of the relationship between allergens and acarians provides insight well beyond allergy, shedding light also on the anti-acarian defenses of many living things, especially humans.
Collapse
Affiliation(s)
- Andrew C. Retzinger
- Department of Emergency Medicine, Camden Clark Medical Center, West Virginia University, Parkersburg, WV, United States
| | - Gregory S. Retzinger
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
9
|
Bai W, Su H, Xu S, Gao Z, Chang Z, Sun X, Liu T. Cyp2e1 protects against OVA-induced allergic rhinitis through the inhibition of Th2 cell activation and differentiation: Mediated by MAFB. Int Immunopharmacol 2024; 132:112003. [PMID: 38603858 DOI: 10.1016/j.intimp.2024.112003] [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: 11/09/2023] [Revised: 03/26/2024] [Accepted: 04/01/2024] [Indexed: 04/13/2024]
Abstract
Allergic rhinitis (AR) is a common allergic disease. Cytochrome P450, family 2, subfamily e, polypeptide 1 (Cyp2e1) is a member of the cytochrome P450 family of enzymes, while its role in AR is still unveiled. In AR mice, T cell-specific overexpression of Cyp2e1 relieved the AR symptoms. Overexpressed-Cyp2e1 restrained the infiltration of eosinophils and mast cells in the nasal mucosa of mice, and the inflammatory cells in nasal lavage fluid (NALF). Cyp2e1 overexpressed mice exhibited decreased goblet cell hyperplasia and mucus secretion as well as decreased MUC5AC expression in nasal mucosa. The epithelial permeability and integrity of nasal mucosa were improved upon Cyp2e1 overexpression in AR mice, as evidenced by decreased fluorescein isothiocyanate-dextran 4 content in serum, increased expression of IL-25, IL-33, and TSLP in NALF, and increased expression of ZO-1 and occluding in nasal mucosa. Cyp2e1 inhibited Th2 immune response by decreasing the expression and secretion of IL-4, IL-5, and IL-13 as well as the expression of GATA-3 in NALF or nasal mucosa. We proved that Cyp2e1 inhibited the differentiation of naïve CD4+ T cells toward the Th2 subtype, which was regulated by MAFB by binding to Cyp2e1 promoter to activate its transcription. Overall, these results show the potential role of Cyp2e1 in alleviating AR symptoms by restraining CD4+ T cells to Th2 cell differentiation. Our findings provide further insight into the AR mechanism.
Collapse
Affiliation(s)
- Weiliang Bai
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Hui Su
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Shengqun Xu
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Zhao Gao
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Ziwen Chang
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Xun Sun
- Department of Immunology, College of Basic Medicine, China Medical University, Shenyang, PR China
| | - Tiancong Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
10
|
Jahanbani F, Sing JC, Maynard RD, Jahanbani S, Dafoe J, Dafoe W, Jones N, Wallace KJ, Rastan A, Maecker HT, Röst HL, Snyder MP, Davis RW. Longitudinal cytokine and multi-modal health data of an extremely severe ME/CFS patient with HSD reveals insights into immunopathology, and disease severity. Front Immunol 2024; 15:1369295. [PMID: 38650940 PMCID: PMC11033372 DOI: 10.3389/fimmu.2024.1369295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Introduction Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) presents substantial challenges in patient care due to its intricate multisystem nature, comorbidities, and global prevalence. The heterogeneity among patient populations, coupled with the absence of FDA-approved diagnostics and therapeutics, further complicates research into disease etiology and patient managment. Integrating longitudinal multi-omics data with clinical, health,textual, pharmaceutical, and nutraceutical data offers a promising avenue to address these complexities, aiding in the identification of underlying causes and providing insights into effective therapeutics and diagnostic strategies. Methods This study focused on an exceptionally severe ME/CFS patient with hypermobility spectrum disorder (HSD) during a period of marginal symptom improvements. Longitudinal cytokine profiling was conducted alongside the collection of extensive multi-modal health data to explore the dynamic nature of symptoms, severity, triggers, and modifying factors. Additionally, an updated severity assessment platform and two applications, ME-CFSTrackerApp and LexiTime, were introduced to facilitate real-time symptom tracking and enhance patient-physician/researcher communication, and evaluate response to medical intervention. Results Longitudinal cytokine profiling revealed the significance of Th2-type cytokines and highlighted synergistic activities between mast cells and eosinophils, skewing Th1 toward Th2 immune responses in ME/CFS pathogenesis, particularly in cognitive impairment and sensorial intolerance. This suggests a potentially shared underlying mechanism with major ME/CFS comorbidities such as HSD, Mast cell activation syndrome, postural orthostatic tachycardia syndrome (POTS), and small fiber neuropathy. Additionally, the data identified potential roles of BCL6 and TP53 pathways in ME/CFS etiology and emphasized the importance of investigating adverse reactions to medication and supplements and drug interactions in ME/CFS severity and progression. Discussion Our study advocates for the integration of longitudinal multi-omics with multi-modal health data and artificial intelligence (AI) techniques to better understand ME/CFS and its major comorbidities. These findings highlight the significance of dysregulated Th2-type cytokines in patient stratification and precision medicine strategies. Additionally, our results suggest exploring the use of low-dose drugs with partial agonist activity as a potential avenue for ME/CFS treatment. This comprehensive approach emphasizes the importance of adopting a patient-centered care approach to improve ME/CFS healthcare management, disease severity assessment, and personalized medicine. Overall, these findings contribute to our understanding of ME/CFS and offer avenues for future research and clinical practice.
Collapse
Affiliation(s)
- Fereshteh Jahanbani
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Justin Cyril Sing
- Department of Molecular Genetics, Donnelly Center, University of Toronto, Toronto, ON, Canada
| | - Rajan Douglas Maynard
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Shaghayegh Jahanbani
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Veterans Affairs (VA) Palo Alto Health Care System, Palo Alto, CA, United States
| | - Janet Dafoe
- ME/CFS Collaborative Research Center at Stanford, Stanford Genome Technology Center, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Whitney Dafoe
- ME/CFS Collaborative Research Center at Stanford, Stanford Genome Technology Center, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Nathan Jones
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Kelvin J. Wallace
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Azuravesta Rastan
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Holden T. Maecker
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Pulmonary and Critical Care Medicine, Institute of Immunity, Transplantation, and Infectious Diseases, Stanford University, Palo Alto, CA, United States
| | - Hannes L. Röst
- Department of Molecular Genetics, Donnelly Center, University of Toronto, Toronto, ON, Canada
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Ronald W. Davis
- ME/CFS Collaborative Research Center at Stanford, Stanford Genome Technology Center, Stanford University School of Medicine, Palo Alto, CA, United States
| |
Collapse
|
11
|
Deng B, Zhao Y, Liu J. Downregulation of lncRNA CDKN2B-AS1 attenuates inflammatory response in mice with allergic rhinitis by regulating miR-98-5p/SOCS1 axis. Funct Integr Genomics 2024; 24:48. [PMID: 38436805 PMCID: PMC10912270 DOI: 10.1007/s10142-024-01318-x] [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: 12/02/2023] [Revised: 01/24/2024] [Accepted: 02/13/2024] [Indexed: 03/05/2024]
Abstract
Long non-coding RNA cyclin-dependent kinase inhibitor 2B antisense RNA 1 (CDKN2B-AS1) in various diseases has been verified. However, the underlying mechanism of CDKN2B-AS1 contributes to the development of allergic rhinitis (AR) remains unknown. To evaluate the impact of CDKN2B-AS1 on AR, BALB/c mice were sensitized by intraperitoneal injection of normal saline containing ovalbumin (OVA) and calmogastrin to establish an AR model. Nasal rubbing and sneezing were documented after the final OVA treatment. The concentrations of IgE, IgG1, and inflammatory elements were quantified using ELISA. Hematoxylin and eosin (H&E) staining and immunofluorescence were used to assess histopathological variations and tryptase expression, respectively. StarBase, TargetScan and luciferase reporter assays were applied to predict and confirm the interactions among CDKN2B-AS1, miR-98-5p, and SOCS1. CDKN2B-AS1, miR-98-5p, and SOCS1 levels were assessed by quantitative real-time PCR (qRT-PCR) or western blotting. Our results revealed that CDKN2B-AS1 was obviously over-expressed in the nasal mucosa of AR patients and AR mice. Down-regulation of CDKN2B-AS1 significantly decreased nasal rubbing and sneezing frequencies, IgE and IgG1 concentrations, and cytokine levels. Furthermore, down-regulation of CDKN2B-AS1 also relieved the pathological changes in the nasal mucosa, and the infiltration of eosinophils and mast cells. Importantly, these results were reversed by the miR-98-5p inhibitor, whereas miR-98-5p directly targeted CDKN2B-AS1, and miR-98-5p negatively regulated SOCS1 level. Our findings demonstrate that down-regulation of CDKN2B-AS1 improves allergic inflammation and symptoms in a murine model of AR through the miR-98-5p/SOCS1 axis, which provides new insights into the latent functions of CDKN2B-AS1 in AR treatment.
Collapse
Affiliation(s)
- Bangyu Deng
- Department of Otolaryngology, First Affiliated Hospital of Soochow University, No.899, Pinghai Road, Suzhou, 215006, Jiangsu, China
- Department of Otolaryngology-Head and Neck Surgery, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yunxia Zhao
- Department of Maternal and Child Health, Suzhou Jinji Lake Health Service Center, Suzhou, Jiangsu, China
| | - Jisheng Liu
- Department of Otolaryngology, First Affiliated Hospital of Soochow University, No.899, Pinghai Road, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
12
|
Oshima T. Functional Dyspepsia: Current Understanding and Future Perspective. Digestion 2023; 105:26-33. [PMID: 37598673 DOI: 10.1159/000532082] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023]
Abstract
BACKGROUND Functional dyspepsia (FD) is a common disorder characterized by chronic or recurrent upper abdominal pain or discomfort without any structural abnormalities in the gastrointestinal tract. FD is categorized into two subgroups based on symptoms: postprandial distress syndrome (PDS) and epigastric pain syndrome. SUMMARY The pathophysiology of FD involves several mechanisms. Delayed gastric emptying is observed in approximately 30% of FD patients but does not correlate with symptom patterns or severity. Impaired gastric accommodation is important in the pathophysiology, particularly for PDS. Visceral hypersensitivity, characterized by heightened sensitivity to normal activities, contributes to the perception of discomfort or pain in FD. Alterations to the duodenal mucosa, including impaired mucosal barrier function and low-grade inflammation, are also implicated in the pathogenesis of FD. Microbial dysbiosis and psychological factors such as stress can further exacerbate symptoms. Treatment options include dietary modifications, establishing a physician-patient relationship, acid suppressants, prokinetics, neuromodulators, and behavioral therapies. Dietary recommendations include eating smaller, more frequent meals, and avoiding trigger foods. Acid suppressants are used as the first-line treatment. Prokinetics and neuromodulators aim to improve gastric motility and central pain processing, respectively. Behavioral therapies, including cognitive behavioral therapy and hypnotherapy, have shown benefits for refractory FD. Severe and refractory cases may require combination therapies or experimental treatments. KEY MESSAGES FD is a disorder of gut-brain interaction involving diverse pathophysiological mechanisms. Individualized treatment based on symptoms and responses to interventions is crucial. Further research is needed to improve the understanding of FD and advance the development of effective therapies.
Collapse
Affiliation(s)
- Tadayuki Oshima
- Department of Gastroenterology, Okazaki City Medical Association Public Health Center, Okazaki, Japan
| |
Collapse
|
13
|
Qi L, Qiu Y, Li S, Yi N, Li C, Teng Z, Li S, Xu X, Lang B, Chen J, Zheng C, Yang Y, Hua J, Wang C, Wu H, Xue Z, Lv B. Single-cell immune profiling reveals broad anti-inflammation response in bipolar disorder patients with quetiapine and valproate treatment. iScience 2023; 26:107057. [PMID: 37534158 PMCID: PMC10391734 DOI: 10.1016/j.isci.2023.107057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/22/2023] [Accepted: 06/01/2023] [Indexed: 08/04/2023] Open
Abstract
Bipolar disorder (BD) is a common mental disorder characterized by manic and depressive episodes. Mood disorders have been associated with immune dysfunction. The combination of quetiapine and valproate has shown positive effects in treating BD, but the impact on immune dynamics remains less understood. Using single-cell RNA sequencing, we observed that B cells exhibited downregulation of inflammation-related genes, while pro-inflammatory mast and eosinophil cells decreased following treatment. Ribosomal peptide production genes were found to be reduced in both B and T cells after treatment. Additionally, our findings suggest that the combined therapy effectively alleviates inflammation by reducing myloid-mediated immune signaling pathways. This study provides valuable insights into the immune atlas and uncovers a potential mechanism for immune disorder alleviation in patients with BD treated with quetiapine and valproate.
Collapse
Affiliation(s)
- Lingbin Qi
- Department of Regenerative Medicine, Translational Center for Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Yan Qiu
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Sujuan Li
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Ning Yi
- Department of Regenerative Medicine, Translational Center for Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Chanyi Li
- Department of Regenerative Medicine, Translational Center for Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Ziwei Teng
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Shiping Li
- Stem Cell and Regenerative Medicine Engineering Research Center of Hunan Province, Hunan Yuanpin Cell Technology Co. Ltd, 102 Dongwu Road, Changsha City, Hunan Province 410100, China
| | - Xuelei Xu
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Bin Lang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Jindong Chen
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
- Department of Psychiatry, Xiangya Boai Rehabilitation Hospital, Changsha 410100, China
| | - Chunbing Zheng
- Stem Cell and Regenerative Medicine Engineering Research Center of Hunan Province, Hunan Yuanpin Cell Technology Co. Ltd, 102 Dongwu Road, Changsha City, Hunan Province 410100, China
| | - Yuan Yang
- Stem Cell and Regenerative Medicine Engineering Research Center of Hunan Province, Hunan Yuanpin Cell Technology Co. Ltd, 102 Dongwu Road, Changsha City, Hunan Province 410100, China
| | - Jiangzhou Hua
- Stem Cell and Regenerative Medicine Engineering Research Center of Hunan Province, Hunan Yuanpin Cell Technology Co. Ltd, 102 Dongwu Road, Changsha City, Hunan Province 410100, China
| | - Cheng Wang
- Stem Cell and Regenerative Medicine Engineering Research Center of Hunan Province, Hunan Yuanpin Cell Technology Co. Ltd, 102 Dongwu Road, Changsha City, Hunan Province 410100, China
| | - Haishan Wu
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhigang Xue
- Department of Regenerative Medicine, Translational Center for Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Bo Lv
- Department of Regenerative Medicine, Translational Center for Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, China
| |
Collapse
|
14
|
Elieh-Ali-Komi D, Metz M, Kolkhir P, Kocatürk E, Scheffel J, Frischbutter S, Terhorst-Molawi D, Fox L, Maurer M. Chronic urticaria and the pathogenic role of mast cells. Allergol Int 2023:S1323-8930(23)00047-3. [PMID: 37210251 DOI: 10.1016/j.alit.2023.05.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 04/24/2023] [Indexed: 05/22/2023] Open
Abstract
The signs and symptoms of chronic urticaria (CU) are caused by the activation and degranulation of skin mast cells (MCs). Recent studies have added to our understanding of how and why skin MCs are involved and different in CU. Also, novel and relevant mechanisms of MC activation in CU have been identified and characterized. Finally, the use of MC-targeted and MC mediator-specific treatments has helped to better define the role of the skin environment, the contribution of specific MC mediators, and the relevance of MC crosstalk with other cells in the pathogenesis of CU. Here, we review these recent findings and their impact on our understanding of CU, with a focus on chronic spontaneous urticaria (CSU). Also, we highlight open questions, issues of controversy, and unmet needs, and we suggest what studies should be performed moving forward.
Collapse
Affiliation(s)
- Daniel Elieh-Ali-Komi
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Allergology and Immunology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany
| | - Martin Metz
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Allergology and Immunology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany
| | - Pavel Kolkhir
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Allergology and Immunology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany
| | - Emek Kocatürk
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Allergology and Immunology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany; Department of Dermatology, Koç University School of Medicine, Istanbul, Turkey
| | - Jörg Scheffel
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Allergology and Immunology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany
| | - Stefan Frischbutter
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Allergology and Immunology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany
| | - Dorothea Terhorst-Molawi
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Allergology and Immunology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany
| | - Lena Fox
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Allergology and Immunology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany
| | - Marcus Maurer
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Allergology and Immunology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany.
| |
Collapse
|
15
|
Franke K, Bal G, Li Z, Zuberbier T, Babina M. Clorfl86/RHEX Is a Negative Regulator of SCF/KIT Signaling in Human Skin Mast Cells. Cells 2023; 12:cells12091306. [PMID: 37174705 PMCID: PMC10177086 DOI: 10.3390/cells12091306] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/20/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023] Open
Abstract
Mast cells (MCs) are key effector cells in allergic and inflammatory diseases, and the SCF/KIT axis regulates most aspects of the cells' biology. Using terminally differentiated skin MCs, we recently reported on proteome-wide phosphorylation changes initiated by KIT dimerization. C1orf186/RHEX was revealed as one of the proteins to become heavily phosphorylated. Its function in MCs is undefined and only some information is available for erythroblasts. Using public databases and our own data, we now report that RHEX exhibits highly restricted expression with a clear dominance in MCs. While expression is most pronounced in mature MCs, RHEX is also abundant in immature/transformed MC cell lines (HMC-1, LAD2), suggesting early expression with further increase during differentiation. Using RHEX-selective RNA interference, we reveal that RHEX unexpectedly acts as a negative regulator of SCF-supported skin MC survival. This finding is substantiated by RHEX's interference with KIT signal transduction, whereby ERK1/2 and p38 both were more strongly activated when RHEX was attenuated. Comparing RHEX and capicua (a recently identified repressor) revealed that each protein preferentially suppresses other signaling modules elicited by KIT. Induction of immediate-early genes strictly requires ERK1/2 in SCF-triggered MCs; we now demonstrate that RHEX diminution translates to this downstream event, and thereby enhances NR4A2, JUNB, and EGR1 induction. Collectively, our study reveals RHEX as a repressor of KIT signaling and function in MCs. As an abundant and selective lineage marker, RHEX may have various roles in the lineage, and the provided framework will enable future work on its involvement in other crucial processes.
Collapse
Affiliation(s)
- Kristin Franke
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Gürkan Bal
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Zhuoran Li
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Torsten Zuberbier
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Magda Babina
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| |
Collapse
|
16
|
Kow ASF, Khoo LW, Tan JW, Abas F, Lee MT, Israf DA, Shaari K, Tham CL. Clinacanthus nutans aqueous leaves extract exerts anti-allergic activity in preclinical anaphylactic models via alternative IgG pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 303:116003. [PMID: 36464074 DOI: 10.1016/j.jep.2022.116003] [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: 03/06/2022] [Revised: 10/31/2022] [Accepted: 11/28/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Allergy is mediated by the crosslinking of immunoglobulins (Ig) -E or -G to their respective receptors, which degranulates mast cells, macrophages, basophils, or neutrophils, releasing allergy-causing mediators. The removal of these mediators such as histamine, platelet-activating factor (PAF) and interleukins (ILs) released by effector cells will alleviate allergy. Clinacanthus nutans (C. nutans), an herbal plant in Southeast Asia, is used traditionally to treat skin rash, an allergic symptom. Previously, we have reported that C. nutans aqueous leaves extract (CNAE) was able to suppress the release of β-hexosaminidase and histamine but not interleukin-4 (IL-4) and tumor necrosis factor-alpha (TNF-α) in the IgE-induced mast cell degranulation model at 5 mg/mL and above. We also found that CNAE could protect rats against ovalbumin-challenged active systemic anaphylaxis (OVA-ASA) through the downregulation and upregulation of certain metabolites using proton nuclear magnetic resonance (1H-NMR) metabolomics approach. AIM OF THE STUDY As allergy could be mediated by both IgE and IgG, we further evaluated the anti-allergy potential of CNAE in both in vitro model of IgG-induced macrophage activation and in vivo anaphylaxis models to further dissect the mechanism of action underlying the anti-allergic properties of CNAE. MATERIAL & METHODS The anti-allergy potential of CNAE was evaluated in in vivo anaphylaxis models of ovalbumin-challenged active systemic anaphylaxis (OVA-ASA) and IgE-challenged passive systemic anaphylaxis (PSA) using Sprague Dawley rats as well as IgG-challenged passive systemic anaphylaxis (IgG-PSA) using C57BL/6 mice. Meanwhile, in vitro model of IgG-induced macrophage activation model was performed using IC-21 macrophages. The release of soluble mediators from both IgE and IgG-mediated pathways were measured using enzyme-linked immunosorbent assay (ELISA). The signaling molecules targeted by CNAE were identified by performing Western blot. RESULTS IgG, platelet-activating factor (PAF) and IL-6 was suppressed by CNAE in OVA-ASA, but not IgE. In addition, CNAE significantly suppressed PAF and IL-6 in IgG-PSA but did not suppress histamine, IL-4 and leukotrienes C4 (LTC4) in IgE-PSA. CNAE also inhibited IL-6 and TNF-α by inhibiting the phosphorylation of ERK1/2 in the IgG-induced macrophage activation model. CONCLUSION Overall, our findings supported that CNAE exerts its anti-allergic properties by suppressing the IgG pathway and its mediators by inhibiting ERK1/2 phosphorylation, thus providing scientific evidence supporting its traditional use in managing allergy.
Collapse
Affiliation(s)
- Audrey Siew Foong Kow
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia; Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, 56000, Malaysia.
| | - Leng Wei Khoo
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, 43400, Malaysia.
| | - Ji Wei Tan
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia; School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Malaysia.
| | - Faridah Abas
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, 43400, Malaysia; Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, Serdang, 43400, Malaysia.
| | - Ming-Tatt Lee
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, 56000, Malaysia; Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan; Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan.
| | - Daud Ahmad Israf
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia.
| | - Khozirah Shaari
- Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, Serdang, 43400, Malaysia.
| | - Chau Ling Tham
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia.
| |
Collapse
|
17
|
Liu P, Liu T, Zhang M, Mo R, Zhou W, Li D, Wu Y. Effects of Avenanthramide on the Small Intestinal Damage through Hsp70-NF-κB Signaling in an Ovalbumin-Induced Food Allergy Model. Int J Mol Sci 2022; 23:ijms232315229. [PMID: 36499554 PMCID: PMC9739943 DOI: 10.3390/ijms232315229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/11/2022] Open
Abstract
A food allergy is caused by an abnormal immune reaction and can induce serious intestinal inflammation and tissue damage. Currently, the avoidance of food allergens is still the most effective way to prevent or reduce allergic symptoms, so the development of new strategies to treat allergies is important. Avenanthramide (AVA) is a bioactive polyphenol derived from oats with a wide range of biological activities; however, it is still not clear whether or how AVA alleviates intestinal damage under allergic situations. The aim of this study was to explore the effect of AVA on the small intestinal damage in an ovalbumin (OVA)-induced food allergy model and its mechanism. In experiment 1, 10 mg/kg bw and 20 mg/kg bw doses of AVA both decreased the serum levels of OVA-specific IgE, histamine, and prostaglandin D induced by OVA. The AVA administration relieved inflammation indicated by the lower serum concentrations of pro-inflammatory cytokines including interleukin-1β, IL-6, and tumor necrosis factor-α. The levels of tight junction proteins including Claudin-1, ZO-1, and Occludin in the jejunum were elevated after AVA administration, accompanied by the improved intestinal morphology. Furthermore, AVA elevated the protein expression of heat shock protein 70 (Hsp70) and inhibited the phosphorylation of nuclear factor kappa-B (NF-κB), thus the apoptozole, which a Hsp70 inhibitor, was applied in experiment 2 to assess the contribution of Hsp70-NF-κB signaling to the effects of AVA. In the experiment 2, the inhibition of Hsp70 signaling treatment abolished the beneficial effects of AVA on the small intestinal damage and other allergic symptoms in mice challenged with OVA. Taken together, our results indicated that AVA exerted an intestinal protection role in the OVA-induced allergy, the mechanism of which was partly mediated by the Hsp70-NF-κB signaling.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yi Wu
- Correspondence: ; Tel.: +86-6273-3588
| |
Collapse
|
18
|
Babina M, Franke K, Bal G. How "Neuronal" Are Human Skin Mast Cells? Int J Mol Sci 2022; 23:ijms231810871. [PMID: 36142795 PMCID: PMC9505265 DOI: 10.3390/ijms231810871] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/05/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022] Open
Abstract
Mast cells are evolutionarily old cells and the principal effectors in allergic responses and inflammation. They are seeded from the yolk sac during embryogenesis or are derived from hematopoietic progenitors and are therefore related to other leukocyte subsets, even though they form a separate clade in the hematopoietic system. Herein, we systematically bundle information from several recent high-throughput endeavors, especially those comparing MCs with other cell types, and combine such information with knowledge on the genes’ functions to reveal groups of neuronal markers specifically expressed by MCs. We focus on recent advances made regarding human tissue MCs, but also refer to studies in mice. In broad terms, genes hyper-expressed in MCs, but largely inactive in other myelocytes, can be classified into subcategories such as traffic/lysosomes (MLPH and RAB27B), the dopamine system (MAOB, DRD2, SLC6A3, and SLC18A2), Ca2+-related entities (CALB2), adhesion molecules (L1CAM and NTM) and, as an overall principle, the transcription factors and modulators of transcriptional activity (LMO4, PBX1, MEIS2, and EHMT2). Their function in MCs is generally unknown but may tentatively be deduced by comparison with other systems. MCs share functions with the nervous system, as they express typical neurotransmitters (histamine and serotonin) and a degranulation machinery that shares features with the neuronal apparatus at the synapse. Therefore, selective overlaps are plausible, and they further highlight the uniqueness of MCs within the myeloid system, as well as when compared with basophils. Apart from investigating their functional implications in MCs, a key question is whether their expression in the lineage is due to the specific reactivation of genes normally silenced in leukocytes or whether the genes are not switched off during mastocytic development from early progenitors.
Collapse
Affiliation(s)
- Magda Babina
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Allergology, Hindenburgdamm 30, 12203 Berlin, Germany
- Correspondence:
| | - Kristin Franke
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Allergology, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Gürkan Bal
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Allergology, Hindenburgdamm 30, 12203 Berlin, Germany
| |
Collapse
|
19
|
Yoon S, Min Y, Park C, Kim D, Heo Y, Kim M, Son E, Ghosh M, Son YO, Hur CG. Innate Immune Response Analysis in Meniscus Xenotransplantation Using Normal and Triple Knockout Jeju Native Pigs. Int J Mol Sci 2022; 23:ijms231810416. [PMID: 36142330 PMCID: PMC9499368 DOI: 10.3390/ijms231810416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Although allogenic meniscus grafting can be immunologically safe, it causes immune rejection due to an imbalanced tissue supply between donor and recipient. Pigs are anatomically and physiologically similar to adult humans and are, therefore, considered to be advantageous xenotransplantation models. However, immune rejection caused by genetic difference damages the donor tissue and can sometimes cause sudden death. Immune rejection is caused by genes; porcine GGTA1, CMAH, and B4GLANT2 are the most common. In this study, we evaluated immune cells infiltrating the pig meniscus transplanted subcutaneously into BALB/c mice bred for three weeks. We compared the biocompatibility of normal Jeju native black pig (JNP) meniscus with that of triple knockout (TKO) JNP meniscus (α-gal epitope, N-glycolylneuraminic acid (Neu5Gc), and Sd (a) epitope knockout using CRISPR-Cas 9). Mast cells, eosinophils, neutrophils, and macrophages were found to have infiltrated the transplant boundary in the sham (without transplantation), normal (normal JNP), and test (TKO JNP) samples after immunohistochemical analysis. When compared to normal and sham groups, TKO was lower. Cytokine levels did not differ significantly between normal and test groups. Because chronic rejection can occur after meniscus transplantation associated with immune cell infiltration, we propose studies with multiple genetic editing to prevent immune rejection.
Collapse
Affiliation(s)
- Seungwon Yoon
- Cronex Co., Jeju-si 63078, Korea
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju-si 63243, Korea
| | - Yunhui Min
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju-si 63243, Korea
| | | | - Dahye Kim
- Division of Animal Genetics and Bioinformatics, The National Institute of Animal Science, RDA, Wanju 55465, Korea
| | - Yunji Heo
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si 63243, Korea
| | - Mangeun Kim
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju-si 63243, Korea
| | | | - Mrinmoy Ghosh
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si 63243, Korea
- Department of Biotechnology, School of Bio, Chemical and Processing Engineering (SBCE), Kalasalingam Academy of Research and Educational, Krishnankoil 626126, India
| | - Young-Ok Son
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju-si 63243, Korea
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si 63243, Korea
- Correspondence: (Y.-O.S.); (C.-G.H.); Tel.: +82-64-754-3331 (Y.-O.S.); +82-64-805-0033 (C.-G.H.)
| | - Chang-Gi Hur
- Cronex Co., Jeju-si 63078, Korea
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju-si 63243, Korea
- Correspondence: (Y.-O.S.); (C.-G.H.); Tel.: +82-64-754-3331 (Y.-O.S.); +82-64-805-0033 (C.-G.H.)
| |
Collapse
|
20
|
Roots of Lithospermum erythrorhizon Alleviated Ovalbumin-Induced Allergic Rhinitis and IgE-triggered Degranulation of RBL-2H3 Cells. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12126116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Lithospermum erythrorhizon (L. erythrorhizon) root is used in traditional medicine for its anti-inflammatory, antibacterial, and antioxidant properties. However, no studies have examined its impact on allergic rhinitis (AR). Here, we explored the protective effects of L. erythrorhizon in immunoglobulin E (IgE)-stimulated RBL-2H3 cells and in an ovalbumin (OVA)-induced AR mouse model. In the latter, we examined nasal mucosal inflammation, allergen-specific cytokine production, and histological changes to the nasal mucosa. In the mouse model, oral administration of an ethanol extract of L. erythrorhizon (LE) led to a marked reduction in rubbing and sneeze frequency, a significant decrease in serum OVA-specific IgE and IgG1 levels, and a significant increase in the IgG2a/IgG1 ratio. LE also reduced expression of interleukin (IL)-4, IL-5, and IL-13 in nasal lavage fluid (NALF), and suppressed inflammatory cell infiltration and epithelial degradation in nasal tissues. In IgE-stimulated RBL-2H3 cells, LE suppressed release of degranulation markers such as β-hexosaminidase and histamine. Based on these findings, we suggest that LE may ameliorate OVA-induced AR by regulating mast cell-mediated inflammatory responses.
Collapse
|
21
|
Zhou L, Huang Y, Han Z, Wang J, Sun N, Zhang R, Dong W, Deng C, Zhuang G. Effects of rosmarinic acid on the inflammatory response in allergic rhinitis rat models after PM2.5 exposure. J Clin Lab Anal 2022; 36:e24316. [PMID: 35285093 PMCID: PMC8993598 DOI: 10.1002/jcla.24316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 02/07/2022] [Accepted: 02/16/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Studies have shown the promising prospects of rosmarinic acid (RosA) for the prevention and treatment of allergic diseases. OBJECTIVE The aim of this study was to investigate the effects of RosA on inflammatory reaction in rat models of allergic rhinitis (AR) after PM2.5 exposure. METHODS Allergic rhinitis rat models were established by ovalbumin sensitization, and PM2.5 was applied at a concentration of 1000 μg/m3 , 3 h a day for 30 consecutive days. RosA was administered via intraperitoneal injection (20 mg/kg/d) for seven consecutive days. Allergic nasal symptoms were recorded. The expressions of interleukin (IL)-4, IL-13, interferon (INF)-γ, and OVA-sIgE were determined by ELISA. Histopathological changes in nasal mucosa were observed by HE staining. mRNA expressions of T-bet and GATA-3 in nasal mucosa were detected by RT-PCR. NF-κBp65 in cell nuclei and IκBα in cytoplasm were analyzed by Western blot. RESULTS PM2.5 exposure worsened allergic nasal symptoms in AR rats, while RosA ameliorated these symptoms. Histopathologically, AR rats exhibited disorganized nasal mucosal epithelium, cell exfoliation, eosinophilic infiltration of lamina propria, gland swelling, and submucosal vascular congestion, which were aggravated by PM2.5 exposure and alleviated by RosA. RosA decreased the expressions of IL-4, IL-13, and increased the level of IFN-γ in PM2.5-exposed AR rats. After RosA intervention, the expressions of GATA-3 mRNA and NF-κBp65 in PM2.5-exposed AR rats were significantly reduced, while those of T-bet mRNA and IκBα were markedly increased. CONCLUSION Rosmarinic acid may alleviate symptoms of AR rat models exposed to PM2.5 through the modulation of the NF-κB pathway and Th1/Th2 balance.
Collapse
Affiliation(s)
- Lingling Zhou
- Department of OtolaryngologyHuadong Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Yu Huang
- Department of OtolaryngologyHuadong Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Zhijin Han
- Department of OtolaryngologyHuadong Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Jinchao Wang
- Department of OtolaryngologyHuadong Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Na Sun
- Department of OtolaryngologyHuadong Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Ruxin Zhang
- Department of OtolaryngologyHuadong Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Weiyang Dong
- Department of Environmental Science and Engineering, Center for Atmospheric Chemistry StudyFudan UniversityShanghaiChina
| | - Congrui Deng
- Department of Environmental Science and Engineering, Center for Atmospheric Chemistry StudyFudan UniversityShanghaiChina
| | - Guoshun Zhuang
- Department of Environmental Science and Engineering, Center for Atmospheric Chemistry StudyFudan UniversityShanghaiChina
| |
Collapse
|
22
|
Apolipoprotein (a)/Lipoprotein(a)-Induced Oxidative-Inflammatory α7-nAChR/p38 MAPK/IL-6/RhoA-GTP Signaling Axis and M1 Macrophage Polarization Modulate Inflammation-Associated Development of Coronary Artery Spasm. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9964689. [PMID: 35096275 PMCID: PMC8793348 DOI: 10.1155/2022/9964689] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 09/21/2021] [Accepted: 11/23/2021] [Indexed: 12/14/2022]
Abstract
Objective. Apolipoprotein (a)/lipoprotein(a) (Lp(a)), a major carrier of oxidized phospholipids, and α7-nicotinic acetylcholine receptor (α7-nAChR) may play an important role in the development of coronary artery spasm (CAS). In CAS, the association between Lp(a) and the α7-nAChR-modulated inflammatory macrophage polarization and activation and smooth muscle cell dysfunction remains unknown. Methods. We investigated the relevance of Lp(a)/α7-nAChR signaling in patient monocyte-derived macrophages and human coronary artery smooth muscle cells (HCASMCs) using expression profile correlation analyses, fluorescence-assisted cell sorting flow cytometry, immunoblotting, quantitative real-time polymerase chain reaction, and clinicopathological analyses. Results. There are increased serum Lp(a) levels (3.98-fold,
) and macrophage population (3.30-fold,
) in patients with CAS compared with patients without CAS. Serum Lp(a) level was positively correlated with high-sensitivity C-reactive protein (
,
), IL-6 (
,
), and α7-nAChR (
,
) in patients with CAS, but not in patients without CAS. Compared with untreated or low-density lipoprotein- (LDL-) treated macrophages, Lp(a)-treated macrophages exhibited markedly enhanced α7-nAChR mRNA expression (
) and activity (
), in vitro and ex vivo. Lp(a) but not LDL preferentially induced CD80+ macrophage (M1) polarization and reduced the inducible nitric oxide synthase expression and the subsequent NO production. While shRNA-mediated loss of α7-nAChR function reduced the Lp(a)-induced CD80+ macrophage pool, both shRNA and anti-IL-6 receptor tocilizumab suppressed Lp(a)-upregulated α7-nAChR, p-p38 MAPK, IL-6, and RhoA-GTP protein expression levels in cultures of patient monocyte-derived macrophages and HCASMCs. Conclusions. Elevated Lp(a) levels upregulate α7-nAChR/IL-6/p38 MAPK signaling in macrophages of CAS patients and HCASMC, suggesting that Lp(a)-triggered inflammation mediates CAS through α7-nAChR/p38 MAPK/IL-6/RhoA-GTP signaling induction, macrophage M1 polarization, and HCASMC activation.
Collapse
|
23
|
Cil M, Leblebisatan G, Leblebisatan S, Barutcu A, Cil MK, Kilinc Y. Deep Vein Thrombosis After a Wild Bee Sting. J Pediatr Hematol Oncol 2022; 44:e241-e242. [PMID: 33512868 DOI: 10.1097/mph.0000000000002072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/14/2020] [Indexed: 11/26/2022]
Abstract
Allergic reactions from insect bites are mostly observed with bee stings. Bee sting reactions can be classified into 3 main headings: local, systemic, and rare reactions. Vascular thrombosis is considered both in rare and systemic reactions. The wild bee venom induces the secretion of many inflammatory mediators, including histamine, phospholipase A1, and thromboxane, leading to vasoconstriction and thrombosis. Inflammatory cytokines also cause endothelial injury and deterioration of the microcirculation. In the literature, rare reactions have been reported including various central and arterial vascular pathologies such as aortic thrombosis, cerebral infarction, and myocardial infarction; however, there is rare publication concerning peripheral deep vein thrombosis (DVT). Although DVT produces good results with effective and rapid treatment, it can be fatal because of causes such as pulmonary embolism in the absence of timely intervention. Herein, for the first time in the literature, we present a pediatric case of peripheral DVT after a wild bee sting.
Collapse
Affiliation(s)
| | | | - Serife Leblebisatan
- Department of Radiology, Adana City Training and Research Hospital, Adana, Turkey
| | | | - Merve K Cil
- Pediatric Infectious Disease, Cukurova University Medical Faculty
| | | |
Collapse
|
24
|
Oh JS, Lee SJ, Choung SY. Lithospermum erythrorhizon Alleviates Atopic Dermatitis-like Skin Lesions by Restoring Immune Balance and Skin Barrier Function in 2.4-Dinitrochlorobenzene-Induced NC/Nga Mice. Nutrients 2021; 13:nu13093209. [PMID: 34579088 PMCID: PMC8470668 DOI: 10.3390/nu13093209] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/08/2023] Open
Abstract
The incidence of atopic dermatitis (AD), a disease characterized by an abnormal immune balance and skin barrier function, has increased rapidly in developed countries. This study investigated the anti-atopic effect of Lithospermum erythrorhizon (LE) using NC/Nga mice induced by 2,4-dinitrochlorobenzene. LE reduced AD clinical symptoms, including inflammatory cell infiltration, epidermal thickness, ear thickness, and scratching behavior, in the mice. Additionally, LE reduced serum IgE and histamine levels, and restored the T helper (Th) 1/Th2 immune balance through regulation of the IgG1/IgG2a ratio. LE also reduced the levels of AD-related cytokines and chemokines, including interleukin (IL)-1β, IL-4, IL-6, tumor necrosis factor-α (TNF-α), thymic stromal lymphopoietin, thymus and activation-regulated chemokine, macrophage-derived chemokine, regulated on activation, normal T cell expressed and secreted, and monocyte chemoattractant protein-1 in the serum. Moreover, LE modulated AD-related cytokines and chemokines expressed and secreted by Th1, Th2, Th17, and Th22 cells in the dorsal skin and splenocytes. Furthermore, LE restored skin barrier function by increasing pro-filaggrin gene expression and levels of skin barrier-related proteins filaggrin, involucrin, loricrin, occludin, and zonula occludens-1. These results suggest that LE is a potential therapeutic agent that can alleviate AD by modulating Th1/Th2 immune balance and restoring skin barrier function.
Collapse
Affiliation(s)
- Jin-Su Oh
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
| | | | - Se-Young Choung
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
- Department of Preventive Pharmacy and Toxicology, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
- Correspondence:
| |
Collapse
|
25
|
Chioncel V, Andrei CL, Brezeanu R, Sinescu C, Avram A, Tatu AL. Some Perspectives on Hypersensitivity to Coronary Stents. Int J Gen Med 2021; 14:4327-4336. [PMID: 34408475 PMCID: PMC8364397 DOI: 10.2147/ijgm.s326679] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 07/22/2021] [Indexed: 11/23/2022] Open
Abstract
The development of coronary stents has represented a revolution in the treatment of coronary heart disease. Beyond their many advantages, stents also have their limitations and complications. Allergic reactions to coronary stents are more common than acknowledged. These stented patients are exposed to foreign substances inserted in direct contact with the coronary intima. Hypersensitivity to stent components and drugs prescribed after stent insertion together with any environmental exposure seem to contribute to these adverse reactions. Patients can present to the hospital with a wide range of symptoms and multiple complications, the most important ones being instent restenosis and stent thrombosis. Although not very common (and not always easy to identify), allergic reactions after coronary or peripheral stents should be taken into account. Careful selection of patients (for elective stent implantation) depending on the propensity to allergies, although hard to achieve, represents a key factor in reducing the number of these complications.
Collapse
Affiliation(s)
- Valentin Chioncel
- Department of Cardio-Thoracic Pathology, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, 050474, Romania
| | - Catalina Liliana Andrei
- Department of Cardio-Thoracic Pathology, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, 050474, Romania
| | - Radu Brezeanu
- Department of Cardio-Thoracic Pathology, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, 050474, Romania
| | - Crina Sinescu
- Department of Cardio-Thoracic Pathology, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, 050474, Romania
| | - Anamaria Avram
- Department of Cardio-Thoracic Pathology, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, 050474, Romania
| | - Alin Laurentiu Tatu
- Medical and Pharmaceutical Research Unit/Competitive, Interdisciplinary Research Integrated Platform "Dunărea de Jos", ReForm-UDJG, Research Centre in the Field of Medical and Pharmaceutical Sciences, Faculty of Medicine and Pharmacy, Clinical Medical Department, "Dunărea de Jos" University of Galati, Galati, 800010, Romania
| |
Collapse
|
26
|
Wang Z, Franke K, Zuberbier T, Babina M. Cytokine Stimulation via MRGPRX2 Occurs with Lower Potency than by FcεRI-aggregation but with Similar Dependence on the ERK1/2 Module in Human Skin Mast Cells. J Invest Dermatol 2021; 142:414-424.e8. [PMID: 34329659 DOI: 10.1016/j.jid.2021.07.153] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 07/04/2021] [Accepted: 07/06/2021] [Indexed: 12/17/2022]
Abstract
Skin mast cells (MCs) contribute to chronic dermatoses that partially rely on MC-derived cytokines. The discovery of MRGPRX2 explains MC-dependent symptoms independently of FcεRI-activation. Here, we investigated whether MRGPRX2 can elicit cytokines, determined its relative potency versus FcεRI and addressed the underlying mechanisms. MRGPRX2-activation by compound 48/80 or Substance P on skin MCs induced TNF-α, IL-8, IL-13, CCL1, CCL2 mRNA and protein, yet induction was typically reduced compared with FcεRI-crosslinking. Generally, cytokine secretion required de-novo-synthesis with maximum accumulation at ≈8 h. Addressing key kinases revealed robust, rapid (1 min), and lasting (30 min) phosphorylation of ERK1/2 following MRGPRX2-ligation, while pp38, and pAKT signals were weaker, and pJNK hardly detectable. The kinase spectrum following FcεRI-aggregation was comparable, but responses considerably delayed. The MEK/ERK pathway was essential for all cytokines examined and four inhibitors of this module gave complete suppression. Variable and weaker contribution was found for PI3K>JNK>p38. Strikingly, cytokine profiles and signaling prerequisites were similar for MRGPRX2 and FcεRI and likely mainly dictated by the MC subset. Collectively, in skin MCs, the physiological producers of MRGPRX2, agonist binding elicits cytokines, yet less efficiently than FcεRI-aggregation. MRGPRX2-associated inflammation may thus be less tissue-destructive than responses to allergic challenge.
Collapse
Affiliation(s)
- Zhao Wang
- Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Dermatology, The Second Affiliated Hospital, Northwest Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Kristin Franke
- Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Torsten Zuberbier
- Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Magda Babina
- Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
27
|
Rahim NA, Jantan I, Said MM, Jalil J, Abd Razak AF, Husain K. Anti-Allergic Rhinitis Effects of Medicinal Plants and Their Bioactive Metabolites via Suppression of the Immune System: A Mechanistic Review. Front Pharmacol 2021; 12:660083. [PMID: 33927634 PMCID: PMC8076953 DOI: 10.3389/fphar.2021.660083] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/10/2021] [Indexed: 12/30/2022] Open
Abstract
Allergic rhinitis (AR) is a common inflammatory condition of the nasal mucosa and it is an immunoglobulin E-mediated disease. The incidence and prevalence of AR globally have been escalating over recent years. Antihistamines, intranasal corticosteroids, decongestants, intranasal anticholinergics, intranasal cromolyn, leukotriene receptor antagonists and immunotherapy have been used in the treatment of AR. However, there is a need to search for more effective and safer remedies as many of the current treatments have reported side effects. Medicinal plants have been used traditionally to relief symptoms of AR but their efficacy and safety have not been scientifically proven. In this review, up-to-date reports of studies on the anti-allergic rhinitis of several medicinal plants and their bioactive metabolites through suppression of the immune system are compiled and critically analyzed. The plant samples were reported to suppress the productions of immunoglobulin E, cytokines and eosinophils and inhibit histamine release. The suppression of cytokines production was found to be the main mechanistic effect of the plants to give symptomatic relief. The prospect of these medicinal plants as sources of lead molecules for development of therapeutic agents to treat AR is highlighted. Several bioactive metabolites of the plants including shikonin, okicamelliaside, warifteine, methylwarifteine, luteolin-7-O-rutinoside, tussilagone, petasin, and mangiferin have been identified as potential candidates for development into anti-allergic rhinitis agents. The data collection was mainly from English language articles published in journals, or studies from EBSCOHOST, Medline and Ovid, Scopus, Springer, and Google Scholar databases from the year 1985-2020. The terms or keywords used to find relevant studies were allergic rhinitis OR pollinosis OR hay fever, AND medicinal plant OR single plant OR single herb OR phytotherapy. This comprehensive review serves as a useful resource for medicinal plants with anti-allergic rhinitis potential, understanding the underlying mechanisms of action and for future exploration to find natural product candidates in the development of novel anti-allergic rhinitis agents.
Collapse
Affiliation(s)
- Nur Amira Rahim
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Ibrahim Jantan
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Malaysia
| | - Mazlina Mohd Said
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Juriyati Jalil
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Amirul Faiz Abd Razak
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Khairana Husain
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
28
|
Almeida TFA, Oliveira SR, de Noronha MS, Moreno A, Mesquita RA, Abreu LG, da Silva TA. Type IV hypersensitivity associated with restorative materials: Clinical report and systematic literature review. J Prosthet Dent 2021; 128:1201-1210. [PMID: 33820631 DOI: 10.1016/j.prosdent.2021.01.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 10/21/2022]
Abstract
STATEMENT OF PROBLEM Type IV hypersensitivity reactions (Type IV HR) are immune responses mediated by antigen-specific effector T cells. PURPOSE The purpose of this clinical report and systematic review was to report the clinicopathological features of Type IV HR in the oral mucosa and to present a systematic literature review of case reports and case series of individuals with Type IV HR in the oral mucosa related to contact with dental materials. MATERIAL AND METHODS The presented clinical lesions were melanotic macules with burning that affected the internal labial mucosa in contact with composite resin veneer crowns. Histopathological and immunohistochemical analysis of the lesion was performed. The systematic literature review was performed based on a search in 4 electronic databases (PubMed/MEDLINE, Scopus, Web of Science, and Ovid). RESULTS Immunohistochemistry showed positivity for CD4, CD8, CD20, CD3, tryptase, and CD117. After conservative treatment, the patient reported improvement of symptoms, and a decrease in the number of inflammatory cells was verified. Twenty-one articles were included in the review. Unlike the present patient, the authors of all the articles recommended radical treatment with the removal of the dental material. CONCLUSIONS Type IV HR in oral mucosa is rare, and the assessment of clinical and histopathological characteristics is essential to perform an accurate diagnosis and provide appropriate treatment.
Collapse
Affiliation(s)
- Tatiana F A Almeida
- Postdoctoral research, Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Sicília R Oliveira
- PhD student, Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Mariana S de Noronha
- PhD student, Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Amália Moreno
- Professor, Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Ricardo A Mesquita
- Professor, Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Lucas G Abreu
- Professor, Department of Child's and Adolescent's Oral Health, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Tarcília A da Silva
- Professor, Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.
| |
Collapse
|
29
|
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
|
30
|
Zou J, Yang Y, Fu Q, Liu H, Zhang C, Liu L, Wang Y, Li Y. Eosinophils Are More Strongly Relevant to Allergic Sensitization Than Basophils in Pediatric Adenotonsillar Hypertrophy. Front Pediatr 2021; 9:598063. [PMID: 33869109 PMCID: PMC8044536 DOI: 10.3389/fped.2021.598063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 03/10/2021] [Indexed: 11/23/2022] Open
Abstract
The relationship between eosinophils/basophils and allergic sensitization is not clear in pediatric adenotonsillar hypertrophy (ATH). The objective of this study is to investigate the relationship between eosinophil/basophil counts and peripheral specific IgE levels, and identify the common allergens in children with ATH. We initially screened 1,031 consecutive children who underwent adenotonsillectomy in our department from June 2018 to June 2019, and finally included 676 children. The eosinophil count, basophil count, and levels of specific IgE were collected. Correlations between two quantitative variables were assessed using the Pearson or Spearman coefficient. Logistic regression analyses were performed to evaluate the odds ratios (ORs) for atopy after controlling for age, sex, vitamin D, BMI, and visiting season. Both the eosinophil and basophil counts in atopic participants were significantly higher compared to non-atopic participants. The eosinophil count correlated with the levels of IgE specific to all allergens, and eosinophilia was independently associated with all tested atopy allergens other than atopy to dander after multivariate adjustment. Additionally, the basophil count correlated with the IgE levels specific to A. alternate and food mix, and basophilia was still significantly associated with atopy to food mix after multivariable adjustment. Furthermore, among allergic participants, D. farinae was the most prevalent allergen, followed by food mix, D. pteronyssinus, and A. alternata. In conclusion, eosinophils were more relevant to allergic sensitization than basophils, with eosinophils being significantly associated with all tested atopy allergens apart from dander, and basophils being associated with atopy to food mix. Furthermore, D. farinae was the most prevalent allergen and may be indicative of desensitization therapy.
Collapse
Affiliation(s)
- Juanjuan Zou
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan, China
| | - Yan Yang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan, China
| | - Qiang Fu
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan, China
| | - Huayang Liu
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan, China
| | - Chao Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Lili Liu
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Yan Wang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan, China
| | - Yanzhong Li
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan, China
| |
Collapse
|
31
|
Abstract
The eosinophil is an enigmatic cell with a continuing ability to fascinate. A considerable history of research endeavor on eosinophil biology stretches from the present time back to the nineteenth century. Perhaps one of the most fascinating aspects of the eosinophil is how accumulating knowledge has changed the perception of its function from passive bystander, modulator of inflammation, to potent effector cell loaded with histotoxic substances through to more recent recognition that it can act as both a positive and negative regulator of complex events in both innate and adaptive immunity. This book consists of chapters written by experts in the field of eosinophil biology that provide comprehensive clearly written protocols for techniques designed to underpin research into the function of the eosinophil in health and disease.
Collapse
Affiliation(s)
- Paige Lacy
- Alberta Respiratory Centre (ARC) Research, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Helene F Rosenberg
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Garry M Walsh
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
32
|
Kim BH, Lee S. Sophoricoside from Sophora japonica ameliorates allergic asthma by preventing mast cell activation and CD4 + T cell differentiation in ovalbumin-induced mice. Biomed Pharmacother 2021; 133:111029. [PMID: 33254020 DOI: 10.1016/j.biopha.2020.111029] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/04/2020] [Accepted: 11/15/2020] [Indexed: 12/24/2022] Open
Abstract
Asthma is a chronic inflammatory lung disorder with continuously increasing prevalence worldwide. Novel strategies are needed to prevent or improve asthma. The aim of this study was to investigate the effects of sophoricoside from Sophora japonica on allergic asthma. The mature seeds of S. japonica contain a large amount of sophoricoside. Sophoricoside reduced allergic and asthmatic symptoms by suppressing airway inflammation and antibody-antigen reaction in mouse models. In particular, sophoricoside suppressed immune cell recruitment into the airway lumens of the lungs and production of pro-inflammatory cytokines in the bronchoalveolar lavage fluid (BALF) of ovalbumin (OVA)-induced mice. It also decreased the amounts of histamine and arachidonic acid metabolites released in OVA-induced mice and antibody-antigen stimulated mast cells. In addition, sophoricoside decreased differentiation of naïve CD4+ T cells into T helper type 1 (Th1), Th2, and Th17 cells. Overall, we demonstrated that sophoricoside improved allergic asthma by suppressing mast cell activation and CD4+ T cell differentiation.
Collapse
Affiliation(s)
- Byung-Hak Kim
- Korea Institute of Science and Technology for Eastern Medicine (KISTEM), NEUMED Inc., Seoul 02440, Republic of Korea.
| | - Sanghyun Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea.
| |
Collapse
|
33
|
Lee D, Park YH, Lee JE, Kim HS, Min KY, Jo MG, Kim HS, Choi WS, Kim YM. Dasatinib Inhibits Lyn and Fyn Src-Family Kinases in Mast Cells to Suppress Type I Hypersensitivity in Mice. Biomol Ther (Seoul) 2020; 28:456-464. [PMID: 32268657 PMCID: PMC7457176 DOI: 10.4062/biomolther.2020.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 12/21/2022] Open
Abstract
Mast cells (MCs) are systemically distributed and secrete several allergic mediators such as histamine and leukotrienes to cause type I hypersensitivity. Dasatinib is a type of anti-cancer agent and it has also been reported to inhibit human basophils. However, dasatinib has not been reported for its inhibitory effects on MCs or type I hypersensitivity in mice. In this study, we examined the inhibitory effect of dasatinib on MCs and MC-mediated allergic response in vitro and in vivo. in vitro, dasatinib inhibited the degranulation of MCs by antigen stimulation in a dose-dependent manner (IC50, ~34 nM for RBL-2H3 cells; ~52 nM for BMMCs) without any cytotoxicity. It also suppressed the secretion of inflammatory cytokines IL-4 and TNF-α by antigen stimulation. Furthermore, dasatinib inhibited MC-mediated passive cutaneous anaphylaxis (PCA) in mice (ED50, ~29 mg/kg). Notably, dasatinib significantly suppressed the degranulation of MCs in the ear tissue. As the mechanism of its effect, dasatinib inhibited the activation of Syk and Syk-mediated downstream signaling proteins, LAT, PLCγ1, and three typical MAP kinases (Erk1/2, JNK, and p38), which are essential for the activation of MCs. Interestingly, in vitro tyrosine kinase assay, dasatinib directly inhibited the activities of Lyn and Fyn, the upstream tyrosine kinases of Syk in MCs. Taken together, dasatinib suppresses MCs and PCA in vitro and in vivo through the inhibition of Lyn and Fyn Src-family kinases. Therefore, we suggest the possibility of repositioning the anti-cancer drug dasatinib as a treatment for various MC-mediated type I hypersensitive diseases.
Collapse
Affiliation(s)
- Dajeong Lee
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Young Hwan Park
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Ji Eon Lee
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
- College of Pharmacy, Duksung Women’s University, Seoul 01369, Republic of Korea
| | - Hyuk Soon Kim
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Keun Young Min
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Min Geun Jo
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Hyung Sik Kim
- Division of Toxicology, College of Pharmacy, Sungkyunkwan University, Suwon 6419, Republic of Korea
| | - Wahn Soo Choi
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Young Mi Kim
- College of Pharmacy, Duksung Women’s University, Seoul 01369, Republic of Korea
| |
Collapse
|
34
|
Tavares LP, Peh HY, Tan WSD, Pahima H, Maffia P, Tiligada E, Levi-Schaffer F. Granulocyte-targeted therapies for airway diseases. Pharmacol Res 2020; 157:104881. [PMID: 32380052 PMCID: PMC7198161 DOI: 10.1016/j.phrs.2020.104881] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 12/24/2022]
Abstract
The average respiration rate for an adult is 12-20 breaths per minute, which constantly exposes the lungs to allergens and harmful particles. As a result, respiratory diseases, which includes asthma, chronic obstructive pulmonary disease (COPD) and acute lower respiratory tract infections (LTRI), are a major cause of death worldwide. Although asthma, COPD and LTRI are distinctly different diseases with separate mechanisms of disease progression, they do share a common feature - airway inflammation with intense recruitment and activation of granulocytes and mast cells. Neutrophils, eosinophils, basophils, and mast cells are crucial players in host defense against pathogens and maintenance of lung homeostasis. Upon contact with harmful particles, part of the pulmonary defense mechanism is to recruit these cells into the airways. Despite their protective nature, overactivation or accumulation of granulocytes and mast cells in the lungs results in unwanted chronic airway inflammation and damage. As such, understanding the bright and the dark side of these leukocytes in lung physiology paves the way for the development of therapies targeting this important mechanism of disease. Here we discuss the role of granulocytes in respiratory diseases and summarize therapeutic strategies focused on granulocyte recruitment and activation in the lungs.
Collapse
Affiliation(s)
- Luciana P Tavares
- ImmuPhar - Immunopharmacology Section Committee of International Union of Basic and Clinical Pharmacology (IUPHAR); Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Hong Yong Peh
- ImmuPhar - Immunopharmacology Section Committee of International Union of Basic and Clinical Pharmacology (IUPHAR); Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, 16 Medical Drive, 117600, Singapore
| | - Wan Shun Daniel Tan
- ImmuPhar - Immunopharmacology Section Committee of International Union of Basic and Clinical Pharmacology (IUPHAR); Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, 16 Medical Drive, 117600, Singapore
| | - Hadas Pahima
- ImmuPhar - Immunopharmacology Section Committee of International Union of Basic and Clinical Pharmacology (IUPHAR); Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Pasquale Maffia
- ImmuPhar - Immunopharmacology Section Committee of International Union of Basic and Clinical Pharmacology (IUPHAR); Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Ekaterini Tiligada
- ImmuPhar - Immunopharmacology Section Committee of International Union of Basic and Clinical Pharmacology (IUPHAR); Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Francesca Levi-Schaffer
- ImmuPhar - Immunopharmacology Section Committee of International Union of Basic and Clinical Pharmacology (IUPHAR); Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
35
|
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: 4.4] [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]
|
36
|
Mangiferin Alleviates Ovalbumin-Induced Allergic Rhinitis via Nrf2/HO-1/NF-κB Signaling Pathways. Int J Mol Sci 2020; 21:ijms21103415. [PMID: 32408566 PMCID: PMC7279452 DOI: 10.3390/ijms21103415] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/06/2020] [Accepted: 05/11/2020] [Indexed: 12/26/2022] Open
Abstract
Mangiferin (MF), extracted from mango trees, is considered to have anti-inflammatory, anti-apoptotic, and antioxidant effects. However, its effects on allergic rhinitis (AR), remain unclear. We investigated the mechanisms underlying the protective action of MF in ovalbumin (OVA)-induced AR models. AR was induced by OVA challenge in BALB/c mice. Prior to this, MF and dexamethasone were administered. Mice were examined for nasal mucosal inflammation, the generation of allergen-specific cytokine response, and histopathological changes in the nasal mucosa and lung tissue. MF ameliorated nasal symptoms and nasal mucosa inflammation in OVA-induced AR and reduced inflammatory cell infiltration and epithelial disruption in these tissues. MF inhibited the overproduction of Th2/Th17 cytokines and transcription factors. MF downregulated the HO-1/Nrf2 pathways, reduced oxidative stress biomarker levels, and the NF-κB signaling pathways were inhibited. MF exerts protective effects in AR by inhibiting NF-κB and activating HO-1/Nrf2 pathways. MF could be used for the treatment of AR.
Collapse
|
37
|
Vitallé J, Terrén I, Orrantia A, Bilbao A, Gamboa PM, Borrego F, Zenarruzabeitia O. The Expression and Function of CD300 Molecules in the Main Players of Allergic Responses: Mast Cells, Basophils and Eosinophils. Int J Mol Sci 2020; 21:ijms21093173. [PMID: 32365988 PMCID: PMC7247439 DOI: 10.3390/ijms21093173] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 04/17/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023] Open
Abstract
Allergy is the host immune response against non-infectious substances called allergens. The prevalence of allergic diseases is increasing worldwide. However, while some drugs counteract the symptomatology caused by allergic reactions, no completely effective treatments for allergic diseases have been developed yet. In this sense, the ability of surface activating and inhibitory receptors to modulate the function of the main effector cells of allergic responses makes these molecules potential pharmacological targets. The CD300 receptor family consists of members with activating and inhibitory capabilities mainly expressed on the surface of immune cells. Multiple studies in the last few years have highlighted the importance of CD300 molecules in several pathological conditions. This review summarizes the literature on CD300 receptor expression, regulation and function in mast cells, basophils and eosinophils, the main players of allergic responses. Moreover, we review the involvement of CD300 receptors in the pathogenesis of certain allergic diseases, as well as their prospective use as therapeutic targets for the treatment of IgE-dependent allergic responses.
Collapse
Affiliation(s)
- Joana Vitallé
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (J.V.); (I.T.); (A.O.); (A.B.); (P.M.G.); (F.B.)
| | - Iñigo Terrén
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (J.V.); (I.T.); (A.O.); (A.B.); (P.M.G.); (F.B.)
| | - Ane Orrantia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (J.V.); (I.T.); (A.O.); (A.B.); (P.M.G.); (F.B.)
| | - Agurtzane Bilbao
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (J.V.); (I.T.); (A.O.); (A.B.); (P.M.G.); (F.B.)
- Pediatrics Service, Cruces University Hospital, 48903 Barakaldo, Spain
| | - Pedro M. Gamboa
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (J.V.); (I.T.); (A.O.); (A.B.); (P.M.G.); (F.B.)
- Allergology Service, Cruces University Hospital, 48903 Barakaldo, Spain
| | - Francisco Borrego
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (J.V.); (I.T.); (A.O.); (A.B.); (P.M.G.); (F.B.)
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Olatz Zenarruzabeitia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (J.V.); (I.T.); (A.O.); (A.B.); (P.M.G.); (F.B.)
- Correspondence: ; Tel.: +34-699-227-735
| |
Collapse
|
38
|
Wang C, Li L, Jiang J, Li L, Li J, Xu C, Jin S, Zhu L, Yan G. Pterostilbene Inhibits FcεRI Signaling through Activation of the LKB1/AMPK Pathway in Allergic Response. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:3456-3465. [PMID: 32096633 DOI: 10.1021/acs.jafc.9b07126] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In this study, the role and mechanism of pterostilbene (Pts) in mast cell degranulation in vitro and in vivo were investigated. The results showed that Pts inhibited mast cell-mediated local passive allergic reactions in mice. In addition, treatment with Pts reduced both histamine release and calcium influx in rat peritoneal mast cells and RBL-2H3 cells and reduced IgE-mediated mast cell activation. Furthermore, the mechanism underlying Pts inhibition of mast cell signaling was probed via studying the effects of Pts on liver kinase B1 (LKB1), including the use of the LKB1 activator metformin and siRNA knockdown of LKB1. The data showed that Pts reduced the release of inflammatory mediators such as tumor necrosis factor-α, interleukin-6, leukotriene C4, and prostaglandin D2 in mast cells by activating the LKB1/adenosine monophosphate (AMP)-activated protein kinase (AMPK) signaling pathway. Furthermore, Pts inhibited phosphorylation of FcεRI and FcεRI-mediated degranulation in RBL-2H3 cells. These effects were attenuated after LKB1 knockdown. Taken together, Pts could inhibit FcεRI signaling through activation of the LKB1/AMPK signaling pathway in IgE-mediated mast cell activation. Thus, Pts might be an effective therapeutic agent for mast cell-mediated allergic diseases.
Collapse
Affiliation(s)
- Chongyang Wang
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji 133002, P. R. China
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
| | - Liangchang Li
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji 133002, P. R. China
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
| | - Jingzhi Jiang
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji 133002, P. R. China
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
| | - Li Li
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji 133002, P. R. China
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
| | - Junfeng Li
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji 133002, P. R. China
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
| | - Chang Xu
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji 133002, P. R. China
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
| | - Shan Jin
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
- Department of Dermatology, Yanbian University Hospital, Yanji 133002, P. R. China
| | - Lianhua Zhu
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
- Department of Dermatology, Yanbian University Hospital, Yanji 133002, P. R. China
| | - Guanghai Yan
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji 133002, P. R. China
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
| |
Collapse
|
39
|
宋 小, 张 俊. [Effect of sublingual immunotherapy on inflammatory factors and autophagy in patients with allergic rhinitis]. LIN CHUANG ER BI YAN HOU TOU JING WAI KE ZA ZHI = JOURNAL OF CLINICAL OTORHINOLARYNGOLOGY, HEAD, AND NECK SURGERY 2020; 34:230-234. [PMID: 32791589 PMCID: PMC10127856 DOI: 10.13201/j.issn.2096-7993.2020.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Indexed: 06/11/2023]
Abstract
Objective:To observe the effect of sublingual immunotherapy on the expression of serum inflammatory factors IL-4 and TNF-α as well as autophagy-associated protein LC3 in nasal mucosa in patients with allergic rhinitis(AR). Method:Forty patients with AR were randomly divided into SLIT group(n=20) and control group(n=20), the SLIT group received a 2-year intervention with a standardized dust mite vaccine SLIT in combination with conventional drugs, the control group received placebo and conventional drug treatment. Blood samples and inferior turbinate mucosa were collected of both groups before and after the treatment; the clinical symptoms, signs and medication scores of the two groups before and after treatment were analyzed; the expressions of serum IL-4 and TNF-α were detected by ELISA before and after treatment; the expression of autophagy-related protein LC3 was detected by Western blot. Result:There were no significant differences in the pre-treatment signs, symptoms, medication scores, age, gender, serum IL-4, TNF-α, and LC3 expression between the SLIT group and the control group(P>0.05). After a 2-year treatment, the symptom scores the of SLIT group were significantly improved compared with the control group; serum levels of IL-4 and TNF-α were significantly decreased in the SLIT group; the expression of autophagy-related protein LC3 in the SLIT group was significantly lower than that in the control group, and the difference was statistically significant(P<0.05). Conclusion:SLIT combined with conventional drug therapy is more effective in improving the symptoms of AR patients than conventional drug therapy. SLIT can reduce the inflammation level and expression of autophagy-related proteins in AR patients to a certain extent.
Collapse
Affiliation(s)
- 小云 宋
- 长沙市第一医院耳鼻咽喉科(长沙,410005)Department of Otolaryngology, the First Hospital of Changsha, Changsha, 410005, China
| | - 俊杰 张
- 长沙市第一医院耳鼻咽喉科(长沙,410005)Department of Otolaryngology, the First Hospital of Changsha, Changsha, 410005, China
| |
Collapse
|
40
|
Taki M, Oshima T, Li M, Sei H, Tozawa K, Tomita T, Fukui H, Watari J, Miwa H. Duodenal low-grade inflammation and expression of tight junction proteins in functional dyspepsia. Neurogastroenterol Motil 2019; 31:e13576. [PMID: 30790378 DOI: 10.1111/nmo.13576] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/29/2019] [Accepted: 02/05/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Duodenal changes in functional dyspepsia (FD) might be related to the development of symptoms. However, relationships among low-grade inflammation, Helicobacter pylori infection, and protein expression by tight junctions (TJs) in the duodenum are unclear. We therefore aimed to determine whether duodenal inflammation and genes associated with TJ proteins are associated with FD. METHODS We evaluated inflammatory cell infiltration of the duodenum, H pylori infection, and genes associated with TJ proteins in duodenal biopsy specimens from 35 patients with FD according to the Rome III diagnostic questionnaire and from 31 asymptomatic controls without structural diseases. We immunohistochemically detected eosinophils and mast cells and counted them. The expression of claudins, occludin, and zonula occludens (ZO)-1 mRNA was evaluated using quantitative RT-PCR. Infection with H pylori was determined by measuring serum antibodies, rapid urease or urea breath tests, and endoscopic findings. RESULTS Sex, age, and H pyloriinfection rates did not differ between patients with FD and controls. The numbers of eosinophils and mast cells were significantly increased in patients with FD compared with controls and were significantly correlated. Inflammatory cell counts in the duodenum were not associated with H pylori infection status. Claudin-3 mRNA expression was increased in the patients with FD. CONCLUSIONS Subtle inflammation identified in the duodenum of patients with FD might be associated with the onset and persistence of dyspeptic symptoms.
Collapse
Affiliation(s)
- Masato Taki
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Tadayuki Oshima
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Min Li
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hiroo Sei
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Katsuyuki Tozawa
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Toshihiko Tomita
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hirokazu Fukui
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Jiro Watari
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hiroto Miwa
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
41
|
Pahima H, Puzzovio PG, Levi-Schaffer F. 2B4 and CD48: A powerful couple of the immune system. Clin Immunol 2019; 204:64-68. [DOI: 10.1016/j.clim.2018.10.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/22/2018] [Accepted: 10/22/2018] [Indexed: 01/10/2023]
|
42
|
The ameliorative effect of AST2017-01 in an ovalbumin-induced allergic rhinitis animal model. Inflamm Res 2019; 68:387-395. [PMID: 30874868 DOI: 10.1007/s00011-019-01226-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/08/2019] [Accepted: 03/09/2019] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE AST2017-01 is developed to be used for treatment and prevention of allergic diseases and composed of processed-Cordyceps militaris and processed-Rumex crispus. But, effect of AST2017-01 remains unclear in an allergic rhinitis (AR). So, this study aimed to explore the effects of AST2017-01 in ovalbumin (OVA)-induced AR animal model. METHODS OVA-induced AR animals were orally administered AST2017-01 and chrysophanol, an active component of AST2017-01 for 10 days. RESULTS In mice with AR, AST2017-01 and chrysophanol markedly decreased number of rubs, IgE, histamine, thymic stromal lymphopoietin, tumor necrosis factor-α, interleukin (IL)-1β, IL-4, IL-5, and IL-13 in serum or nasal mucosa tissues. Moreover, activities and protein levels of caspase-1 were markedly diminished by oral administration of AST2017-01 and chrysophanol. Declines of macrophage inflammatory protein-2, intercellular adhesion molecules-1, eosinophil, and mast cells were also noted in nasal mucosa tissues of AST2017-01 and chrysophanol groups. CONCLUSIONS Taken together, these findings indicate that AST2017-01 has an anti-allergic effect as a therapeutic agent or functional food for treating and preventing AR.
Collapse
|
43
|
Anti-Inflammatory Effects of Cold Thermal Therapy on Allergic Skin Inflammation Induced by Trimellitic Anhydride in BALB/c Mice. Mediators Inflamm 2019; 2019:1936769. [PMID: 30833826 PMCID: PMC6369506 DOI: 10.1155/2019/1936769] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/14/2018] [Accepted: 12/06/2018] [Indexed: 02/04/2023] Open
Abstract
Cold and hot thermal therapies are widely used as a traditional therapy in many cultures and are often prescribed in the treatment of various musculoskeletal and neurological conditions which present themselves to primary care physicians. However, there are no reports that investigated either the effects of cold and hot thermal therapies on the skin inflammation of trimellitic anhydride- (TMA-) induced dermatitis-like contact hypersensitivity (CHS) mouse model, or the mechanism of thermal therapy on allergic skin inflammation. Therefore, in this study, to reveal the anti-inflammatory effect of thermal therapy and its mechanism on TMA-induced CHS, we analyzed ear-swelling response (ear edema), vascular permeability, serum IgE levels, histological examination, and histamine and Th2 cytokine levels. Cold thermal therapy reduced the ear-swelling response, the vascular permeability, the serum IgE levels, and the infiltration of eosinophils and mast cells as well as the mast cell degranulation. To determine the mechanism by which cold thermal therapy inhibits allergic skin inflammation, detailed studies were carried out revealing that cold thermal therapy suppressed IL-4 and IL-5 secretion and mast cell activation. These results indicated that cold thermal therapy cures skin inflammation of TMA-induced CHS by decreasing Th2 cytokine release, especially IL-4 and IL-5, and mast cell activation. These data suggest that new insight into the mechanism of robust therapeutic effects of cold thermal therapy against allergic dermatitis, and cold thermal therapy may prove to be a useful therapeutic modality on allergic inflammatory diseases as traditional use as well as Th2- or mast cell-mediated allergic responses.
Collapse
|
44
|
Lianto P, Ogutu FO, Zhang Y, He F, Che H. Inhibitory effects of quail egg on mast cells degranulation by suppressing PAR2-mediated MAPK and NF-kB activation. Food Nutr Res 2018; 62:1084. [PMID: 30083085 PMCID: PMC6060182 DOI: 10.29219/fnr.v62.1084] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 06/20/2018] [Accepted: 06/22/2018] [Indexed: 12/22/2022] Open
Abstract
Background Quail egg (QE) has been reported to possess an anti-allergic and anti-inflammatory activity. We have demonstrated that whole QE was able to attenuate the allergic symptoms in food allergy-induced EoE murine model, but whether QE albumen or QE yolk plays a more important role still remains unclear. Objective In this current study, we investigated the suppressive role of QE in mast cell degranulation and cytokine production of the effect phase response. Method A passive cutaneous anaphylaxis (PCA) mouse model was used to confirm the anti-allergic effect of QE. Besides, HMC-1 cell model was used to study its suppressive role in more detail. In this in vitro study, we divided QE into three groups: whole QE, QE albumen, and QE yolk. The effect of QE treatment on mast cell degranulation and intracellular calcium influx was investigated. Moreover, the effect of QE allergy- related mediators, genes, and proteins were also assessed by ELISA, RT-PCR, and western blotting. Results and discussion Our data showed that the extent of mast cell degranulation-mediated ear vascular permeability in IgE-mediated PCA mice treated with whole QE (17 mg/kg) was decreased significantly up to 43.31 ± 0.42% reduction. HMC-1 cell-based immunological assay in vitro indicated that QE, particularly its albumen, acted as a 'mast cell stabilizer'. Under the concentration of 70 μg/mL, QE albumen effectively suppressed the releases of β-hexosaminidase, histamine, and tryptase, as well as Th2 and pro-inflammatory cytokine production; reached 30 up to 50% reduction. Besides, QE albumen was also able to significantly modulate the upregulation of IL-10 up to 58.30 ± 5.9%. Interestingly, our data indicated that QE yolk still had a significant inhibitory effect on modulating Th2 cytokines in its highest concentration (100 μg/mL), while QE albumen showed no inhibitory effect. Western blot analysis showed QE albumen effectively down-regulated the expressions of calcium-related protein (TRPC1, Orai1, STIM1, PLC-γ and IP3R), facilitated the reduction of PAR-2 and induced the reduction of phosphorylation of JNK, IKKα, p50 and p65 protein expressions. Conclusion As confirmed by PCA and HMC-1 cell-based immunology assay, QE albumen and QE yolk may work together through exerting anti-allergy activity and can be used as a potential anti-allergic nutrient in the future.
Collapse
Affiliation(s)
- Priscilia Lianto
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P.R. China.,College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P.R. China; and
| | - Fredrick O Ogutu
- Food Technology Division of Kenya Industrial Research and Development Institute, South C - Popo Rd., Off Mombasa Rd., PO Box 30650-00100, Nairobi, Kenya
| | - Yani Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P.R. China; and
| | - Feng He
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P.R. China; and
| | - Huilian Che
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P.R. China.,College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P.R. China; and
| |
Collapse
|
45
|
Leptin stimulates tissue rat mast cell pro-inflammatory activity and migratory response. Inflamm Res 2018; 67:789-799. [PMID: 30019195 PMCID: PMC6096628 DOI: 10.1007/s00011-018-1171-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/22/2018] [Accepted: 07/13/2018] [Indexed: 12/11/2022] Open
Abstract
Objective The aim of this study was to determine whether leptin, a member of the adipocytokines involved in immune and inflammatory response regulation, may influence some aspects of mast cell biology. Materials and methods Experiments were done in vitro on fully mature tissue rat mast cells isolated from the peritoneal cavity, and leptin was used at concentrations 0.001–100 ng/ml. The effect of leptin on mast cell degranulation (histamine release assay), intracellular Ca2+ level (fluorimetry), pro-inflammatory mediator release (ELISA technique), surface receptor expression (flow cytometry and confocal microscopy), and migration (Boyden microchamber assay) was estimated. Results Leptin was found to stimulate mast cells to degranulation and histamine release. It induced the intracellular Ca2+ increase, as well. In response to leptin stimulation, mast cells generated and released cysLTs and chemokine CCL3. Leptin-induced upregulation of CYSLTR1 and CYSLTR2 surface expression was observed. Moreover, this adipocytokine stimulated mast cells to migratory response, even in the absence of extracellular matrix (ECM) proteins. Conclusions Our observations clearly documented that leptin promotes the pro-inflammatory activity of mast cells, and it thereby engages these cells in the inflammatory processes.
Collapse
|
46
|
Kang YM, Chung KS, Kook IH, Kook YB, Bae H, Lee M, An HJ. Inhibitory effects of bee venom on mast cell-mediated allergic inflammatory responses. Int J Mol Med 2018. [PMID: 29532852 DOI: 10.3892/ijmm.2018.3558] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Although bee venom (BV) is a toxin that causes bee stings to be painful, it has been widely used clinically for the treatment of certain immune‑associated diseases. BV has been used traditionally for the treatment of chronic inflammatory diseases. In this regard, the present study analyzed the effect of BV on the regulation of inflammatory mediator production by mast cells and their allergic inflammatory responses in an animal model. HMC‑1 cells were treated with BV prior to stimulation with phorbol‑12‑myristate 13‑acetate plus calcium ionophore A23187 (PMACI). The production of allergy‑associated pro‑inflammatory mediators was examined, and the underlying mechanisms were investigated. Furthermore, to investigate whether BV exhibits anti‑inflammatory effects associated with anti‑allergic effects in vivo, a compound 48/80‑induced anaphylaxis model was used. BV inhibited histamine release, mRNA expression and production of cytokines in the PMACI‑stimulated HMC‑1 cells. Furthermore, the inhibitory effects of BV on mitogen‑activated protein kinase (MAPK), MAPK kinase, signal transducer and activator of transcription 3 (STAT3) and Akt were demonstrated. The present study also investigated the ability of BV to inhibit compound 48/80‑induced systemic anaphylaxis in vivo. BV protected the mice against compound 48/80‑induced anaphylactic‑associated mortality. Furthermore, BV suppressed the mRNA expression levels of pro‑inflammatory cytokines, and suppressed the activation of MAPK and STAT3 in this model. These results provide novel insights into the possible role of BV as a modulator for mast cell‑mediated allergic inflammatory disorders.
Collapse
Affiliation(s)
- Yun-Mi Kang
- Department of Pharmacology, College of Korean Medicine, Sangji University, Wonju, Gangwon‑do 26339, Republic of Korea
| | - Kyung-Sook Chung
- Catholic Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - In-Hoon Kook
- Department of Pharmacology, College of Korean Medicine, Sangji University, Wonju, Gangwon‑do 26339, Republic of Korea
| | - Yoon-Bum Kook
- Department of Prescription, College of Korean Medicine, Sangji University, Wonju, Gangwon‑do 26339, Republic of Korea
| | - Hyunsu Bae
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Dongdaemoon‑Gu, Seoul 02447, Republic of Korea
| | - Minho Lee
- Catholic Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hyo-Jin An
- Department of Pharmacology, College of Korean Medicine, Sangji University, Wonju, Gangwon‑do 26339, Republic of Korea
| |
Collapse
|
47
|
Skullcapflavone II attenuates ovalbumin-induced allergic rhinitis through the blocking of Th2 cytokine production and mast cell histamine release. Int Immunopharmacol 2017; 52:77-84. [PMID: 28886581 DOI: 10.1016/j.intimp.2017.08.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/25/2017] [Accepted: 08/28/2017] [Indexed: 12/24/2022]
Abstract
Allergic rhinitis is a common heterogeneous chronic upper airway disorder and is an IgE-mediated inflammation characterized by one or more nasal symptoms such as sneezing, itching, nasal discharge, rhinorrhea, post nasal drainage and nasal blockage. In the present study, the effects of skullcapflavone II (SCFII) on upper airway inflammation, Th2 cytokines, and NF-κB signaling in an ovalbumin (OVA)-induced allergic rhinitis (AR) murine model in vivo were investigated. OVA-induced AR mice increased nasal symptoms, eosinophils and mast cells infiltration into nasal cavity, OVA-specific IgE/IgG1 and histamine in serum, Th2 cytokines including IL-13 and GATA3, and NF-κB signaling in NALF and lung homogenate. Interestingly, treatment of SCFII reduced the levels of OVA-specific IgE/IgG1 and histamine in serum, of Th2 cytokines and of NF-κB signaling in the NALF and the lung homogenate, and histopathological changes in the nasal tissue and the lung. Also, dexamethasone suppressed such increases. The results of this study suggested that SCFII may ameliorate allergic inflammation of upper airway in AR mice model by blocking the Th2 cytokine production, the NF-κB signal pathway and the mast cell histamine release. Taken together, we suggest that SCFII may be used as a therapeutic agent for patients with Th2-mediated or mast cell-mediated allergic diseases.
Collapse
|
48
|
Gangwar RS, Minai-Fleminger Y, Seaf M, Gutgold A, Shikotra A, Barber C, Chauhan A, Holgate S, Bradding P, Howarth P, Eliashar R, Berkman N, Levi-Schaffer F. CD48 on blood leukocytes and in serum of asthma patients varies with severity. Allergy 2017; 72:888-895. [PMID: 27859399 DOI: 10.1111/all.13082] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND CD48 is a membrane receptor (mCD48) on eosinophils and mast cells and exists in a soluble form (sCD48). CD48 has a pivotal role in murine asthma and in the proinflammatory interactions of mast cells with eosinophils via its ligand CD244. Thus, CD48 might be important in human asthma. METHODS Therefore, two separate cohorts (IL and UK) comprising mild, moderate, and severe asthma and healthy volunteers were evaluated for blood leukocyte mCD48 expression and sCD48 in serum. Asthmatic bronchial biopsies were immunostained for CD48. sCD48 effect on CD244-dependent eosinophil activation was evaluated. RESULTS Eosinophil mCD48 expression was significantly elevated in moderate while downregulated in severe asthma. mCD48 expression on B, T, and NK cells and monocytes in severe asthma was significantly increased. sCD48 levels were significantly higher in mild while reduced in severe asthma. sCD48 optimal cutoff values for differentiating asthma from health were identified as >1482 pg/ml (IL) and >1619 pg/ml (UK). In asthmatic bronchial biopsies, mCD48 was expressed predominantly by eosinophils. sCD48 inhibited anti-CD244-induced eosinophil activation. CONCLUSIONS mCD48 and sCD48 are differentially expressed in the peripheral blood of asthma patients of varying severity. sCD48 inhibits CD244-mediated eosinophil activation. These findings suggest that CD48 may play an important role in human asthma.
Collapse
Affiliation(s)
- R. S. Gangwar
- Pharmacology & Experimental Therapeutics Unit; Institute for Drug Research; School of Pharmacy; Faculty of Medicine; The Hebrew University of Jerusalem; Jerusalem Israel
| | - Y. Minai-Fleminger
- Pharmacology & Experimental Therapeutics Unit; Institute for Drug Research; School of Pharmacy; Faculty of Medicine; The Hebrew University of Jerusalem; Jerusalem Israel
| | - M. Seaf
- Pharmacology & Experimental Therapeutics Unit; Institute for Drug Research; School of Pharmacy; Faculty of Medicine; The Hebrew University of Jerusalem; Jerusalem Israel
| | - A. Gutgold
- Pharmacology & Experimental Therapeutics Unit; Institute for Drug Research; School of Pharmacy; Faculty of Medicine; The Hebrew University of Jerusalem; Jerusalem Israel
| | - A. Shikotra
- Department of Infection, Immunity and Inflammation; Institute for Lung Health; University of Leicester; Leicester UK
| | - C. Barber
- Clinical and Experimental Sciences; Faculty of Medicine; University of Southampton; Southampton UK
- NIHR Respiratory Biomedical Research Unit; Southampton General Hospital; Southampton UK
| | - A. Chauhan
- Portsmouth Hospitals NHS Trust; Portsmouth UK
| | - S. Holgate
- Clinical and Experimental Sciences; Faculty of Medicine; University of Southampton; Southampton UK
- NIHR Respiratory Biomedical Research Unit; Southampton General Hospital; Southampton UK
| | - P. Bradding
- Department of Infection, Immunity and Inflammation; Institute for Lung Health; University of Leicester; Leicester UK
| | - P. Howarth
- Clinical and Experimental Sciences; Faculty of Medicine; University of Southampton; Southampton UK
- NIHR Respiratory Biomedical Research Unit; Southampton General Hospital; Southampton UK
| | - R. Eliashar
- Department of Otolaryngology/Head and Neck Surgery; Hadassah Hebrew University Medical Center; Jerusalem Israel
| | - N. Berkman
- Institute of Pulmonary Medicine; Hadassah Hebrew University Medical Center; Jerusalem Israel
| | - F. Levi-Schaffer
- Pharmacology & Experimental Therapeutics Unit; Institute for Drug Research; School of Pharmacy; Faculty of Medicine; The Hebrew University of Jerusalem; Jerusalem Israel
| |
Collapse
|
49
|
Sibilano R, Gaudenzio N, DeGorter MK, Reber LL, Hernandez JD, Starkl PM, Zurek OW, Tsai M, Zahner S, Montgomery SB, Roers A, Kronenberg M, Yu M, Galli SJ. A TNFRSF14-FcɛRI-mast cell pathway contributes to development of multiple features of asthma pathology in mice. Nat Commun 2016; 7:13696. [PMID: 27982078 PMCID: PMC5171877 DOI: 10.1038/ncomms13696] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 10/26/2016] [Indexed: 01/07/2023] Open
Abstract
Asthma has multiple features, including airway hyperreactivity, inflammation and remodelling. The TNF superfamily member TNFSF14 (LIGHT), via interactions with the receptor TNFRSF14 (HVEM), can support TH2 cell generation and longevity and promote airway remodelling in mouse models of asthma, but the mechanisms by which TNFSF14 functions in this setting are incompletely understood. Here we find that mouse and human mast cells (MCs) express TNFRSF14 and that TNFSF14:TNFRSF14 interactions can enhance IgE-mediated MC signalling and mediator production. In mouse models of asthma, TNFRSF14 blockade with a neutralizing antibody administered after antigen sensitization, or genetic deletion of Tnfrsf14, diminishes plasma levels of antigen-specific IgG1 and IgE antibodies, airway hyperreactivity, airway inflammation and airway remodelling. Finally, by analysing two types of genetically MC-deficient mice after engrafting MCs that either do or do not express TNFRSF14, we show that TNFRSF14 expression on MCs significantly contributes to the development of multiple features of asthma pathology.
Collapse
Affiliation(s)
- Riccardo Sibilano
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Nicolas Gaudenzio
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Marianne K. DeGorter
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Laurent L. Reber
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Immunology, Unit of Antibodies in Therapy and Pathology, INSERM U1222, Institut Pasteur, Paris 75015, France
| | - Joseph D. Hernandez
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Philipp M. Starkl
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences and Department of Medicine I, Research Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Oliwia W. Zurek
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Mindy Tsai
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Sonja Zahner
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA
| | - Stephen B. Montgomery
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Axel Roers
- Institute for Immunology, Technische Universität Dresden, Dresden 01307, Germany
| | - Mitchell Kronenberg
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA
| | - Mang Yu
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Stephen J. Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Microbiology and Immunology and Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
50
|
Gangwar RS, Landolina N, Arpinati L, Levi-Schaffer F. Mast cell and eosinophil surface receptors as targets for anti-allergic therapy. Pharmacol Ther 2016; 170:37-63. [PMID: 27773785 DOI: 10.1016/j.pharmthera.2016.10.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Roopesh Singh Gangwar
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Nadine Landolina
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Ludovica Arpinati
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Francesca Levi-Schaffer
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel.
| |
Collapse
|