1
|
Abdel-Mageed HM. Atopic dermatitis: a comprehensive updated review of this intriguing disease with futuristic insights. Inflammopharmacology 2025; 33:1161-1187. [PMID: 39918744 PMCID: PMC11914373 DOI: 10.1007/s10787-025-01642-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 01/10/2025] [Indexed: 03/19/2025]
Abstract
Atopic dermatitis (AD) is a paradigmatic prevalent, long-lasting, and inflammatory skin condition with a diverse range of clinical manifestations. The etiology and clinical symptoms of AD are influenced by complex pathophysiological processes, which involve a strong genetic component, epidermal dysfunction, and immunological dysregulation, and a strong influence of other physiological and environmental factors. The FDA has approved targeted and well-tolerated immunomodulators including biologics like dupilumab and crisaborole, and small molecules such as baricitinib, as novel therapies for AD. They effectively treat AD but are too expensive for most patients. The review provides an update on the state of knowledge of AD pathogenesis, discusses the available diagnostic and scoring indices, and provides a scientific foundation for treatment methods for AD. This review also presents data on clinical efficacy of innovative treatments' considering recent guidelines, emphasizing the newest medications and ongoing trials. Finally, the new implication of artificial intelligence (AI) in AD management is explored, where AI can speed up diagnosis and therapy. The PubMed, Google Scholar, and ScienceDirect databases were used for this review.
Collapse
Affiliation(s)
- Heidi M Abdel-Mageed
- Molecular Biology Department, National Research Centre, El Behoth St, Dokki, Giza, Egypt.
| |
Collapse
|
2
|
Zhou X, Dunham D, Sindher SB, Long A, Fernandes A, Chang I, Assa’ad A, Pongracic J, Spergel JM, Tam J, Tilles S, Wang J, Boyd SD, Chinthrajah RS, Nadeau KC. HLA-DR + regulatory T cells and IL-10 are associated with success or failure of desensitization outcomes. Allergy 2025; 80:762-774. [PMID: 39291303 PMCID: PMC11893263 DOI: 10.1111/all.16311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/14/2024] [Accepted: 08/05/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Omalizumab (XOLAIR®)-assisted multi-food oral immunotherapy (mOIT) has been shown to safely, effectively, and rapidly desensitize patients with multiple food allergies. In our clinical trial (NCT02626611) on omalizumab-assisted mOIT, different desensitization outcomes (success or failure of desensitization) were observed following a period of either continued or discontinued mOIT. However, the association between the immunological changes induced by omalizumab-assisted mOIT and desensitization outcomes has not yet been fully elucidated. In this study, due to the key roles of regulatory T (Treg) cells and the type 2 helper T cell (Th2) pathway in immune tolerance to food allergens, we aimed to characterize their association with the desensitization outcomes of omalizumab-assisted mOIT. METHODS Mass cytometry and multiplex cytokine assays were performed on blood samples obtained from participants with allergies to peanut, cashew, or milk in our phase 2 clinical study (NCT02626611). Comprehensive statistical and bioinformatic analyses were conducted on high-dimensional cytometry-based single-cell data and high-throughput multiplex cytokine data. RESULTS Our results demonstrated that the frequency of HLA-DR+ Treg cells, and the production of Th2 cytokines (IL-4, IL-5, IL-13, and IL-9) as well as the immunoregulatory cytokine IL-10 by peripheral blood mononuclear cells (PBMCs) was significantly increased in cultures with allergen compared to cultures with media alone at baseline (Week 0). We also observed increased frequency of allergen responsive HLA-DR+ Treg cells and enhanced production of IL-10 by PBMCs in participants who achieved successful desensitization compared to those with failure of desensitization. However, the production of Th2 cytokines by PBMCs did not show significant differences between participants with different desensitization outcomes (success vs. failure of desensitization), despite omalizumab-assisted mOIT inducing a significant reduction in the production of Th2 cytokines. CONCLUSIONS We demonstrated that the frequency of HLA-DR+ Treg cells and IL-10 cytokine production by PBMCs are associated with desensitization outcomes of omalizumab-assisted mOIT. These findings suggest potential immunological parameters that could be targeted to enhance desensitization success rates.
Collapse
Affiliation(s)
- Xiaoying Zhou
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Diane Dunham
- Sean N. Parker Center for Allergy and Asthma Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sayantani B Sindher
- Sean N. Parker Center for Allergy and Asthma Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrew Long
- Sean N. Parker Center for Allergy and Asthma Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrea Fernandes
- Sean N. Parker Center for Allergy and Asthma Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Iris Chang
- Sean N. Parker Center for Allergy and Asthma Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Amal Assa’ad
- Division of Allergy and Immunology, Cincinnati Children’s Medical Center, Cincinnati, OH, USA
| | - Jacqueline Pongracic
- Division of Allergy and Immunology, the Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA
| | - Jonathan M Spergel
- Division of Allergy and Immunology, The Children’s Hospital of Philadelphia Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan Tam
- Division of Clinical Immunology and Allergy, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Stephen Tilles
- Seattle Allergy and Asthma Research Institute, Seattle, WA, USA
- University of Washington, Seattle, WA, USA
| | - Julie Wang
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott D. Boyd
- Sean N. Parker Center for Allergy and Asthma Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Human Immune Monitoring Center, Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - R. Sharon Chinthrajah
- Sean N. Parker Center for Allergy and Asthma Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Kari C. Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
3
|
AlYafie R, Velayutham D, van Panhuys N, Jithesh PV. The genetics of hyper IgE syndromes. Front Immunol 2025; 16:1516068. [PMID: 40040707 PMCID: PMC11876172 DOI: 10.3389/fimmu.2025.1516068] [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: 10/23/2024] [Accepted: 01/29/2025] [Indexed: 03/06/2025] Open
Abstract
Hyper IgE syndromes (HIES) form a rare group of primary immunodeficiency disorders (PIDs) distinguished by persistent skin abscesses, dermatitis, allergies, and infections, in addition to their characteristic high serum IgE levels. Autosomal dominant (AD) and autosomal recessive (AR) genetic defects have been reported in HIES. From a clinical perspective, AD-HIES cases generally exhibit several non-immunologic features, including connective tissue, dental and skeletal abnormalities, whilst AR-HIES conditions have a higher incidence of neurologic complications and cutaneous viral infections. Genetic defects associated with HIES lead to impaired immune signaling, affecting pathways crucial for immune cell development, function, and immune response to pathogens/allergens. As a result, HIES patients are predisposed to recurrent bacterial and/or fungal infections, as well as atopic allergic responses. In many cases, the exact biological mechanisms responsible for the variations observed in the clinical phenotypes between the two inherited forms of HIES are still unclear. In this review, we describe the genetic basis of HIES with a distinction between the AR-HIES and AD-HIES forms, to better comprehend the different underlying molecular mechanisms, a distinction which is imperative for the accurate diagnosis, management, and development of targeted therapies for HIES patients.
Collapse
Affiliation(s)
- Randa AlYafie
- College of Health and Life Sciences, Hamad bin Khalifa University, Doha, Qatar
- Laboratory of Immunoregulation, Research Department, Sidra Medicine, Doha, Qatar
| | - Dinesh Velayutham
- College of Health and Life Sciences, Hamad bin Khalifa University, Doha, Qatar
| | - Nicholas van Panhuys
- College of Health and Life Sciences, Hamad bin Khalifa University, Doha, Qatar
- Laboratory of Immunoregulation, Research Department, Sidra Medicine, Doha, Qatar
| | | |
Collapse
|
4
|
Plichta J, Panek M. Role of the TGF-β cytokine and its gene polymorphisms in asthma etiopathogenesis. FRONTIERS IN ALLERGY 2025; 6:1529071. [PMID: 39949968 PMCID: PMC11821632 DOI: 10.3389/falgy.2025.1529071] [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: 11/15/2024] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Transforming growth factor beta (TGF-β) is a pluripotent cytokine expressed by all cells of the human body which plays important roles in maintaining homeostasis and allowing for proper individual development. Disturbances in TGF-β signaling contribute to the development of many diseases and disorders, including cancer and organ fibrosis. One of the diseases with the best-characterized correlation between TGF-β action and etiopathogenesis is asthma. Asthma is the most common chronic inflammatory disease of the lower and upper respiratory tract, characterized by bronchial hyperresponsiveness to a number of environmental factors, leading to bronchospasm and reversible limitation of expiratory flow. TGF-β, in particular TGF-β1, is a key factor in the etiopathogenesis of asthma. TGF-β1 concentration in bronchoalveolar lavage fluid samples is elevated in atopic asthma, and TGF-β expression is increased in asthmatic bronchial samples. The expression of all TGF-β isoforms is affected by a number of single nucleotide polymorphisms found in the genes encoding these cytokines. Some of the SNPs that alter the level of TGF-β expression may be associated with the occurrence and severity of symptoms of asthma and other diseases. The TGF-β gene polymorphisms, which are the subject of this paper, are potential diagnostic factors. If properly used, these polymorphisms can facilitate the early and precise diagnosis of asthma, allowing for the introduction of appropriate therapy and reduction of asthma exacerbation frequency.
Collapse
Affiliation(s)
- Jacek Plichta
- Department of Internal Medicine, Asthma and Allergology, Medical University of Lodz, Lodz, Poland
| | | |
Collapse
|
5
|
Satitsuksanoa P, van de Veen W, Tan G, Lopez J, Wirz O, Jansen K, Sokolowska M, Mirer D, Globinska A, Boonpiyathad T, Schneider SR, Barletta E, Spits H, Chang I, Babayev H, Tahralı İ, Deniz G, Yücel EÖ, Kıykım A, Boyd SD, Akdis CA, Nadeau K, Akdis M. Allergen-specific B cell responses in oral immunotherapy-induced desensitization, remission, and natural outgrowth in cow's milk allergy. Allergy 2025; 80:161-180. [PMID: 38989779 PMCID: PMC11724240 DOI: 10.1111/all.16220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 06/04/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Antigen-specific memory B cells play a key role in the induction of desensitization and remission to food allergens in oral immunotherapy and in the development of natural tolerance (NT). Here, we characterized milk allergen Bos d 9-specific B cells in oral allergen-specific immunotherapy (OIT) and in children spontaneously outgrowing cow's milk allergy (CMA) due to NT. METHODS Samples from children with CMA who received oral OIT (before, during, and after), children who naturally outgrew CMA (NT), and healthy individuals were received from Stanford biobank. Bos d 9-specific B cells were isolated by flow cytometry and RNA-sequencing was performed. Protein profile of Bos d 9-specific B cells was analyzed by proximity extension assay. RESULTS Increased frequencies of circulating milk allergen Bos d 9-specific B cells were observed after OIT and NT. Milk-desensitized subjects showed the partial acquisition of phenotypic features of remission, suggesting that desensitization is an earlier stage of remission. Within these most significantly expressed genes, IL10RA and TGFB3 were highly expressed in desensitized OIT patients. In both the remission and desensitized groups, B cell activation-, Breg cells-, BCR-signaling-, and differentiation-related genes were upregulated. In NT, pathways associated with innate immunity characteristics, development of marginal zone B cells, and a more established suppressor function of B cells prevail that may play a role in long-term tolerance. The analyses of immunoglobulin heavy chain genes in specific B cells demonstrated that IgG2 in desensitization, IgG1, IgA1, IgA2, IgG4, and IgD in remission, and IgD in NT were predominating. Secreted proteins from allergen-specific B cells revealed higher levels of regulatory cytokines, IL-10, and TGF-β after OIT and NT. CONCLUSION Allergen-specific B cells are essential elements in regulating food allergy towards remission in OIT-received and naturally resolved individuals.
Collapse
Affiliation(s)
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of ZürichDavosSwitzerland
| | - Ge Tan
- Swiss Institute of Allergy and Asthma Research (SIAF), University of ZürichDavosSwitzerland
- Functional Genomics Center Zürich, ETH ZürichZürichSwitzerland
| | - Juan‐Felipe Lopez
- Swiss Institute of Allergy and Asthma Research (SIAF), University of ZürichDavosSwitzerland
| | - Oliver Wirz
- Swiss Institute of Allergy and Asthma Research (SIAF), University of ZürichDavosSwitzerland
- Department of PathologyStanford UniversityStanfordCaliforniaUSA
| | - Kirstin Jansen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of ZürichDavosSwitzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of ZürichDavosSwitzerland
- Christine Kühne–Center for Allergy Research and Education (CK‐CARE)DavosSwitzerland
| | - David Mirer
- Swiss Institute of Allergy and Asthma Research (SIAF), University of ZürichDavosSwitzerland
| | - Anna Globinska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of ZürichDavosSwitzerland
| | - Tadech Boonpiyathad
- Swiss Institute of Allergy and Asthma Research (SIAF), University of ZürichDavosSwitzerland
| | - Stephan R. Schneider
- Swiss Institute of Allergy and Asthma Research (SIAF), University of ZürichDavosSwitzerland
| | - Elena Barletta
- Swiss Institute of Allergy and Asthma Research (SIAF), University of ZürichDavosSwitzerland
- Swiss Institute of BioinformaticsLaussaneSwitzerland
| | - Hergen Spits
- Department of Experimental ImmunologyAcademic Medical Center of the University of AmsterdamAmsterdamthe Netherlands
| | - Iris Chang
- Sean N. Parker Center for Allergy and Asthma ResearchStanfordCaliforniaUSA
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of ZürichDavosSwitzerland
| | - İlhan Tahralı
- Department of ImmunologyAziz Sancar Institute of Experimental Medicine, Istanbul UniversityIstanbulTurkey
| | - Gunnur Deniz
- Department of ImmunologyAziz Sancar Institute of Experimental Medicine, Istanbul UniversityIstanbulTurkey
| | - Esra Özek Yücel
- Division of Pediatrics, Department of Pediatric Allergy and Immunology, Istanbul Faculty of MedicineIstanbul UniversityIstanbulTurkey
- Department of Pediatric Allergy and Immunology, Cerrahpasa Medical FacultyIstanbul University‐CerrahpasaIstanbulTurkey
| | - Ayca Kıykım
- Department of Pediatric Allergy and Immunology, Cerrahpasa Medical FacultyIstanbul University‐CerrahpasaIstanbulTurkey
| | - Scott D. Boyd
- Department of PathologyStanford UniversityStanfordCaliforniaUSA
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of ZürichDavosSwitzerland
- Christine Kühne–Center for Allergy Research and Education (CK‐CARE)DavosSwitzerland
| | - Kari Nadeau
- Sean N. Parker Center for Allergy and Asthma ResearchStanfordCaliforniaUSA
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of ZürichDavosSwitzerland
| |
Collapse
|
6
|
Frischmeyer-Guerrerio PA, Young FD, Aktas ON, Haque T. Insights into the clinical, immunologic, and genetic underpinnings of food allergy. Immunol Rev 2024; 326:162-172. [PMID: 39034662 PMCID: PMC11436304 DOI: 10.1111/imr.13371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
The last few decades have seen striking changes in the field of food allergy. The prevalence of the disease has risen dramatically in many parts of the globe, and management of the condition has undergone major revision. While delayed introduction of common allergenic foods during infancy was advised for many years, the learning early about peanut allergy (LEAP) trial and other studies led to a major shift in infant feeding practices, with deliberate early introduction of these foods now recommended. Additionally, the Food and Drug Administration approved the first treatment for food allergy in 2020-a peanut oral immunotherapy (OIT) product that likely represents just the beginning of new immunotherapy-based and other treatments for food allergy. Our knowledge of the environmental and genetic factors contributing to the pathogenesis of food allergy has also undergone transformational advances. Here, we will discuss our efforts to improve the clinical care of patients with food allergy and our understanding of the immunological mechanisms contributing to this common disease.
Collapse
Affiliation(s)
- Pamela A Frischmeyer-Guerrerio
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Fernanda D Young
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Ozge N Aktas
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Tamara Haque
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
7
|
Bastyte D, Tamasauskiene L, Stakaitiene I, Briede K, Ugenskiene R, Valiukeviciene S, Gradauskiene B. Relation of T Cell Profile with Vitamin D Receptor and Vitamin D-Binding Protein Gene Polymorphisms in Atopy. Int J Mol Sci 2024; 25:9021. [PMID: 39201708 PMCID: PMC11354884 DOI: 10.3390/ijms25169021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/15/2024] [Accepted: 08/18/2024] [Indexed: 09/03/2024] Open
Abstract
Atopic diseases, including atopic dermatitis (AD) and allergic asthma (AA), are characterized by complex immune responses involving various T cells subsets and their cytokine profiles. It is assumed that single nucleotide polymorphisms (SNPs) in the Vitamin D receptor (VDR) gene and the Vitamin D-binding protein (GC) gene are related to the action of Vitamin D and, consequently, play a role in regulating the immune response. However, there is not enough data to unequivocally support the hypothesis about the relationship between T cells profile and VDR or GC SNPs. Two hundred sixty-six subjects (aged > 18 years) were involved in the study: 100 patients with mild or moderate AD, 85 patients with mild or moderate AA, and 81 healthy individuals. Blood cell counts were determined by standard methods. Flow cytometric analysis was used to evaluate CD4+ T-helper (Th) cell subtypes: Th2, Th1, Th17, and T regulatory (Treg) cells in peripheral blood. Measurements of cytokines, total immunoglobulin E (IgE), and Vitamin D levels in serum were evaluated by ELISA. Significantly higher levels of Th1, Th2, and Th17 cells, along with lower levels of Tregs, were found in patients with atopic diseases compared to healthy individuals. Additionally, higher serum levels of interleukin (IL) 5, IL-17A, and transforming growth factor-β1 (TGF-β1), as well as lower levels of IL-10, were observed in patients with atopic diseases than in control. The study established associations between VDR SNPs and immune profiles: the AA genotype of rs731236 was associated with increased Th2 and Th17 cells and a higher Th1/Th2 ratio; the GG genotype of rs731236 was linked to decreased serum IL-10 and TGF-β1 levels; and the TT genotype of rs11168293 was associated with increased IL-10 levels. Additionally, the GG genotype of GC gene SNP rs4588 was associated with reduced Th2 and Th17 lymphocytes, while the TT genotype of rs4588 was linked to decreased IL-10 levels. Furthermore, the CC genotype of rs7041 was associated with higher levels of Th2, Th17, IL-10, and IL-35, as well as reduced levels of TGF-β1, while the GG genotype of rs3733359 was associated with reduced IL-10 levels. In conclusion, our study demonstrates that the Vitamin D receptor gene single nucleotide polymorphisms rs731236 and rs11168293, along with polymorphisms in the Vitamin D-binding protein gene (rs4588, rs7041, rs3733359), are significantly associated with variations in T cell profiles in atopy. These variations may play a crucial role in promoting inflammation and provide insight into the genetic factors contributing to the pathogenesis of atopy.
Collapse
Affiliation(s)
- Daina Bastyte
- Department of Immunology and Allergology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (D.B.); (L.T.)
- Laboratory of Immunology, Department of Immunology and Allergology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Laura Tamasauskiene
- Department of Immunology and Allergology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (D.B.); (L.T.)
- Laboratory of Immunology, Department of Immunology and Allergology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Ieva Stakaitiene
- Department of Genetics and Molecular Medicine, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Kamilija Briede
- Department of Skin and Venereal Diseases, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Rasa Ugenskiene
- Department of Genetics and Molecular Medicine, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Skaidra Valiukeviciene
- Department of Skin and Venereal Diseases, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Brigita Gradauskiene
- Department of Immunology and Allergology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (D.B.); (L.T.)
| |
Collapse
|
8
|
Fakhimahmadi A, Roth-Walter F, Hofstetter G, Wiederstein M, Jensen SA, Berger M, Szepannek N, Bianchini R, Pali-Schöll I, Jensen-Jarolim E, Hufnagl K. Mould allergen Alt a 1 spiked with the micronutrient retinoic acid reduces Th2 response and ameliorates Alternaria allergy in BALB/c mice. Allergy 2024. [PMID: 38818808 DOI: 10.1111/all.16181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/29/2024] [Accepted: 05/20/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND We investigated the biological function of the mould allergen Alt a 1 as a carrier of micronutrients, such as the vitamin A metabolite retinoic acid (RA) and the influence of RA binding on its allergenicity in vitro and in vivo. METHODS Alt a 1-RA complex formation was analyzed in silico and in vitro. PBMCs from Alternaria-allergic donors were stimulated with Alt a 1 complexed with RA (holo-Alt a 1) or empty apo-Alt a 1 and analyzed for cytokine production and CD marker expression. Serum IgE-binding and crosslinking assays to apo- and holo-protein were correlated to B-cell epitope analysis. Female BALB/c mice already sensitized to Alt a 1 were intranasally treated with apo-Alt a 1, holo-Alt a 1 or RA alone before measuring anaphylactic response, serum antibody levels, splenic cytokines and CD marker expression. RESULTS In silico docking calculations and in vitro assays showed that the extent of RA binding depended on the higher quaternary state of Alt a 1. Holo-Alt a 1 loaded with RA reduced IL-13 released from PBMCs and CD3+CD4+CRTh2 cells. Complexing Alt a 1 to RA masked its IgE B-cell epitopes and reduced its IgE-binding capacity. In a therapeutic mouse model of Alternaria allergy nasal application of holo-Alt a 1, but not of apo-Alt a 1, significantly impeded the anaphylactic response, impaired splenic antigen-presenting cells and induced IL-10 production. CONCLUSION Holo-Alt a 1 binding to RA was able to alleviate Th2 immunity in vitro, modulate an ongoing Th2 response and prevent anaphylactic symptoms in vivo, presenting a novel option for improving allergen-specific immunotherapy in Alternaria allergy.
Collapse
Affiliation(s)
- Aila Fakhimahmadi
- Messerli Research Institute, Department of Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Franziska Roth-Walter
- Messerli Research Institute, Department of Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Gerlinde Hofstetter
- Messerli Research Institute, Department of Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Markus Wiederstein
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | - Sebastian A Jensen
- Messerli Research Institute, Department of Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
- AllergyCare Allergy Diagnosis Center, Private Clinic Döbling, Vienna, Austria
| | - Markus Berger
- Messerli Research Institute, Department of Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Nathalie Szepannek
- Messerli Research Institute, Department of Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Rodolfo Bianchini
- Messerli Research Institute, Department of Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Isabella Pali-Schöll
- Messerli Research Institute, Department of Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Erika Jensen-Jarolim
- Messerli Research Institute, Department of Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- AllergyCare Allergy Diagnosis Center, Private Clinic Döbling, Vienna, Austria
- Biomedical International R+D GmbH, Vienna, Austria
| | - Karin Hufnagl
- Messerli Research Institute, Department of Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- AllergyCare Allergy Diagnosis Center, Private Clinic Döbling, Vienna, Austria
| |
Collapse
|
9
|
Shafi T, Rasool Wani R, Hussain S, Bhat IA, Makhdoomi R, Bashir SA, Hassan I, Shah ZA. Investigating dysregulation of TGF-β1/SMAD3 signaling in atopic dermatitis: a molecular and immunohistochemical analysis. Clin Exp Immunol 2024; 216:192-199. [PMID: 38066678 PMCID: PMC11036103 DOI: 10.1093/cei/uxad130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/07/2023] [Accepted: 12/02/2023] [Indexed: 04/24/2024] Open
Abstract
Atopic dermatitis (AD) is a persistent and recurring inflammatory condition affecting the skin. An expanding corpus of evidence indicates the potential participation of transforming growth factor-β1 (TGF-β1) in the modulation of inflammation and tissue remodeling in AD. The primary objective of this study was to examine the aberrant modulation of TGF-β1/small mothers against decapentaplegic homolog 3 (SMAD3) signaling through a comprehensive analysis of their molecular and protein expression profiles. The study encompassed an aggregate of 37 participants, which included 25 AD patients and 12 controls. The assessment of mRNA and protein levels of TGF-β1 and SMAD3 was conducted utilizing quantitative real-time PCR and immunohistochemistry (IHC), whereas serum IgE and vitamin D levels were estimated by ELISA and chemiluminescence, respectively. Quantitative analysis demonstrated a 2.5-fold upregulation of TGF-β1 mRNA expression in the lesional AD skin (P < 0.0001). IHC also exhibited a comparable augmented pattern, characterized by moderate to strong staining intensities. In addition, TGF-β1 mRNA showed an association with vitamin D deficiency in serum (P < 0.02), and its protein expression was linked with the disease severity (P < 0.01) Furthermore, a significant decrease in the expression of the SMAD3 gene was observed in the affected skin (P = 0.0004). This finding was further confirmed by evaluating the protein expression and phosphorylation of SMAD3, both of which exhibited a decrease. These findings suggest that there is a dysregulation in the TGF-β1/SMAD3 signaling pathway in AD. Furthermore, the observed augmentation in mRNA and protein expression of TGF-β1, along with its correlation with the disease severity, holds considerable clinical significance and emphasizes its potential role in AD pathogenesis.
Collapse
Affiliation(s)
- Tabasum Shafi
- Department of Immunology and Molecular Medicine, SKIMS, Srinagar, India-190011
| | - Roohi Rasool Wani
- Department of Immunology and Molecular Medicine, SKIMS, Srinagar, India-190011
| | - Showkat Hussain
- Department of Immunology and Molecular Medicine, SKIMS, Srinagar, India-190011
| | - Imtiyaz A Bhat
- Department of Immunology and Molecular Medicine, SKIMS, Srinagar, India-190011
| | | | - Sheikh Adil Bashir
- Department of Plastic and Reconstructive Surgery, SKIMS, Srinagar, India-190011
| | - Iffat Hassan
- Department of Dermatology, Venereology, and Leprosy, GMC, Srinagar, India-190010
| | - Zafar A Shah
- Department of Immunology and Molecular Medicine, SKIMS, Srinagar, India-190011
| |
Collapse
|
10
|
Afshari M, Kolackova M, Rosecka M, Čelakovská J, Krejsek J. Unraveling the skin; a comprehensive review of atopic dermatitis, current understanding, and approaches. Front Immunol 2024; 15:1361005. [PMID: 38500882 PMCID: PMC10944924 DOI: 10.3389/fimmu.2024.1361005] [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: 12/24/2023] [Accepted: 02/14/2024] [Indexed: 03/20/2024] Open
Abstract
Atopic dermatitis, also known as atopic eczema, is a chronic inflammatory skin disease characterized by red pruritic skin lesions, xerosis, ichthyosis, and skin pain. Among the social impacts of atopic dermatitis are difficulties and detachment in relationships and social stigmatization. Additionally, atopic dermatitis is known to cause sleep disturbance, anxiety, hyperactivity, and depression. Although the pathological process behind atopic dermatitis is not fully known, it appears to be a combination of epidermal barrier dysfunction and immune dysregulation. Skin is the largest organ of the human body which acts as a mechanical barrier to toxins and UV light and a natural barrier against water loss. Both functions face significant challenges due to atopic dermatitis. The list of factors that can potentially trigger or contribute to atopic dermatitis is extensive, ranging from genetic factors, family history, dietary choices, immune triggers, and environmental factors. Consequently, prevention, early clinical diagnosis, and effective treatment may be the only resolutions to combat this burdensome disease. Ensuring safe and targeted drug delivery to the skin layers, without reaching the systemic circulation is a promising option raised by nano-delivery systems in dermatology. In this review, we explored the current understanding and approaches of atopic dermatitis and outlined a range of the most recent therapeutics and dosage forms brought by nanotechnology. This review was conducted using PubMed, Google Scholar, and ScienceDirect databases.
Collapse
Affiliation(s)
- Moeina Afshari
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czechia
| | - Martina Kolackova
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czechia
| | - Michaela Rosecka
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czechia
| | - Jarmila Čelakovská
- Department of Dermatology and Venereology, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czechia
| | - Jan Krejsek
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czechia
| |
Collapse
|
11
|
Milner JD. ERBIN and phosphoglucomutase 3 deficiency. Curr Opin Immunol 2023; 84:102353. [PMID: 37369151 PMCID: PMC11583051 DOI: 10.1016/j.coi.2023.102353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 02/14/2023] [Accepted: 05/12/2023] [Indexed: 06/29/2023]
Abstract
ERBIN and phosphoglucomutase 3 (PGM3) mutations both lead to rare primary atopic disorders characterized by allergic disease and connective tissue abnormalities, though each disorder has its own rather unique pattern of multisystem presentations. Pathway studies show how ERBIN mutations allow for enhanced TGFb signaling, and prevent STAT3 from negative-regulating TGFb signaling. This likely explains many elements of clinical overlap between disorders of STAT3 and TGFb signaling. The excessive TGFb signaling leading to increased IL-4 receptor expression also provides the rationale for precision-based therapy blocking the IL-4 receptor to treat the atopic disease. The mechanism by which PGM3 deficiency leads to atopic phenotypes is not well understood, nor is the broad variability in disease penetrance and expressivity, though preliminary studies suggest an overlap with IL-6 receptor signaling defects.
Collapse
Affiliation(s)
- Joshua D Milner
- Department of Pediatrics, Columbia University Irving Medical Center, USA.
| |
Collapse
|
12
|
Li YK, Wang HY, Chen Y, Shi XQ, Zhang XX, Li K, Fu WP, Sun C. Single‑nucleotide polymorphism rs6592645 confers asthma risk through regulating LRRC32 expression. Exp Ther Med 2023; 26:451. [PMID: 37614425 PMCID: PMC10443064 DOI: 10.3892/etm.2023.12150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/23/2023] [Indexed: 08/25/2023] Open
Abstract
Asthma is a complex disease, often with evident genetic predisposition; for example, the single-nucleotide polymorphism (SNP) rs7130588 was significantly associated with asthma by genome-wide association study (GWAS). Analysis of 1000 Genomes Project data suggests that there is another SNP, rs6592645, in complete linkage disequilibrium with rs7130588 and should present the same signal in GWAS. However, the causal SNP and the mechanism for the association between rs7130588 and asthma remain to be elucidated. In the presents study, results from dual-luciferase assays indicated that the A/G alleles of rs7130588 failed to present significantly different reporter gene expression. By contrast, A allele of rs6592645 presented a significant increase in relative luciferase activity than G allele, thus suggesting that rs6592645 may be a causal SNP. Using chromosome conformation capture, the enhancer region containing rs6592645 was observed to interact with promoter region of leucine-rich repeat-containing 32 (LRRC32). Gene expression quantification suggested that LRRC32 expression is significantly increased in lung tissue of patients with asthma and is dependent on the genotype of this locus, thus verifying that LRRC32 may be involved in asthma onset and that rs6592645 can regulate LRRC32 expression. Through chromatin immunoprecipitation, transcription factor 3 (TCF3) was identified to bind to rs6592645 surrounding region and the interaction between TCF3 and rs6592645 surrounding region was investigated. Results from the present study may improve our understanding of the mechanism by which the genetic variation in this locus might influence asthma susceptibility.
Collapse
Affiliation(s)
- Yi-Kun Li
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, P.R. China
| | - Hong-Yan Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, P.R. China
| | - Ying Chen
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, P.R. China
| | - Xiao-Qian Shi
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, P.R. China
| | - Xin-Xin Zhang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, P.R. China
| | - Ke Li
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, P.R. China
| | - Wei-Ping Fu
- Department of Respiratory Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Chang Sun
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, P.R. China
| |
Collapse
|
13
|
Pontikas A, Antonatos C, Evangelou E, Vasilopoulos Y. Candidate Gene Association Studies in Atopic Dermatitis in Participants of European and Asian Ancestry: A Systematic Review and Meta-Analysis. Genes (Basel) 2023; 14:1456. [PMID: 37510360 PMCID: PMC10379179 DOI: 10.3390/genes14071456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/12/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Atopic dermatitis (AD) has been extensively investigated for genetic associations utilizing both candidate gene approaches and genome-wide scans. Here, we comprehensively evaluated the available literature to determine the association of candidate genes in AD to gain additional insight into the etiopathogenesis of the disease. We systematically screened all studies that explored the association between polymorphisms and AD risks in cases of European and Asian ancestry and synthesized the available evidence through a random-effects meta-analysis. We identified 99 studies that met our inclusion/exclusion criteria that examined 17 candidate loci in Europeans and 14 candidate genes in Asians. We confirmed the significant associations between FLG variants in both European and Asian populations and AD risk, while synthesis of the available data revealed novel loci mapped to IL18 and TGFB1 genes in Europeans and IL12RB1 and MIF in Asians that have not yet been identified by genome-wide association studies. Our findings provide comprehensive evidence for AD risk loci in cases of both European and Asian ancestries, validating previous associations as well as revealing novel loci that could imply previously unexplored biological pathways.
Collapse
Affiliation(s)
- Alexandros Pontikas
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Charalabos Antonatos
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, 45110 Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, 45110 Ioannina, Greece
- Department of Epidemiology & Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK
| | - Yiannis Vasilopoulos
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| |
Collapse
|
14
|
Shafi T, Rasool R, Ayub S, Bhat IA, Gull A, Hussain S, Hassan Shah I, Shah ZA. Analysis of intronic SNP (rs4147358) and expression of SMAD3 gene in Atopic Dermatitis: A case-control study. Immunobiology 2023; 228:152390. [PMID: 37100019 DOI: 10.1016/j.imbio.2023.152390] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/10/2023] [Accepted: 04/18/2023] [Indexed: 04/28/2023]
Abstract
BACKGROUND Atopic Dermatitis (AD) is a multifactorial cutaneous disorder associated with chronic inflammation of the skin. Growing evidence points to TGF-β/SMAD signaling as a key player in mediating inflammation and the subsequent tissue remodeling, often resulting in fibrosis. This study investigates the role of a core transcription factor involved in TGF-β signaling i.e., SMAD3 genetic variants (rs4147358) in AD predisposition and its association with SMAD3 mRNA expression, serum IgE levels, and sensitization to various allergens in AD patients. METHODS A total of 246 subjects including 134 AD cases and 112 matched healthy controls were genotyped for SMAD3 intronic SNP by PCR-RFLP. mRNA expression of SMAD3 was determined by quantitative Real-Time PCR (qRT-PCR), Vitamin-D levels by chemiluminescence, and total serum IgE levels by ELISA. In-vivo allergy testing was performed for the evaluation of allergic reactions to house dust mites (HDM) and food allergens. RESULTS A significantly higher frequency of mutant genotype AA (cases: 19.4% vs controls: 8.9%) (OR = 2.8, CI = 1.2 - 6.7, p = 0.01) was observed in AD cases. The mutant allele 'A' also showed a 1.9-fold higher risk for AD compared to the wild allele 'C' indicating that the carriers of the A allele have a higher risk for AD predisposition (OR-1.9, CI = 1.3-2.8, p < 0.001). In addition, quantitative analysis of SMAD3 mRNA in peripheral blood showed 2.8-fold increased expression in AD cases as compared to healthy controls. Stratification analysis revealed the association of the mutant AA genotype with deficient serum Vitamin D levels (p = 0.02) and SMAD3 mRNA overexpression with HDM sensitization (p = 0.03). Furthermore, no significant association of genotypes with SMAD3 mRNA expression was observed. CONCLUSION Our study indicates that SMAD3 intronic SNP bears a significant risk of AD development. Moreover, overexpression of SMAD3 mRNA and its association with HDM sensitization highlights the possible role of this gene in AD pathogenesis.
Collapse
Affiliation(s)
- Tabasum Shafi
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| | - Roohi Rasool
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India.
| | - Sakeena Ayub
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| | - Imtiyaz A Bhat
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| | - Ayaz Gull
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| | - Showkat Hussain
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| | - Iffat Hassan Shah
- Department of Dermatology, Venereology, and Leprosy, GMC- Srinagar 190010, India
| | - Zafar A Shah
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| |
Collapse
|
15
|
Shafi T, Rasool R, Ayub S, Bhat IA, Shah IH, Hussain S, Shah ZA, Baba SM, Makhdoomi R, Bashir SA. Unveiling the TGF- β1 paradox: Significant implication of TGF- β1 promoter variants and its mRNA and protein expression in atopic dermatitis. Mol Immunol 2023; 157:214-224. [PMID: 37084506 DOI: 10.1016/j.molimm.2023.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/23/2023]
Abstract
BACKGROUND Atopic Dermatitis (AD) is a chronic inflammatory skin disorder with evidence of lichenification in later stages. There is mounting evidence supporting the role of TGF- β1 in mediating inflammation as well as subsequent tissue remodeling, often resulting in fibrosis. Given the role of genetic variants in the differential expression of TGF-β1 in various diseases, this study seeks to ascertain the role of TGF-β1 promoter variants (rs1800469 and rs1800468) in AD susceptibility, as well as their association with TGF- β1 mRNA expression, TGF- β1 serum levels and skin prick test positivity in Atopic Dermatitis patients. METHODS An aggregate of 246 subjects including 134 AD cases and 112 matched healthy controls were genotyped for TGF-β1 promoter polymorphisms by PCR-RFLP. TGF- β1 mRNA was quantified by quantitative Real-Time PCR (qRT-PCR), Vitamin-D levels by chemiluminescence, and serum TGF- β1, and total IgE levels were determined by ELISA. In-vivo allergy testing was performed for the evaluation of allergic reactions to house dust mites and food allergens. RESULTS A higher frequency of TT genotypes of rs1800469 (OR = 7.7, p = 0.0001) and GA+AA genotypes of rs1800468 (OR-4.4, p < 0.0001) were observed in AD cases than those in controls. Haplotype analysis demonstrated that TG haplotype carriers had an increased risk of AD (p = 0.013). Quantitative analysis revealed a significant upregulation of both mRNA (p = 0.0002) and serum levels (p < 0.0001) of TGF- β1 with a substantial positive correlation between them (Correlation coefficient=0.504; p = 0.01). Moreover, serum TGF-β1 levels were associated with quality of life (p = 0.03), the severity of the disease (p = 0.03), and House dust mite allergy (p = 0.01) whereas TGF-β1 mRNA levels positively correlated with disease severity(p = 0.02). Stratification analysis revealed that the TT genotype of rs1800469 was associated with higher IgE levels (p = 0.01) and eosinophil percentage(p = 0.007) whereas the AA genotype of rs1800468 correlated with elevated serum IgE levels (p = 0.01). Besides, no significant association of genotypes with mRNA and serum expression of TGF-β1 was observed. CONCLUSION Our study indicates that TGF-β1 promoter SNPs bear a significant risk of AD development. Moreover, upregulation of TGF-β1 mRNA and serum levels and their association with disease severity, quality of life, and HDM allergy suggests its role as a diagnostic/prognostic biomarker that could help in the development of new therapeutic and prevention strategies.
Collapse
Affiliation(s)
- Tabasum Shafi
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| | - Roohi Rasool
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India.
| | - Sakeena Ayub
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| | - Imtiyaz A Bhat
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| | - Iffat Hassan Shah
- Department of Dermatology, Venereology, and Leprosy, GMC- Srinagar 190010, India
| | - Showkat Hussain
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| | - Zafar A Shah
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| | - Shahid M Baba
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| | | | - Sheikh Adil Bashir
- Department of Plastic and Reconstructive Surgery, SKIMS, Srinagar 190011, India
| |
Collapse
|
16
|
Li R, Wang R, Zhong S, Asghar F, Li T, Zhu L, Zhu H. TGF-β1-overexpressing mesenchymal stem cells reciprocally regulate Th17/Treg cells by regulating the expression of IFN-γ. Open Life Sci 2021; 16:1193-1202. [PMID: 34761110 PMCID: PMC8565592 DOI: 10.1515/biol-2021-0118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/22/2021] [Accepted: 09/27/2021] [Indexed: 12/23/2022] Open
Abstract
Transforming growth factor (TGF)-β1 and mesenchymal stromal cells (MSCs) are two effective immunosuppressive agents for organ transplantation technology. This study aims to explore the molecular mechanism of TGF-β1-overexpressed MSCs on T cell immunosuppression. To achieve that, BM-MSCs were isolated from canine bone marrow, and their osteogenic differentiation and surface markers were detected. The TGF-β1 gene was transferred into lentivirus and modified MSCs (TGF-β1/MSCs) by lentivirus transfection. Furthermore, TGF-β1/MSCs were co-cultured with T cells to investigate their effect on differentiation and immune regulation. Results showed that TGF-β1/MSCs significantly downregulated the proportion of CD4+ CD8+ T cells in lymphocytes and significantly upregulated the proportion of CD4+ CD25+ T cells. Moreover, TGF-β1/MSCs significantly upregulated the expression of IL-10 in CD4+ T cells and downregulated the expression of IL-17A, IL-21, and IL-22. Meanwhile, interferon-γ (IFN-γ) neutralizing antibody blocked the effects of TGF-β1/MSCs on the differentiation inhibition of Th17. Overall, our results confirm the strong immunosuppressive effect of TGF-β1/MSCs in vitro and demonstrate that IFN-γ mediates the immunosuppressive effect of TGF-β1/MSC.
Collapse
Affiliation(s)
- Ruixue Li
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650101, China
| | - Renyong Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650101, China
| | - Shijie Zhong
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650101, China
| | - Farhan Asghar
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650101, China
| | - Tiehan Li
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650101, China
| | - Lei Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650101, China
| | - Hong Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650101, China
| |
Collapse
|
17
|
Intestinal immunoregulation: lessons from human mendelian diseases. Mucosal Immunol 2021; 14:1017-1037. [PMID: 33859369 DOI: 10.1038/s41385-021-00398-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 02/04/2023]
Abstract
The mechanisms that maintain intestinal homeostasis despite constant exposure of the gut surface to multiple environmental antigens and to billions of microbes have been scrutinized over the past 20 years with the goals to gain basic knowledge, but also to elucidate the pathogenesis of inflammatory bowel diseases (IBD) and to identify therapeutic targets for these severe diseases. Considerable insight has been obtained from studies based on gene inactivation in mice as well as from genome wide screens for genetic variants predisposing to human IBD. These studies are, however, not sufficient to delineate which pathways play key nonredundant role in the human intestinal barrier and to hierarchize their respective contribution. Here, we intend to illustrate how such insight can be derived from the study of human Mendelian diseases, in which severe intestinal pathology results from single gene defects that impair epithelial and or hematopoietic immune cell functions. We suggest that these diseases offer the unique opportunity to study in depth the pathogenic mechanisms leading to perturbation of intestinal homeostasis in humans. Furthermore, molecular dissection of monogenic intestinal diseases highlights key pathways that might be druggable and therapeutically targeted in common forms of IBD.
Collapse
|
18
|
Hsu YSO, Lu KL, Fu Y, Wang CW, Lu CW, Lin YF, Chang WC, Yeh KY, Hung SI, Chung WH, Chen CB. The Roles of Immunoregulatory Networks in Severe Drug Hypersensitivity. Front Immunol 2021; 12:597761. [PMID: 33717075 PMCID: PMC7953830 DOI: 10.3389/fimmu.2021.597761] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
The immunomodulatory effects of regulatory T cells (Tregs) and co-signaling receptors have gained much attention, as they help balance immunogenic and immunotolerant responses that may be disrupted in autoimmune and infectious diseases. Drug hypersensitivity has a myriad of manifestations, which ranges from the mild maculopapular exanthema to the severe Stevens-Johnson syndrome (SJS), toxic epidermal necrolysis (TEN), and drug reaction with eosinophilia and systemic symptoms/drug-induced hypersensitivity syndrome (DRESS/DIHS). While studies have identified high-risk human leukocyte antigen (HLA) allotypes, the presence of the HLA allotype at risk is not sufficient to elicit drug hypersensitivity. Recent studies have suggested that insufficient regulation by Tregs may play a role in severe hypersensitivity reactions. Furthermore, immune checkpoint inhibitors, such as anti-CTLA-4 or anti-PD-1, in cancer treatment also induce hypersensitivity reactions including SJS/TEN and DRESS/DIHS. Taken together, mechanisms involving both Tregs as well as coinhibitory and costimulatory receptors may be crucial in the pathogenesis of drug hypersensitivity. In this review, we summarize the currently implicated roles of co-signaling receptors and Tregs in delayed-type drug hypersensitivity in the hope of identifying potential pharmacologic targets.
Collapse
Affiliation(s)
- Yun-Shiuan Olivia Hsu
- Department of Medical Education, Chang Gung Memorial Hospital, Linkou, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kun-Lin Lu
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Yun Fu
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chuang-Wei Wang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Memorial Hospital, Chang Gung Immunology Consortium, Linkou, Taiwan
| | - Chun-Wei Lu
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Taiwan
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Yu-Fen Lin
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan
- Department of Nursing, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Wen-Cheng Chang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Kun-Yun Yeh
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Shuen-Iu Hung
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Memorial Hospital, Chang Gung Immunology Consortium, Linkou, Taiwan
| | - Wen-Hung Chung
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Memorial Hospital, Chang Gung Immunology Consortium, Linkou, Taiwan
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan
- Department of Dermatology, Chang Gung Hospital, Xiamen, China
- Department of Dermatology, Beijing Tsinghua Chang Gung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Department of Dermatology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chun-Bing Chen
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Memorial Hospital, Chang Gung Immunology Consortium, Linkou, Taiwan
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan
- Department of Dermatology, Chang Gung Hospital, Xiamen, China
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
- College of Medicine, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
19
|
Pierce SK, Schwartzberg PL, Shah NN, Taylor N. Women in immunology: 2020 and beyond. Nat Immunol 2020; 21:254-258. [PMID: 32094649 PMCID: PMC11549733 DOI: 10.1038/s41590-020-0618-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 01/24/2020] [Indexed: 12/27/2022]
Abstract
Women have been at the forefront of tremendous achievements in immunology in the past decade. However, disparities still exist, limiting upward potential and further advancements. As four NIH intramural women scientists who care deeply about scientific progress and the progress of women in our field, we review ongoing challenges and discuss potential approaches to help advance the promotion of women in the sciences.
Collapse
Affiliation(s)
- Susan K Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA.
| | - Pamela L Schwartzberg
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA.
| | - Nirali N Shah
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA.
| | - Naomi Taylor
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA.
- IGMM, Université de Montpellier, CNRS, Montpellier, France.
| |
Collapse
|