1
|
James AE, Abdalgani M, Khoury P, Freeman AF, Milner JD. T H2-driven manifestations of inborn errors of immunity. J Allergy Clin Immunol 2024; 154:245-254. [PMID: 38761995 DOI: 10.1016/j.jaci.2024.05.007] [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: 10/31/2023] [Revised: 04/25/2024] [Accepted: 05/06/2024] [Indexed: 05/20/2024]
Abstract
Monogenic lesions in pathways critical for effector functions responsible for immune surveillance, protection against autoinflammation, and appropriate responses to allergens and microorganisms underlie the pathophysiology of inborn errors of immunity (IEI). Variants in cytokine production, cytokine signaling, epithelial barrier function, antigen presentation, receptor signaling, and cellular processes and metabolism can drive autoimmunity, immunodeficiency, and/or allergic inflammation. Identification of these variants has improved our understanding of the role that many of these proteins play in skewing toward TH2-related allergic inflammation. Early-onset or atypical atopic disease, often in conjunction with immunodeficiency and/or autoimmunity, should raise suspicion for an IEI. This becomes a diagnostic dilemma if the initial clinical presentation is solely allergic inflammation, especially when the prevalence of allergic diseases is becoming more common. Genetic sequencing is necessary for IEI diagnosis and is helpful for early recognition and implementation of targeted treatment, if available. Although genetic evaluation is not feasible for all patients with atopy, identifying atopic patients with molecular immune abnormalities may be helpful for diagnostic, therapeutic, and prognostic purposes. In this review, we focus on IEI associated with TH2-driven allergic manifestations and classify them on the basis of the affected molecular pathways and predominant clinical manifestations.
Collapse
Affiliation(s)
- Alyssa E James
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Manar Abdalgani
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Paneez Khoury
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Alexandra F Freeman
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| | - Joshua D Milner
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| |
Collapse
|
2
|
Simpson EL, Silverberg JI, Nosbaum A, Winthrop K, Guttman-Yassky E, Hoffmeister KM, Egeberg A, Valdez H, Fan H, Farooqui SA, Chan G, Alderfer J, Romero W, Chittuluru K. Integrated Safety Update of Abrocitinib in 3802 Patients with Moderate-to-Severe Atopic Dermatitis: Data from More than 5200 Patient-Years with Up to 4 Years of Exposure. Am J Clin Dermatol 2024; 25:639-654. [PMID: 38888681 PMCID: PMC11193687 DOI: 10.1007/s40257-024-00869-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Abrocitinib, an oral, once-daily, Janus kinase 1-selective inhibitor, is efficacious in moderate-to-severe atopic dermatitis with a manageable long-term safety profile. OBJECTIVE We aimed to provide updated integrated long-term safety results for abrocitinib from available data accrued up to a maximum of almost 4 years in patients with moderate-to-severe atopic dermatitis from the JADE clinical development program. METHODS Analysis included 3802 patients (exposure: 5213.9 patient-years) from the phase II monotherapy study (NCT02780167) and the phase III studies JADE MONO-1 (NCT03349060), JADE MONO-2 (NCT03575871), JADE TEEN (NCT03796676), JADE COMPARE (NCT03720470), JADE DARE (NCT04345367; 200 mg only), JADE REGIMEN (NCT03627767), and JADE EXTEND (NCT03422822; data cutoff 25 September, 2021). Data from patients receiving one or more doses of abrocitinib 200 mg or 100 mg were pooled in a consistent-dose cohort (patients were allocated to receive the same abrocitinib dose throughout exposure in the qualifying parent study and/or long-term study) or a variable-dose cohort (patients received open-label abrocitinib 200 mg; responders were randomized to abrocitinib 200 mg, 100 mg, or placebo, and could then receive abrocitinib 200 mg plus topical corticosteroids as rescue therapy). Incidence rates of adverse events of special interest were assessed. Cox regression analysis of risk factors for herpes zoster and serious infections was performed. RESULTS Overall, this safety analysis of long-term data up to a maximum of ~ 4 years of abrocitinib exposure does not indicate any changes from the previously reported risk profile. The most frequent serious infections (per Medical Dictionary for Regulatory Activities preferred term) with consistent-dose abrocitinib 200 mg and 100 mg were herpes zoster (0.5% and 0.2%), pneumonia (0.2% with either dose), and herpes simplex (0.1% with either dose). Risk factors for herpes zoster were a history of herpes zoster, abrocitinib 200-mg dose, age ≥ 65 years, absolute lymphocyte count < 1 × 103/mm3 before the event, and residing in Asia. For serious infections, > 100 kg body weight was a risk factor. Incidence rate/100 patient-years (95% confidence interval) with the consistent abrocitinib 200-mg and 100-mg dose combined was higher in older (aged ≥ 65 years) patients versus younger (aged 18 to < 65 years) patients for serious adverse events (17.6 [11.7‒25.4] vs 6.7 [5.8‒7.8]), malignancy excluding non-melanoma skin cancer (2.4 [0.6‒6.0] vs 0.1 [0.0‒0.4]), non-melanoma skin cancer (2.4 [0.6‒6.1] vs 0.2 [0.1‒0.4]), lymphopenia (3.5 [1.3‒7.6] vs 0.1 [0.0‒0.3]), and venous thromboembolism (1.7 [0.4‒5.1] vs 0.1 [0.0‒0.3]). Incident rate/100 patient-years (95% confidence interval) of non-melanoma skin cancer with the consistent abrocitinib 200-mg and 100-mg dose combined was higher in current/former smokers (0.9 [0.4‒1.6]) vs never-smokers (0.0 [0.0‒0.1]). CONCLUSIONS This safety update showed a consistent profile for abrocitinib with no new safety signals and continues to support that abrocitinib has a manageable long-term safety profile in patients with moderate-to-severe atopic dermatitis. Risk of specific adverse events was higher in certain patient populations, especially those aged ≥ 65 years. [Video abstract available.] CLINICAL TRIAL REGISTRATION: NCT02780167; study start date: April, 2016; primary completion date: March, 2017; study completion date: April, 2017. NCT03349060; study start date: 7 December, 2017; study completion date: 26 March, 2019. NCT03575871; study start date: 29 June, 2018; study completion date: 13 August, 2019. NCT03720470; study start date: 29 October, 2018; primary completion date: 27 December, 2019; study completion date: 6 March, 2020. NCT03796676; study start date: 18 February, 2019; study completion date: 8 April, 2020. NCT03627767; study start date: 11 June, 2018; primary completion date: 2 September, 2020; study completion date: 7 October, 2020. NCT04345367; study start date: 11 June, 2020; primary completion date: 16 December, 2020; study completion date: 13 July, 2021. NCT03422822; study start date: 8 March, 2018; study completion date: ongoing (estimated completion date: 31 January, 2026).
Collapse
Affiliation(s)
- Eric L Simpson
- Department of Dermatology, Oregon Health and Science University, Portland, OR, USA
| | - Jonathan I Silverberg
- Department of Dermatology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Audrey Nosbaum
- Hospices Civils de Lyon, Allergologie et Immunologie Clinique, Centre Hospitalier Lyon Sud, Pierre-Bénite, France
| | - Kevin Winthrop
- Department of Dermatology, Oregon Health and Science University, Portland, OR, USA
| | | | - Karin M Hoffmeister
- Versiti, Translational Glycomics Center, Blood Research Institute, Milwaukee, WI, USA
- Medical College of Wisconsin, Milwaukee, WI, USA
| | - Alexander Egeberg
- Bispebjerg University Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Ryguła I, Pikiewicz W, Kaminiów K. Novel Janus Kinase Inhibitors in the Treatment of Dermatologic Conditions. Molecules 2023; 28:8064. [PMID: 38138551 PMCID: PMC10745734 DOI: 10.3390/molecules28248064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/10/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Janus kinase inhibitors, also known as JAK inhibitors, JAKinibs or JAKi, are a new group of disease-modifying drugs. They work by inhibiting enzymes involved in the transmission of information from receptors located in the cell membrane to the cell interior, specifically to the cell nucleus, thus disrupting the JAK-STAT pathway. This pathway plays a role in key cellular processes such as the immune response and cell growth. This feature is used in the treatment of patients with rheumatological, gastroenterological and hematological diseases. Recently, it has been discovered that JAK-STAT pathway inhibitors also show therapeutic potential against dermatological diseases such as atopic dermatitis, psoriasis, alopecia areata and acquired vitiligo. Studies are underway to use them in the treatment of several other dermatoses. Janus kinase inhibitors represent a promising class of drugs for the treatment of skin diseases refractory to conventional therapy. The purpose of this review is to summarize the latest knowledge on the use of JAKi in dermatological treatment.
Collapse
Affiliation(s)
- Izabella Ryguła
- Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland;
| | - Wojciech Pikiewicz
- Department of Medical and Health Sciences, Collegium Medicum—Faculty of Medicine, WSB University, 41-300 Dąbrowa Górnicza, Poland;
| | - Konrad Kaminiów
- Department of Medical and Health Sciences, Collegium Medicum—Faculty of Medicine, WSB University, 41-300 Dąbrowa Górnicza, Poland;
| |
Collapse
|
4
|
Traidl S, Harries L, Kienlin P, Begemann G, Roesner LM, Werfel T. Dupilumab strengthens herpes simplex virus type 1-specific immune responses in atopic dermatitis. J Allergy Clin Immunol 2023; 152:1460-1469.e5. [PMID: 37660986 DOI: 10.1016/j.jaci.2023.08.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/22/2023] [Accepted: 08/03/2023] [Indexed: 09/05/2023]
Abstract
BACKGROUND Impaired virus clearance in a subgroup of atopic dermatitis (AD) patients can lead to severe herpes simplex virus (HSV) infections called eczema herpeticum (EH). We recently identified a type 2 skewed viral immune response in EH patients. Clinical data suggest a reduced incidence of EH in AD patients treated with dupilumab, although immunologic investigations of this phenomenon are still lacking. OBJECTIVE We examined the impact of dupilumab on the HSV type 1 (HSV-1) specific immune response in AD, focusing on patients with (ADEH+) and without (ADEH-) a history of EH. METHODS Sera and peripheral blood mononuclear cells were collected from ADEH+ and ADEH- patients, a subgroup of whom was receiving dupilumab treatment, and healthy controls. Serum samples were tested for IgE against HSV-1 glycoprotein D (n = 85). Peripheral blood mononuclear cells were stimulated with HSV peptides, and activated CD4+ and CD8+ cells were characterized by flow cytometry after magnetic enrichment via CD154 or CD137 (n = 60). Cytokine production of HSV-1-reactive T-cell lines (n = 33) and MHC-I tetramer+ (HSV-1-UL25) CD8+ T cells was investigated by bead assay and intracellular cytokine staining (n = 21). RESULTS We confirmed that HSV-1-specific IgE is elevated in ADEH+ patients. During dupilumab treatment, the IgE levels were significantly decreased, reaching levels of healthy controls. HSV-1-specific TC1 frequencies were elevated in ADEH- patients treated with dupilumab compared to dupilumab-negative patients. There were no changes in the frequencies of HSV-1-specific TH cells while receiving dupilumab therapy. AD patients receiving dupilumab exhibited elevated IFN-γ and reduced IL-4 production in HSV-1-UL25-epitope-specific T cells compared to dupilumab-negative patients. CONCLUSION Dupilumab may improve the HSV-1-specific immune response in AD as a result of an increased type I immune response and a reduction of HSV-1-specific IgE.
Collapse
Affiliation(s)
- Stephan Traidl
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany.
| | - Leonard Harries
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Petra Kienlin
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Gabriele Begemann
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Lennart M Roesner
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Thomas Werfel
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
5
|
Simpson EL, Schlievert PM, Yoshida T, Lussier S, Boguniewicz M, Hata T, Fuxench Z, De Benedetto A, Ong PY, Ko J, Calatroni A, Rudman Spergel AK, Plaut M, Quataert SA, Kilgore SH, Peterson L, Gill AL, David G, Mosmann T, Gill SR, Leung DYM, Beck LA. Rapid reduction in Staphylococcus aureus in atopic dermatitis subjects following dupilumab treatment. J Allergy Clin Immunol 2023; 152:1179-1195. [PMID: 37315812 PMCID: PMC10716365 DOI: 10.1016/j.jaci.2023.05.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) is an inflammatory disorder characterized by dominant type 2 inflammation leading to chronic pruritic skin lesions, allergic comorbidities, and Staphylococcus aureus skin colonization and infections. S aureus is thought to play a role in AD severity. OBJECTIVES This study characterized the changes in the host-microbial interface in subjects with AD following type 2 blockade with dupilumab. METHODS Participants (n = 71) with moderate-severe AD were enrolled in a randomized (dupilumab vs placebo; 2:1), double-blind study at Atopic Dermatitis Research Network centers. Bioassays were performed at multiple time points: S aureus and virulence factor quantification, 16s ribosomal RNA microbiome, serum biomarkers, skin transcriptomic analyses, and peripheral blood T-cell phenotyping. RESULTS At baseline, 100% of participants were S aureus colonized on the skin surface. Dupilumab treatment resulted in significant reductions in S aureus after only 3 days (compared to placebo), which was 11 days before clinical improvement. Participants with the greatest S aureus reductions had the best clinical outcomes, and these reductions correlated with reductions in serum CCL17 and disease severity. Reductions (10-fold) in S aureus cytotoxins (day 7), perturbations in TH17-cell subsets (day 14), and increased expression of genes relevant for IL-17, neutrophil, and complement pathways (day 7) were also observed. CONCLUSIONS Blockade of IL-4 and IL-13 signaling, very rapidly (day 3) reduces S aureus abundance in subjects with AD, and this reduction correlates with reductions in the type 2 biomarker, CCL17, and measures of AD severity (excluding itch). Immunoprofiling and/or transcriptomics suggest a role for TH17 cells, neutrophils, and complement activation as potential mechanisms to explain these findings.
Collapse
Affiliation(s)
- Eric L Simpson
- Department of Dermatology, Oregon Health and Science University, Portland, Ore
| | | | - Takeshi Yoshida
- Department of Dermatology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | | | - Mark Boguniewicz
- Division of Allergy-Immunology, Department of Pediatrics, National Jewish Health and University of Colorado School of Medicine, Denver, Colo
| | - Tissa Hata
- Department of Dermatology, University of California, San Diego, Calif
| | - Zelma Fuxench
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pa
| | - Anna De Benedetto
- Department of Dermatology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Peck Y Ong
- Department of Pediatrics, University Southern California, Los Angeles, Calif
| | - Justin Ko
- Department of Dermatology, Stanford University, Stanford, Calif
| | | | - Amanda K Rudman Spergel
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Marshall Plaut
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Sally A Quataert
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY
| | - Samuel H Kilgore
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa
| | - Liam Peterson
- Department of Dermatology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Ann L Gill
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY
| | | | - Tim Mosmann
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY
| | - Steven R Gill
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY
| | - Donald Y M Leung
- Division of Allergy-Immunology, Department of Pediatrics, National Jewish Health and University of Colorado School of Medicine, Denver, Colo.
| | - Lisa A Beck
- Department of Dermatology, University of Rochester School of Medicine and Dentistry, Rochester, NY.
| |
Collapse
|
6
|
Inborn Errors of Immunity Predisposing to Herpes Simplex Virus Infections of the Central Nervous System. Pathogens 2023; 12:pathogens12020310. [PMID: 36839582 PMCID: PMC9961685 DOI: 10.3390/pathogens12020310] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023] Open
Abstract
Herpesvirus infections can lead to a number of severe clinical manifestations, particularly when involving the central nervous system (CNS), causing encephalitis and meningitis. However, understanding of the host factors conferring increased susceptibility to these diseases and their complications remains incomplete. Previous studies have uncovered defects in the innate Toll-like receptor 3 pathway and production of type I interferon (IFN-I) in children and adults that predispose them to herpes simplex encephalitis. More recently, there is accumulating evidence for an important role of IFN-independent cell-autonomous intrinsic mechanisms, including small nucleolar RNAs, RNA lariat metabolism, and autophagy, in restricting herpesvirus replication and conferring protection against CNS infection. The present review first describes clinical manifestations of HSV infection with a focus on neurological complications and then summarizes the host-pathogen interactions and innate immune pathways responsible for sensing herpesviruses and triggering antiviral responses and immunity. Next, we review the current landscape of inborn errors of immunity and the underlying genetic defects and disturbances of cellular immune pathways that confer increased susceptibility to HSV infection in CNS. Ultimately, we discuss some of the present outstanding unanswered questions relating to inborn errors of immunity and HSV CNS infection together with some perspectives and future directions for research in the pathogenesis of these severe diseases in humans.
Collapse
|
7
|
Broderick C, Ziehfreund S, van Bart K, Arents B, Eyerich K, Weidinger S, Rastrick J, Zink A, Flohr C. Biomarkers associated with the development of comorbidities in patients with atopic dermatitis: A systematic review. Allergy 2023; 78:84-120. [PMID: 36366871 PMCID: PMC10107168 DOI: 10.1111/all.15578] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 10/06/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
Biomarkers associated with the development of comorbidities in atopic dermatitis (AD) patients have been reported, but have not yet been systematically reviewed. Seven electronic databases were searched, from database inception to September 2021. English language randomized controlled trials, prospective and retrospective cohort, and case-control studies that investigated the association between a biomarker and the development of comorbidities in AD patients were included. Two authors independently screened the records for eligibility, one extracted all data, and critically appraised the quality of studies and risk of bias. Fifty six articles met the inclusion criteria, evaluating 146 candidate biomarkers. The most frequently reported biomarkers were filaggrin mutations and allergen specific-IgE. Promising biomarkers include specific-IgE and/or skin prick tests predicting the development of asthma, and genetic polymorphisms predicting the occurrence of eczema herpeticum. The identified studies and biomarkers were highly heterogeneous, and associated with predominately moderate-to-high risk of bias across multiple domains. Overall, findings were inconsistent. High-quality studies assessing biomarkers associated with the development of comorbidities in people with AD are lacking. Harmonized datasets and independent validation studies are urgently needed.
Collapse
Affiliation(s)
- Conor Broderick
- Unit for Population-Based Dermatology Research, School of Basic and Medical Biosciences, St John's Institute of Dermatology, King's College London, London, UK
| | - Stefanie Ziehfreund
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany
| | - Karin van Bart
- Royal College of Physicians, National Guideline Centre, London, UK
| | - Bernd Arents
- Dutch Association for People with Atopic Dermatitis, Nijkerk, The Netherlands
| | - Kilian Eyerich
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany.,Division of Dermatology and Venerology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Stephan Weidinger
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Alexander Zink
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany.,Division of Dermatology and Venerology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Carsten Flohr
- Unit for Population-Based Dermatology Research, School of Basic and Medical Biosciences, St John's Institute of Dermatology, King's College London, London, UK
| | | |
Collapse
|
8
|
Xu G, Gao Y, Pan T, Li S, Zhang Y, Guo J, Tian Z, Xu J, Li Y, Li X. Dynamic immune ecosystem of dengue infection revealed by single-cell sequencing. J Leukoc Biol 2022; 112:1621-1631. [PMID: 35766188 DOI: 10.1002/jlb.6ma0622-738rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 06/06/2022] [Indexed: 01/04/2023] Open
Abstract
Dengue is the most common human arboviral disease worldwide, which can result in severe complications. A dysfunctional immune response in dengue infective patients is a recurrent theme impacting symptoms and mortality, but the heterogeneity and dynamics of immune infiltrates during dengue infection remain poorly characterized. Here, we identified the immune cell types in scRNA-seq data from 13127 cells of 10 dengue infective patients and discovered the dynamic immune ecosystems of dengue infection. Notably, genes that exhibited higher expression in specific cell types play important roles in response to virus infection in a module manner. Transcription factors (TFs) are the major regulators (i.e., PAX5, IRF7, KLF4, and IRF8) that can potentially regulate infection-related genes. We demonstrated that the dynamic rewired regulatory network during dengue infection. Moreover, our data revealed the complex cell-cell communications from control to fever and severe dengue patients and prevalent cell-cell communication rewiring was observed. We further identified the IFN-II and CXCL signaling pathways that medicated the communications and play important roles in dengue infection. Together, our comprehensive analysis of dynamic immune ecosystem of dengue infection provided novel insights for understanding the pathogenesis of and developing effective therapeutic strategies for dengue infection.
Collapse
Affiliation(s)
- Gang Xu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China
| | - Yueying Gao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China
| | - Tao Pan
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China
| | - Si Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China
| | - Ya Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China
| | - Jing Guo
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China
| | - Zhanyu Tian
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China
| | - Juan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yongsheng Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China
| | - Xia Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China.,College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
9
|
Varzari A, Deyneko IV, Bruun GH, Dembic M, Hofmann W, Cebotari VM, Ginda SS, Andresen BS, Illig T. Candidate genes and sequence variants for susceptibility to mycobacterial infection identified by whole-exome sequencing. Front Genet 2022; 13:969895. [PMID: 36338958 PMCID: PMC9632272 DOI: 10.3389/fgene.2022.969895] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/04/2022] [Indexed: 11/13/2022] Open
Abstract
Inborn errors of immunity are known to influence susceptibility to mycobacterial infections. The aim of this study was to characterize the genetic profile of nine patients with mycobacterial infections (eight with BCGitis and one with disseminated tuberculosis) from the Republic of Moldova using whole-exome sequencing. In total, 12 variants in eight genes known to be associated with Mendelian Susceptibility to Mycobacterial Disease (MSMD) were detected in six out of nine patients examined. In particular, a novel splice site mutation c.373–2A>C in STAT1 gene was found and functionally confirmed in a patient with disseminated tuberculosis. Trio analysis was possible for seven out of nine patients, and resulted in 23 candidate variants in 15 novel genes. Four of these genes - GBP2, HEATR3, PPP1R9B and KDM6A were further prioritized, considering their elevated expression in immune-related tissues. Compound heterozygosity was found in GBP2 in a single patient, comprising a maternally inherited missense variant c.412G>A/p.(Ala138Thr) predicted to be deleterious and a paternally inherited intronic mutation c.1149+14T>C. Functional studies demonstrated that the intronic mutation affects splicing and the level of transcript. Finally, we analyzed pathogenicity of variant combinations in gene pairs and identified five patients with putative oligogenic inheritance. In summary, our study expands the spectrum of genetic variation contributing to susceptibility to mycobacterial infections in children and provides insight into the complex/oligogenic disease-causing mode.
Collapse
Affiliation(s)
- Alexander Varzari
- Laboratory of Human Genetics, Chiril Draganiuc Institute of Phthisiopneumology, Kishinev, Moldova
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
- *Correspondence: Alexander Varzari,
| | - Igor V. Deyneko
- Laboratory of Functional Genomics, Timiryazev Institute of Plant Physiology Russian Academy of Sciences, Moscow, Russia
| | - Gitte Hoffmann Bruun
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
- The Villum Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - Maja Dembic
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
- The Villum Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
| | - Winfried Hofmann
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Victor M. Cebotari
- Municipal Hospital of Phthisiopneumology, Department of Pediatrics, Kishinev, Moldova
| | - Sergei S. Ginda
- Laboratory of Immunology and Allergology, Chiril Draganiuc Institute of Phthisiopneumology, Kishinev, Moldova
| | - Brage S. Andresen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
- The Villum Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - Thomas Illig
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| |
Collapse
|
10
|
The Genetics of Eczema Herpeticum. Clin Rev Allergy Immunol 2022; 63:390-397. [DOI: 10.1007/s12016-022-08953-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2022] [Indexed: 11/03/2022]
|
11
|
Werfel T, Irvine AD, Bangert C, Seneschal J, Grond S, Cardillo T, Brinker D, Zhong J, Riedl E, Wollenberg A. An integrated analysis of herpes virus infections from eight randomised clinical studies of baricitinib in adults with moderate to severe atopic dermatitis. J Eur Acad Dermatol Venereol 2022; 36:1486-1496. [DOI: 10.1111/jdv.18193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/13/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Thomas Werfel
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy Hannover Medical School Hannover Germany
- Cluster of Excellence RESIST (EXC 2155) Hannover Medical School Hannover Germany
| | - Alan D. Irvine
- Department of Clinical Medicine Trinity College Dublin Ireland
| | - Christine Bangert
- Department of Dermatology Medical University of Vienna Vienna Austria
| | - Julien Seneschal
- Department of Dermatology and Pediatric Dermatology and National Reference Center for Rare Skin Disorders, Hôpital Saint‐André Bordeaux France
| | | | | | | | | | | | - Andreas Wollenberg
- Department of Dermatology and Allergy, University Hospital Ludwig Maximillian University Munich Germany
- Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Department of Dermatology Brussels Belgium
| |
Collapse
|
12
|
Smith KL, Dai D, Modi BP, Sara R, Garabedian E, Marsh RA, Puck J, Secord E, Sullivan KE, Turvey SE, Biggs CM, the USIDNET Consortium. Inborn Errors of Immunity Associated With Type 2 Inflammation in the USIDNET Registry. Front Immunol 2022; 13:831279. [PMID: 35273610 PMCID: PMC8902297 DOI: 10.3389/fimmu.2022.831279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/27/2022] [Indexed: 12/27/2022] Open
Abstract
Background Monogenic conditions that disrupt proper development and/or function of the immune system are termed inborn errors of immunity (IEIs), also known as primary immunodeficiencies. Patients with IEIs often suffer from other manifestations in addition to infection, and allergic inflammation is an increasingly recognized feature of these conditions. Methods We performed a retrospective analysis of IEIs presenting with allergic inflammation as reported in the USIDNET registry. Our inclusion criteria comprised of patients with a reported monogenic cause for IEI where reported lab eosinophil and/or IgE values were available for the patient prior to them receiving potentially curative therapy. Patients were excluded if we were unable to determine the defective gene underlying their IEI. Patients were classified as having eosinophilia or elevated IgE when their record included at least 1 eosinophil count or IgE value that was greater than the age stratified upper limit of normal. We compared the proportion of patients with eosinophilia or elevated IgE with the proportion of samples in a reference population that fall above the upper limit of normal (2.5%). Results The query submitted to the USIDNET registry identified 1409 patients meeting inclusion criteria with a monogenic cause for their IEI diagnosis, of which 975 had eosinophil counts and 645 had IgE levels obtained prior to transplantation or gene therapy that were available for analysis. Overall, 18.8% (183/975) of the patients evaluated from the USIDNET registry had eosinophilia and 20.9% (135/645) had an elevated IgE. IEIs caused by defects in 32 genes were found to be significantly associated with eosinophilia and/or an elevated IgE level, spanning 7 of the 10 IEI categories according to the International Union of Immunological Societies classification. Conclusion Type 2 inflammation manifesting as eosinophilia or elevated IgE is found in a broad range of IEIs in the USIDNET registry. Our findings suggest that allergic immune dysregulation may be more widespread in IEIs than previously reported.
Collapse
Affiliation(s)
- Kelsey L. Smith
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
- British Columbia (BC) Children’s Hospital, Vancouver, BC, Canada
| | - Darlene Dai
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
- British Columbia (BC) Children’s Hospital, Vancouver, BC, Canada
| | - Bhavi P. Modi
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
- British Columbia (BC) Children’s Hospital, Vancouver, BC, Canada
| | - Rahnuma Sara
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
- British Columbia (BC) Children’s Hospital, Vancouver, BC, Canada
| | - Elizabeth Garabedian
- National Human Genome Research Institute, Bethesda, MD, United States
- National Institutes of Health, Bethesda, MD, United States
| | - Rebecca A. Marsh
- Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, OH, United States
| | - Jennifer Puck
- Division of Allergy/Immunology and Blood and Marrow Transplantation, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | | | - Kathleen E. Sullivan
- Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Stuart E. Turvey
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
- British Columbia (BC) Children’s Hospital, Vancouver, BC, Canada
| | - Catherine M. Biggs
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
- British Columbia (BC) Children’s Hospital, Vancouver, BC, Canada
- St Paul’s Hospital, Vancouver, BC, Canada
| | | |
Collapse
|
13
|
Bosma AL, Ascott A, Iskandar R, Farquhar K, Matthewman J, Langendam MW, Mulick A, Abuabara K, Williams HC, Spuls PI, Langan SM, Middelkamp-Hup MA. Classifying atopic dermatitis: a systematic review of phenotypes and associated characteristics. J Eur Acad Dermatol Venereol 2022; 36:807-819. [PMID: 35170821 PMCID: PMC9307020 DOI: 10.1111/jdv.18008] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 12/10/2021] [Accepted: 01/18/2022] [Indexed: 11/28/2022]
Abstract
Atopic dermatitis is a heterogeneous disease, accompanied by a wide variation in disease presentation and the potential to identify many phenotypes that may be relevant for prognosis and treatment. We aimed to systematically review previously reported phenotypes of atopic dermatitis and any characteristics associated with them. Ovid EMBASE, Ovid MEDLINE and Web of Science were searched from inception till the 12th of February 2021 for studies attempting to classify atopic dermatitis. Primary outcomes are atopic dermatitis phenotypes and characteristics associated with them in subsequent analyses. A secondary outcome is the methodological approach used to derive them. In total, 8,511 records were found. By focusing only on certain clinical phenotypes, 186 studies were eligible for inclusion. The majority of studies were hospital-based (59%, 109/186) and cross-sectional (76%, 141/186). The number of included patients ranged from seven to 526,808. Data-driven approaches to identify phenotypes were only used in a minority of studies (7%, 13/186). Ninety-one studies (49%) investigated a phenotype based on disease severity. A phenotype based on disease trajectory, morphology and eczema herpeticum was investigated in 56 (30%), 22 (12%) and 11 (6%) studies, respectively. Thirty-six studies (19%) investigated morphological characteristics in other phenotypes. Investigated associated characteristics differed between studies. In conclusion, we present an overview of phenotype definitions used in literature for severity, trajectory, morphology and eczema herpeticum, including associated characteristics. There is a lack of uniform and consistent use of atopic dermatitis phenotypes across studies.
Collapse
Affiliation(s)
- A L Bosma
- Department of Dermatology, UMC, location Academic Medical Center, University of Amsterdam, Amsterdam Public Health, Infection and Immunity, Amsterdam, The Netherlands
| | - A Ascott
- Department of Dermatology, University Hospitals Sussex NHS Foundation Trust, Worthing, United Kingdom
| | - R Iskandar
- Faculty of Epidemiology and Population Health, School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - J Matthewman
- Department of Non-communicable disease epidemiology, School of Hygiene and Tropical Medicine, London, United Kingdom
| | - M W Langendam
- Department of Epidemiology and Data Science, UMC, location Amsterdam Medical Center, University of Amsterdam, Amsterdam Public Health research institute, Amsterdam, The Netherlands
| | - A Mulick
- Faculty of Epidemiology and Population Health, School of Hygiene and Tropical Medicine, London, United Kingdom
| | - K Abuabara
- Department of Dermatology, University of California San Francisco, United States
| | - H C Williams
- Centre of Evidence-Based Dermatology, University of Nottingham, United Kingdom
| | - P I Spuls
- Department of Dermatology, UMC, location Academic Medical Center, University of Amsterdam, Amsterdam Public Health, Infection and Immunity, Amsterdam, The Netherlands
| | - S M Langan
- Department of Dermatology, UMC, location Academic Medical Center, University of Amsterdam, Amsterdam Public Health, Infection and Immunity, Amsterdam, The Netherlands.,Faculty of Epidemiology and Population Health, School of Hygiene and Tropical Medicine, London, United Kingdom
| | - M A Middelkamp-Hup
- Department of Dermatology, UMC, location Academic Medical Center, University of Amsterdam, Amsterdam Public Health, Infection and Immunity, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Traidl S, Roesner L, Zeitvogel J, Werfel T. Eczema herpeticum in atopic dermatitis. Allergy 2021; 76:3017-3027. [PMID: 33844308 DOI: 10.1111/all.14853] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 03/29/2021] [Accepted: 04/02/2021] [Indexed: 02/06/2023]
Abstract
Atopic dermatitis (AD) is one of the most common chronic inflammatory skin diseases leading to pruritic skin lesions. A subset of AD patients exhibits a disseminated severe HSV infection called eczema herpeticum (EH) that can cause life-threatening complications. This review gives an overview of the clinical picture, and characteristics of the patients as well as the diagnosis and therapy of EH. A special focus lies on the pathophysiological hallmarks identified so far that predispose for EH. This aspect covers genetic aberrations, immunological changes, and environmental influences displaying a complex multifactorial situation, which is not completely understood. Type 2 skewing of virus-specific T cells in ADEH+ patients has been implicated in immune profile abnormalities, along with impaired functions of dendritic cells and natural killer cells. Furthermore, aberrations in interferon pathway-related genes such as IFNG and IFNGR1 have been identified to increase the risk of EH. IL-4, IL-25, and thymic stromal lymphopoietin (TSLP) are overexpressed in EH, whereas antimicrobial peptides like human β-defensins and LL-37 are reduced. Concerning the epidermal barrier, single nucleotide polymorphisms (SNPs) in skin barrier proteins such as filaggrin were identified in ADEH+ patients. A dysbalance of the skin microbiome also contributes to EH due to an increase of Staphylococcus aureus, which provides a supporting role to the viral infection via secreted toxins such as α-toxin. The risk of EH is reduced in AD patients treated with dupilumab. Further research is needed to identify and specifically target risk factors for EH in AD patients.
Collapse
Affiliation(s)
- Stephan Traidl
- Division of Immunodermatology and Allergy Research Department of Dermatology and Allergy Hannover Medical School Hannover Germany
- Cluster of Excellence RESIST (EXC 2155) Hannover Medical School Hannover Germany
| | - Lennart Roesner
- Division of Immunodermatology and Allergy Research Department of Dermatology and Allergy Hannover Medical School Hannover Germany
- Cluster of Excellence RESIST (EXC 2155) Hannover Medical School Hannover Germany
| | - Jana Zeitvogel
- Division of Immunodermatology and Allergy Research Department of Dermatology and Allergy Hannover Medical School Hannover Germany
- Cluster of Excellence RESIST (EXC 2155) Hannover Medical School Hannover Germany
| | - Thomas Werfel
- Division of Immunodermatology and Allergy Research Department of Dermatology and Allergy Hannover Medical School Hannover Germany
- Cluster of Excellence RESIST (EXC 2155) Hannover Medical School Hannover Germany
| |
Collapse
|
15
|
Multiethnic genome-wide and HLA association study of total serum IgE level. J Allergy Clin Immunol 2021; 148:1589-1595. [PMID: 34536413 PMCID: PMC8665111 DOI: 10.1016/j.jaci.2021.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/26/2021] [Accepted: 09/01/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Total serum IgE (tIgE) is an important intermediate phenotype of allergic disease. Whole genome genetic association studies across ancestries may identify important determinants of IgE. OBJECTIVE We aimed to increase understanding of genetic variants affecting tIgE production across the ancestry and allergic disease spectrum by leveraging data from the National Heart, Lung and Blood Institute Trans-Omics for Precision Medicine program; the Consortium on Asthma among African-ancestry Populations in the Americas (CAAPA); and the Atopic Dermatitis Research Network (N = 21,901). METHODS We performed genome-wide association within strata of study, disease, and ancestry groups, and we combined results via a meta-regression approach that models heterogeneity attributable to ancestry. We also tested for association between HLA alleles called from whole genome sequence data and tIgE, assessing replication of associations in HLA alleles called from genotype array data. RESULTS We identified 6 loci at genome-wide significance (P < 5 × 10-9), including 4 loci previously reported as genome-wide significant for tIgE, as well as new regions in chr11q13.5 and chr15q22.2, which were also identified in prior genome-wide association studies of atopic dermatitis and asthma. In the HLA allele association study, HLA-A∗02:01 was associated with decreased tIgE level (Pdiscovery = 2 × 10-4; Preplication = 5 × 10-4; Pdiscovery+replication = 4 × 10-7), and HLA-DQB1∗03:02 was strongly associated with decreased tIgE level in Hispanic/Latino ancestry populations (PHispanic/Latino discovery+replication = 8 × 10-8). CONCLUSION We performed the largest genome-wide association study and HLA association study of tIgE focused on ancestrally diverse populations and found several known tIgE and allergic disease loci that are relevant in non-European ancestry populations.
Collapse
|
16
|
Bin L, Malley C, Taylor P, Preethi Boorgula M, Chavan S, Daya M, Mathias M, Shankar G, Rafaels N, Vergara C, Potee J, Campbell M, Hanifin JM, Simpson E, Schneider LC, Gallo RL, Hata T, Paller AS, De Benedetto A, Beck LA, Ong PY, Guttman‐Yassky E, Richers B, Baraghoshi D, Ruczinski I, Barnes KC, Leung DYM, Mathias RA. Whole genome sequencing identifies novel genetic mutations in patients with eczema herpeticum. Allergy 2021; 76:2510-2523. [PMID: 33548076 DOI: 10.1111/all.14762] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 12/11/2020] [Accepted: 01/04/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Eczema herpeticum (EH) is a rare complication of atopic dermatitis (AD) caused by disseminated herpes simplex virus (HSV) infection. The role of rare and/or deleterious genetic variants in disease etiology is largely unknown. This study aimed to identify genes that harbor damaging genetic variants associated with HSV infection in AD with a history of recurrent eczema herpeticum (ADEH+). METHODS Whole genome sequencing (WGS) was performed on 49 recurrent ADEH+ (≥3 EH episodes), 491 AD without a history of eczema herpeticum (ADEH-) and 237 non-atopic control (NA) subjects. Variants were annotated, and a gene-based approach (SKAT-O) was used to identify genes harboring damaging genetic variants associated with ADEH+. Genes identified through WGS were studied for effects on HSV responses and keratinocyte differentiation. RESULTS Eight genes were identified in the comparison of recurrent ADEH+to ADEH-and NA subjects: SIDT2, CLEC7A, GSTZ1, TPSG1, SP110, RBBP8NL, TRIM15, and FRMD3. Silencing SIDT2 and RBBP8NL in normal human primary keratinocytes (NHPKs) led to significantly increased HSV-1 replication. SIDT2-silenced NHPKs had decreased gene expression of IFNk and IL1b in response to HSV-1 infection. RBBP8NL-silenced NHPKs had decreased gene expression of IFNk, but increased IL1b. Additionally, silencing SIDT2 and RBBP8NL also inhibited gene expression of keratinocyte differentiation markers keratin 10 (KRT10) and loricrin (LOR). CONCLUSION SIDT2 and RBBP8NL participate in keratinocyte's response to HSV-1 infection. SIDT2 and RBBP8NL also regulate expression of keratinocyte differentiation genes of KRT10 and LOR.
Collapse
Affiliation(s)
- Lianghua Bin
- Department of Pediatrics National Jewish Health Denver CO USA
| | - Claire Malley
- Division of Allergy and Clinical Immunology Johns Hopkins University Baltimore MD USA
| | - Patricia Taylor
- Department of Pediatrics National Jewish Health Denver CO USA
| | | | - Sameer Chavan
- Department of Medicine University of Colorado Aurora CO USA
| | - Michelle Daya
- Department of Medicine University of Colorado Aurora CO USA
| | - Malaika Mathias
- Division of Allergy and Clinical Immunology Johns Hopkins University Baltimore MD USA
| | - Gautam Shankar
- Division of Allergy and Clinical Immunology Johns Hopkins University Baltimore MD USA
| | | | | | | | | | | | - Eric Simpson
- Oregon Health & Science University Portland OR USA
| | | | - Richard L. Gallo
- Department of Dermatology University of California San Diego CA USA
| | - Tissa Hata
- Department of Dermatology University of California San Diego CA USA
| | - Amy S. Paller
- Northwestern University Feinberg School of Medicine Chicago IL USA
| | | | - Lisa A. Beck
- University of Rochester Medical Center Rochester NY USA
| | - Peck Y. Ong
- Children’s Hospital Los Angeles University of Southern California Los Angeles CA USA
| | | | | | | | - Ingo Ruczinski
- Department of Biostatistics Bloomberg School of Public Health Johns Hopkins University Baltimore MD USA
| | | | | | - Rasika A. Mathias
- Division of Allergy and Clinical Immunology Johns Hopkins University Baltimore MD USA
| |
Collapse
|
17
|
Brockman R, Leitenberger S. Review of Scabies Infestation and Selected Common Cutaneous Infections. Pediatr Rev 2021; 42:21-32. [PMID: 33386302 DOI: 10.1542/pir.2018-0305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Ross Brockman
- Department of Dermatology, Oregon Health and Science University, Portland, OR
| | - Sabra Leitenberger
- Department of Dermatology, Oregon Health and Science University, Portland, OR
| |
Collapse
|
18
|
Solimani F, Meier K, Ghoreschi K. Emerging Topical and Systemic JAK Inhibitors in Dermatology. Front Immunol 2019; 10:2847. [PMID: 31849996 PMCID: PMC6901833 DOI: 10.3389/fimmu.2019.02847] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 11/19/2019] [Indexed: 12/12/2022] Open
Abstract
Accumulating data on cellular and molecular pathways help to develop novel therapeutic strategies in skin inflammation and autoimmunity. Examples are psoriasis and atopic dermatitis, two clinically and immunologically well-defined disorders. Here, the elucidation of key pathogenic factors such as IL-17A/IL-23 on the one hand and IL-4/IL-13 on the other hand profoundly changed our therapeutic practice. The knowledge on intracellular pathways and governing factors is shifting our attention to new druggable molecules. Multiple cytokine receptors signal through Janus kinases (JAKs) and associated signal transducer and activators of transcription (STATs). Inhibition of JAKs can simultaneously block the function of multiple cytokines. Therefore, JAK inhibitors (JAKi) are emerging as a new class of drugs, which in dermatology can either be used systemically as oral drugs or locally in topical formulations. Inhibition of JAKs has been shown to be effective in various skin disorders. The first oral JAKi have been recently approved for the treatment of rheumatoid arthritis and psoriatic arthritis. Currently, multiple inhibitors of the JAK/STAT pathway are being investigated for skin diseases like alopecia areata, atopic dermatitis, dermatomyositis, graft-versus-host-disease, hidradenitis suppurativa, lichen planus, lupus erythematosus, psoriasis, and vitiligo. Here, we aim to discuss the immunological basis and current stage of development of JAKi in dermatology.
Collapse
Affiliation(s)
- Farzan Solimani
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Katharina Meier
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
19
|
Eichenfield LF, Bieber T, Beck LA, Simpson EL, Thaçi D, de Bruin-Weller M, Deleuran M, Silverberg JI, Ferrandiz C, Fölster-Holst R, Chen Z, Graham NMH, Pirozzi G, Akinlade B, Yancopoulos GD, Ardeleanu M. Infections in Dupilumab Clinical Trials in Atopic Dermatitis: A Comprehensive Pooled Analysis. Am J Clin Dermatol 2019; 20:443-456. [PMID: 31066001 PMCID: PMC6533236 DOI: 10.1007/s40257-019-00445-7] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Patients with moderate-to-severe atopic dermatitis (AD) have increased infection risk, including skin infections and systemic infections. Immunomodulators (e.g., anti-tumor necrosis factors, anti-interleukin [anti-IL]-23, anti-IL-17, Janus kinase inhibitors) increase risk of infections. Dupilumab (a monoclonal antibody blocking the shared receptor component for IL-4 and IL-13) is approved for inadequately controlled moderate-to-severe AD and for moderate-to-severe eosinophilic or oral corticosteroid-dependent asthma. OBJECTIVE The aim was to determine the impact of dupilumab on infection rates in patients with moderate-to-severe AD. METHODS This analysis pooled data from seven randomized, placebo-controlled dupilumab trials in adults with moderate-to-severe AD. Exposure-adjusted analyses assessed infection rates. RESULTS Of 2932 patients, 1091 received placebo, 1095 dupilumab 300 mg weekly, and 746 dupilumab 300 mg every 2 weeks. Treatment groups had similar infection rates overall per 100 patient-years (placebo, 155; dupilumab weekly, 150; dupilumab every 2 weeks, 156; dupilumab combined, 152), and similar non-skin infection rates. Serious/severe infections were reduced with dupilumab (risk ratio 0.43; p < 0.05), as were bacterial and other non-herpetic skin infections (risk ratio 0.44; p < 0.001). Although herpesviral infection rates overall were slightly higher with dupilumab than placebo, clinically important herpesviral infections (eczema herpeticum, herpes zoster) were less common with dupilumab (risk ratio 0.31; p < 0.01). Systemic anti-infective medication use was lower with dupilumab. CONCLUSIONS Dupilumab is associated with reduced risk of serious/severe infections and non-herpetic skin infections and does not increase overall infection rates versus placebo in patients with moderate-to-severe AD. CLINICALTRIALS. GOV IDENTIFIERS NCT01548404, NCT02210780, NCT01859988, NCT02277743, NCT02277769, NCT02260986, and NCT02755649.
Collapse
MESH Headings
- Adult
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal, Humanized
- Clinical Trials, Phase II as Topic
- Clinical Trials, Phase III as Topic
- Dermatitis, Atopic/complications
- Dermatitis, Atopic/diagnosis
- Dermatitis, Atopic/drug therapy
- Double-Blind Method
- Humans
- Incidence
- Injections, Subcutaneous
- Placebos/administration & dosage
- Placebos/adverse effects
- Randomized Controlled Trials as Topic
- Severity of Illness Index
- Skin Diseases, Infectious/epidemiology
- Skin Diseases, Infectious/etiology
- Skin Diseases, Infectious/prevention & control
- Treatment Outcome
Collapse
Affiliation(s)
- Lawrence F Eichenfield
- Departments of Dermatology and Pediatrics, University of California, San Diego, San Diego, CA, 92123, USA.
- Division of Pediatric and Adolescent Dermatology, Rady Children's Hospital, 3020 Children's Way, Mail Code 5092, San Diego, CA, 92123, USA.
| | - Thomas Bieber
- Department of Dermatology and Allergy, Christine Kühne-Center for Allergy Research and Education, University of Bonn, Bonn, Germany
| | - Lisa A Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY, USA
| | - Eric L Simpson
- Department of Dermatology, Oregon Health and Science University, Portland, OR, USA
| | - Diamant Thaçi
- Comprehensive Center for Inflammation Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | | | - Mette Deleuran
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Carlos Ferrandiz
- Servicio de Dermatología, Hospital Universitario Germans Trias i Pujol, Badalona Universidad Autónoma de Barcelona, Barcelona, Spain
| | | | - Zhen Chen
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | | | | | | | | |
Collapse
|
20
|
Brunner PM. Early immunologic changes during the onset of atopic dermatitis. Ann Allergy Asthma Immunol 2019; 123:152-157. [PMID: 30953783 DOI: 10.1016/j.anai.2019.03.033] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/18/2019] [Accepted: 03/29/2019] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Atopic dermatitis (AD), which is commonly called eczema, is the most common chronic inflammatory skin disease. The pipeline of new targeted treatments is currently expanding, a development that is largely based on our increasing understanding of disease mechanisms. Mechanistic insights have long been based on long-standing adult AD. Recently, studies also investigated early pediatric AD at disease onset, and revealed several differences in barrier and immune properties when compared with long-standing adult AD. This review focuses on immunological changes very early in life that predispose to the development of AD, and summarizes characteristics of the molecular AD phenotype in this age group. DATA SOURCES Review of published literature. STUDY SELECTIONS Studies investigating human AD at disease onset in newborns, toddlers, and young children, in comparison with adults with long-standing disease. RESULTS Already in cord blood, increased Th2 and decreased Th1 levels were found to increase the risk of AD development. Both pediatric and adult AD share Th2/Th22 activation and defects in lipid barrier deposition and tight junction formation, but Th1 activation and epidermal differentiation complex defects are largely absent in pediatric AD. CONCLUSION Immune changes predisposing to AD development are present very early in life. During the first months of disease, AD shows various differences in immune and barrier properties from long-standing adult AD, which might necessitate tailored treatment approaches depending on the age of the patient.
Collapse
|
21
|
Hailey-Hailey Disease With Coexistent Herpes Virus Infection: Insights Into the Diagnostic Conundrum of Herpetic/Pseudoherpetic Features in Cutaneous Acantholytic Disorders. Am J Dermatopathol 2019; 40:749-753. [PMID: 28475517 DOI: 10.1097/dad.0000000000000902] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The specific histopathologic diagnosis of a primary acantholytic disorder takes into account the distribution and extent of acantholysis, presence or absence of dyskeratosis, nature of the dermal inflammatory cell infiltrate, and immunofluorescence findings. Herpes virus infection is a common cause of secondary acantholysis where distinctive viral cytopathic changes aid in making it a clear-cut diagnosis in majority of cases. We present a case of coexistence of Hailey-Hailey disease and herpes simplex virus infection to compare and contrast their histopathologic features. This is imperative because acantholytic cells from primary acantholytic disorders may occasionally show cytological features traditionally associated with herpes virus infection (pseudoherpetic changes). The objective of this article is to create a greater awareness of pseudoherpetic changes and also to explore the clinical significance of coexistence of a primary acantholytic disorder and herpes virus infection, as in this case.
Collapse
|
22
|
Progress in Understanding Atopic Dermatitis. J Invest Dermatol 2018; 138:e93-e95. [PMID: 30466540 DOI: 10.1016/j.jid.2018.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 10/02/2018] [Indexed: 01/20/2023]
|
23
|
Guan WJ, Li JC, Liu F, Zhou J, Liu YP, Ling C, Gao YH, Li HM, Yuan JJ, Huang Y, Chen CL, Chen RC, Zhang X, Zhong NS. Next-generation sequencing for identifying genetic mutations in adults with bronchiectasis. J Thorac Dis 2018; 10:2618-2630. [PMID: 29997923 PMCID: PMC6006054 DOI: 10.21037/jtd.2018.04.134] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 04/19/2018] [Indexed: 11/06/2022]
Abstract
BACKGROUND Defective airway host-defense (e.g., altered mucus properties, ciliary defects) contributes to the pathogenesis of bronchiectasis. This study aims to determine whether genetic mutations associated with defective airway host-defense are implicated in the pathogenesis of bronchiectasis. METHODS Based on the systematic screening of 32 frequently reported bronchiectasis-associated genes, we performed next-generation sequencing (NGS) on peripheral blood samples from 192 bronchiectasis patients and 100 healthy subjects. The variant distribution frequency and pathogenicity of mutations were analyzed. RESULTS We identified 162 rare variants in 192 bronchiectasis patients, and 85 rare variants among 100 healthy subjects. Among bronchiectasis patients, 25 (15.4%), 117 (72.2%) and 18 (11.1%) rare variants were associated with cystic fibrosis transmembrane receptor (CFTR), epithelial sodium channel, and primary ciliary dyskinesia genes, respectively. Biallelic CFTR variants were detected in four bronchiectasis patients but none of the healthy subjects. Carriers of homozygous p.M470 plus at least one CFTR rare variant were detected in 6.3% of bronchiectasis patients (n=12) and in 1.0% of healthy subjects (n=1, P=0.039). Twenty-six patients (16 with idiopathic and 6 with post-infectious bronchiectasis) harbored biallelic variants. Bronchiectasis patients with biallelic DNAH5 variants, or biallelic CFTR variants plus an epithelial sodium channel variant, tended to have greater disease severity. CONCLUSIONS Genetic mutations leading to impaired host-defense might have implicated in the pathogenesis of bronchiectasis. Genetic screening may be a useful tool for unraveling the underlying causes of bronchiectasis, and offers molecular information which is complementary to conventional etiologic assessment for bronchiectasis.
Collapse
Affiliation(s)
- Wei-Jie Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou 510120, China
| | - Jia-Cheng Li
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Fang Liu
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Jian Zhou
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Ya-Ping Liu
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Chao Ling
- Laboratory of Clinical Genetics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yong-Hua Gao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hui-Min Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
| | - Jing-Jing Yuan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
| | - Yan Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
| | - Chun-Lan Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
| | - Rong-Chang Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
| | - Xue Zhang
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Nan-Shan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
| |
Collapse
|
24
|
Abstract
Barrier sites such as the skin play a critical role in immune defense. They must maintain homeostasis with commensals and rapidly detect and limit pathogen invasion. This is accomplished in part through the production of endogenous antimicrobial peptides and proteins, which can be either constitutive or inducible. Here, we focus particularly on the control of innate antiviral proteins and present the basic aspects of their regulation in the skin by interferons (IFNs), IFN-independent immunity, and environmental factors. We also discuss the activity and (dys-)function of antiviral proteins in the context of skin-tropic viruses and highlight the relevance of the innate antiviral pathway as a potential therapeutic avenue for vulnerable patient populations and skin diseases with high risk for virus infections.
Collapse
|
25
|
Khanolkar A, Kirschmann DA, Caparelli EA, Wilks JD, Cerullo JM, Bergerson JRE, Jennings LJ, Fuleihan RL. CD4 T cell-restricted IL-2 signaling defect in a patient with a novel IFNGR1 deficiency. J Allergy Clin Immunol 2018; 141:435-439.e7. [PMID: 28927822 DOI: 10.1016/j.jaci.2017.08.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 08/08/2017] [Accepted: 08/21/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Aaruni Khanolkar
- Department of Pathology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill; Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Ill; Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill.
| | - Dawn A Kirschmann
- Department of Pathology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill
| | - Edward A Caparelli
- Department of Pathology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill
| | - Jeffrey D Wilks
- Department of Pathology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill
| | - Jillian M Cerullo
- Department of Pathology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill
| | - Jenna R E Bergerson
- Division of Allergy and Immunology, Ann and Robert H. Lurie Children's Hospital of Chicago, Feinberg School of Medicine, Chicago, Ill; Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill
| | - Lawrence J Jennings
- Department of Pathology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill; Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Ramsay L Fuleihan
- Division of Allergy and Immunology, Ann and Robert H. Lurie Children's Hospital of Chicago, Feinberg School of Medicine, Chicago, Ill; Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill
| |
Collapse
|
26
|
Yazd NKK, Patel RR, Dellavalle RP, Dunnick CA. Genetic Risk Factors for Development of Atopic Dermatitis: a Systematic Review. CURRENT DERMATOLOGY REPORTS 2017. [DOI: 10.1007/s13671-017-0199-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
27
|
van de Vosse E, van Dissel JT. IFN-γR1 defects: Mutation update and description of the IFNGR1 variation database. Hum Mutat 2017; 38:1286-1296. [PMID: 28744922 DOI: 10.1002/humu.23302] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/20/2017] [Accepted: 07/23/2017] [Indexed: 12/29/2022]
Abstract
IFN-γ signaling is essential for the innate immune defense against mycobacterial infections. IFN-γ signals through the IFN-γ receptor, which consists of a tetramer of two IFN-γR1 chains in complex with two IFN-γR2 chains, where IFN-γR1 is the ligand-binding chain of the interferon-γ receptor and IFN-γR2 is the signal-transducing chain of the IFN-γ receptor. Germline mutations in the gene IFNGR1 encoding the IFN-γR1 cause a primary immunodeficiency that mainly leads to mycobacterial infections. Here, we review the molecular basis of this immunodeficiency in the 130 individuals described to date, and report mutations in five new individuals, bringing the total number to 135 individuals from 98 kindreds. Forty unique IFNGR1 mutations have been reported and they exert either an autosomal dominant or an autosomal recessive effect. Mutations resulting in premature stopcodons represent the majority of IFNGR1 mutations (60%; 24 out of 40), followed by amino acid substitutions (28%, 11 out of 40). All known mutations, as well as 287 other variations, have been deposited in the online IFNGR1 variation database (www.LOVD.nl/IFNGR1). In this article, we review the function of IFN-γR1 and molecular genetics of human IFNGR1.
Collapse
Affiliation(s)
- Esther van de Vosse
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Jaap T van Dissel
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
28
|
Ma CA, Stinson JR, Zhang Y, Abbott JK, Weinreich MA, Hauk PJ, Reynolds PR, Lyons JJ, Nelson CG, Ruffo E, Dorjbal B, Glauzy S, Yamakawa N, Arjunaraja S, Voss K, Stoddard J, Niemela J, Zhang Y, Rosenzweig SD, McElwee JJ, DiMaggio T, Matthews HF, Jones N, Stone KD, Palma A, Oleastro M, Prieto E, Bernasconi AR, Dubra G, Danielian S, Zaiat J, Marti MA, Kim B, Cooper MA, Romberg N, Meffre E, Gelfand EW, Snow AL, Milner JD. Germline hypomorphic CARD11 mutations in severe atopic disease. Nat Genet 2017; 49:1192-1201. [PMID: 28628108 PMCID: PMC5664152 DOI: 10.1038/ng.3898] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 05/18/2017] [Indexed: 12/13/2022]
Abstract
Few monogenic causes for severe manifestations of common allergic diseases have been identified. Through next-generation sequencing on a cohort of patients with severe atopic dermatitis with and without comorbid infections, we found eight individuals, from four families, with novel heterozygous mutations in CARD11, which encodes a scaffolding protein involved in lymphocyte receptor signaling. Disease improved over time in most patients. Transfection of mutant CARD11 expression constructs into T cell lines demonstrated both loss-of-function and dominant-interfering activity upon antigen receptor-induced activation of nuclear factor-κB and mammalian target of rapamycin complex 1 (mTORC1). Patient T cells had similar defects, as well as low production of the cytokine interferon-γ (IFN-γ). The mTORC1 and IFN-γ production defects were partially rescued by supplementation with glutamine, which requires CARD11 for import into T cells. Our findings indicate that a single hypomorphic mutation in CARD11 can cause potentially correctable cellular defects that lead to atopic dermatitis.
Collapse
Affiliation(s)
- Chi A Ma
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jeffrey R Stinson
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Yuan Zhang
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jordan K Abbott
- Immunodeficiency Diagnosis and Treatment Program, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Michael A Weinreich
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Pia J Hauk
- Immunodeficiency Diagnosis and Treatment Program, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Paul R Reynolds
- Immunodeficiency Diagnosis and Treatment Program, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Jonathan J Lyons
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Celeste G Nelson
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Elisa Ruffo
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Batsukh Dorjbal
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Salomé Glauzy
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Natsuko Yamakawa
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Swadhinya Arjunaraja
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Kelsey Voss
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Jennifer Stoddard
- Immunology Service, Department of Laboratory Medicine, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Julie Niemela
- Immunology Service, Department of Laboratory Medicine, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Yu Zhang
- Human Immunological Disease Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sergio D Rosenzweig
- Immunology Service, Department of Laboratory Medicine, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Joshua J McElwee
- Merck Research Laboratories, Merck and Co., Inc., Boston, Massachusetts, USA
| | - Thomas DiMaggio
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Helen F Matthews
- Human Immunological Disease Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Nina Jones
- Clinical Research Directorate/Clinical Monitoring Research Program, Leidos Biomedical Research, Inc., NCI Campus at Frederick, Frederick, Maryland, USA
| | - Kelly D Stone
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Alejandro Palma
- Servicio de Immunología y Reumatología, Hospital Nacional de Pediatría Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Matías Oleastro
- Servicio de Immunología y Reumatología, Hospital Nacional de Pediatría Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Emma Prieto
- Servicio de Immunología y Reumatología, Hospital Nacional de Pediatría Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Andrea R Bernasconi
- Servicio de Immunología y Reumatología, Hospital Nacional de Pediatría Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Geronimo Dubra
- Servicio de Immunología y Reumatología, Hospital Nacional de Pediatría Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Silvia Danielian
- Servicio de Immunología y Reumatología, Hospital Nacional de Pediatría Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Jonathan Zaiat
- Servicio de Immunología y Reumatología, Hospital Nacional de Pediatría Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Marcelo A Marti
- Servicio de Immunología y Reumatología, Hospital Nacional de Pediatría Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Brian Kim
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Megan A Cooper
- Department of Pediatrics, Division of Rheumatology and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Neil Romberg
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Eric Meffre
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Erwin W Gelfand
- Immunodeficiency Diagnosis and Treatment Program, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Andrew L Snow
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Joshua D Milner
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
29
|
Rowe RK, Gill MA. Effects of Allergic Sensitization on Antiviral Immunity: Allergen, Virus, and Host Cell Mechanisms. Curr Allergy Asthma Rep 2017; 17:9. [PMID: 28233152 DOI: 10.1007/s11882-017-0677-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Multiple clinical and epidemiological studies demonstrate links between allergic sensitization and virus-induced atopic disease exacerbations. This review summarizes the recent findings regarding allergen, viral, and host cellular mechanisms relevant to these observations. RECENT FINDINGS Recent studies have focused on the molecular pathways and genetic influences involved in allergen-mediated inhibition of innate antiviral immune responses. Multiple tissue and cell types from atopic individuals across the atopy spectrum exhibit deficient interferon responses to a variety of virus infections. Impairment in barrier function, viral RNA and DNA recognition by intracellular sensing molecules, and dysregulation of signaling components are broadly affected by allergic sensitization. Finally, genetic predisposition by numerous nucleotide polymorphisms also impacts immune pathways and potentially contributes to virus-associated atopic disease pathogenesis. Allergen-virus interactions in the setting of atopy involve complex tissue and cellular mechanisms. Future studies defining the pathways underlying these interactions could uncover potential therapeutic targets. Available data suggest that therapies tailored to restore specific components of antiviral responses will likely lead to improved clinical outcomes in allergic disease.
Collapse
Affiliation(s)
- Regina K Rowe
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9063, USA
| | - Michelle A Gill
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9063, USA. .,Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
30
|
Nomura T, Kabashima K. Advances in atopic dermatitis in 2015. J Allergy Clin Immunol 2017; 138:1548-1555. [PMID: 27931536 DOI: 10.1016/j.jaci.2016.10.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/31/2016] [Accepted: 10/12/2016] [Indexed: 12/15/2022]
Abstract
This review aims to highlight recently published articles on atopic dermatitis (AD). Updated are the insights into epidemiology, pathology, diagnostics, and therapy. Epidemiologic studies have revealed a positive correlation between AD and systemic conditions, such as rheumatoid arthritis, inflammatory bowel disease, and neonatal adiposity. Pathologic findings highlight the involvement of novel barrier factors (desmoplakin and claudin), novel immune cell subsets (pathogenic effector TH2 cells and group 2 innate lymphoid cells), and differential skewing of helper T cells (eg, TH17 dominance in Asians with AD). As diagnostics, noninvasive examinations of the transepidermal water loss of neonates, the density of epidermal Staphylococcus species, and the gut flora might prognosticate the onset of AD. As for therapy, various methods are proposed, including conventional (petrolatum and UV) and molecule-oriented regimens targeting Janus kinase, signal transducer and activator of transcription 3, extracellular signal-regulated kinase, sirtuin 1, or aryl hydrocarbon receptor.
Collapse
Affiliation(s)
- Takashi Nomura
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Experimental Therapeutics, Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan.
| | - Kenji Kabashima
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Singapore Immunology Network (SIgN) and Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore; PRESTO, Japan Science and Technology Agency, Saitama, Japan
| |
Collapse
|
31
|
Welsch K, Holstein J, Laurence A, Ghoreschi K. Targeting JAK/STAT signalling in inflammatory skin diseases with small molecule inhibitors. Eur J Immunol 2017; 47:1096-1107. [DOI: 10.1002/eji.201646680] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/08/2017] [Accepted: 05/24/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Katharina Welsch
- Department of Dermatology; University Medical Center, Eberhard Karls University Tübingen; Germany
| | - Julia Holstein
- Department of Dermatology; University Medical Center, Eberhard Karls University Tübingen; Germany
| | - Arian Laurence
- Department of Haemato-Oncology, Northern Centre for Cancer Care; Newcastle University; UK
| | - Kamran Ghoreschi
- Department of Dermatology; University Medical Center, Eberhard Karls University Tübingen; Germany
| |
Collapse
|
32
|
Huang YJ, Marsland BJ, Bunyavanich S, O'Mahony L, Leung DYM, Muraro A, Fleisher TA. The microbiome in allergic disease: Current understanding and future opportunities-2017 PRACTALL document of the American Academy of Allergy, Asthma & Immunology and the European Academy of Allergy and Clinical Immunology. J Allergy Clin Immunol 2017; 139:1099-1110. [PMID: 28257972 PMCID: PMC5899886 DOI: 10.1016/j.jaci.2017.02.007] [Citation(s) in RCA: 223] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/02/2017] [Accepted: 02/07/2017] [Indexed: 12/15/2022]
Abstract
PRACTALL is a joint initiative of the American Academy of Allergy, Asthma & Immunology and the European Academy of Allergy and Clinical Immunology to provide shared evidence-based recommendations on cutting-edge topics in the field of allergy and immunology. PRACTALL 2017 is focused on what has been established regarding the role of the microbiome in patients with asthma, atopic dermatitis, and food allergy. This is complemented by outlining important knowledge gaps regarding its role in allergic disease and delineating strategies necessary to fill these gaps. In addition, a review of progress in approaches used to manipulate the microbiome will be addressed, identifying what has and has not worked to serve as a baseline for future directions to intervene in allergic disease development, progression, or both.
Collapse
Affiliation(s)
- Yvonne J Huang
- Division of Pulmonary and Critical Care, Department of Internal Medicine, University of Michigan, Ann Arbor, Mich
| | - Benjamin J Marsland
- Service de Pneumologie, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Supinda Bunyavanich
- Division of Allergy and Immunology, Departments of Pediatrics and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Liam O'Mahony
- Molecular Immunology, Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland
| | - Donald Y M Leung
- Division of Allergy and Immunology, Department of Pediatrics, National Jewish Health, Denver, Colo
| | - Antonella Muraro
- Food Allergy Referral Centre Veneto Region, Department of Women and Child Health, Padua General University Hospital, Padua, Italy
| | - Thomas A Fleisher
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Md.
| |
Collapse
|
33
|
Temporal and Racial Differences Associated with Atopic Dermatitis Staphylococcusaureus and Encoded Virulence Factors. mSphere 2016; 1:mSphere00295-16. [PMID: 27981233 PMCID: PMC5143412 DOI: 10.1128/msphere.00295-16] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 11/18/2016] [Indexed: 11/25/2022] Open
Abstract
Monitoring pathogen emergence provides insight into how pathogens adapt in the human population. Secreted virulence factors, important contributors to infections, may differ in a manner dependent on the strain and host. Temporal changes of Staphylococcus aureus toxigenic potential, for example, in encoding toxic shock syndrome toxin 1 (TSST-1), contributed to an epidemic of TSS with significant health impact. This study monitored changes in atopic dermatitis (AD) S. aureus isolates and demonstrated both temporal and host infection differences according to host race based on secreted superantigen potential. The current temporal increase in enterotoxin gene cluster superantigen prevalence and lack of the gene encoding TSST-1 in AAs predict differences in infection types and presentations. Atopic dermatitis (AD) is an inflammatory skin condition strongly associated with Staphylococcus aureus colonization and infection. S. aureus strains shift in populations in ~10-year intervals depending on virulence factors. Shifts in S. aureus virulence factors may in part explain the racial differences observed in the levels of prevalence and severity of AD. AD S. aureus isolates collected from 2011 to 2014 (103 isolates) and in 2008 (100 isolates) were examined for the prevalence of genes encoding superantigens (SAgs). The strains from 2011 to 2014 were obtained from AD patients as a part of the National Institute of Allergy and Infectious Diseases (NIAID) Atopic Dermatitis Research Network (ADRN). The prevalence of SAg genes was investigated temporally and racially. The enterotoxin gene cluster (EGC) was more prevalent in the 2011–2014 AD isolates than in the 2008 AD isolates. The prevalences of virulence factor genes were similar in European American (EA) and Mexican American (MA) patients but differed in 6 of 22 SAg genes between EA and African American (AA) or MA and AA isolates; notably, AA isolates lacked tstH, the gene encoding toxic shock syndrome toxin 1 (TSST-1). The presence of tstH and sel-p (enterotoxin-like P) was associated with decreased clinical severity and increased blood eosinophils, respectively. The EGC is becoming more prevalent, consistent with the previously observed 10 years of cycling of S. aureus strains. Race-specific S. aureus selection may account for differences in virulence factor profiles. The lack of TSST-1-positive (TSST-1+) AD S. aureus in AA is consistent with the lack of AAs acquiring TSST-1-associated menstrual toxic shock syndrome (TSS). IMPORTANCE Monitoring pathogen emergence provides insight into how pathogens adapt in the human population. Secreted virulence factors, important contributors to infections, may differ in a manner dependent on the strain and host. Temporal changes of Staphylococcus aureus toxigenic potential, for example, in encoding toxic shock syndrome toxin 1 (TSST-1), contributed to an epidemic of TSS with significant health impact. This study monitored changes in atopic dermatitis (AD) S. aureus isolates and demonstrated both temporal and host infection differences according to host race based on secreted superantigen potential. The current temporal increase in enterotoxin gene cluster superantigen prevalence and lack of the gene encoding TSST-1 in AAs predict differences in infection types and presentations.
Collapse
|
34
|
Interleukins (from IL-1 to IL-38), interferons, transforming growth factor β, and TNF-α: Receptors, functions, and roles in diseases. J Allergy Clin Immunol 2016; 138:984-1010. [DOI: 10.1016/j.jaci.2016.06.033] [Citation(s) in RCA: 450] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 12/25/2022]
|
35
|
Abstract
PURPOSE OF REVIEW The review will examine recent advances in our understanding of atopic dermatitis and how these mechanisms provide a framework for new approaches to the management of this common skin disease. RECENT FINDINGS The mechanisms by which epithelial skin barrier and immune responses contribute to the complex clinical phenotypes found in atopic dermatitis are being elucidated. Atopic dermatitis often precedes food allergy because reduced skin barrier function allows environmental food allergens to penetrate the skin leading to systemic allergen sensitization. There is increasing evidence that atopic dermatitis is a systemic disease. New treatments are focused on intervention in polarized immune responses leading to allergic diseases. This includes antagonism of IL-4 and IL-13 effects. Prevention strategies involve maintaining normal skin barrier function with emollients to prevent allergens and microbes from penetrating the skin. SUMMARY Recent work on the pathogenesis of atopic dermatitis has important implications for its clinical management, including the development of effective barrier creams and biologicals targeting specific polarized immune pathways resulting in skin inflammation.
Collapse
Affiliation(s)
- Donald Y M Leung
- Edelstein Family Chair of Pediatric Allergy-Immunology, National Jewish Health, Denver, Colorado, USA and Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The 2nd Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
36
|
Molecular Mechanisms of Cutaneous Inflammatory Disorder: Atopic Dermatitis. Int J Mol Sci 2016; 17:ijms17081234. [PMID: 27483258 PMCID: PMC5000632 DOI: 10.3390/ijms17081234] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/21/2016] [Accepted: 07/26/2016] [Indexed: 02/06/2023] Open
Abstract
Atopic dermatitis (AD) is a multifactorial inflammatory skin disease resulting from interactions between genetic susceptibility and environmental factors. The pathogenesis of AD is poorly understood, and the treatment of recalcitrant AD is still challenging. There is accumulating evidence for new gene polymorphisms related to the epidermal barrier function and innate and adaptive immunity in patients with AD. Newly-found T cells and dendritic cell subsets, cytokines, chemokines and signaling pathways have extended our understanding of the molecular pathomechanism underlying AD. Genetic changes caused by environmental factors have been shown to contribute to the pathogenesis of AD. We herein present a review of the genetics, epigenetics, barrier dysfunction and immunological abnormalities in AD with a focus on updated molecular biology.
Collapse
|
37
|
Wawrzyniak P, Akdis CA, Finkelman FD, Rothenberg ME. Advances and highlights in mechanisms of allergic disease in 2015. J Allergy Clin Immunol 2016; 137:1681-1696. [PMID: 27090934 DOI: 10.1016/j.jaci.2016.02.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 02/03/2016] [Accepted: 02/17/2016] [Indexed: 01/08/2023]
Abstract
This review highlights some of the advances in mechanisms of allergic disease, particularly anaphylaxis, including food allergy, drug hypersensitivity, atopic dermatitis (AD), allergic conjunctivitis, and airway diseases. During the last year, a mechanistic advance in food allergy was achieved by focusing on mechanisms of allergen sensitization. Novel biomarkers and treatment for mastocytosis were presented in several studies. Novel therapeutic approaches in the treatment of atopic dermatitis and psoriasis showed that promising supplementation of the infant's diet in the first year of life with immunoactive prebiotics might have a preventive role against early development of AD and that therapeutic approaches to treat AD in children might be best directed to the correction of a TH2/TH1 imbalance. Several studies were published emphasizing the role of the epithelial barrier in patients with allergic diseases. An impaired skin barrier as a cause for sensitization to food allergens in children and its relationship to filaggrin mutations has been an important development. Numerous studies presented new approaches for improvement of epithelial barrier function and novel biologicals used in the treatment of inflammatory skin and eosinophilic diseases. In addition, novel transcription factors and signaling molecules that can develop as new possible therapeutic targets have been reported.
Collapse
Affiliation(s)
- Paulina Wawrzyniak
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland, Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland, Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland.
| | - Fred D Finkelman
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, University of Cincinnati College of Medicine, and the Department of Medicine, Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
38
|
Brar K, Leung DYM. Recent considerations in the use of recombinant interferon gamma for biological therapy of atopic dermatitis. Expert Opin Biol Ther 2016; 16:507-14. [PMID: 26694988 DOI: 10.1517/14712598.2016.1135898] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Atopic dermatitis (AD) is the most common inflammatory skin disease in the general population. There are different endophenotypes of AD that likely have a unique immune and molecular basis, such as those who are predisposed to eczema herpeticum, or Staphylococcus aureus infections. AREAS COVERED In this review, we highlight the endophenotypes of AD where reduced interferon gamma expression may be playing a role. Additionally, we review the potential role of recombinant interferon gamma therapy in the treatment of atopic dermatitis and the particular phenotypes that may benefit from this treatment. EXPERT OPINION Recombinant interferon gamma treatment will likely benefit the pediatric population with AD, as well as those with susceptibilities for skin infections. Future studies are needed to elucidate whether IFN-γ may reduce the prevalence of skin infection in AD.
Collapse
Affiliation(s)
- Kanwaljit Brar
- a Division of Pediatric Allergy-Immunology, Department of Pediatrics , National Jewish Health , Denver , CO , USA
| | - Donald Y M Leung
- a Division of Pediatric Allergy-Immunology, Department of Pediatrics , National Jewish Health , Denver , CO , USA.,b Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology , The State Key Clinical Specialty in Allergy, The Second Affiliated Hospital of Guangzhou Medical University , Guangzhou , China
| |
Collapse
|