1
|
Vidal-Quist JC, Ortego F, Rombauts S, Hernández-Crespo P. The genome-wide response of Dermatophagoides pteronyssinus to cystatin A, a peptidase inhibitor from human skin, sheds light on its digestive physiology and allergenicity. INSECT MOLECULAR BIOLOGY 2024; 33:662-677. [PMID: 38878274 DOI: 10.1111/imb.12931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/19/2024] [Indexed: 11/06/2024]
Abstract
The digestive physiology of house dust mites (HDMs) is particularly relevant for their allergenicity since many of their allergens participate in digestion and are excreted into faecal pellets, a main source of exposure for allergic subjects. To gain insight into the mite dietary digestion, the genome of the HDM Dermatophagoides pteronyssinus was screened for genes encoding peptidases (n = 320), glycosylases (n = 77), lipases and esterases (n = 320), peptidase inhibitors (n = 65) and allergen-related proteins (n = 52). Basal gene expression and transcriptional responses of mites to dietary cystatin A, a cysteine endopeptidase inhibitor with previously shown antinutritional effect on mites, were analysed by RNAseq. The ingestion of cystatin A resulted in significant regulation of different cysteine endopeptidase and glycosylase genes. One Der p 1-like and two cathepsin B-like cysteine endopeptidase genes of high basal expression were induced, which suggests their prominent role in proteolytic digestion together with major allergen Der p 1. A number of genes putatively participating in the interaction of mites with their microbiota and acquired by horizontal gene transfer were repressed, including genes encoding the peptidase Der p 38, two 1,3-beta-glucanases, a lysozyme and a GH19 chitinase. Finally, the disruption of mite digestion resulted in the regulation of up to 17 allergen and isoallergen genes. Altogether, our results shed light on the putative role of specific genes in digestion and illustrate the connection between the digestive physiology of HDM and allergy.
Collapse
Affiliation(s)
- José Cristian Vidal-Quist
- Entomología Aplicada a la Agricultura y la Salud, Departamento de Biotecnología Microbiana y de Plantas, Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, Madrid, Spain
| | - Félix Ortego
- Entomología Aplicada a la Agricultura y la Salud, Departamento de Biotecnología Microbiana y de Plantas, Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, Madrid, Spain
| | - Stephane Rombauts
- Center for Plant Systems Biology, VIB, Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Pedro Hernández-Crespo
- Entomología Aplicada a la Agricultura y la Salud, Departamento de Biotecnología Microbiana y de Plantas, Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, Madrid, Spain
| |
Collapse
|
2
|
Qin QZ, Tang J, Wang CY, Xu ZQ, Tian M. Construction by artificial intelligence and immunovalidation of hypoallergenic mite allergen Der f 36 vaccine. Front Immunol 2024; 15:1325998. [PMID: 38601166 PMCID: PMC11004385 DOI: 10.3389/fimmu.2024.1325998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 03/12/2024] [Indexed: 04/12/2024] Open
Abstract
Background The house dust mite (HDM) is widely recognized as the most prevalent allergen in allergic diseases. Allergen-specific immunotherapy (AIT) has been successfully implemented in clinical treatment for HDM. Hypoallergenic B-cell epitope-based vaccine designed by artificial intelligence (AI) represents a significant progression of recombinant hypoallergenic allergen derivatives. Method The three-dimensional protein structure of Der f 36 was constructed using Alphafold2. AI-based tools were employed to predict B-cell epitopes, which were subsequently verified through IgE-reaction testing. Hypoallergenic Der f 36 was then synthesized, expressed, and purified. The reduced allergenicity was assessed by enzyme-linked immunosorbent assay (ELISA), immunoblotting, and basophil activation test. T-cell response to hypoallergenic Der f 36 and Der f 36 was evaluated based on cytokine expression in the peripheral blood mononuclear cells (PBMCs) of patients. The immunogenicity was evaluated and compared through rabbit immunization with hypoallergenic Der f 36 and Der f 36, respectively. The inhibitory effect of the blocking IgG antibody on the specific IgE-binding activity and basophil activation of Der f 36 allergen was also examined. Results The final selected non-allergic B-cell epitopes were 25-48, 57-67, 107-112, 142-151, and 176-184. Hypoallergenic Der f 36 showed significant reduction in IgE-binding activity. The competitive inhibition of IgE-binding to Der f 36 was investigated using the hypoallergenic Der f 36, and only 20% inhibition could be achieved, which is greatly reduced when compared with inhibition by Der f 36 (98%). The hypoallergenic Der f 36 exhibited a low basophil-stimulating ratio similar to that of the negative control, and it could induce an increasing level of IFN-γ but not Th2 cytokines IL-5 and IL-13 in PBMCs. The vaccine-specific rabbit blocking IgG antibodies could inhibit the patients' IgE binding and basophil stimulation activity of Derf 36. Conclusion This study represents the first application of an AI strategy to facilitate the development of a B-cell epitope-based hypoallergenic Der f 36 vaccine, which may become a promising immunotherapy for HDM-allergic patients due to its reduced allergenicity and its high immunogenicity in inducing blocking of IgG.
Collapse
Affiliation(s)
- Qiao-Zhi Qin
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Pediatric Department, Northern Jiangsu People’s Hospital, Yangzhou, China
| | - Jian Tang
- Department of Pharmacy, The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Cai-Yun Wang
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Zhi-Qiang Xu
- Research Division of Clinical Pharmacology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, China
| | - Man Tian
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
3
|
Dominguez J, Holmes SK, Bartone RD, Tisch LJ, Tighe RM, Bonner JC, Payne CK. House Dust Mite Extract Forms a Der p 2 Corona on Multi-Walled Carbon Nanotubes: Implications for Allergic Airway Disease. ENVIRONMENTAL SCIENCE. NANO 2024; 11:324-335. [PMID: 38577066 PMCID: PMC10990074 DOI: 10.1039/d3en00666b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Multi-walled carbons nanotubes (MWCNTs) are used in materials for the construction, automotive, and aerospace industries. Workers and consumers are exposed to these materials via inhalation. Existing recommended exposure limits are based on MWCNT exposures that do not take into account more realistic co-exposures. Our goal was to understand how a common allergen, house dust mites, interacts with pristine MWCNTs and lung fluid proteins. We used gel electrophoresis, western blotting, and proteomics to characterize the composition of the allergen corona formed from house dust mite extract on the surface of MWCNTs. We found that the corona is dominated by der p 2, a protein associated with human allergic responses to house dust mites. Der p 2 remains adsorbed on the surface of the MWCNTs following subsequent exposures to lung fluid proteins. The high concentration of der p 2, localized on surface of MWCNTs, has important implications for house dust mite-induced allergies and asthma. This research provides a detailed characterization of the complex house dust mite-lung fluid protein coronas for future cellular and in vivo studies. These studies will help to address the molecular and biochemical mechanisms underlying the exacerbation of allergic lung disease by nanomaterials.
Collapse
Affiliation(s)
- Judith Dominguez
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina, USA 27708
| | - Samantha K. Holmes
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina, USA 27708
| | - Ryan D. Bartone
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA 27695
| | - Logan J. Tisch
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA 27695
| | - Robert M. Tighe
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA 27710
| | - James C. Bonner
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA 27695
| | - Christine K. Payne
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina, USA 27708
| |
Collapse
|
4
|
Hubert J, Vrtala S, Sopko B, Dowd SE, He Q, Klimov PB, Harant K, Talacko P, Erban T. Predicting Blomia tropicalis allergens using a multiomics approach. Clin Transl Allergy 2023; 13:e12302. [PMID: 37876035 PMCID: PMC10542617 DOI: 10.1002/clt2.12302] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/01/2023] [Accepted: 09/07/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND The domestic mite Blomia tropicalis is a major source of allergens in tropical and subtropical regions. Despite its great medical importance, the allergome of this mite has not been sufficiently studied. Only 14 allergen groups have been identified in B. tropicalis thus far, even though early radioimmunoelectrophoresis techniques (27 uncharacterized allergen complexes) and comparative data based on 40 allergen groups officially recognized by the World Health Organization (WHO)/IUIS in domestic astigmatid mites suggest the presence of a large set of additional allergens. METHODS Here, we employ a multiomics approach to assess the allergome of B. tropicalis using genomic and transcriptomic sequence data and perform highly sensitive protein abundance quantification. FINDINGS Among the 14 known allergen groups, we confirmed 13 (one WHO/IUIS allergen, Blo t 19, was not found) and identified 16 potentially novel allergens based on sequence similarity. These data indicate that B. tropicalis shares 27 known/deduced allergen groups with pyroglyphid house dust mites (genus Dermatophagoides). Among these groups, five allergen-encoding genes are highly expressed at the transcript level: Blo t 1, Blo t 5, Blo t 21 (known), Blo t 15, and Blo t 18 (predicted). However, at the protein level, a different set of most abundant allergens was found: Blo t 2, 10, 11, 20 and 21 (mite bodies) or Blo t 3, 4, 6 and predicted Blo t 13, 14 and 36 (mite feces). INTERPRETATION We report the use of an integrated omics method to identify and predict an array of mite allergens and advanced, label-free proteomics to determine allergen protein abundance. Our research identifies a large set of novel putative allergens and shows that the expression levels of allergen-encoding genes may not be strictly correlated with the actual allergenic protein abundance in mite bodies.
Collapse
Affiliation(s)
- Jan Hubert
- Crop Research InstitutePragueCzechia
- Department of Microbiology, Nutrition and DieteticsFaculty of Agrobiology, Food and Natural ResourcesCzech University of Life Sciences PraguePragueCzechia
| | - Susanne Vrtala
- Department of Pathophysiology and Allergy ResearchCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | | | - Scot E. Dowd
- MR DNA (Molecular Research LP)ShallowaterTexasUSA
| | - Qixin He
- Purdue UniversityLilly Hall of Life SciencesWest LafayetteIndianaUSA
| | - Pavel B. Klimov
- Purdue UniversityLilly Hall of Life SciencesWest LafayetteIndianaUSA
| | - Karel Harant
- Proteomics Core FacilityFaculty of ScienceCharles UniversityBIOCEVVestecCzechia
- Institute for Environmental StudiesFaculty of ScienceCharles UniversityPragueCzechia
| | - Pavel Talacko
- Proteomics Core FacilityFaculty of ScienceCharles UniversityBIOCEVVestecCzechia
| | | |
Collapse
|
5
|
Zhou Y, Li Q, Pan R, Wang Q, Zhu X, Yuan C, Cai F, Gao Y, Cui Y. Regulatory roles of three miRNAs on allergen mRNA expression in Tyrophagus putrescentiae. Allergy 2022; 77:469-482. [PMID: 34570913 DOI: 10.1111/all.15111] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Tyrophagus putresecentiae is an important mite species in rural and urban environments, causing sensitization and allergic disease. While evidence suggests that microRNAs (miRNAs) may regulate the expression of allergen-encoding genes, no study has directly investigated this possibility. Here, this gap was addressed by profiling miRNAs and elucidating their target allergen messenger RNAs (mRNAs) in this mite species. METHODS Small RNA and transcriptome libraries were constructed for eggs, larvae, nymphs, and adults. After deep miRNA and whole-transcriptome sequencing were performed, the miRNA and allergen-encoding mRNA regulatory networks were explored. RESULTS A total of 540 miRNAs were identified, including 155 with expression levels differing significantly across the four mite developmental stages (p < .01), 59 of which were novel. The mRNA expression for allergens was higher for Tyr p 1 in adults than in other developmental stages; Tyr p 2-5, 7, 10, 13, 33, and 34 in immature stages; and Tyr p 28, 35, and 36 in eggs and adults. A combined miRNA and transcriptome bioinformatics analysis showed that allergen Tyr p 3 was regulated by miRNA PC-5p-5698441_1, Tyr p 4 was regulated by PC-5p-7050653_1, and Tyr p 34 was regulated by PC-5p-5534223_1 and PC-5p-5698441_1. These three allergen mRNA and three miRNAs were identified using qRT-PCR, and their regulatory roles were confirmed by double-fluorescent reporter gene system and site-directed mutagenesis technology. CONCLUSIONS For the first time, allergen mRNA expression and miRNAs were profiled throughout the life cycle for an allergen-producing mite, and the results showed that miRNAs bind to target allergen mRNAs to regulate their expression.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Pediatrics Laboratory The Affiliated Wuxi Children's Hospital of Nanjing Medical University Wuxi China
| | - Qingqing Li
- Department of Clinical Laboratory The Affiliated Wuxi People's Hospital of Nanjing Medical University Wuxi China
| | - Ruilin Pan
- Department of Clinical Laboratory The Affiliated Wuxi People's Hospital of Nanjing Medical University Wuxi China
| | - Qiong Wang
- Department of Clinical Laboratory The Affiliated Wuxi People's Hospital of Nanjing Medical University Wuxi China
| | - Xuming Zhu
- Department of Clinical Laboratory The Affiliated Wuxi People's Hospital of Nanjing Medical University Wuxi China
| | - Cunyin Yuan
- Department of Clinical Laboratory The Affiliated Wuxi People's Hospital of Nanjing Medical University Wuxi China
| | - Fangfang Cai
- Department of Clinical Laboratory The Affiliated Wuxi People's Hospital of Nanjing Medical University Wuxi China
| | - Ya‐dong Gao
- Department of Allergology Zhongnan Hospital of Wuhan University Wuhan China
| | - Yubao Cui
- Department of Clinical Laboratory The Affiliated Wuxi People's Hospital of Nanjing Medical University Wuxi China
| |
Collapse
|
6
|
Nony E, Moingeon P. Proteomics in support of immunotherapy: contribution to model-based precision medicine. Expert Rev Proteomics 2021; 19:33-42. [PMID: 34937491 DOI: 10.1080/14789450.2021.2020653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Proteomics encompasses a wide and expanding range of methods to identify, characterize, and quantify thousands of proteins from a variety of biological samples, including blood samples, tumors, and tissues. Such methods are supportive of various forms of immunotherapy applied to chronic conditions such as allergies, autoimmune diseases, cancers, and infectious diseases. AREAS COVERED In support of immunotherapy, proteomics based on mass spectrometry has multiple specific applications related to (i) disease modeling and patient stratification, (ii) antigen/ autoantigen/neoantigen/ allergen identification, (iii) characterization of proteins and monoclonal antibodies used for immunotherapeutic or diagnostic purposes, (iv) identification of biomarkers and companion diagnostics and (v) monitoring by immunoproteomics of immune responses elicited in the course of the disease or following immunotherapy. EXPERT OPINION Proteomics contributes as an enabling technology to an evolution of immunotherapy toward a precision medicine approach aiming to better tailor treatments to patients' specificities in multiple disease areas. This trend is favored by a better understanding through multi-omics profiling of both the patient's characteristics, his/her immune status as well as of the features of the immunotherapeutic drug.
Collapse
Affiliation(s)
- Emmanuel Nony
- Protein Sciences Department, Institut de Recherches Servier, Croissy Sur Seine, France
| | - Philippe Moingeon
- Center for Therapeutic Innovation, Immuno-inflammatory Disease, Institut de Recherches Servier, Croissy Sur Seine, France
| |
Collapse
|
7
|
Abu Khweek A, Joldrichsen MR, Kim E, Attia Z, Krause K, Daily K, Estfanous S, Hamilton K, Badr A, Anne MNK, Eltobgy M, Corps KN, Carafice C, Zhang X, Gavrilin MA, Boyaka PN, Amer AO. Caspase-11 regulates lung inflammation in response to house dust mites. Cell Immunol 2021; 370:104425. [PMID: 34800762 PMCID: PMC8714054 DOI: 10.1016/j.cellimm.2021.104425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 08/15/2021] [Accepted: 08/17/2021] [Indexed: 10/20/2022]
Abstract
Asthma is an inflammatory lung disorder characterized by mucus hypersecretion, cellular infiltration, and bronchial hyper-responsiveness. House dust mites (HDM) are the most prevalent cause of allergic sensitization. Canonical and noncanonical inflammasomes are multiprotein complexes that assemble in response to pathogen or danger-associated molecular patterns (PAMPs or DAMPs). Murine caspase-11 engages the noncanonical inflammasome. We addressed the role of caspase-11 in mediating host responses to HDM and subsequent allergic inflammation using caspase-11-/- mice, which lack caspase-11 while express caspase-1. We found that HDM induce caspase-11 expression in vitro. The presence of IL-4 and IL-13 promote caspase-11 expression. Additionally, caspase-11-/- macrophages show reduced release of IL-6, IL-12, and KC, and express lower levels of costimulatory molecules (e.g., CD40, CD86 and MHCII) in response to HDM stimulation. Notably, HDM sensitization of caspase-11-/- mice resulted in similar levels of IgE responses and hypothermia in response to nasal HDM challenge compared to WT. However, analysis of cell numbers and cytokines in bronchiolar alveolar lavage fluid (BALF) and histopathology of representative lung segments demonstrate altered inflammatory responses and reduced neutrophilia in the airways of the caspase-11-/- mice. These findings indicate that caspase-11 regulates airway inflammation in response to HDM exposure.
Collapse
Affiliation(s)
- Arwa Abu Khweek
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA; Department of Biology and Biochemistry, Birzeit University, West Bank, Palestine
| | - Marisa R Joldrichsen
- Department of Veterinary Biosciences, The Ohio State University, Columbus OH 43210, USA
| | - Eunsoo Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus OH 43210, USA
| | - Zayed Attia
- Department of Veterinary Biosciences, The Ohio State University, Columbus OH 43210, USA
| | - Kathrin Krause
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Kylene Daily
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Shady Estfanous
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Kaitlin Hamilton
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Asmaa Badr
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Midhun N K Anne
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Mostafa Eltobgy
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Kara N Corps
- Department of Veterinary Biosciences, The Ohio State University, Columbus OH 43210, USA
| | - Cierra Carafice
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Xiaoli Zhang
- Center for Biostatistics, The Ohio State University, Columbus OH 43210, USA
| | - Mikhail A Gavrilin
- Department of Internal Medicine, The Ohio State University, Columbus OH 43210, USA
| | - Prosper N Boyaka
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA; Department of Veterinary Biosciences, The Ohio State University, Columbus OH 43210, USA; Infectious Diseases Institute, The Ohio State University, Columbus OH 43210, USA.
| | - Amal O Amer
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA.
| |
Collapse
|
8
|
Erban T, Klimov PB, Harant K, Talacko P, Nesvorna M, Hubert J. Label-free proteomic analysis reveals differentially expressed Wolbachia proteins in Tyrophagus putrescentiae: Mite allergens and markers reflecting population-related proteome differences. J Proteomics 2021; 249:104356. [PMID: 34438106 DOI: 10.1016/j.jprot.2021.104356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/12/2021] [Accepted: 08/19/2021] [Indexed: 12/21/2022]
Abstract
Tyrophagus putrescentiae is an astigmatid mite of great economic, medical and veterinary importance. The microbiome, especially intracellular bacteria, may affect allergy/allergen expression. We targeted Wolbachia proteins, allergen comparisons and markers in Wolbachia-mite interactions in three mite populations. A decoy database was constructed by proteogenomics using the T. putrescentiae draft genome, Wolbachia transcriptome assembly and current T. putrescentiae-related sequences in GenBank. Among thousands of mite-derived proteins, 18 Wolbachia proteins were reliably identified. We suggest that peroxiredoxin, bacterioferritin, ankyrin repeat domain-containing protein and DegQ family serine endoprotease indicate a higher-level bacterium-bacterium-host interaction. We produced evidence that the host-Wolbachia interaction is modulated through pattern recognition receptors (PRRs), mannose-binding lectins/mannose receptors, the cholinergic anti-inflammatory pathway with TNF-α, and others. We observed Tyr p 3 suppression in mites with Wolbachia, linking trypsin to PRR modulation. Nine out of the 12 current WHO/IUIS official allergens were reliably identified, but the remaining three allergens, Tyr p 1, 8 and 35, were detected as only trace hits. This study provides numerous markers for further Wolbachia-host interaction research. For accuracy, mite allergens should be considered according to abundance in species, but mite populations/strains, as well as their microbiome structure, may be key factors. SIGNIFICANCE: The astigmatid mites occurring in homes are significant producers of allergens that are highly dangerous to humans and domesticated animals. Mites are tightly associated with microorganisms that affect their biology and consequently allergy signatures. Mite populations were found to be infected with certain intracellular bacteria, but some populations lacked an intracellular bacterium. Our previous research showed that some populations of Tyrophagus putrescentiae are infected with Wolbachia, but some populations host additional bacteria of interest. Thus, there are not only interactions between the mites and Wolbachia but also likely an additional level of interaction that can be found in the interaction between different bacteria in the mites. These "higher-level" signatures and consequences that bacteria affect, including allergen production, are not understood in mites. In this study, we identified Wolbachia-specific proteins in mites for the first time. This study provides Wolbachia- and mite-derived markers that can be clues for describing "higher-level" mite-bacterium-bacterium interactions. Indeed, the microbiome contribution to allergies can potentially be derived directly from bacterial proteins, especially if they are abundant.
Collapse
Affiliation(s)
- Tomas Erban
- Crop Research Institute, Drnovska 507/73, Prague 6-Ruzyne CZ-16106, Czechia.
| | - Pavel B Klimov
- School of Natural Sciences, Bangor University, Bangor LL57 2 UW, UK; Institute of Biology, University of Tyumen, Pirogova 3, 625043 Tyumen, Russia
| | - Karel Harant
- Proteomics Core Facility, Faculty of Science, Charles University, BIOCEV, Prumyslova 595, Vestec CZ-25242, Czechia; Institute for Environmental Studies, Faculty of Science, Charles University, Benatska 2, Prague 2 CZ-128 01, Czechia
| | - Pavel Talacko
- Proteomics Core Facility, Faculty of Science, Charles University, BIOCEV, Prumyslova 595, Vestec CZ-25242, Czechia
| | - Marta Nesvorna
- Crop Research Institute, Drnovska 507/73, Prague 6-Ruzyne CZ-16106, Czechia
| | - Jan Hubert
- Crop Research Institute, Drnovska 507/73, Prague 6-Ruzyne CZ-16106, Czechia
| |
Collapse
|
9
|
Chen J, Cai Z, Fan D, Hu J, Hou Y, He Y, Zhang Z, Zhao Z, Gao P, Hu W, Sun J, Li J, Ji K. Chromosome-level assembly of Dermatophagoides farinae genome and transcriptome reveals two novel allergens Der f 37 and Der f 39. World Allergy Organ J 2021; 14:100590. [PMID: 34659625 PMCID: PMC8487952 DOI: 10.1016/j.waojou.2021.100590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 01/06/2023] Open
Abstract
Accurate house dust mite (HDM) genome and transcriptome data would promote our understanding of HDM allergens. We sought to assemble chromosome-level genome and precise transcriptome profiling of Dermatophagoides farinae and identify novel allergens. In this study, genetic material extracted from HDM bodies and eggs were sequenced. Short-reads from next generation sequencing (NGS) and long-reads from PacBio/Nanopore sequencing were used to construct the D. farinae nuclear genome, transcriptome, and mitochondrial genome. The candidate homologs were screened through aligning our assembled transcriptome data with amino acid sequences in the WHO/IUIS database. Our results showed that compared with the D. farinae draft genome, bacterial DNA content in the presently developed sequencing reads was dramatically reduced (from 22.9888% to 1.5585%), genome size was corrected (from 53.55 Mb to 58.77 Mb), and the contig N50 was increased (from 8.54 kb to 9365.49 kb). The assembled genome has 10 contigs with minimal microbial contamination, 33 canonical allergens and 2 novel allergens. Eight homologs (≥50% homology) were cloned; 2 bound HDM allergic-sera and were identified as allergens (Der f 37 and Der f 39). In conclusion, a chromosome-level genome, transcriptome and mitochondrial genome of D. farinae was generated to support allergen identification and development of diagnostics and immunotherapeutic vaccines.
Collapse
Affiliation(s)
- Jiajie Chen
- Department of Biochemistry and Molecular Biology, Laboratory Department of Pinghu Hospital, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Zelang Cai
- Department of Biochemistry and Molecular Biology, Laboratory Department of Pinghu Hospital, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | | | - Jiayu Hu
- Department of Biochemistry and Molecular Biology, Laboratory Department of Pinghu Hospital, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Yibo Hou
- Department of Biochemistry and Molecular Biology, Laboratory Department of Pinghu Hospital, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Yongsen He
- Department of Biochemistry and Molecular Biology, Laboratory Department of Pinghu Hospital, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Zhen Zhang
- Department of Biochemistry and Molecular Biology, Laboratory Department of Pinghu Hospital, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Zhenfu Zhao
- Department of Biochemistry and Molecular Biology, Laboratory Department of Pinghu Hospital, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Pan Gao
- Shenzhen University General Hospital, Shenzhen, 518060, China
| | - Wanzhen Hu
- Shenzhen University General Hospital, Shenzhen, 518060, China
| | - Jinlyu Sun
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
- Corresponding author.
| | - Jiang Li
- EasyATCG L.L.C, Shenzhen, 518000, China
- Corresponding author.
| | - Kunmei Ji
- Department of Biochemistry and Molecular Biology, Laboratory Department of Pinghu Hospital, Health Science Center, Shenzhen University, Shenzhen, 518060, China
- Corresponding author.
| |
Collapse
|
10
|
Vidal-Quist JC, Ortego F, Hernández-Crespo P. Contribution of cysteine and serine proteases to proteolytic digestion in an allergy-eliciting house dust mite. JOURNAL OF INSECT PHYSIOLOGY 2021; 133:104285. [PMID: 34284041 DOI: 10.1016/j.jinsphys.2021.104285] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/13/2021] [Accepted: 07/13/2021] [Indexed: 06/13/2023]
Abstract
The digestive physiology of house dust mites (HDM) is of interest to understand their allergenicity towards humans since many of their allergens are digestive enzymes and/or are excreted into airborne fecal pellets. The aim of this study is to provide insight on the biochemical basis of proteolytic digestion in Dermatophagoides pteronyssinus, the most widespread HDM species. First, assays using non-specific protein substrates on purified fecal and body extracts determined that body-associated activity is almost exclusively dependent on cysteine proteases, and specifically on major allergen Der p 1. By contrast, cysteine and serine proteases contributed similarly to the activity estimated on fecal extracts. Second, the screening of group-specific peptide-based protease inhibitors followed by ingestion bioassays revealed that the human skin-derived cysteine protease inhibitor cystatin A produces a significant reduction in mite feeding (i.e. excreted guanine), and triggers the overproduction of Der p 1 (3-fold increase by ELISA). Noteworthy, the inhibition of cysteine proteases by cystatin A also resulted in a reduction in three non-target serine protease activities. Further incubation of these extracts with exogenous Der p 1, but not with other commercial cysteine proteases, restored trypsin (Der p 3) and chymotrypsin (Der p 6) activities, indicating that Der p 1 is responsible for their activation in vivo. Finally, the role of serine proteases on the mite's digestive physiology is discussed based on their remarkable activity in fecal extracts and the autocoprophagic behavior reported in mites in this study.
Collapse
Affiliation(s)
- José Cristian Vidal-Quist
- Laboratorio de Entomología Aplicada a la Agricultura y la Salud, Centro de Investigaciones Biológicas Margarita Salas CSIC, Spain.
| | - Félix Ortego
- Laboratorio de Entomología Aplicada a la Agricultura y la Salud, Centro de Investigaciones Biológicas Margarita Salas CSIC, Spain
| | - Pedro Hernández-Crespo
- Laboratorio de Entomología Aplicada a la Agricultura y la Salud, Centro de Investigaciones Biológicas Margarita Salas CSIC, Spain
| |
Collapse
|
11
|
Incorvaia C, Al‐Ahmad M, Ansotegui IJ, Arasi S, Bachert C, Bos C, Bousquet J, Bozek A, Caimmi D, Calderón MA, Casale T, Custovic A, De Blay F, Demoly P, Devillier P, Didier A, Fiocchi A, Fox AT, Gevaert P, Gomez M, Heffler E, Ilina N, Irani C, Jutel M, Karagiannis E, Klimek L, Kuna P, O'Hehir R, Kurbacheva O, Matricardi PM, Morais‐Almeida M, Mosges R, Novak N, Okamoto Y, Panzner P, Papadopoulos NG, Park H, Passalacqua G, Pawankar R, Pfaar O, Schmid‐Grendelmeier P, Scurati S, Tortajada‐Girbés M, Vidal C, Virchow JC, Wahn U, Worm M, Zieglmayer P, Canonica GW. Personalized medicine for allergy treatment: Allergen immunotherapy still a unique and unmatched model. Allergy 2021; 76:1041-1052. [PMID: 32869882 DOI: 10.1111/all.14575] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 12/17/2022]
Abstract
The introduction of personalized medicine (PM) has been a milestone in the history of medical therapy, because it has revolutionized the previous approach of treating the disease with that of treating the patient. It is known today that diseases can occur in different genetic variants, making specific treatments of proven efficacy necessary for a given endotype. Allergic diseases are particularly suitable for PM, because they meet the therapeutic success requirements, including a known molecular mechanism of the disease, a diagnostic tool for such disease, and a treatment blocking the mechanism. The stakes of PM in allergic patients are molecular diagnostics, to detect specific IgE to single-allergen molecules and to distinguish the causative molecules from those merely cross-reactive, pursuit of patient's treatable traits addressing genetic, phenotypic, and psychosocial features, and omics, such as proteomics, epi-genomics, metabolomics, and breathomics, to forecast patient's responsiveness to therapies, to detect biomarker and mediators, and to verify the disease control. This new approach has already improved the precision of allergy diagnosis and is likely to significantly increase, through the higher performance achieved with the personalized treatment, the effectiveness of allergen immunotherapy by enhancing its already known and unique characteristics of treatment that acts on the causes.
Collapse
Affiliation(s)
| | - Mona Al‐Ahmad
- Microbiology Department Faculty of Medicine Kuwait University Kuwait
- Drug Allergy Unit Department of Allergy Al‐Rashed Allergy Center Kuwait
| | | | - Stefania Arasi
- Department of Allergy Bambino Gesu' Childrens' Hospital IRCCS Rome Italy
| | - Claus Bachert
- Upper Airways Research Laboratory ENT Dept Ghent University Hospital Ghent Belgium
- Karolinska Institutet Stockholm Sweden
- Department of ENT Diseases Karolinska University Hospital Stockholm Sweden
| | - Catherine Bos
- Stallergenes Greer Medical Affairs Department Antony France
| | - Jean Bousquet
- University Hospital Montpellier France – MACVIA‐France Montpellier France
| | - Andrzéj Bozek
- Clinical Department of Internal Disease, Dermatology and Allergology Medical University of Silesia Katowice Poland
| | - Davide Caimmi
- Department of Pulmonology and Addictology Arnaud de Villeneuve Hospital Montpellier University Montpellier France
| | - Moises A. Calderón
- Imperial College London – National Heart and Lung Institute Royal Brompton Hospital NHS London UK
| | - Thomas Casale
- Division of Allergy/Immunology University of South Florida Tampa FL USA
| | - Adnan Custovic
- Centre for Respiratory Medicine and Allergy Institute of Inflammation and Repair University of Manchester and University Hospital of South Manchester Manchester UK
| | - Frédéric De Blay
- Allergy Division Chest Diseases Department Strasbourg University Hospital Strasbourg France
| | - Pascal Demoly
- Department of Pulmonology and Addictology Arnaud de Villeneuve Hospital Montpellier University Montpellier France
- Sorbonne Université UMR‐S 1136 INSERM IPLESP EPAR Team Paris France
| | - Philippe Devillier
- Laboratoire de Recherche en Pharmacologie Respiratoire Pôle des Maladies des Voies Respiratoires Hôpital Foch Université Paris‐Saclay Suresnes France
| | - Alain Didier
- Respiratory Disease Dept Larrey Hospital University Hospital of Toulouse Paul Sabatier University Toulouse France
| | - Alessandro Fiocchi
- Department of Allergy Bambino Gesu' Childrens' Hospital IRCCS Rome Italy
| | - Adam T. Fox
- Department of Paediatric Allergy Guy's & St Thomas' Hospitals NHS Foundation Trust London UK
| | - Philippe Gevaert
- Upper Airways Research Laboratory ENT Dept Ghent University Hospital Ghent Belgium
| | | | - Enrico Heffler
- Personalized Medicine, Asthma & Allergy – Humanitas Clinical and Research Center IRCCS Rozzano Italy
- Department of Biomedical Science Humanitas University Pieve Emanuele Italy
| | - Natalia Ilina
- Federal Institute of Immunology of Russia Moscow Russia
| | - Carla Irani
- Department of Internal Medicine and Clinical Immunology Faculty of Medicine Hotel Dieu de France Hospital Saint Joseph University Beirut Lebanon
| | - Marek Jutel
- Department of Clinical Immunology Wrocław Medical University Wrocław Poland
| | | | - Ludger Klimek
- Center for Rhinology and Allergology Wiesbaden Germany
| | - Piotr Kuna
- Division of Internal Medicine, Asthma and Allergy Barlicki University Hospital Medical University of Lodz Lodz Poland
| | - Robin O'Hehir
- Alfred Hospital and Monash University Melbourne Australia
| | - Oxana Kurbacheva
- National Research Center – Institute of Immunology Federal Medical‐Biological Agency of Russia Moscow Russia
| | - Paolo M. Matricardi
- Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine Charité – University Medicine Berlin Berlin Germany
| | - Mario Morais‐Almeida
- Immunoallergy Department of CUF‐Descobertas Hospital Lisbon Portugal
- CUF‐Infante Santo Hospital Lisbon Portugal
| | - Ralph Mosges
- Faculty of Medicine Institute of Medical Statistics and Computational Biology University of Cologne Cologne Germany
- CRI – Clinical Research International Ltd. Cologne Germany
| | - Natalija Novak
- Department of Dermatology and Allergy University Hospital Bonn Bonn Germany
| | - Yoshitaka Okamoto
- Department of Otorhinolaryngology Chiba University Hospital Chiba Japan
| | - Petr Panzner
- Department of Immunology and Allergology Faculty of Medicine in Pilsen Charles University in Prague Pilsen Czech Republic
| | - Nikolaos G. Papadopoulos
- Division of Infection, Immunity & Respiratory Medicine Royal Manchester Children's Hospital University of Manchester Manchester UK
- Allergy Department 2nd Pediatric Clinic Athens General Children's Hospital "P&A Kyriakou" University of Athens Athens Greece
| | - Hae‐Sim Park
- Department of Allergy and Clinical Immunology Ajou University School of Medicine Suwon South Korea
| | - Giovanni Passalacqua
- Allergy and Respiratory Diseases Ospedale Policlino San Martino – University of Genoa Genoa Italy
| | - Ruby Pawankar
- Department of Pediatrics Nippon Medical School Tokyo Japan
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery Section of Rhinology and Allergy University Hospital Marburg Philipps‐Universität Marburg Marburg Germany
| | | | - Silvia Scurati
- Stallergenes Greer Medical Affairs Department Antony France
| | - Miguel Tortajada‐Girbés
- Pediatric Pulmonology and Allergy Unit Department of Pediatrics Dr. Peset University Hospital Valencia Spain
- Department of Pediatrics, Obstetrics and Gynecology University of Valencia Valencia Spain
- IVI Foundation Valencia Spain
| | - Carmen Vidal
- Allergy Service Complejo Hospitalario Universitario de Santiago Santiago de Compostela Spain
| | - J. Christian Virchow
- Department of Pneumology/Intensive Care Medicine University of Rostock Rostock Germany
| | - Ulrich Wahn
- Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine Charité – University Medicine Berlin Berlin Germany
| | - Margitta Worm
- Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine Charité – University Medicine Berlin Berlin Germany
| | | | - Giorgio W. Canonica
- Personalized Medicine, Asthma & Allergy – Humanitas Clinical and Research Center IRCCS Rozzano Italy
- Department of Biomedical Science Humanitas University Pieve Emanuele Italy
| |
Collapse
|
12
|
Abu Khweek A, Kim E, Joldrichsen MR, Amer AO, Boyaka PN. Insights Into Mucosal Innate Immune Responses in House Dust Mite-Mediated Allergic Asthma. Front Immunol 2020; 11:534501. [PMID: 33424827 PMCID: PMC7793902 DOI: 10.3389/fimmu.2020.534501] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 09/01/2020] [Indexed: 01/09/2023] Open
Abstract
The prevalence of asthma has been rising steadily for several decades, and continues to be a major public health and global economic burden due to both direct and indirect costs. Asthma is defined as chronic heterogeneous inflammatory diseases characterized by airway obstruction, mucus production and bronchospasm. Different endotypes of asthma are being recognized based on the distinct pathophysiology, genetic predisposition, age, prognosis, and response to remedies. Mucosal innate response to environmental triggers such as pollen, cigarette smoke, fragrances, viral infection, and house dust mite (HDM) are now recognized to play an important role in allergic asthma. HDM are the most pervasive allergens that co-habitat with us, as they are ubiquitous in-house dusts, mattress and bedsheets, and feed on a diet of exfoliated human skin flakes. Dermatophagoides pteronyssinus, is one among several HDM identified up to date. During the last decade, extensive studies have been fundamental in elucidating the interactions between HDM allergens, the host immune systems and airways. Moreover, the paradigm in the field of HDM-mediated allergy has been shifted away from being solely a Th2-geared to a complex response orchestrated via extensive crosstalk between the epithelium, professional antigen presenting cells (APCs) and components of the adaptive immunity. In fact, HDM have several lessons to teach us about their allergenicity, the complex interactions that stimulate innate immunity in initiating and perpetuating the lung inflammation. Herein, we review main allergens of Dermatophagoides pteronyssinus and their interactions with immunological sentinels that promote allergic sensitization and activation of innate immunity, which is critical for the development of the Th2 biased adaptive immunity to HDM allergens and development of allergic asthma.
Collapse
Affiliation(s)
- Arwa Abu Khweek
- Department of Biology and Biochemistry, Birzeit University, Birzeit, Palestine.,Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States.,Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Eunsoo Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Marisa R Joldrichsen
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Amal O Amer
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States.,The Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States
| | - Prosper N Boyaka
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States.,Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States.,The Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
13
|
Pinto L, Torres C, Gil C, Santos HM, Capelo JL, Borges V, Gomes JP, Silva C, Vieira L, Poeta P, Igrejas G. Multiomics Substrates of Resistance to Emerging Pathogens? Transcriptome and Proteome Profile of a Vancomycin-Resistant Enterococcus faecalis Clinical Strain. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2020; 24:81-95. [PMID: 32073998 DOI: 10.1089/omi.2019.0164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Antibiotic resistance and hospital acquired infections are on the rise worldwide. Vancomycin-resistant enterococci have been reported in clinical settings in recent decades. In this multiomics study, we provide comprehensive proteomic and transcriptomic analyses of a vancomycin-resistant Enterococcus faecalis clinical isolate from a patient with a urinary tract infection. The previous genotypic profile of the strain C2620 indicated the presence of antibiotic resistance genes characteristic of the vanB cluster. To further investigate the transcriptome of this pathogenic strain, we used whole genome sequencing and RNA-sequencing to detect and quantify the genes expressed. In parallel, we used two-dimensional gel electrophoresis followed by MALDI-TOF/MS (Matrix-assisted laser desorption/ionization-Time-of-flight/Mass spectrometry) to identify the proteins in the proteome. We studied the membrane and cytoplasm subproteomes separately. From a total of 207 analysis spots, we identified 118 proteins. The protein list was compared to the results obtained from the full transcriptome assay. Several genes and proteins related to stress and cellular response were identified, as well as some linked to antibiotic and drug responses, which is consistent with the known state of multiresistance. Even though the correlation between transcriptome and proteome data is not yet fully understood, the use of multiomics approaches has proven to be increasingly relevant to achieve deeper insights into the survival ability of pathogenic bacteria found in health care facilities.
Collapse
Affiliation(s)
- Luís Pinto
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Veterinary Science Department, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Carmen Torres
- Área de Bioquímica y Biología Molecular, Universidad de La Rioja, Logroño, Spain
| | - Concha Gil
- Departamento de Microbiologia II, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Hugo M Santos
- LAQV-REQUIMTE, Faculty of Science and Technology, Nova University of Lisbon, Lisbon, Portugal
| | - José Luís Capelo
- LAQV-REQUIMTE, Faculty of Science and Technology, Nova University of Lisbon, Lisbon, Portugal
| | - Vítor Borges
- Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health, Lisbon, Portugal
| | - João Paulo Gomes
- Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health, Lisbon, Portugal
| | - Catarina Silva
- Innovation and Technology Unit, Department of Human Genetics, National Institute of Health, Lisbon, Portugal
| | - Luís Vieira
- Innovation and Technology Unit, Department of Human Genetics, National Institute of Health, Lisbon, Portugal
| | - Patrícia Poeta
- Veterinary Science Department, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,LAQV-REQUIMTE, Faculty of Science and Technology, Nova University of Lisbon, Lisbon, Portugal
| | - Gilberto Igrejas
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,LAQV-REQUIMTE, Faculty of Science and Technology, Nova University of Lisbon, Lisbon, Portugal
| |
Collapse
|
14
|
Abu Khweek A, Amer AO. Pyroptotic and non-pyroptotic effector functions of caspase-11. Immunol Rev 2020; 297:39-52. [PMID: 32737894 PMCID: PMC7496135 DOI: 10.1111/imr.12910] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 12/16/2022]
Abstract
Innate immune cells, epithelial cells, and many other cell types are capable of detecting infection or tissue injury, thus mounting regulated immune response. Inflammasomes are highly sophisticated and effective orchestrators of innate immunity. These oligomerized multiprotein complexes are at the center of various innate immune pathways, including modulation of the cytoskeleton, production and maturation of cytokines, and control of bacterial growth and cell death. Inflammasome assembly often results in caspase‐1 activation, which is an inflammatory caspase that is involved in pyroptotic cell death and release of inflammatory cytokines in response to pathogen patterns and endogenous danger stimuli. However, the nature of stimuli and inflammasome components are diverse. Caspase‐1 activation mediated release of mature IL‐1β and IL‐18 in response to canonical stimuli initiated by NOD‐like receptor (NLR), and apoptosis‐associated speck‐like protein containing a caspase recruitment domain (ASC). On the other hand, caspase‐11 delineates a non‐canonical inflammasome that promotes pyroptotic cell death and non‐pyroptotic functions in response to non‐canonical stimuli. Caspase‐11 in mice and its homologues in humans (caspase‐4/5) belong to caspase‐1 family of cysteine proteases, and play a role in inflammation. Knockout mice provided new genetic tools to study inflammatory caspases and revealed the role of caspase‐11 in mediating septic shock in response to lethal doses of lipopolysaccharide (LPS). Recognition of LPS mediates caspase‐11 activation, which promotes a myriad of downstream effects that include pyroptotic and non‐pyroptotic effector functions. Therefore, the physiological functions of caspase‐11 are much broader than its previously established roles in apoptosis and cytokine maturation. Inflammation induced by exogenous or endogenous agents can be detrimental and, if excessive, can result in organ and tissue damage. Consequently, the existence of sophisticated mechanisms that tightly regulate the specificity and sensitivity of inflammasome pathways provides a fine‐tuning balance between adequate immune response and minimal tissue damage. In this review, we summarize effector functions of caspase‐11.
Collapse
Affiliation(s)
- Arwa Abu Khweek
- Department of Biology and Biochemistry, Birzeit University, West Bank, Palestine
| | - Amal O Amer
- Department of Microbial Infection and Immunity, Infectious Disease Institute, College of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
15
|
Abstract
House dust mites are an unsurpassed cause of atopic sensitization and allergic illness throughout the world. The major allergenic dust mites Dermatophagoides pteronyssinus, Dermatophagoides farinae, Euroglyphus maynei, and Blomia tropicalis are eight-legged members of the Arachnid class. Their approximately 3-month lifespan comprises egg, larval, protonymph, tritonymph, and adult stages, with adults, about one fourth to one third of a millimeter in size, being at the threshold of visibility. The geographic and seasonal distributions of dust mites are determined by their need for adequate humidity, while their distribution within substrates is further determined by their avoidance of light. By contacting the epithelium of the eyes, nose, lower airways, skin, and gut, the allergen-containing particles of dust mites can induce sensitization and atopic symptoms in those organs. Various mite allergens, contained primarily in mite fecal particles but also in shed mite exoskeletons and decaying mite body fragments, have properties that include proteolytic activity, homology with the lipopolysaccharide-binding component of Toll-like receptor 4, homology with other invertebrate tropomyosins, and chitin-cleaving and chitin-binding activity. Mite proteases have direct epithelial effects including the breaching of tight junctions and the stimulation of protease-activated receptors, the latter inducing pruritus, epithelial dysfunction, and cytokine release. Other components, including chitin, unmethylated mite and bacterial DNA, and endotoxin, activate pattern recognition receptors of the innate immune system and act as adjuvants promoting sensitization to mite and other allergens. Clinical conditions resulting from mite sensitization and exposure include rhinitis, sinusitis, conjunctivitis, asthma, and atopic dermatitis. Systemic allergy symptoms can also occur from the ingestion of cross-reacting invertebrates, such as shrimp or snail, or from the accidental ingestion of mite-contaminated foods. Beyond their direct importance as a major allergen source, an understanding of dust mites leads to insights into the nature of atopy and of allergic sensitization in general.
Collapse
|
16
|
Erban T, Klimov P, Talacko P, Harant K, Hubert J. Proteogenomics of the house dust mite, Dermatophagoides farinae: Allergen repertoire, accurate allergen identification, isoforms, and sex-biased proteome differences. J Proteomics 2020; 210:103535. [DOI: 10.1016/j.jprot.2019.103535] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/28/2019] [Accepted: 09/26/2019] [Indexed: 12/15/2022]
|
17
|
Food allergomics based on high-throughput and bioinformatics technologies. Food Res Int 2019; 130:108942. [PMID: 32156389 DOI: 10.1016/j.foodres.2019.108942] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/14/2022]
Abstract
Food allergy is a serious food safety problem worldwide, and the investigation of food allergens is the foundation of preventing and treating them, but relevant knowledge is far from sufficient. With the advent of the "big data era", it has been possible to investigate food allergens by high-throughput methods, proposing the concept of allergomics. Allergomics is the discipline studying the repertoire of allergens, which has relatively higher throughput and is faster and more sensitive than conventional methods. This review introduces the basis of allergomics and summarizes its major strategies and applications. Particularly, strategies based on immunoblotting, phage display, allergen microarray, and bioinformatics are reviewed in detail, and the advantages and limitations of each strategy are discussed. Finally, further development of allergomics is predicted. This provides basic theories and recent advances in food allergomics research, which could be insightful for both food allergy research and practical applications.
Collapse
|
18
|
Waldron R, McGowan J, Gordon N, Mitchell EB, Fitzpatrick DA, Doyle S. Characterisation of three novel β-1,3 glucanases from the medically important house dust mite Dermatophagoides pteronyssinus (airmid). INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2019; 115:103242. [PMID: 31520716 DOI: 10.1016/j.ibmb.2019.103242] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/09/2019] [Accepted: 09/09/2019] [Indexed: 06/10/2023]
Abstract
The European house dust mite, Dermatophagoides pteronyssinus is a major source of airborne allergens worldwide and is found in half of European homes. Interactions between microbes and house dust mites (HDM) are considered important factors that allow them to persist in the home. Laboratory studies indicate the European HDM, D. pteronyssinus is a mycophagous mite, capable of utilising a variety of fungi for nutrients, however specific mycolytic digestive enzymes are unknown. Our previous work identified a number of putative glycosyl hydrolases present in the predicted proteome of D. pteronyssinus airmid and validated the expression of 42 of these. Of note, three GH16 proteins with predicted β-1,3 glucanase activity were found to be consistently present in the mite body and excretome. Here, we performed an extensive bioinformatic, proteomic and biochemical study to characterize three-novel β-1,3 glucanases from this medically important house dust mite. The genes encoding novel β-1,3 glucanases designated Glu1, Glu2 and Glu3 were identified in D. pteronyssinus airmid, each exhibited more than 59% amino acid identity to one another. These enzymes are encoded by Glu genes present in a tri-gene cluster and protein homologs are found in other acari. The patchy phyletic distribution of Glu proteins means their evolutionary history remains elusive, however horizontal gene transfer cannot be completely excluded. Recombinant Glu1 and Glu2 exhibit hydrolytic activity toward laminarin, pachyman and barley glucan. Excreted β-1,3 glucanase activity was increased in response to D. pteronyssinus airmid feeding on baker's yeast. Active β-1,3 glucanases are expressed and excreted in the faeces of D. pteronyssinus airmid indicating they are digestive enzymes capable of breaking down β-1,3 glucans of fungi present in house dust.
Collapse
Affiliation(s)
- Rose Waldron
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland; Airmid Healthgroup Ltd., Trinity Enterprise Campus, Dublin, Ireland
| | - Jamie McGowan
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland; Human Health Research Institute, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Natasha Gordon
- Airmid Healthgroup Ltd., Trinity Enterprise Campus, Dublin, Ireland
| | - E Bruce Mitchell
- Airmid Healthgroup Ltd., Trinity Enterprise Campus, Dublin, Ireland
| | - David A Fitzpatrick
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland; Human Health Research Institute, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Sean Doyle
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland.
| |
Collapse
|
19
|
Santos da Silva E, Marques Ponte JC, Barbosa da Silva M, Silva Pinheiro C, Carvalho Pacheco LG, Ferreira F, Briza P, Alcantara-Neves NM. Proteomic Analysis Reveals Allergen Variability among Breeds of the Dust Mite Blomia tropicalis. Int Arch Allergy Immunol 2019; 180:159-172. [PMID: 31563904 DOI: 10.1159/000501964] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/06/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The dawn of the "omics" technologies has changed allergy research, increasing the knowledge and identification of new allergens. However, these studies have been almost restricted to Dermatophagoides spp. Although Blomia tropicalis has long been established as a clinically important source of allergens, a thorough proteomic characterization is still lacking for this dust mite. OBJECTIVE To increase knowledge of B. tropicalis allergens through proteomic analysis. METHODS Eleven in-bred lineages of B. tropicalis were obtained from 11 unique different pregnant females. Their somatic extracts were analyzed and compared with a commercially available extract by liquid chromatography tandem mass spectrometry (LC-MS/MS). RESULTS Considerable differences in the protein expression profiles were found among the breeds, and most of them displayed higher expression levels of major allergens than the commercially available extract. Blo t 2 was the most prominent allergenic protein in the analyzed extracts. Six identified allergens and 14 isoforms have not yet been recognized by IUIS. Conversely, 3 previously recognized B. tropicalis allergens were not found. CONCLUSIONS The clear impact of inbreeding on allergen content shown by our study leads us to conclude that the quantification and/or identification of allergens from in-bred lines should be routinely considered for mite cultivation in order to select breeds with higher amounts of major allergens. In this sense, LC-MS/MS may be a useful method to achieve this quality control for research and commercial purposes.
Collapse
Affiliation(s)
- Eduardo Santos da Silva
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.,Department of Biosciences, University of Salzburg, Salzburg, Austria.,Programa de Pós-Graduação em Biotecnologia da Rede Nordeste de Biotecnologia (RENORBIO), Natal, Brazil
| | - João Carlos Marques Ponte
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.,Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Márcia Barbosa da Silva
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.,Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Carina Silva Pinheiro
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Luis Gustavo Carvalho Pacheco
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Fatima Ferreira
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Peter Briza
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Neuza Maria Alcantara-Neves
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil, .,Programa de Pós-Graduação em Biotecnologia da Rede Nordeste de Biotecnologia (RENORBIO), Natal, Brazil,
| |
Collapse
|
20
|
Teufelberger AR, Bröker BM, Krysko DV, Bachert C, Krysko O. Staphylococcus aureus Orchestrates Type 2 Airway Diseases. Trends Mol Med 2019; 25:696-707. [PMID: 31176612 DOI: 10.1016/j.molmed.2019.05.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 12/15/2022]
Abstract
Staphylococcus aureus persistently colonizes the nostrils of one-third of the population but colonizes the sinus mucosa in up to 90% of patients with nasal polyps, implying a possible role in airway disease. Recent findings give new mechanistic insights into the ability of S. aureus to trigger type 2 inflammatory responses in the upper and lower airways. This novel concept of a S. aureus-driven chronic airway inflammatory disease suggests a new understanding of disease triggers. This article reviews the role of S. aureus in chronic inflammatory airway diseases and discusses possible therapeutic approaches to target S. aureus.
Collapse
Affiliation(s)
- Andrea R Teufelberger
- Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University, Ghent, Belgium
| | - Barbara M Bröker
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Dmitri V Krysko
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia; Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent, Belgium
| | - Claus Bachert
- Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University, Ghent, Belgium; Division of ENT Diseases, CLINTEC, Karolinska Institute, Stockholm, Sweden
| | - Olga Krysko
- Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University, Ghent, Belgium.
| |
Collapse
|
21
|
Waldron R, McGowan J, Gordon N, McCarthy C, Mitchell EB, Fitzpatrick DA. Proteome and allergenome of the European house dust mite Dermatophagoides pteronyssinus. PLoS One 2019; 14:e0216171. [PMID: 31042761 PMCID: PMC6493757 DOI: 10.1371/journal.pone.0216171] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/15/2019] [Indexed: 11/18/2022] Open
Abstract
The European house dust mite Dermatophagoides pteronyssinus is of significant medical importance as it is a major elicitor of allergic illnesses. In this analysis we have undertaken comprehensive bioinformatic and proteomic examination of Dermatophagoides pteronyssinus airmid, identified 12,530 predicted proteins and validated the expression of 4,002 proteins. Examination of homology between predicted proteins and allergens from other species revealed as much as 2.6% of the D. pteronyssinus airmid proteins may cause an allergenic response. Many of the potential allergens have evidence for expression (n = 259) and excretion (n = 161) making them interesting targets for future allergen studies. Comparative proteomic analysis of mite body and spent growth medium facilitated qualitative assessment of mite group allergen localisation. Protein extracts from house dust contain a substantial number of uncharacterised D. pteronyssinus proteins in addition to known and putative allergens. Novel D. pteronyssinus proteins were identified to be highly abundant both in house dust and laboratory cultures and included numerous carbohydrate active enzymes that may be involved in cuticle remodelling, bacteriophagy or mycophagy. These data may have clinical applications in the development of allergen-specific immunotherapy that mimic natural exposure. Using a phylogenomic approach utilising a supermatrix and supertree methodologies we also show that D. pteronyssinus is more closely related to Euroglyphus maynei than Dermatophagoides farinae.
Collapse
Affiliation(s)
- Rose Waldron
- Department of Biology, National University of Ireland Maynooth, Co. Kildare, Ireland
- Airmid Healthgroup Ltd., Trinity Enterprise Campus, Dublin, Ireland
| | - Jamie McGowan
- Department of Biology, National University of Ireland Maynooth, Co. Kildare, Ireland
- Human Health Research Institute, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Natasha Gordon
- Airmid Healthgroup Ltd., Trinity Enterprise Campus, Dublin, Ireland
| | - Charley McCarthy
- Department of Biology, National University of Ireland Maynooth, Co. Kildare, Ireland
- Human Health Research Institute, Maynooth University, Maynooth, Co. Kildare, Ireland
| | | | - David A. Fitzpatrick
- Department of Biology, National University of Ireland Maynooth, Co. Kildare, Ireland
- Human Health Research Institute, Maynooth University, Maynooth, Co. Kildare, Ireland
| |
Collapse
|
22
|
Muth T, Hartkopf F, Vaudel M, Renard BY. A Potential Golden Age to Come-Current Tools, Recent Use Cases, and Future Avenues for De Novo Sequencing in Proteomics. Proteomics 2018; 18:e1700150. [PMID: 29968278 DOI: 10.1002/pmic.201700150] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/23/2018] [Indexed: 01/15/2023]
Abstract
In shotgun proteomics, peptide and protein identification is most commonly conducted using database search engines, the method of choice when reference protein sequences are available. Despite its widespread use the database-driven approach is limited, mainly because of its static search space. In contrast, de novo sequencing derives peptide sequence information in an unbiased manner, using only the fragment ion information from the tandem mass spectra. In recent years, with the improvements in MS instrumentation, various new methods have been proposed for de novo sequencing. This review article provides an overview of existing de novo sequencing algorithms and software tools ranging from peptide sequencing to sequence-to-protein mapping. Various use cases are described for which de novo sequencing was successfully applied. Finally, limitations of current methods are highlighted and new directions are discussed for a wider acceptance of de novo sequencing in the community.
Collapse
Affiliation(s)
- Thilo Muth
- Bioinformatics Unit (MF 1), Department for Methods Development and Research Infrastructure, Robert Koch Institute, 13353, Berlin, Germany
| | - Felix Hartkopf
- Bioinformatics Unit (MF 1), Department for Methods Development and Research Infrastructure, Robert Koch Institute, 13353, Berlin, Germany
| | - Marc Vaudel
- K.G. Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, 5020, Bergen, Norway.,Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, 5020, Bergen, Norway
| | - Bernhard Y Renard
- Bioinformatics Unit (MF 1), Department for Methods Development and Research Infrastructure, Robert Koch Institute, 13353, Berlin, Germany
| |
Collapse
|
23
|
Reginald K, Tan CL, Chen S, Yuen L, Goh SY, Chew FT. Characterization of Der f 22 - a paralogue of the major allergen Der f 2. Sci Rep 2018; 8:11743. [PMID: 30082894 PMCID: PMC6079044 DOI: 10.1038/s41598-018-30224-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/26/2018] [Indexed: 11/30/2022] Open
Abstract
We previously identified an expressed sequence tag clone, Der f 22, showing 41% amino acid identity to published Der f 2, and show that both genes are possible paralogues. The objective of this study was to characterize the genomic, proteomic and immunological functions Der f 22 and Der f 2. The full-length sequence of Der f 2 and Der f 22 coded for mature proteins of 129 and 135 amino acids respectively, both containing 6 cysteine residues. Phylogenetic analysis of known group 2 allergens and their homologues from our expressed sequence tag library showed that Der f 22 is a paralogue of Der f 2. Both Der f 2 and Der f 22 were single gene products with one intron. Both allergens showed specific IgE-binding to over 40% of the atopic patients, with limited of cross-reactivity. Both allergens were detected at the gut region of D. farinae by immunostaining. Der f 22 is an important allergen with significant IgE reactivity among the atopic population, and should be considered in the diagnostic panel and evaluated as future hypoallergen vaccine therapeutic target.
Collapse
Affiliation(s)
- Kavita Reginald
- Centre for Virus and Vaccine Research, Sunway University, Bandar Sunway, 47500, Selangor, Malaysia.,Department of Biological Sciences, Sunway University, Bandar Sunway, 47500, Selangor, Malaysia
| | - Chye Ling Tan
- Cell Cycle Control in Skin Epidermis, Institute of Medical Biology, A*STAR, 138648, Singapore, Singapore
| | - Simin Chen
- Allergy and Molecular Immunology Laboratory, Department of Biological Science, National University of Singapore, 117543, Singapore, Singapore
| | - Liling Yuen
- Allergy and Molecular Immunology Laboratory, Department of Biological Science, National University of Singapore, 117543, Singapore, Singapore
| | - Sock Yong Goh
- Allergy and Molecular Immunology Laboratory, Department of Biological Science, National University of Singapore, 117543, Singapore, Singapore
| | - Fook Tim Chew
- Allergy and Molecular Immunology Laboratory, Department of Biological Science, National University of Singapore, 117543, Singapore, Singapore.
| |
Collapse
|
24
|
Cui Y, Wang Q, Jia H. Consideration of methods for identifying mite allergens. Clin Transl Allergy 2018; 8:14. [PMID: 29719717 PMCID: PMC5921985 DOI: 10.1186/s13601-018-0200-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 03/24/2018] [Indexed: 12/14/2022] Open
Abstract
House dust mites are small arthropods that produce proteins—found in their feces, body parts, and eggs—that are major triggers of human allergies worldwide. The goal of this review is to describe the current methods used to identify these allergens. A literature search for allergen identification methods employed between 1995 and 2016 revealed multiple techniques that can be broadly grouped into discovery and confirmation phases. The discovery phase employs screening for mite proteins that can bind IgEs in sera from animals or patients allergic to dust mites. The confirmation phase employs biochemical methods to isolate either native or recombinant mite proteins, confirms the IgE binding of the purified allergens, and uses either in vitro or in vivo assays to demonstrate that the purified antigen can stimulate an immune response. The methods used in the two phases are defined and their strengths and weaknesses are discussed. The majority of HDM-allergic patients may respond to just a small subset of proteins, but new protein discovery methods are still warranted in order to develop a complete panel of HDM allergens for component resolved diagnosis and patient-tailored therapies.
Collapse
Affiliation(s)
- Yubao Cui
- Department of Clinical Laboratory, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299, Qingyang Road, Wuxi, 214023 Jiangsu Province People's Republic of China
| | - Qiong Wang
- Department of Clinical Laboratory, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299, Qingyang Road, Wuxi, 214023 Jiangsu Province People's Republic of China
| | - Haoyuan Jia
- Department of Clinical Laboratory, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299, Qingyang Road, Wuxi, 214023 Jiangsu Province People's Republic of China
| |
Collapse
|
25
|
Liu XY, Yang KY, Wang MQ, Kwok JSL, Zeng X, Yang Z, Xiao XJ, Lau CPY, Li Y, Huang ZM, Ba JG, Yim AKY, Ouyang CY, Ngai SM, Chan TF, Leung ELH, Liu L, Liu ZG, Tsui SKW. High-quality assembly of Dermatophagoides pteronyssinus genome and transcriptome reveals a wide range of novel allergens. J Allergy Clin Immunol 2018; 141:2268-2271.e8. [PMID: 29305317 DOI: 10.1016/j.jaci.2017.11.038] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/17/2017] [Accepted: 11/24/2017] [Indexed: 02/07/2023]
Affiliation(s)
- Xiao-Yu Liu
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, School of Medicine, Shenzhen University, China
| | - Kevin Yi Yang
- School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China; Hong Kong Bioinformatics Centre, the Chinese University of Hong Kong, Hong Kong, China
| | - Ming-Qiang Wang
- School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China; Hong Kong Bioinformatics Centre, the Chinese University of Hong Kong, Hong Kong, China
| | - Jamie Sui-Lam Kwok
- School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China; Hong Kong Bioinformatics Centre, the Chinese University of Hong Kong, Hong Kong, China
| | - Xi Zeng
- School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China; Hong Kong Bioinformatics Centre, the Chinese University of Hong Kong, Hong Kong, China
| | - Zhiyuan Yang
- School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China; Hong Kong Bioinformatics Centre, the Chinese University of Hong Kong, Hong Kong, China
| | - Xiao-Jun Xiao
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, School of Medicine, Shenzhen University, China
| | - Carol Po-Ying Lau
- School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China
| | - Ying Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, China
| | - Zhi-Ming Huang
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, School of Medicine, Shenzhen University, China
| | - Jin-Ge Ba
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, School of Medicine, Shenzhen University, China
| | | | - Chun-Yan Ouyang
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, School of Medicine, Shenzhen University, China
| | - Sai-Ming Ngai
- School of Life Sciences, the Chinese University of Hong Kong, Hong Kong
| | - Ting-Fung Chan
- School of Life Sciences, the Chinese University of Hong Kong, Hong Kong
| | - Elaine Lai-Han Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, China
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, China
| | - Zhi-Gang Liu
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, School of Medicine, Shenzhen University, China.
| | - Stephen Kwok-Wing Tsui
- School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China; Hong Kong Bioinformatics Centre, the Chinese University of Hong Kong, Hong Kong, China; Centre for Microbial Genomics and Proteomics, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|