1
|
Li C, Han T, Zhong P, Zhang Y, Zhao T, Wang S, Wang X, Tian Y, Gong G, Liu Y, Huang L, Lu Y, Wang Z. α2,6-linked sialylated oligosaccharides riched in goat milk alleviate food allergy by regulating the gut flora and mucin O-glycosylation. Carbohydr Polym 2025; 350:123049. [PMID: 39647952 DOI: 10.1016/j.carbpol.2024.123049] [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: 09/09/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 12/10/2024]
Abstract
The nutritious goat milk has low allergenicity. Oligosaccharides represent one of the crucial functional constituents in goat milk, which are structurally similar to human milk oligosaccharides (HMOs). Currently, the anti-allergic activity of GMOs has not been reported. In this study, GMOs were efficiently separated into neutral (NGMOs) and sialylated (SGMOs) fractions, following by qualitative and quantitative analysis at the isomer level using online LC-MS/MS. Fifteen NGMOs and 28 SGMOs were detected in goat milk, with 10 SGMOs reported for the first time. Distinctly, α2,6-linked SGMOs were 3.9 times more abundant in goat milk than in bovine milk, with the total relative content of 6'SL, 3'SLN and 6'NGL in SGMOs approach to 60%, which is more similar to HMOs. Orally administering GMOs, especially α2,6-linked sialylated oligosaccharides, significantly alleviated food allergy in ovalbumin-induced BALB/c mice. SGMOs restored the balance of Lachnospiraceae, Erysipelotrichaceae, and Bacteroidaceae, reconstructed the intestinal mucosal barrier, especially restored the levels of fucosylation, sialylation, and sulfation of mucin O-glycans, increased the expression of four core type 2 O-glycans (F1H2N2, F2H2N2, S1F2H2N2, and A1F1H2N2) significantly. This is the first comprehensive study of the anti-allergic activity of GMOs, and the results lay the foundation for the development of GMOs-based natural anti-allergic components.
Collapse
Affiliation(s)
- Cheng Li
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Tianjiao Han
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Peiyun Zhong
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Yuyang Zhang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Tong Zhao
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Shukai Wang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Xiaoqin Wang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Yang Tian
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Guiping Gong
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Yuxia Liu
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Linjuan Huang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Yu Lu
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China.
| | - Zhongfu Wang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China.
| |
Collapse
|
2
|
Kato Y, Morikawa T, Fujieda S. Comprehensive review of pollen-food allergy syndrome: Pathogenesis, epidemiology, and treatment approaches. Allergol Int 2025; 74:42-50. [PMID: 39278756 DOI: 10.1016/j.alit.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/06/2024] [Accepted: 08/18/2024] [Indexed: 09/18/2024] Open
Abstract
Pollen-food allergy syndrome (PFAS) is caused by cross-reaction of a specific pollen antigen with the corresponding food allergen in sensitized individuals. The manifestations are usually limited to oral symptoms; however, sometimes, rhinitis, respiratory and skin symptoms, and anaphylactic shock may occur. In PFAS pathogenesis, when food containing protein antigens (pan-allergens) with high homology to pollen antigens is ingested, mast cells bound to pollen antigen-specific IgE distributed in the oral mucosa cross-react with the food antigen, causing a local type I allergic reaction. The prevalence of PFAS depends on the geographic conditions, such as the type and amount of pollen in the area. PFAS is prevalent in all regions owing to the wide variety of pollen antigens implicated in the disease, such as alder and grass pollen, even outside of the birch habitat area. Basic research on PFAS is expected to significantly contribute to elucidating the pathogenesis and development of therapeutic strategies for PFAS. Currently, effective treatment for patients with PFAS that allows safe consumption of raw foods is lacking, and avoiding the intake of causative foods is the basis of prevention. Furthermore, allergen immunotherapy for PFAS has not yet been established, but various attempts are underway to develop it into a novel treatment strategy. This review highlights the current research landscape on the pathophysiology, epidemiology, and clinical aspects of PFAS. We outline the research gaps that should be addressed to improve the outcomes of patients with PFAS.
Collapse
Affiliation(s)
- Yukinori Kato
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medical Science, University of Fukui, Fukui, Japan.
| | - Taiyo Morikawa
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medical Science, University of Fukui, Fukui, Japan
| | - Shigeharu Fujieda
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medical Science, University of Fukui, Fukui, Japan
| |
Collapse
|
3
|
Liu YJ, Wang HY, Wang R, Yu J, Shi JJ, Chen RY, Yang GJ, Chen J. IgE-FcεRI protein-protein interaction as a therapeutic target against allergic asthma: An updated review. Int J Biol Macromol 2025; 284:138099. [PMID: 39608548 DOI: 10.1016/j.ijbiomac.2024.138099] [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: 10/08/2024] [Revised: 11/16/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
In the last decade, research has clarified the binding interactions between immunoglobulin E (IgE) and its high-affinity receptor, the FcεRI alpha chain (FcεRI). The formation of the IgE-FcεRI complex is crucial in the context of atopic allergies, linking allergen recognition to cellular activation and disease manifestation. Consequently, pharmacological strategies that disrupt these interactions are vital for managing atopic conditions. Historically, the complexity of the IgE-FcεRI binding process and challenges in producing functional recombinant derivatives has complicated data interpretation. However, advancements in structural biology, protein engineering, and immunological studies have enhanced our understanding of these protein-protein interactions (PPI), facilitating the development of more effective therapies. The introduction of anti-IgE therapies underscores the significance of the IgE-FcεRI PPI in allergic asthma. IgE, that is present locally and systemically, serves as a sensory mechanism in the adaptive immune response, particularly in mast cells (MCs) and basophils. When bound to FcεRI, IgE enables rapid memory responses to allergens, but dysregulation can lead to severe allergic asthma. Thus, the reactivity of IgE sensors can be finely modulated using various IgE-associated molecules. This review explores the mechanisms underlying IgE-dependent MC activation and its regulation by these molecules, including the latest therapeutic candidates under investigation.
Collapse
Affiliation(s)
- Yan-Jun Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China
| | - Hui-Ying Wang
- Department of Allergy and Clinical Immunology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310009, China
| | - Ran Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China
| | - Jing Yu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China
| | - Jin-Jin Shi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China
| | - Ru-Yi Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China
| | - Guan-Jun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China.
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China.
| |
Collapse
|
4
|
Cañete PF, Yu D. Follicular T cells and the control of IgE responses. Allergol Int 2025; 74:13-19. [PMID: 39455298 DOI: 10.1016/j.alit.2024.09.007] [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: 09/08/2024] [Accepted: 09/13/2024] [Indexed: 10/28/2024] Open
Abstract
Atopy is considered the epidemic of the 21st century, and while decades of research have established a direct link between Th2 cells driving pathogenic IgE-mediated allergic disease, only in the past years have T follicular helper (Tfh) cells emerged as pivotal drivers of these responses. In this review, we will examine the molecular mechanisms governing the IgE response, with a particular emphasis on the key cytokines and signaling pathways. We will discuss the exclusion of IgE-producing B cells from germinal centers and explore the recently established role of follicular T cell function and heterogeneity in driving or curtailing these immune responses. Additionally, we will assess the current state of major monoclonal antibodies and allergen immunotherapies designed to counteract Th2-driven inflammation, as well as reflect on the need to investigate how these biologics impact Tfh cell activity, differentiation, and function, as these insights could pave the way for much-needed therapeutic innovation in the treatment of allergic diseases.
Collapse
Affiliation(s)
- Pablo F Cañete
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia; Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| | - Di Yu
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia; Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
5
|
Kang YM, Kim HM, Lee J, Baek JS, Lee M, An HJ. Indole-3-carbinol alleviates allergic skin inflammation via periostin/thymic stromal lymphopoietin suppression in atopic dermatitis. Chin Med 2024; 19:177. [PMID: 39722037 DOI: 10.1186/s13020-024-01042-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Atopic dermatitis (AD) is a chronic multifactorial inflammatory skin disorder with a complex etiology. Despite its increasing prevalence, treatment of AD is still limited. Indole-3-carbinol (I3C) is found in cruciferous vegetables and is formed when these vegetables are cut, chewed, or cooked; it exerts diverse pharmacological activities. METHODS HaCaT keratinocytes stimulated with tumor necrosis factor-α and interferon-γ mixture and NC/Nga mice stimulated with 2,4-dinitrochlorobenzen (DNCB) were used for AD models, in vitro and in vivo, respectively. RESULTS The results showed that I3C reduced the expression of pro-inflammatory cytokines, thymic stromal lymphopoietin (TSLP), and periostin in in vitro model. Oral administration of I3C alleviated AD-like skin inflammatory symptoms, including serum IgE levels, epidermal thickening, inflammatory cell infiltration, transepidermal water loss, and scratching behavior. Moreover, I3C decreased the expression of TSLP and periostin and recovered the expression of skin barrier proteins by regulating Aryl Hydrocarbon Receptor and inhibiting the mitogen-activated protein kinase and nuclear factor-κB pathways in the skin of DNCB-induced AD mice. CONCLUSIONS I3C is suggested as a potential therapeutic alternative for the treatment of AD by repressing allergic inflammatory pathways.
Collapse
Affiliation(s)
- Yun-Mi Kang
- Department of Herbology, College of Korean Medicine, Sangji University, Wonju, Gangwon-Do, 26339, Republic of Korea
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), 70 Cheomdan-Ro, Dong-Gu, Daegu, 41062, Republic of Korea
| | - Hye-Min Kim
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Herbology, College of Korean Medicine, Sangji University, Wonju, Gangwon-Do, 26339, Republic of Korea
| | - Junho Lee
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jong-Suep Baek
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Minho Lee
- Department of Life Science, Dongguk University-Seoul, Ilsandong-Gu, Goyang-Si, 10326, Gyeonggi-do, Republic of Korea.
| | - Hyo-Jin An
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
- Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
6
|
Ye M, Deng G, Liu Q, Jiang X, Wang T, Tan G, Ai J, Liu H. SO 2 activates Th17 cells through the JAK1,2/STAT3 signaling pathway. Int Immunopharmacol 2024; 143:113263. [PMID: 39353391 DOI: 10.1016/j.intimp.2024.113263] [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: 04/08/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVE To investigate the effect of SO2 on Th1/Th2/Th17 cells in allergic rhinitis (AR) and the role of JAK1, 2/STAT3 signaling pathways.To Provide potential directions for the treatment of AR. METHODS Fifteen AR patients were enrolled as the experimental group, while 15 healthy volunteers served as the normal control group. After collecting venous blood, peripheral blood mononuclear cells (PBMCs) were isolated and cultured, followed by the addition of SO2 derivatives and the JAK inhibitor Ruxolitinib. Flow cytometry was employed to assess alterations in the Th1/Th2 and Th17/Treg cell balance upon stimulation with SO2 and Ruxolitinib. qRT-PCR was utilized to detect the expression of Th1-related cytokines IL-2 and IFN-γ, Th2-related cytokines IL-4 and IL-5, Th17-related cytokines IL-17A and RORγt, as well as genes JAK1, JAK2, and STAT3. Flow cytometric cytokine analysis was conducted for quantitative assessment of the expression levels of inflammation-related cytokines in PBMC culture supernatants after stimulation. In addition, we stimulated the Jurkat T lymphocyte cell line with SO2 derivatives, added Ruxolitinib as an inhibitor, and used Western blot analysis to further determine the effects of SO2 on Th cells and the role of the JAK1,2/STAT3 signaling pathway in this process. RESULTS Stimulation with SO2 derivatives upregulated the expression levels of Th2 cells and associated cytokines, as well as Th1 cells and associated cytokines. both AR patients and healthy individuals displayed increased percentages of Th17 cells and Th17/Treg ratios in PBMCs. The expression of IL-17A, RORγt, and IL-6 was also elevated. Under SO2 stimulation, the expression of JAK1, JAK2, STAT3, and RORγt in Jurkat cells increased. Moreover, after the application of Ruxolitinib, the JAK/STAT signaling pathway was inhibited. This led to a reduction in Th17 cells and IL-17A levels in both AR patients and healthy individuals, as well as a decrease in RORγt expression in Jurkat cells. Additionally, the expression of IL-5 decreased in healthy individuals. CONCLUSION SO2 exposure exacerbated Th1/Th2/Th17 inflammation in AR patients and induced Th1 and Th17 inflammation in healthy individuals. The stimulatory effect of SO2 on Th17 cell differentiation could be inhibited by Ruxolitinib. This suggests that the Th17 inflammation induced by SO2 stimulation may be related to the activation of the JAK/STAT signaling pathway, and this has been confirmed in the Jurkat cell line.
Collapse
Affiliation(s)
- Maoyu Ye
- Department of Otorhinolaryngology-Head and Neck Surgery, Third Xiangya Hospital, Central South University, China
| | - Guohao Deng
- Department of Otorhinolaryngology-Head and Neck Surgery, Third Xiangya Hospital, Central South University, China
| | - Qian Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Soochow University, China
| | - Xian Jiang
- Department of Otorhinolaryngology-Head and Neck Surgery, Third Xiangya Hospital, Central South University, China
| | - Tiansheng Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Third Xiangya Hospital, Central South University, China
| | - Guolin Tan
- Department of Otorhinolaryngology-Head and Neck Surgery, Third Xiangya Hospital, Central South University, China
| | - Jingang Ai
- Department of Otorhinolaryngology-Head and Neck Surgery, Third Xiangya Hospital, Central South University, China.
| | - Honghui Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, Third Xiangya Hospital, Central South University, China.
| |
Collapse
|
7
|
Long L, Xu XL, Duan YF, Long L, Chen JY, Yin YH, Zhu YG, Huang Q. Extracellular Vesicles Are Prevalent and Effective Carriers of Environmental Allergens in Indoor Dust. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024. [PMID: 39711517 DOI: 10.1021/acs.est.4c10056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
The global incidence of allergic diseases is rising and poses a substantial threat to human health. Allergenic proteins released by various allergenic species play a critical role in the pathogenesis of allergic diseases and have been widely detected in the environmental matrix. However, the release, presence and interaction of environmental allergens with human body remain to be elucidated. In this study, we reported the widespread of allergen-harboring extracellular vesicles (EVs) in indoor dust from 75 households across five provinces in China. Particle size and abundance of EVs were correlated with specific environmental factors. EVs showed long persistence and high resistance to environmental stress. Metagenomics and metaproteomics data revealed that most indoor allergenic species released allergens within the EVs into dust. A higher abundance of allergenic species and their derived EVs was observed in urban areas compared to rural areas. ELISA confirmed the allergenic activity of the EV-associated allergens. Allergens are common components and even markers of EVs, as evidenced by the data compilation of various allergenic species. The proportion of EV-associated allergens varied across species. EVs facilitated allergen entry into epithelial cells. Intranasally administered EVs can be rapidly transported to the lungs and gastrointestinal tract. EV-associated allergens exhibited higher allergenicity compared with non-EV allergens. Our findings elucidate a vesicle pathway through which environmental allergens are released, persist, and trigger allergic responses within EVs.
Collapse
Affiliation(s)
- Lu Long
- Xiamen Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xue-Li Xu
- Xiamen Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi-Fang Duan
- Xiamen Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Li Long
- Health Management Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jing-Yu Chen
- Xiamen Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Yu-Han Yin
- Xiamen Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Yong-Guan Zhu
- Xiamen Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Qiansheng Huang
- Xiamen Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| |
Collapse
|
8
|
Plum T, Feyerabend TB, Rodewald HR. Beyond classical immunity: Mast cells as signal converters between tissues and neurons. Immunity 2024; 57:2723-2736. [PMID: 39662090 DOI: 10.1016/j.immuni.2024.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/15/2024] [Accepted: 11/12/2024] [Indexed: 12/13/2024]
Abstract
Mast cells are regarded as effectors in immune defense against parasites and venoms and play an essential role in the pathology of allergic diseases. More recently, mast cells have been shown to receive stimuli derived from type 2 immunity, tissue damage, stress, and inflammation. Mast cells then rapidly convert these diverse signals into appropriate, organ-specific protective reflexes that can limit inflammation or reduce tissue damage. In this review, we consider functions of mast cells in sensations-such as pain, itch, and nausea-arising from tissue insults and inflammation and the ensuing protective responses. In light of emerging data highlighting the involvement of mast cells in neuroimmune communication, we also propose that mast cells are "signal converters" linking immunological and tissue states with nervous system responses.
Collapse
Affiliation(s)
- Thomas Plum
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany.
| | - Thorsten B Feyerabend
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany.
| |
Collapse
|
9
|
Yang X, Sima Y, Zhao J, Zhang J, Wang X, Zhang L. Risk and protective factors for coronavirus disease 2019 (COVID-19) in allergic rhinitis patients: a national survey in China. FRONTIERS IN ALLERGY 2024; 5:1479493. [PMID: 39720613 PMCID: PMC11666529 DOI: 10.3389/falgy.2024.1479493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/18/2024] [Indexed: 12/26/2024] Open
Abstract
Background Several epidemiological studies have shown that allergic rhinitis (AR) patients are more susceptible to coronavirus disease 2019 (COVID-19). Objective We aim to investigate the risk factors for COVID-19 in AR patients. Methods A retrospective nationwide cohort study was conducted based on a questionnaire survey in China. The baseline characteristics, region of residence, smoking and drinking status, comorbidities, vaccination status and previous infection information were obtained. Allergen test results, the SARS-CoV-2 nucleic acid test, and antigen detection results were collected. Information on AR and comorbid medication use pre-SARS-CoV-2 infection was also collected. Binary logistic regression and analysis of covariance (different adjusted models) were conducted. Results In all, 830 AR patients were included; 627 patients (75.54%) were infected with SARS-CoV-2. AR comorbid with allergic conjunctivitis (AC) was a protective factor [OR: 0.525 (95% CI = 0.296-0.929), P = 0.027] against SARS-CoV-2 infection, while AR comorbid with food allergy was a risk factor [OR: 6.404 (95% CI = 1.349-30.402), P = 0.0195]. Although fewer patients received four doses of the vaccine, the results showed a significant protective effect against SARS-CoV-2 infection in AR patients [OR: 0.093 (95% CI = 0.025-0.348), P = 0.0004]. Underweight was a protective factor against COVID-19 [OR: 0.287 (95% CI = 0.147-0.562), P = 0.0003] after full multivariable adjustment. Overweight was associated with a 2.071-fold higher risk for COVID-19 compared with normal weight [(95% CI = 1.045-4.105), P = 0.0370]. Additionally, house dust mite (HDM)-specific allergies were also protective against COVID-19 [OR: 0.537 (95% CI = 0.290-0.996), P = 0.0484]. Conclusions This study revealed underlying protective and risk factors, which might be used to improve the management of AR and COVID-19.
Collapse
Affiliation(s)
- Xiaozhe Yang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Yutong Sima
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Jinming Zhao
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Jing Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Xiangdong Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Ouyang XL, Yuan ZL, Chen XB, Gan HW, Guo SH, Cai J, Zhong JJ. Can aged Camellia oleifera Abel oil truly be used to treat atopic dermatitis? Front Pharmacol 2024; 15:1449994. [PMID: 39697554 PMCID: PMC11652156 DOI: 10.3389/fphar.2024.1449994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Atopic dermatitis is an inflammatory skin condition characterized by erythema, eruption, lichenification, and pruritus. Aged Camellia oleifera Abel oil, an effective empirical plant oil utilized by the Gannan Hakka people in China to alleviate the symptoms of atopic dermatitis. However, no scientific studies have been reported to prove whether this oil is truly effective. We conducted this study to confirm whether aged C. oleifera oil could alleviate the symptoms of 2,4-dinitrochlorobenzene (DNCB)-induced atopic dermatitis in mice. Differences in the thickness and weight of the right and left ears were measured. ELISA was used to determine the serum levels of the inflammatory factors IL-4, IgE, IFN-γ, and TNF-α. HE staining was performed to observe inflammatory cell infiltration in the mouse skin lesions. In addition, the metabolites of aged C. oleifera oils were analyzed, and molecular docking was used to assess the binding affinity of the major metabolites to filaggrin, a protein involved in skin barrier function. Animal studies showed that aged C. oleifera oil significantly improved the symptoms of atopic dermatitis. HE staining and measurement of inflammatory factor levels revealed similar results. A total of 41 metabolites were tentatively identified in the oil, with fatty acids emerging as the major metabolites. Molecular docking confirmed that the three most abundant fatty acids, i.e., oleic acid, n-hexadecanoic acid, and octadecanoic acid, bind well to filaggrin. Our results suggest that aged C. oleifera oils can be used to ameliorate the symptoms of atopic dermatitis. Fatty acids may be the major active metabolites responsible for the observed therapeutic effects by reducing transdermal water loss, increasing skin hydration, alleviating DNCB-induced skin barrier alterations, and eliminating itchy scratching caused by dry skin.
Collapse
Affiliation(s)
- Xi-Lin Ouyang
- Department of Pharmacy, Gannan Healthcare Vocational College, Ganzhou, China
| | - Zhang-Lin Yuan
- Department of Pharmacy, Gannan Healthcare Vocational College, Ganzhou, China
| | - Xiao-Bing Chen
- Department of Pharmacy, Gannan Healthcare Vocational College, Ganzhou, China
| | - Hong-Wan Gan
- Department of Dermatology, Ganzhou People’s Hospital, Ganzhou, China
| | - Sen-Hui Guo
- Department of Pharmacy, Gannan Healthcare Vocational College, Ganzhou, China
| | - Juan Cai
- Department of Pharmacy, Gannan Healthcare Vocational College, Ganzhou, China
| | - Jing-Jing Zhong
- Department of Dermatology, Ganzhou People’s Hospital, Ganzhou, China
| |
Collapse
|
11
|
Brabenec L, Gupta S, Eichwald T, Rafei M, Talbot S. Decoding the neuroimmune axis in the atopic march: mechanisms and implications. Curr Opin Immunol 2024; 91:102507. [PMID: 39579588 DOI: 10.1016/j.coi.2024.102507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/25/2024]
Abstract
The immune and nervous systems have co-evolved complex mechanisms to sense environmental dangers and orchestrate a concerted response to safeguard tissue and mobilize host defenses. This sophisticated interplay, marked by a shared repertoire of receptors and ligands, influences disease pathogenesis. Neuro-immune interactions in allergic diseases are pivotal for symptom development, from anaphylaxis to chronic conditions like asthma and atopic dermatitis. This review explores the neuro-immune interplay within the atopic march, emphasizing its role in host defense, inflammation resolution, and tissue repair. We delve into the multifaceted functions of nociceptors in orchestrating type 2 immune responses and the progression of allergic disorders, focusing on key regulators such as CGRP-RAMP1 and SP-MRGPRB2/A2. Additionally, we discuss the potential of nociceptor neuron-blocking drugs to target neuro-immunity, offering the possibility of reversing the progression of the atopic march. Altogether, we underscore the need for targeted interventions to disrupt the pathological processes and enhance therapeutic outcomes at various stages of the atopic march.
Collapse
Affiliation(s)
- Laura Brabenec
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Stockholm County, Sweden
| | - Surbhi Gupta
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Tuany Eichwald
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Stockholm County, Sweden; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Moutih Rafei
- Department of Pharmacology and Physiology, Université de Montreal, Montreal, Quebec, Canada
| | - Sebastien Talbot
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Stockholm County, Sweden; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
12
|
Zhou X, Simonin EM, Jung YS, Galli SJ, Nadeau KC. Role of allergen immunotherapy and biologics in allergic diseases. Curr Opin Immunol 2024; 91:102494. [PMID: 39357079 PMCID: PMC11609009 DOI: 10.1016/j.coi.2024.102494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 09/01/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024]
Abstract
The rise in the prevalence of allergic diseases has become a global health burden. Allergic diseases are a group of immune-mediated disorders characterized by IgE-mediated conditions resulting from a type 2 helper T cell (Th2)-skewed immune response. This review aims to comprehensively summarize recent research on the roles of allergen immunotherapy (AIT) and biologics in allergic diseases. Specifically, we review the mechanisms of AIT and biologics in modulating innate and adaptive immunity involved in allergic disease pathogenesis, as well as their safety and efficacy in the treatment of allergic diseases. We also discuss current new AIT strategies such as recombinant allergen-based vaccines and allergen extract nanoencapsulation. Further research is needed to understand immune tolerance mechanisms beyond the Th2 pathway and to characterize immunological changes in responders and nonresponders to AIT or biologics. This additional research may uncover new targets for monitoring treatment responses and developing personalized treatment strategies for allergic diseases.
Collapse
Affiliation(s)
- Xiaoying Zhou
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Elisabeth M Simonin
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Youn S Jung
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Stephen J Galli
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA; Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
13
|
Giannetti MP. Epidemiology, Risk Factors, and Management of Biphasic Anaphylaxis. Curr Allergy Asthma Rep 2024; 24:651-656. [PMID: 39259441 DOI: 10.1007/s11882-024-01179-1] [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] [Accepted: 08/28/2024] [Indexed: 09/13/2024]
Abstract
PURPOSE OF REVIEW Biphasic anaphylaxis is a variant of anaphylaxis characterized by recurrence of symptoms after initial resolution of anaphylaxis. It was first described in the mid 1990s by Popa and Lerner. Our understanding of the pathophysiology and epidemiology of the condition has advanced considerably since then. The purpose of this manuscript is to review the literature surrounding biphasic anaphylaxis while highlighting key works and recent advances. RECENT FINDINGS Prior studies have estimated biphasic anaphylaxis occurs in 0.4-20% of anaphylaxis episodes. The wide range may be related to differences in anaphylaxis diagnostic criteria which was inconsistent across studies. Recently identified risk factors for occurrence of biphasic anaphylaxis include severe initial symptoms including hypotension or hypoxia, delay in epinephrine use, and greater than one dose of epinephrine required to treat symptoms. Despite our progress to better understand biphasic anaphylaxis, there remain gaps in the literature. This article aims to review the recent literature including, epidemiology, risk factors, and management of biphasic anaphylaxis.
Collapse
Affiliation(s)
- Matthew P Giannetti
- Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, 60 Fenwood Road Hale Building for Transformational Medicine, 5th floor, Boston, MA, 02132, USA.
- Harvard Medical School, Boston, MA, 02132, USA.
| |
Collapse
|
14
|
Zhang EL, Van Petten J, Eitzen G. RhoGDI in RBL-2H3 cells acts as a negative regulator of Rho GTPase signaling to inhibit granule exocytosis. J Leukoc Biol 2024; 116:1498-1514. [PMID: 38943612 PMCID: PMC11599123 DOI: 10.1093/jleuko/qiae150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 05/28/2024] [Accepted: 06/27/2024] [Indexed: 07/01/2024] Open
Abstract
Mast cells are hematopoietic-derived immune cells that possess numerous cytoplasmic granules containing immune mediators such as cytokines and histamine. Antigen stimulation triggers mast cell granule exocytosis, releasing granule contents in a process known as degranulation. We have shown that Rho GTPase signaling is an essential component of granule exocytosis, however, the proteins that regulate Rho GTPases during this process are not well defined. Here we examined the role of Rho guanine-nucleotide dissociation inhibitors (RhoGDIs) in regulating Rho GTPase signaling using RBL-2H3 cells as a mast cell model. We found that RBL-2H3 cells express two RhoGDI isoforms which are primarily localized to the cytosol. Knockdown of RhoGDI1 and RhoGDI2 greatly reduced the levels of all Rho GTPases tested: RhoA, RhoG, Rac1, Rac2, and Cdc42. The reduction in Rho GTPase levels was accompanied by an increase in their membrane-localized fraction and an elevation in the levels of active Rho GTPases. All RhoGDI knockdown strains had altered resting cell morphology, although each strain was activation competent when stimulated. Live cell imaging revealed that the RhoGDI1/2 double knockdown (DKD) strain maintained its activated state for prolonged periods of time compared to the other strains. Only the RhoGDI1/2 DKD strain showed a significant increase in granule exocytosis. Conversely, RhoGDI overexpression in RBL-2H3 cells did not noticeably affect Rho GTPases or degranulation. Based on these results, RhoGDIs act as negative regulators of Rho GTPases during mast cell degranulation, and inhibit exocytosis by sequestering Rho GTPases in the cytosol.
Collapse
Affiliation(s)
- Eric L Zhang
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada
| | - Jennifer Van Petten
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada
| | - Gary Eitzen
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
15
|
Hu H, Qin QZ, Zheng W, Xu ZQ, Chen X. Construction of a Hybrid Vaccine Based on Der f 35-Derived Peptides with Reduced Allergenicity. Int Arch Allergy Immunol 2024:1-17. [PMID: 39591953 DOI: 10.1159/000541815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/03/2024] [Indexed: 11/28/2024] Open
Abstract
INTRODUCTION House dust mite is the primary trigger of allergic respiratory diseases worldwide, and allergen-specific immunotherapy (AIT) is the only disease-modifying treatment in the clinic. The use of allergen molecules instead of extracts is a promising strategy in AIT. In this study, we constructed a peptide hybrid vaccine against the major mite allergen Der f 35 and verified its hypoallergenicity, making it to be a promising candidate for AIT of mite allergy. METHODS The gene encoding Der f 35 was retrieved and synthesized. The hypoallergenic peptide fragments derived from the B-cell epitopes were synthesized based on the predicted profiles of B-cell or T helper-cell epitopes in Der f 35, they were verified by immunoglobulin E (IgE)-reaction test and fused to non-allergenic protein carrier to form the hybrid vaccine. Both the wild-type Der f 35 and the designed vaccine were expressed in Escherichia coli and purified by chromatography; their IgE-binding activity was compared by indirect enzyme-linked immunosorbent assay (ELISA), Western blot, inhibition ELISA, and basophil activation test (BAT). The blocking immunoglobulin G (IgG) against the designed vaccine was raised in rabbits and its ability to inhibit IgE binding of Der f 35 was evaluated by ELISA. The vaccine's effects on peripheral blood mononuclear cells (PBMCs) were investigated. RESULTS A total of 29 out of 60 (48.33%) IgE-positive sera against Der f 35 were screened. Five peptide fragments (residue 39-42, 60-67, 73-107, 111-118, 126-143) from Der f 35 were selected as candidates, in which four peptides exhibited almost no IgE reactivity and the fragment 73-107 had weak reactions. Only 5.98-24.02% inhibition rates could be achieved by the peptides when compared with Der f 35 (97.32%). The designed vaccine migrated at approximately 30 kDa by SDS-PAGE. The IgE-ELISA revealed a significant reduction in IgE-binding activity to the vaccine when compared to wild-type Der f 35 (p < 0.0001); the decreased allergenicity was further confirmed by IgE-Western blot, inhibition ELISA, and BAT, respectively. The IgE-reactivity of Der f 35 could be blocked by the vaccine-induced IgG (p < 0.01). The levels of IL-5 and IL-13 from PBMCs were significantly decreased after stimulation by the vaccine than that by Der f 35 (p < 0.05). CONCLUSION The designed B-cell epitope vaccine of Der f 35 showed greatly diminished allergenicity and Th2 activity. It could be an effective and safe candidate to prevent allergic adverse reactions during the immunotherapy of mite allergy and merits the further study.
Collapse
Affiliation(s)
- Haoyang Hu
- School of Medicine, Nantong University, Nantong, China
| | - Qiao-Zhi Qin
- Pediatric Department, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Wei Zheng
- Department of Pharmacy, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Zhi-Qiang Xu
- Department of Pharmacy, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiang Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
16
|
Pogorelov D, Bode SFN, He X, Ramiro-Garcia J, Hedin F, Ammerlaan W, Konstantinou M, Capelle CM, Zeng N, Poli A, Domingues O, Montamat G, Hunewald O, Ciré S, Baron A, Longworth J, Demczuk A, Bazon ML, Casper I, Klimek L, Neuberger-Castillo L, Revets D, Guyonnet L, Delhalle S, Zimmer J, Benes V, Codreanu-Morel F, Lehners-Weber C, Weets I, Alper P, Brenner D, Gutermuth J, Guerin C, Morisset M, Hentges F, Schneider R, Shamji MH, Betsou F, Wilmes P, Glaab E, Cosma A, Goncalves J, Hefeng FQ, Ollert M. Multiomics approaches disclose very-early molecular and cellular switches during insect-venom allergen-specific immunotherapy: an observational study. Nat Commun 2024; 15:10266. [PMID: 39592626 PMCID: PMC11599746 DOI: 10.1038/s41467-024-54684-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Allergen-specific immunotherapy (AIT) induces immune tolerance, showing the highest success rate (>95%) for insect venom while a much lower chance for pollen allergy. However, the molecular switches leading to successful durable tolerance restoration remain elusive. The primary outcome of this observational study is the comprehensive immunological cellular characterization during the AIT initiation phase, whereas the secondary outcomes are the serological and Th2-cell-type-specific transcriptomic analyses. Here we apply a multilayer-omics approach to reveal dynamic peripheral immune landscapes during the AIT-initiation phase in venom allergy patients (VAP) versus pollen-allergic and healthy controls. Already at baseline, VAP exhibit altered abundances of several cell types, including classical monocytes (cMono), CD4+ hybrid type 1-type 17 cells (Th1-Th17 or Th1/17) and CD8+ counterparts (Tc1-Tc17 or Tc1/17). At 8-24 h following AIT launch in VAP, we identify a uniform AIT-elicited pulse of late-transitional/IL-10-producing B cells, IL-6 signaling within Th2 cells and non-inflammatory serum-IL-6 levels. Sequential induction of activation and survival protein markers also immediately occur. A disequilibrium between serum IL-6 and cMono in VAP baseline is restored at day seven following AIT launch. Our longitudinal analysis discovers molecular switches during initiation-phase insect-venom AIT that secure long-term outcomes. Trial number: NCT02931955.
Collapse
Affiliation(s)
- Dimitrii Pogorelov
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Center of Allergy & Environment, Technical University of Munich, Munich, Germany
| | - Sebastian Felix Nepomuk Bode
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
- Department of General Pediatrics, Adolescent Medicine and Neonatology, Medical Centre-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Xin He
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Javier Ramiro-Garcia
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Fanny Hedin
- National Cytometry Platform, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Wim Ammerlaan
- Integrated BioBank of Luxembourg, Luxembourg Institute of Health, Dudelange, Luxembourg
| | - Maria Konstantinou
- National Cytometry Platform, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Christophe M Capelle
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Ni Zeng
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Aurélie Poli
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Olivia Domingues
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Guillem Montamat
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Oliver Hunewald
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Séverine Ciré
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Alexandre Baron
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Joseph Longworth
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Agnieszka Demczuk
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Murilo Luiz Bazon
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Ingrid Casper
- Center for Rhinology and Allergology, Wiesbaden, Germany
| | - Ludger Klimek
- Center for Rhinology and Allergology, Wiesbaden, Germany
| | | | - Dominique Revets
- National Cytometry Platform, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Lea Guyonnet
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Cytometry Platform, Institut Curie; Innovative Therapies in Haemostasis, INSERM, Université de Paris, Paris, France
| | - Sylvie Delhalle
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Jacques Zimmer
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Vladimir Benes
- Genomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Françoise Codreanu-Morel
- National Unit of Immunology-Allergology, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - Christiane Lehners-Weber
- National Unit of Immunology-Allergology, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - Ilse Weets
- Department of Clinical Biology/ Research Group Experimental Pharmacology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Pinar Alper
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Dirk Brenner
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
- Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, Odense, Denmark
| | - Jan Gutermuth
- Department of Dermatology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Coralie Guerin
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Cytometry Platform, Institut Curie; Innovative Therapies in Haemostasis, INSERM, Université de Paris, Paris, France
| | - Martine Morisset
- National Unit of Immunology-Allergology, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
- Allergy Unit, Angers University Hospital, Angers, France
| | - François Hentges
- National Unit of Immunology-Allergology, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - Reinhard Schneider
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Mohamed H Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, Imperial College London, London, UK
| | - Fay Betsou
- Integrated BioBank of Luxembourg, Luxembourg Institute of Health, Dudelange, Luxembourg
- CRBIP, Institut Pasteur, Université Paris Cité, Paris, France
| | - Paul Wilmes
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Antonio Cosma
- National Cytometry Platform, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Jorge Goncalves
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Feng Q Hefeng
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg.
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg.
- Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
17
|
Lim HJ, Han KM, Kim SH, Ryu SK, You JR, Yoon JH, Kwon E, Kim JE, Kang BC. Antigenicity evaluation of lac color and exploratory study for identifying potential biomarkers of anaphylaxis. Lab Anim Res 2024; 40:40. [PMID: 39587638 PMCID: PMC11590302 DOI: 10.1186/s42826-024-00229-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/23/2024] [Accepted: 11/07/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Lac color, a natural red dye derived from the larvae of laccifer lacca kerr, is one of the most commonly used substances in food. To date, no studies have reported on the antigenicity of lac color and the other biomarkers that can determine anaphylactic reactions. To address this, we evaluated the antigenicity of lac color through active systemic anaphylaxis (ASA) in addition to identifying potential biomarkers performing exploratory studies. For ASA test, Guinea pigs (n = 5) were sensitized with 0(negative control), 4 mg/kg of lac color, 4 mg/kg of lac color + FCA, and 5 mg/kg of ovalbumin + FCA (positive control) 3 times a week for three weeks. Fourteen days after the last sensitization, animals were challenged intravenously weekly for two weeks. Hematological and histopathological analyses were performed and compared to control groups. RESULTS In the ASA test, all lac color groups showed mild symptoms such as nose rubbing, urination, and evacuation, which are insufficient indicators of anaphylaxis. Exploratory studies identified several biomarkers: decreased platelet count, and increased basophil count; distention in the lung, and redness on the inner wall of trachea; mononuclear inflammatory cell infiltration (MICI) in the ear, and heart hemorrhage. When these biomarkers were applied to the ASA test of lac color, in comparison to the negative control group, the positive control group (ovalbumin + FCA) showed a significant over 60-fold reduction in platelet count and nearly threefold higher basophil count compared to other groups. Furthermore, only positive control group exhibited full lung distention and severe redness on the inner wall of the trachea. Mononuclear inflammatory cell infiltration (MICI) in the ear was about three times higher, and heart hemorrhage was only present in the positive control group compared to others. None of the lac color groups were different from the negative control group (p > 0.05), whereas the positive control group was significantly different (p < 0.05). CONCLUSIONS Our study concludes that lac color, at the tested concentrations, does not induce antigenicity in the guinea pig model, providing valuable safety data. Furthermore, the biomarkers identified in this study offer a supportive approach to evaluating the immunogenicity of substances in future research.
Collapse
Affiliation(s)
- Hyun-Jin Lim
- Laboratory Animal Medicine, Graduate School of Translational Medicine, College of Medicine, Seoul National University, 103, Daehak-ro Jongno-gu, Seoul, Republic of Korea
| | - Kang Min Han
- Laboratory Animal Medicine, Graduate School of Translational Medicine, College of Medicine, Seoul National University, 103, Daehak-ro Jongno-gu, Seoul, Republic of Korea
- Department of Pathology, CHA Ilsan Medical Center, Goyang-si, Republic of Korea
| | - Seung-Hyun Kim
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Soo-Kyung Ryu
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ji-Ran You
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jung-Hee Yoon
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Euna Kwon
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ji-Eun Kim
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Byeong-Cheol Kang
- Laboratory Animal Medicine, Graduate School of Translational Medicine, College of Medicine, Seoul National University, 103, Daehak-ro Jongno-gu, Seoul, Republic of Korea.
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
18
|
Badewy R, Glogauer M, Connor KL, Sgro M, Lai JY, Bazinet RP, Tenenbaum HC, Azarpazhooh A. The unrevealed links: periodontal health, human milk composition, and infant gut microbiome dynamics. Prim Health Care Res Dev 2024; 25:e62. [PMID: 39540631 DOI: 10.1017/s1463423624000215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
AIM This review aims to identify the mechanistic relationships related to periodontal diseases and its possible association with changes in human milk composition and the composition and function of infants' gut microbiome. BACKGROUND Maternal health conditions, especially inflammatory, are associated with altered human milk composition. It is not known whether maternal oral inflammatory diseases, including periodontal diseases, deleteriously affect human milk composition. METHODS A narrative review was conducted according to SANRA, the Scale for the Assessment of Narrative Review Articles, guidelines. PubMed, Google Scholar, and Cochrane database of systematic reviews were searched from September 2019 up to December 2023 using keywords such as breast/human milk, maternal health/infections, and periodontal diseases. Reference lists of relevant articles were also screened. Our primary outcome of interest was human milk composition (i.e., any changes in macronutrients, immunological components, etc.). Secondary outcomes included changes in human milk microbiome and subsequent changes in the infant gut microbiome. Outcomes were synthesized using a narrative approach where the existing evidence and current literature were summarized. No risk of bias assessment of the studies was performed in this review. FINDINGS The search yielded no studies investigating the relationship between periodontal diseases in nursing mothers and changes in human milk composition. However, a dose-response relationship exists between the severity of periodontal diseases and the risk of adverse pregnancy outcomes such as preterm birth. Mastitis and diabetes affected milk lipids. Immunoglobulin A (sIgA) was increased in mastitis, whereas reduced concentrations were reported in diabetes. Potential biological pathways through which periodontal diseases can negatively affect human milk composition include the systemic dissemination of inflammatory cytokines like IL-6, PGE2, and tumor necrosis factor (TNF)-β that can be up-regulated by bacterial by-products. This biological plausibility needs to be investigated, given the potentially negative impact on the quality of human milk that could be caused by periodontal inflammation.
Collapse
Affiliation(s)
- Rana Badewy
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
- Department of Dental Oncology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Kristin L Connor
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada
| | - Michael Sgro
- Department of Pediatrics, and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Pediatrics, Division of Neonatology, University of Toronto, Toronto, ON, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jim Yuan Lai
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Richard P Bazinet
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Howard C Tenenbaum
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
- Department of Dentistry, Centre for Advanced Dental Research and Care, Mount Sinai Hospital, Toronto, ON, Canada
| | - Amir Azarpazhooh
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
- Department of Dentistry, Centre for Advanced Dental Research and Care, Mount Sinai Hospital, Toronto, ON, Canada
| |
Collapse
|
19
|
Ferri I, Canala B, Rossi L. Unravelling the Role of Chitin and Chitosan in Prebiotic Activity and Correlation With Cancer: A Narrative Review. Nutr Rev 2024:nuae168. [PMID: 39530850 DOI: 10.1093/nutrit/nuae168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
This review describes the state of the art regarding the prebiotic role of chitin and the interactions of chitin and chitosan with cancer cells. Chitin is the second most abundant polysaccharide in nature and a constitutive component of crustacean shells and the exoskeleton of insects. Chitosan is the deacetylated form of chitin, which is obtained by chemical processing or the enzymatic activity of deacetylases found in microorganisms and insects. Edible insects have recently been introduced in Western countries, thus raising concerns regarding food safety and due to their chitin content and the release of chitosan during the digestive process. The roles of insect chitin and chitosan in the gastrointestinal tract, microbiome modulation, and cancer have been widely investigated. Several in vitro and in vivo studies have shown the possible microbiota modulation of chitin and its relevant communication with the immune system, thus confirming its prebiotic activity. No evidence has been provided on the cancerogenic activity of chitin; however, studies have suggested that chitin has a cytotoxic effect on cancer cell lines. Chitosan has been confirmed to exhibit apoptotic and cytotoxic activities on cancer cells in several in vitro studies on cancer cell lines and in vivo models. In conclusion, the literature does not show a direct connection between the presence of chitin or chitosan and the onset of cancer. However, cytotoxic and apoptotic activities in relation to cancerous lines have been demonstrated.
Collapse
Affiliation(s)
- Irene Ferri
- Department of Veterinary Medicine and Animal Sciences (DIVAS), University of Milan, Lodi 26900, Italy
| | - Benedetta Canala
- Department of Veterinary Medicine and Animal Sciences (DIVAS), University of Milan, Lodi 26900, Italy
| | - Luciana Rossi
- Department of Veterinary Medicine and Animal Sciences (DIVAS), University of Milan, Lodi 26900, Italy
| |
Collapse
|
20
|
Choi MY, Jo MG, Min KY, Kim B, Kim Y, Choi WS. Antimicrobial Peptide Pro10-1D Exhibits Anti-Allergic Activity: A Promising Therapeutic Candidate. Int J Mol Sci 2024; 25:12138. [PMID: 39596204 PMCID: PMC11594534 DOI: 10.3390/ijms252212138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Although antimicrobial peptides (AMPs) exhibit a range of biological functions, reports on AMPs with therapeutic effects in allergic disorders are limited. In this study, we investigated the anti-allergic effects of Pro10-1D, a 10-meric AMP derived from insect defensin protaetiamycine. Our findings demonstrate that Pro10-1D effectively inhibits antigen-induced degranulation of mast cells (MCs) with IC50 values of approximately 11.6 μM for RBL-2H3 cells and 2.7 μM for bone marrow-derived MCs. Furthermore, Pro10-1D suppressed the secretion of cytokines with IC50 values of approximately 2.8 μM for IL-4 and approximately 8.6 μM for TNF-α. Mechanistically, Pro10-1D inhibited the Syk-LAT-PLCγ1 signaling pathway in MCs and decreased the activation of mitogen-activated protein kinases (MAPKs). Pro10-1D demonstrated a dose-dependent reduction in IgE-mediated passive cutaneous anaphylaxis in mice with an ED50 value of approximately 7.6 mg/kg. Further investigation revealed that Pro10-1D significantly reduced the activity of key kinases Fyn and Lyn, which are critical in the initial phase of the FcεRI-mediated signaling pathway, with IC50 values of approximately 22.6 μM for Fyn and approximately 1.5 μM for Lyn. Collectively, these findings suggest that Pro10-1D represents a novel therapeutic candidate for the treatment of IgE-mediated allergic disorders by targeting the Lyn/Fyn Src family kinases in MCs.
Collapse
Affiliation(s)
- Min Yeong Choi
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Min Geun Jo
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Keun Young Min
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Byeongkwon Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Yangmee Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Wahn Soo Choi
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
- Institute of Biomedical Sciences & Technology, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
21
|
Fernandes AMS, da Silva ES, Silva RC, Silveira EF, Santiago LF, de Andrade Belitardo EMM, Alves VDS, Bôas DSV, de Freitas LAR, Ferreira F, Jacquet A, Pacheco LGC, Alcantara-Neves NM, Pinheiro CS. Therapeutic potential of a novel hybrid protein: Mitigating allergy and airway remodeling in chronic asthma models induced by Dermatophagoides pteronyssinus. Mol Immunol 2024; 175:121-131. [PMID: 39357098 DOI: 10.1016/j.molimm.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/23/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND The house-dust mite Dermatophagoides pteronyssinus is a key trigger of allergic asthma. Therefore, it is essential to develop new vaccines that can alter inflammatory processes and airway remodeling. The goal of this study was to test the hypoallergenic and immunogenic characteristics of the hypoallergen rDer p 2231 in a murine model of chronic asthma induced by D. pteronyssinus. METHODS For this, we measured the levels of IgE, IgG1, IgG2a, and cytokines produced by mice receiving the rDer p 2231 protein. Histopathological parameters of the chronic inflammatory response were also investigated by assessing inflammation and airway remodeling. RESULTS rDer p 2231 given as a therapeutic vaccine, led to a reduction in the production of IgE, eosinophils, and neutrophils, a lower activity of eosinophilic peroxidase in the airways, and an increase in the production of IgG1 and IgG2a antibodies. IgG antibodies blocked IgE binding to parental allergens in sera from atopic patients. Splenocytes, BALF, and lung from mice treated with rDer p 2231 secreted higher levels of Th1 and regulatory cytokines, as well as reduced levels of Th2 cytokines. Histopathological investigation of the lower airways demonstrated reductions in the thickness of the bronchiolar smooth muscle layer, in the subepithelial fibrosis, and in the goblet cells hyperplasia. CONCLUSIONS Our preclinical studies suggest that rDer p 2231 is a promising candidate for the treatment of D. pteronyssinus allergy, as the hypoallergen has demonstrated the ability to reduce IgE production, induce specific blocking antibodies, restore and balance Th1/Th2 immune responses, and significantly reduce airway remodeling factors. However, additional clinical studies are needed to more accurately assess the efficacy and safety of rDer p 2231 as a vaccine against D. pteronyssinus-induced allergy.
Collapse
Affiliation(s)
| | - Eduardo Santos da Silva
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil; Post-Graduate Program in Immunology (PPGIm) of the Federal University of Bahia, Salvador, BA, Brazil.
| | - Raphael Chagas Silva
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil; Post-Graduate Program in Immunology (PPGIm) of the Federal University of Bahia, Salvador, BA, Brazil.
| | - Elisânia Fontes Silveira
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil.
| | - Leonardo Freire Santiago
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil.
| | | | - Vítor Dos Santos Alves
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil.
| | - Deise Souza Vilas Bôas
- Post-Graduate Program in Immunology (PPGIm) of the Federal University of Bahia, Salvador, BA, Brazil; Laboratory of Histotechnology, Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil.
| | - Luiz Antônio Rodrigues de Freitas
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FioCruz), Salvador, BA, Brazil; Department of Pathology and Forensic Medicine, School of Medicine, Federal University of Bahia, Salvador, BA, Brazil.
| | - Fatima Ferreira
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria.
| | - Alain Jacquet
- Department of Biochemistry, Chulalongkorn University, Bangkok, Thailand.
| | - Luis Gustavo Carvalho Pacheco
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil.
| | - Neuza Maria Alcantara-Neves
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil; Post-Graduate Program in Immunology (PPGIm) of the Federal University of Bahia, Salvador, BA, Brazil.
| | - Carina Silva Pinheiro
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil; Post-Graduate Program in Immunology (PPGIm) of the Federal University of Bahia, Salvador, BA, Brazil.
| |
Collapse
|
22
|
Terl M, Diamant Z, Kosturiak R, Jesenak M. Choosing the right biologic treatment for individual patients with severe asthma - Lessons learnt from Picasso. Respir Med 2024; 234:107766. [PMID: 39181277 DOI: 10.1016/j.rmed.2024.107766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 07/07/2024] [Accepted: 08/10/2024] [Indexed: 08/27/2024]
Abstract
Severe asthma represents a true challenge for clinicians from two basic perspectives, i.e.: a rational assessment of the underlying endo/phenotype and the subsequent selection of the best fitted (personalized) and effective treatment. Even though asthma is a heterogeneous disease, in the majority of therapy-compliant patients, it is possible to achieve (almost) complete disease control or even remission through conventional and quite uniform step-based pharmacotherapy, even without phenotyping. However, the absence of deeper assessment of individual patients revealed its handicap to its fullest extent during the first years of the new millennium upon the launch of biological therapeutics for patients with the most severe forms of asthma. The introduction of differentially targeted biologics into clinical practice became a challenge in terms of understanding and recognizing the etiopathogenetic heterogeneity of the asthmatic inflammation, pheno/endotyping, and, consequently, to choose the right biologic for the right patient. The answers to the following three questions should lead to correct identification of the dominant pheno/endotype: Is it really (severe) asthma? Is it eosinophilic asthma? If eosinophilic, is it (predominantly) allergen-driven? The identification of the best achievable and relevant alliance between endotypes and phenotypes ("euphenotypes") should be based not only on the assessment of the actual clinical characteristics and laboratory biomarkers, but more importantly, on the evaluation of their development and changes over time. In the current paper, we present a pragmatic three-step approach to severe asthma diagnosis and management.
Collapse
Affiliation(s)
- Milan Terl
- Department of Pneumology and Phthisiology, University Hospital and Faculty of Medicine in Pilsen, Charles University Prague, Czech Republic
| | - Zuzana Diamant
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands; Dept Microbiology Immunology & Transplantation, KU Leuven, Catholic University of Leuven, Belgium; Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Radovan Kosturiak
- Outpatient Clinic for Clinical Immunology and Allergology, Nitra, Slovak Republic; Department of Paediatrics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Teaching Hospital in Martin, Martin, Slovak Republic.
| | - Milos Jesenak
- Department of Paediatrics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Teaching Hospital in Martin, Martin, Slovak Republic; Department of Clinical Immunology and Allergology, University Teaching Hospital in Martin, Martin, Slovak Republic; Department of Pulmonology and Phthisiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Teaching Hospital in Martin, Martin, Slovak Republic.
| |
Collapse
|
23
|
Eggel A, Pennington LF, Jardetzky TS. Therapeutic monoclonal antibodies in allergy: Targeting IgE, cytokine, and alarmin pathways. Immunol Rev 2024; 328:387-411. [PMID: 39158477 PMCID: PMC11659931 DOI: 10.1111/imr.13380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
The etiology of allergy is closely linked to type 2 inflammatory responses ultimately leading to the production of allergen-specific immunoglobulin E (IgE), a key driver of many allergic conditions. At a high level, initial allergen exposure disrupts epithelial integrity, triggering local inflammation via alarmins including IL-25, IL-33, and TSLP, which activate type 2 innate lymphoid cells as well as other immune cells to secrete type 2 cytokines IL-4, IL-5 and IL-13, promoting Th2 cell development and eosinophil recruitment. Th2 cell dependent B cell activation promotes the production of allergen-specific IgE, which stably binds to basophils and mast cells. Rapid degranulation of these cells upon allergen re-exposure leads to allergic symptoms. Recent advances in our understanding of the molecular and cellular mechanisms underlying allergic pathophysiology have significantly shaped the development of therapeutic intervention strategies. In this review, we highlight key therapeutic targets within the allergic cascade with a particular focus on past, current and future treatment approaches using monoclonal antibodies. Specific targeting of alarmins, type 2 cytokines and IgE has shown varying degrees of clinical benefit in different allergic indications including asthma, chronic spontaneous urticaria, atopic dermatitis, chronic rhinosinusitis with nasal polyps, food allergies and eosinophilic esophagitis. While multiple therapeutic antibodies have been approved for clinical use, scientists are still working on ways to improve on current treatment approaches. Here, we provide context to understand therapeutic targeting strategies and their limitations, discussing both knowledge gaps and promising future directions to enhancing clinical efficacy in allergic disease management.
Collapse
Affiliation(s)
- Alexander Eggel
- Department for BioMedical ResearchUniversity of BernBernSwitzerland
- Department of Rheumatology and ImmunologyUniversity Hospital BernBernSwitzerland
| | | | - Theodore S. Jardetzky
- Department of Structural BiologyStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
24
|
Lucas M, Freitas M, Silva AMS, Fernandes E, Ribeiro D. Styrylchromones: Biological Activities and Structure-Activity Relationship. ChemMedChem 2024:e202400782. [PMID: 39480961 DOI: 10.1002/cmdc.202400782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/02/2024]
Abstract
Styrylchromones (SC) are a group of oxygen-containing heterocyclic compounds, which are characterized by the attachment of a styryl group to the chromone core. SC can be found in nature or can be chemically synthesized in the laboratory. As their presence in nature is scarce, the synthetic origin is the most common. Two types of SC are known: 2-styrylchromones and 3-styrylchromones. However, 2-styrylchromones are the most common, being more broadly found in nature and whose chemical synthesis is more commonly described. A wide variety of SC has been described in the literature, with different substituents in different positions, the majority of which are distributed on the A- and/or B-rings. Over the years, several biological activities have been attributed to SC. This work presents a comprehensive review of the biological activities attributed to SC and their structure-activity relationship, based on a published literature search, since 1989. The following biological activities are thoroughly reviewed and discussed in this review: antioxidant, antiallergic, antiviral, antibacterial, antifungal, anti-inflammatory and antitumoral, affinity and selectivity for A3 adenosine receptors, neuroprotective, and α-glucosidase inhibition. In general, SC are composed by a promising scaffold with great potential for the development of new drugs.
Collapse
Affiliation(s)
- Mariana Lucas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313, Porto, Portugal
| | - Marisa Freitas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313, Porto, Portugal
| | - Artur M S Silva
- LAQV-REQUIMTE & QOPNA, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313, Porto, Portugal
| | - Daniela Ribeiro
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313, Porto, Portugal
- Faculty of Agrarian Sciences and Environment, University of the Azores, Rua Capitão João d'Ávila-Pico da Urze, 9700-042, Angra do Heroísmo, Açores, Portugal
| |
Collapse
|
25
|
Kubo M, Harada Y, Sasaki T. The role of dendritic cells in the instruction of helper T cells in the allergic march. Int Immunol 2024; 36:559-566. [PMID: 39162776 DOI: 10.1093/intimm/dxae050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/18/2024] [Indexed: 08/21/2024] Open
Abstract
Allergy is a complex array of diseases influenced by innate and adaptive immunity, genetic polymorphisms, and environmental triggers. Atopic dermatitis is a chronic inflammatory skin disease characterized by barrier defects and immune dysregulation, sometimes leading to asthma and food allergies because of the atopic march. During atopic skin inflammation, Langerhans cells and dendritic cells (DCs) in the skin capture and deliver allergen information to local lymph nodes. DCs are essential immune sensors coordinating immune reactions by capturing and presenting antigens to T cells. In the context of allergic responses, DCs play a crucial role in instructing two types of helper T cells-type 2 helper T (Th2) cells and follicular helper T (TFH) cells-in allergic responses and IgE antibody responses. In skin sensitization, the differentiation and function of Th2 cells and TFH cells are influenced by skin-derived factors, including epithelial cytokines, chemokines, and signalling pathways to modify the function of migratory DCs and conventional DCs. In this review, we aim to understand the specific mechanisms involving DCs in allergic responses to provide insights into the pathogenesis of allergic diseases and potential therapeutic strategies.
Collapse
Affiliation(s)
- Masato Kubo
- Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, Noda-shi, Chiba 278-0022, Japan
- Laboratory for Cytokine Regulation, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Kanagawa 230-0045, Japan
| | - Yasuyo Harada
- Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, Noda-shi, Chiba 278-0022, Japan
| | - Takanori Sasaki
- Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, Noda-shi, Chiba 278-0022, Japan
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
26
|
Mekala JR, Nalluri HP, Reddy PN, S B S, N S SK, G V S D SK, Dhiman R, Chamarthy S, Komaragiri RR, Manyam RR, Dirisala VR. Emerging trends and therapeutic applications of monoclonal antibodies. Gene 2024; 925:148607. [PMID: 38797505 DOI: 10.1016/j.gene.2024.148607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 04/02/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Monoclonal antibodies (mAbs) are being used to prevent, detect, and treat a broad spectrum of malignancies and infectious and autoimmune diseases. Over the past few years, the market for mAbs has grown exponentially. They have become a significant part of many pharmaceutical product lines, and more than 250 therapeutic mAbs are undergoing clinical trials. Ever since the advent of hybridoma technology, antibody-based therapeutics were realized using murine antibodies which further progressed into humanized and fully human antibodies, reducing the risk of immunogenicity. Some of the benefits of using mAbs over conventional drugs include a drastic reduction in the chances of adverse reactions, interactions between drugs, and targeting specific proteins. While antibodies are very efficient, their higher production costs impede the process of commercialization. However, their cost factor has been improved by developing biosimilar antibodies, which are affordable versions of therapeutic antibodies. Along with biosimilars, innovations in antibody engineering have helped to design bio-better antibodies with improved efficacy than the conventional ones. These novel mAb-based therapeutics are set to revolutionize existing drug therapies targeting a wide spectrum of diseases, thereby meeting several unmet medical needs. In the future, mAbs generated by applying next-generation sequencing (NGS) are expected to become a powerful tool in clinical therapeutics. This article describes the methods of mAb production, pre-clinical and clinical development of mAbs, approved indications targeted by mAbs, and novel developments in the field of mAb research.
Collapse
Affiliation(s)
- Janaki Ramaiah Mekala
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522502, Guntur, Andhra Pradesh, INDIA.
| | - Hari P Nalluri
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India
| | - Prakash Narayana Reddy
- Department of Microbiology, Dr. V.S. Krishna Government College, Visakhapatnam 530013, India
| | - Sainath S B
- Department of Biotechnology, Vikrama Simhapuri University, Nellore 524320, AP, India
| | - Sampath Kumar N S
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India
| | - Sai Kiran G V S D
- Santhiram Medical College and General Hospital, Nandyal, Kurnool 518501, AP, India
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Sciences, National Institute of Technology Rourkela-769008, India
| | - Sahiti Chamarthy
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522502, Guntur, Andhra Pradesh, INDIA
| | - Raghava Rao Komaragiri
- Department of CSE, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522302, Andhra Pradesh, INDIA
| | - Rajasekhar Reddy Manyam
- Amrita School of Computing, Amrita Vishwa Vidyapeetham, Amaravati Campus, Amaravati, Andhra Pradesh, India
| | - Vijaya R Dirisala
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India.
| |
Collapse
|
27
|
Galletta F, Gambadauro A, Foti Randazzese S, Passanisi S, Sinatra V, Caminiti L, Zirilli G, Manti S. Pathophysiology of Congenital High Production of IgE and Its Consequences: A Narrative Review Uncovering a Neglected Setting of Disorders. Life (Basel) 2024; 14:1329. [PMID: 39459629 PMCID: PMC11509725 DOI: 10.3390/life14101329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/06/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Elevated serum IgE levels serve as a critical marker for uncovering hidden immunological disorders, particularly inborn errors of immunity (IEIs), which are often misdiagnosed as common allergic conditions. IgE, while typically associated with allergic diseases, plays a significant role in immune defense, especially against parasitic infections. However, extremely high levels of IgE can indicate more severe conditions, such as Hyper-IgE syndromes (HIES) and disorders with similar features, including Omenn syndrome, Wiskott-Aldrich syndrome, and IPEX syndrome. Novel insights into the genetic mutations responsible for these conditions highlight their impact on immune regulation and the resulting clinical features, including recurrent infections, eczema, and elevated IgE. This narrative review uniquely integrates recent advances in the genetic understanding of IEIs and discusses how these findings impact both diagnosis and treatment. Additionally, emerging therapeutic strategies, such as hematopoietic stem cell transplantation (HSCT) and gene therapies, are explored, underscoring the potential for personalized treatment approaches. Emphasizing the need for precise diagnosis and tailored interventions aims to enhance patient outcomes and improve the quality of care for those with elevated IgE levels and associated immunological disorders.
Collapse
Affiliation(s)
| | | | | | - Stefano Passanisi
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age ‘Gaetano Barresi’, University of Messina, 98124 Messina, Italy; (F.G.); (A.G.); (S.F.R.); (V.S.); (L.C.); (G.Z.)
| | | | | | | | - Sara Manti
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age ‘Gaetano Barresi’, University of Messina, 98124 Messina, Italy; (F.G.); (A.G.); (S.F.R.); (V.S.); (L.C.); (G.Z.)
| |
Collapse
|
28
|
Shirvani P, Shirvani A, Holick MF. Decoding the Genetic Basis of Mast Cell Hypersensitivity and Infection Risk in Hypermobile Ehlers-Danlos Syndrome. Curr Issues Mol Biol 2024; 46:11613-11629. [PMID: 39451569 PMCID: PMC11506785 DOI: 10.3390/cimb46100689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Hypermobile Ehlers-Danlos syndrome (hEDS) is a connective tissue disorder marked by joint hypermobility, skin hyperextensibility, and tissue fragility. Recent studies have linked hEDS with mast cell activation syndrome (MCAS), suggesting a genetic interplay affecting immune regulation and infection susceptibility. This study aims to decode the genetic basis of mast cell hypersensitivity and increased infection risk in hEDS by identifying specific genetic variants associated with these conditions. We conducted whole-genome sequencing (WGS) on 18 hEDS participants and 7 first-degree relatives as controls, focusing on identifying genetic variants associated with mast cell dysregulation. Participants underwent clinical assessments to document hEDS symptoms and mast cell hypersensitivity, with particular attention to past infections and antihistamine response. Our analysis identified specific genetic variants in MT-CYB, HTT, MUC3A, HLA-B and HLA-DRB1, which are implicated in hEDS and MCAS. Protein-protein interaction (PPI) network analysis revealed significant interactions among identified variants, highlighting their involvement in pathways related to antigen processing, mucosal protection, and collagen synthesis. Notably, 61.1% of the hEDS cohort reported recurrent infections compared to 28.5% in controls, and 72.2% had documented mast cell hypersensitivity versus 14.2% in controls. These findings provide a plausible explanation for the complex interplay between connective tissue abnormalities and immune dysregulation in hEDS. The identified genetic variants offer insights into potential therapeutic targets for modulating mast cell activity and improving patient outcomes. Future research should validate these findings in larger cohorts and explore the functional implications of these variants to develop effective treatment strategies for hEDS and related mast cell disorders.
Collapse
Affiliation(s)
| | - Arash Shirvani
- Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Michael F. Holick
- Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
29
|
Xie C, Yang J, Gul A, Li Y, Zhang R, Yalikun M, Lv X, Lin Y, Luo Q, Gao H. Immunologic aspects of asthma: from molecular mechanisms to disease pathophysiology and clinical translation. Front Immunol 2024; 15:1478624. [PMID: 39439788 PMCID: PMC11494396 DOI: 10.3389/fimmu.2024.1478624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
In the present review, we focused on recent translational and clinical discoveries in asthma immunology, facilitating phenotyping and stratified or personalized interventions for patients with this condition. The immune processes behind chronic inflammation in asthma exhibit marked heterogeneity, with diverse phenotypes defining discernible features and endotypes illuminating the underlying molecular mechanisms. In particular, two primary endotypes of asthma have been identified: "type 2-high," characterized by increased eosinophil levels in the airways and sputum of patients, and "type 2-low," distinguished by increased neutrophils or a pauci-granulocytic profile. Our review encompasses significant advances in both innate and adaptive immunities, with emphasis on the key cellular and molecular mediators, and delves into innovative biological and targeted therapies for all the asthma endotypes. Recognizing that the immunopathology of asthma is dynamic and continuous, exhibiting spatial and temporal variabilities, is the central theme of this review. This complexity is underscored through the innumerable interactions involved, rather than being driven by a single predominant factor. Integrated efforts to improve our understanding of the pathophysiological characteristics of asthma indicate a trend toward an approach based on disease biology, encompassing the combined examination of the clinical, cellular, and molecular dimensions of the disease to more accurately correlate clinical traits with specific disease mechanisms.
Collapse
Affiliation(s)
- Cong Xie
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Jingyan Yang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Aman Gul
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
- Department of Respiratory Medicine, Uyghur Medicines Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China
- College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Yifan Li
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Rui Zhang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Maimaititusun Yalikun
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Xiaotong Lv
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhan Lin
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qingli Luo
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Huijuan Gao
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
30
|
Siti Sarah CO, Mohd Ashari NS. Exploration of Allergic Rhinitis: Epidemiology, Predisposing Factors, Clinical Manifestations, Laboratory Characteristics, and Emerging Pathogenic Mechanisms. Cureus 2024; 16:e71409. [PMID: 39539885 PMCID: PMC11558229 DOI: 10.7759/cureus.71409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2024] [Indexed: 11/16/2024] Open
Abstract
Allergic rhinitis (AR) is a widespread allergic condition, with its prevalence continuing to rise globally. This disease has a significant impact on patients' quality of life. Understanding the underlying pathophysiology is important to develop better-targeted therapies. For decades, the primary assumption has been that an allergy is caused by unbalanced and overactive immunological responses against allergens, driven mainly by activated T helper 2 (Th2) cells and due to aberrant T-regulatory cells. The more recent hypothesis that is gaining attention relies on the dysregulation of the epithelial barrier, which might result in allergen uptake as a primary defect in the pathogenesis of allergic reactions. The nasal epithelial barrier is considered a crucial first line of defense in the upper airway, as it protects the host's immune system from exposure to allergens. Thus, this review will discuss AR's epidemiology, predisposing factors, clinical manifestations, laboratory characteristics, and pathogenic mechanisms.
Collapse
Affiliation(s)
- Che Othman Siti Sarah
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, MYS
| | - Noor Suryani Mohd Ashari
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, MYS
| |
Collapse
|
31
|
Fernández-Gallego N, Castillo-González R, Moreno-Serna L, García-Cívico AJ, Sánchez-Martínez E, López-Sanz C, Fontes AL, Pimentel LL, Gradillas A, Obeso D, Neuhaus R, Ramírez-Huesca M, Ruiz-Fernández I, Nuñez-Borque E, Carrasco YR, Ibáñez B, Martín P, Blanco C, Barbas C, Barber D, Rodríguez-Alcalá LM, Villaseñor A, Esteban V, Sánchez-Madrid F, Jiménez-Saiz R. Allergic inflammation triggers dyslipidemia via IgG signalling. Allergy 2024; 79:2680-2699. [PMID: 38864116 DOI: 10.1111/all.16187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Allergic diseases begin early in life and are often chronic, thus creating an inflammatory environment that may precede or exacerbate other pathologies. In this regard, allergy has been associated to metabolic disorders and with a higher risk of cardiovascular disease, but the underlying mechanisms remain incompletely understood. METHODS We used a murine model of allergy and atherosclerosis, different diets and sensitization methods, and cell-depleting strategies to ascertain the contribution of acute and late phase inflammation to dyslipidemia. Untargeted lipidomic analyses were applied to define the lipid fingerprint of allergic inflammation at different phases of allergic pathology. Expression of genes related to lipid metabolism was assessed in liver and adipose tissue at different times post-allergen challenge. Also, changes in serum triglycerides (TGs) were evaluated in a group of 59 patients ≥14 days after the onset of an allergic reaction. RESULTS We found that allergic inflammation induces a unique lipid signature that is characterized by increased serum TGs and changes in the expression of genes related to lipid metabolism in liver and adipose tissue. Alterations in blood TGs following an allergic reaction are independent of T-cell-driven late phase inflammation. On the contrary, the IgG-mediated alternative pathway of anaphylaxis is sufficient to induce a TG increase and a unique lipid profile. Lastly, we demonstrated an increase in serum TGs in 59 patients after undergoing an allergic reaction. CONCLUSION Overall, this study reveals that IgG-mediated allergic inflammation regulates lipid metabolism.
Collapse
Affiliation(s)
- Nieves Fernández-Gallego
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Raquel Castillo-González
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Immunology, Ophthalmology and Ear, Nose and Throat (ENT), Universidad Complutense de Madrid, Madrid, Spain
| | - Lucía Moreno-Serna
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Antonio J García-Cívico
- Department of Basic Medical Sciences, Faculty of Medicine, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Elisa Sánchez-Martínez
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Celia López-Sanz
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Ana Luiza Fontes
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Lígia L Pimentel
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Ana Gradillas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - David Obeso
- Department of Basic Medical Sciences, Faculty of Medicine, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - René Neuhaus
- Department of Basic Medical Sciences, Faculty of Medicine, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | | | | | - Emilio Nuñez-Borque
- Department of Allergy and Immunology, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Yolanda R Carrasco
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Cardiology, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Pilar Martín
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Blanco
- Department of Allergy, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Coral Barbas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Domingo Barber
- Department of Basic Medical Sciences, Faculty of Medicine, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Luis M Rodríguez-Alcalá
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Alma Villaseñor
- Department of Basic Medical Sciences, Faculty of Medicine, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Vanesa Esteban
- Department of Allergy and Immunology, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Faculty of Medicine and Biomedicine, Universidad Alfonso X El Sabio, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Rodrigo Jiménez-Saiz
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
- Department of Medicine, McMaster Immunology Research Centre (MIRC), Schroeder Allergy and Immunology Research Institute (SAIRI), McMaster University, Hamilton, Ontario, Canada
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria (UFV), Madrid, Spain
| |
Collapse
|
32
|
Chen VCH, Wang TN, Kuo CJ, Yang YH, Lee CP, Stewart R, Wu SI. Associations of asthma and psychiatric comorbidities on suicide mortality among community adolescents: A 20-year cohort study. Psychiatry Res 2024; 340:116087. [PMID: 39182318 DOI: 10.1016/j.psychres.2024.116087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/01/2024] [Accepted: 07/14/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND The role of psychiatric comorbidity as a confounder between asthma and subsequent suicide mortality in adolescents remained unclarified. METHODS This study used a 20-year community-based cohort in Taiwan. Adolescents aged 11 to 16 from 123 schools were classified into three subgroups: current asthma (symptoms present in the past year), previous asthma (history of asthma but no symptoms in the past year), and no asthma. The mortality and medical care utilizations until the end of follow-up in 2015 were obtained. Cox proportional hazard and competing risk models were performed. Different adjustment models that included covariates of demographic status, allergy, cigarette smoking, psychiatric diagnoses, alcohol or substance misuse, and attention deficit and hyperactivity disorders were compared. RESULTS During the follow-up, 285 out of 153,526 participants died from suicide. The crude hazard ratio for suicide was 1.95 (95 % CI=1.46∼2.60) in the current asthma subgroup and 2.01 (1.36∼2.97) in the previous asthma subgroup. The adjusted hazard ratios (aHR) attenuated to 1.67 (1.25∼2.24) and 1.72 (1.16∼2.54) respectively after further adjustment for all mental disorders, ADHD, substance, and alcohol use disorders. CONCLUSIONS Our adjustment analyses stratified by different models highlight evidence of asthma as an independent risk factor that predicts suicide among adolescents. Depression and mental disorders were potential confounders and identifications of asthma and psychiatric disorders might help decrease suicide risk.
Collapse
Affiliation(s)
- Vincent Chin-Hung Chen
- Department of Psychiatry, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan; School of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Tsu-Nai Wang
- Department of Public Health, College of Health Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chian-Jue Kuo
- Taipei City Psychiatric Center, Taipei City Hospital, Taipei, Taiwan
| | - Yao-Hsu Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan; Center of Excellence for Chang-Gung Research Datalink, Chang Gung Memorial Hospital, Chiayi, Taiwan; School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chuan-Pin Lee
- Center of Excellence for Chang-Gung Research Datalink, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Robert Stewart
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; South London and Maudsley NHS Foundation Trust, London, UK
| | - Shu-I Wu
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan; Section of Psychiatry, MacKay Memorial Hospital, Taipei, Taiwan.
| |
Collapse
|
33
|
Wang R, Lan C, Benlagha K, Camara NOS, Miller H, Kubo M, Heegaard S, Lee P, Yang L, Forsman H, Li X, Zhai Z, Liu C. The interaction of innate immune and adaptive immune system. MedComm (Beijing) 2024; 5:e714. [PMID: 39286776 PMCID: PMC11401974 DOI: 10.1002/mco2.714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 08/11/2024] [Accepted: 08/11/2024] [Indexed: 09/19/2024] Open
Abstract
The innate immune system serves as the body's first line of defense, utilizing pattern recognition receptors like Toll-like receptors to detect pathogens and initiate rapid response mechanisms. Following this initial response, adaptive immunity provides highly specific and sustained killing of pathogens via B cells, T cells, and antibodies. Traditionally, it has been assumed that innate immunity activates adaptive immunity; however, recent studies have revealed more complex interactions. This review provides a detailed dissection of the composition and function of the innate and adaptive immune systems, emphasizing their synergistic roles in physiological and pathological contexts, providing new insights into the link between these two forms of immunity. Precise regulation of both immune systems at the same time is more beneficial in the fight against immune-related diseases, for example, the cGAS-STING pathway has been found to play an important role in infections and cancers. In addition, this paper summarizes the challenges and future directions in the field of immunity, including the latest single-cell sequencing technologies, CAR-T cell therapy, and immune checkpoint inhibitors. By summarizing these developments, this review aims to enhance our understanding of the complexity interactions between innate and adaptive immunity and provides new perspectives in understanding the immune system.
Collapse
Affiliation(s)
- Ruyuan Wang
- Department of Thyroid and Breast Surgery Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Caini Lan
- Cancer Center Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Kamel Benlagha
- Alloimmunity, Autoimmunity and Transplantation Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160 Paris France
| | - Niels Olsen Saraiva Camara
- Department of Immunology Institute of Biomedical Sciences University of São Paulo (USP) São Paulo São Paulo Brazil
| | - Heather Miller
- Coxiella Pathogenesis Section, Laboratory of Bacteriology Rocky Mountain Laboratories National Institute of Allergy and Infectious Diseases, National Institutes of Health Hamilton Montana USA
| | - Masato Kubo
- Division of Molecular Pathology Research Institute for Biomedical Sciences (RIBS) Tokyo University of Science Noda Chiba Japan
| | - Steffen Heegaard
- Department of Ophthalmology Rigshospitalet Hospital Copenhagen University Copenhagen Denmark
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong China
| | - Lu Yang
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology Wuhan Hubei China
| | - Huamei Forsman
- Department of Laboratory Medicine Institute of Biomedicine, University of Gothenburg Gothenburg Sweden
| | - Xingrui Li
- Department of Thyroid and Breast Surgery Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Zhimin Zhai
- Department of Hematology The Second Hospital of Anhui Medical University Hefei China
| | - Chaohong Liu
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology Wuhan Hubei China
| |
Collapse
|
34
|
Oh SJ, Nguyen TT, Seo Y, Park HJ, Ahn JS, Shin YY, Kang BJ, Jang M, Park J, Jeong JH, Kim HS. Sustained release of stem cell secretome from nano-villi chitosan microspheres for effective treatment of atopic dermatitis. Int J Biol Macromol 2024; 277:134344. [PMID: 39089545 DOI: 10.1016/j.ijbiomac.2024.134344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 07/01/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Canine atopic dermatitis (AD) arises from hypersensitive immune reactions. AD symptoms entail severe pruritus and skin inflammation, with frequent relapses. Consequently, AD patients require continuous management, imposing financial burdens and mental fatigue on pet owners. In this study, we aimed to investigate the therapeutic relevance of secretome from canine adipose tissue-derived mesenchymal stem cells (MSCs), especially after encapsulation in nano-villi chitosan microspheres (CS-MS) to expect improved efficacy. Conditioned media (CM) from MSCs significantly inhibited the proliferation of splenocytes, induced the generation of regulatory T cells, and decreased mast cell degranulation. We found that beneficial soluble factors known to reduce AD symptoms, including transforming growth factor-beta 1, were detectable after sequential concentration and lyophilization of CM. The CS-MS, developed by a phase inversion regeneration method, showed high loading and sustained release of the secretome. Local injection of secretome-loaded CS-MS (ST/SC-MS) effectively reduced clinical severity compared to groups treated with secretome. Histological analysis revealed that ST/SC-MS potently suppressed epidermal hyperplasia, immunocyte infiltration and mast cell activation in the lesion. Taken together, this study presents a novel therapeutic approach exhibiting more potent and prolonged immunoregulatory efficacy of MSC secretome for canine AD treatment.
Collapse
Affiliation(s)
- Su-Jeong Oh
- Department of Oral Biochemistry, Dental and Life Science Institute, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Tiep Tien Nguyen
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea; Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yoojin Seo
- Department of Oral Biochemistry, Dental and Life Science Institute, Pusan National University, Yangsan 50612, Republic of Korea
| | - Hee-Jeong Park
- Department of Oral Biochemistry, Dental and Life Science Institute, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ji-Su Ahn
- Department of Oral Biochemistry, Dental and Life Science Institute, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ye Young Shin
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co. Ltd., Seoul 08590, Republic of Korea
| | - Byung-Jae Kang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea; BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Min Jang
- Department of Veterinary Surgery, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Junhyeung Park
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Hyung-Sik Kim
- Department of Oral Biochemistry, Dental and Life Science Institute, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea.
| |
Collapse
|
35
|
Patel AV, Stevens A, Shahzad H, Iyer AI, Ragland D, Cvetanovich G, Bishop JY, Rauck RC. The effect of allergies on outcomes following shoulder arthroplasty: A national database analysis of 154,478 patients. Shoulder Elbow 2024; 16:527-533. [PMID: 39493408 PMCID: PMC11528767 DOI: 10.1177/17585732231194615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/24/2023] [Indexed: 11/05/2024]
Abstract
Background The objective of this study is to investigate the effect of allergies on complications following total shoulder arthroplasty (TSA). Materials and Methods All data were collected by using the PearlDiver national database to identify patients who had undergone TSA (anatomic or reverse) between 1 January 2010 and 31 October 2021. Patients were stratified into two groups: allergies and no allergies. Comparisons were made regarding complications and revision surgeries. Complications were compared at 30 and 90 days. Revisions were compared at 1, 5 and 10 years postoperatively. Results This study identified 28,182 patients with allergies and 126,296 patients without allergies. Patients with allergies were more likely to require revision surgery at all time points analyzed (p < 0.001). Patients with allergies were more likely to have sepsis within 30 (OR 1.53 [1.30-1.80], p < 0.001) and 90 days (1.71 [1.51-1.94], p < 0.001) postoperatively. Patients with allergies were more likely to experience a wound complication within 30 (1.89 [1.58-2.26], p < 0.001) and 90 days (1.81 [1.58-2.08], p < 0.001). The allergy group experienced higher rates of prosthetic joint infections (PJI) (2.14 [1.81-2.54], p < 0.001) and implant complications at 90 days (1.52 [1.42-1.62], p < 0.001). Discussion Patients with allergies were more likely to require revision surgery, experience wound complications, sepsis and PJI following TSA.
Collapse
Affiliation(s)
- Akshar V Patel
- Department of Orthopaedics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Andrew Stevens
- Department of Orthopaedics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Hania Shahzad
- Department of Orthopaedics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Amogh I Iyer
- Department of Orthopaedics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Dashaun Ragland
- Department of Orthopaedics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Gregory Cvetanovich
- Department of Orthopaedics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Julie Y Bishop
- Department of Orthopaedics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Ryan C Rauck
- Department of Orthopaedics, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
36
|
Scotland BL, Dharmaraj S, Cottingham AL, Truong N, Chapoval SP, Keegan AD, Pearson RM. Impact of antigen loading in tolerogenic nanoparticles to mitigate Th2-mediated allergic lung inflammation. Drug Deliv Transl Res 2024; 14:2930-2944. [PMID: 38862755 PMCID: PMC11608010 DOI: 10.1007/s13346-024-01632-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2024] [Indexed: 06/13/2024]
Abstract
Allergic disease is a major global health concern that imposes significant life-altering and economic burdens on affected individuals. However, there is still no cure. Polymer-based nanoparticles (NP) have shown the potential to induce antigen (Ag)-specific immune tolerance in various Th1/17 and Th2-mediated immune disorders including autoimmunity and allergy. Common methods by which Ags are associated with NPs are through surface conjugation or encapsulation. However, these Ag delivery strategies can be associated with several caveats that dampen their effectiveness such as uncontrolled Ag loading, a high Ag burst release, and an increased immune recognition profile. We previously developed Ag-polymer conjugate NPs (acNPs) to overcome those noted limitations, while allowing for controlled delivery of precise quantities of Ag to innate immune cells for Ag-specific CD4 T cell modulation. Here, we utilized ovalbumin (OVA) protein-poly(lactic-co-glycolic acid) (PLGA) conjugate NPs (acNP-OVA) to elucidate the impact of Ag loading on the induction of Th2 tolerance using a prophylactic and therapeutic OVA/ALUM-induced mouse model of allergic lung inflammation (ALI) in comparison to Ag-encapsulated PLGA NPs (NP(Ag)). We demonstrate that acNP-OVA formulations reduced OVA-specific IgE and inhibited Th2 cytokine secretions in an Ag loading-dependent manner when administered prophylactically. Administration of acNP-OVA to pre-sensitized mice did not affect OVA-specific IgE and Th2 cytokines tended to be reduced, however, there was no clear Ag loading dependency. acNP-OVA with medium-to-low Ag loadings were well tolerated, while formulations with high Ag loadings, including NP(Ag) resulted in anaphylaxis. Overall, our results clarify the relationship between Ag loading and Ag-specific IgE and Th2 cytokine responses in a murine model of ALI, which provides insight useful for future design of tolerogenic NP-based immunotherapies.
Collapse
Affiliation(s)
- Brianna L Scotland
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD, 21201, USA
| | - Shruti Dharmaraj
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD, 21201, USA
| | - Andrea L Cottingham
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD, 21201, USA
| | - Nhu Truong
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD, 21201, USA
| | - Svetlana P Chapoval
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD, 21201, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, 800 W. Baltimore Street, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA
| | - Achsah D Keegan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD, 21201, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, 800 W. Baltimore Street, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA
| | - Ryan M Pearson
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD, 21201, USA.
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD, 21201, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA.
| |
Collapse
|
37
|
Ashour A, Amen Y, Allam AE, Mwakalukwa R, Niwa Y, Shimizu K. The peels of sacred Lotus (Nelumbo nucifera) rhizomes suppress allergic reactions by inhibiting the A23187-induced degranulation in rat basophilic leukemia (RBL2H3) cells. Fitoterapia 2024; 178:106153. [PMID: 39089597 DOI: 10.1016/j.fitote.2024.106153] [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: 06/04/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
We isolated ten compounds from methanolic extract of the peels of sacred lotus (Nelumbo nucifera) rhizomes which were identified as β-sitosterol linoleate 1, β-sitosterol 2, lupeol 3, stigmasterol 3-O-β-D-glucoside 4, oleanolic acid 5, betulinic acid 6, pinoresinol 7, 4-hydroxybenzoic acid 8, catechin 9 and gallocatechin 10. All of the isolated compounds from the peels of sacred lotus rhizomes are reported for the first time, and were investigated for their anti-allergic activity. We found that three of them, stigmasterol 3-O-β-D-glucoside 4, oleanolic acid 5 and pinoresinol 7, were capable of inhibiting A23187-induced degranulation in RBL-2H3 cells with IC50 values 0.18 ± 0.01 mM, 0.28 ± 0.06 mM, and 0.27 ± 0.01 mM, respectively. With an exception to 4, compounds 5 and 7 achieved the anti-allergic effect without affecting the cells viability even at higher concentrations with their selectivity indices (SI) being >5. By reducing A23187-induced degranulation, it is suggestive of a mechanism attenuation of Ca2+ elevation. Our findings suggest that, the peels of sacred lotus rhizomes would be beneficial for providing an inexpensive source for the production of bioactive compounds with anti-allergic effect.
Collapse
Affiliation(s)
- Ahmed Ashour
- Department of Pharmacognosy, Faculty of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Yhiya Amen
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Division of Systematic Forest and Forest Products Sciences, Department of Agro-Environmental Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, 812-8581, Japan
| | - Ahmed E Allam
- Division of Systematic Forest and Forest Products Sciences, Department of Agro-Environmental Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, 812-8581, Japan; Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Rogers Mwakalukwa
- Division of Systematic Forest and Forest Products Sciences, Department of Agro-Environmental Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, 812-8581, Japan; Department of Pharmacognosy, School of Pharmacy, Muhimbili University of Health and Allied Sciences, Tanzania
| | - Yasuharu Niwa
- Division of Systematic Forest and Forest Products Sciences, Department of Agro-Environmental Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, 812-8581, Japan
| | - Kuniyoshi Shimizu
- Division of Systematic Forest and Forest Products Sciences, Department of Agro-Environmental Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, 812-8581, Japan.
| |
Collapse
|
38
|
Stevenson M, Hebron ML, Liu X, Balaraman K, Wolf C, Moussa C. c-KIT inhibitors reduce pathology and improve behavior in the Tg(SwDI) model of Alzheimer's disease. Life Sci Alliance 2024; 7:e202402625. [PMID: 39009412 PMCID: PMC11249953 DOI: 10.26508/lsa.202402625] [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] [Received: 01/30/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
Treatments for Alzheimer's disease have primarily focused on removing brain amyloid plaques to improve cognitive outcomes in patients. We developed small compounds, known as BK40143 and BK40197, and we hypothesize that these drugs alleviate microglial-mediated neuroinflammation and induce autophagic clearance of neurotoxic proteins to improve behavior in models of neurodegeneration. Specificity binding assays of BK40143 and BK40197 showed primary binding to c-KIT/Platelet Derived Growth Factor Receptors (PDGFR)α/β, whereas BK40197 also differentially binds to FYVE finger-containing phosphoinositide kinase (PIKFYVE). Both compounds penetrate the CNS, and treatment with these drugs inhibited the maturation of peripheral mast cells in transgenic mice, correlating with cognitive improvements on measures of memory and anxiety. In the brain, microglial activation was profoundly attenuated and amyloid-beta and tau were reduced via autophagy. Multi-kinase inhibition, including c-KIT, exerts multifunctional effects to reduce neurodegenerative pathology via autophagy and microglial activity and may represent a potential therapeutic option for neurodegeneration.
Collapse
Affiliation(s)
- Max Stevenson
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington DC, USA
| | - Michaeline L Hebron
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington DC, USA
| | - Xiaoguang Liu
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington DC, USA
| | - Kaluvu Balaraman
- Medicinal Chemistry Shared Resource, Department of Chemistry, Georgetown University Medical Center, Washington DC, USA
| | - Christian Wolf
- Medicinal Chemistry Shared Resource, Department of Chemistry, Georgetown University Medical Center, Washington DC, USA
| | - Charbel Moussa
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington DC, USA
| |
Collapse
|
39
|
Zhang Y, Xie Y, Zhang X, Duan C, Ma J, Wang Y, Wu Y, Shan N, Cheng K, Zhuang R, Bian K. CD226 implicated in Akt-dependent apoptosis of CD4 + T cell contributes to asthmatic pathogenesis. Cell Death Dis 2024; 15:705. [PMID: 39349422 PMCID: PMC11442704 DOI: 10.1038/s41419-024-07080-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 10/02/2024]
Abstract
Asthma is a chronic airway inflammatory disease in which CD4+ T cell dysregulation occurs. Here, we investigated the molecular role and clinical significance of CD226, a costimulatory molecule of T lymphocytes, in the development of allergic asthma. Our results revealed that the expression of CD226 was significantly increased in CD4+ effector T cells, especially in T helper (Th) 2 cells and Th17 cells in patients with asthma. Moreover, CD4+ T cell-specific Cd226-knockout mice were generated and together with littermates were challenged with ovalbumin (OVA) to establish a model of allergic asthma. We found that CD226 deficiency in CD4+ T cells mitigated lung inflammation, IgE production, and eosinophil infiltration and reduced airway remodeling in experimental allergic asthma. However, the impact of CD226 on asthma was independent of Treg cell modulation. Through RNA-seq data analysis, the apoptosis pathway was screened. Mechanistically, CD226 deletion promoted CD4+ T cell late apoptosis via the activation of Caspase-3 in an Akt-dependent manner. Furthermore, blocking CD226 signaling with a recombinant fusion protein attenuated asthma features in mice and achieved a good therapeutic effect. Overall, this study revealed a unique role of CD226 in CD4+ T cell regulation in asthma pathogenesis. Therefore, targeting CD226 may provide new insights into the clinical treatment of asthma.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Otolaryngology Head and Neck Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yang Xie
- Department of Otolaryngology Head and Neck Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xuexin Zhang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chujun Duan
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Jingchang Ma
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuling Wang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yilin Wu
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Niqi Shan
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Kun Cheng
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ran Zhuang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Ka Bian
- Department of Otolaryngology Head and Neck Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
40
|
Wang Y, Zhang L, Shi B, Luo J. Trends and research foci in immunoregulatory mechanisms of allergic rhinitis: a bibliometric analysis (2014-2024). Front Immunol 2024; 15:1443954. [PMID: 39380999 PMCID: PMC11458462 DOI: 10.3389/fimmu.2024.1443954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
Background This study aims to provide a comprehensive bibliometric analysis of research trends, hotspots, and future directions in the immunoregulatory mechanisms of allergic rhinitis (AR) from 2014 to 2024. Methods Data were sourced from the Web of Science Core Collection (WoSCC), covering articles and reviews published between April 1, 2014, and March 31, 2024. The search terms included "Allergic Rhinitis," "AR," and related terms along with specific keywords related to immune cells and inflammatory mediators. Bibliometric tools such as CiteSpace, VOSviewer, and SCImago Graphica were used to analyze institutional cooperation networks, keyword co-occurrence, citation bursts, and research topic evolution. Microsoft Excel 2019 was employed to display annual publication trends. Results A total of 2200 papers met the inclusion and exclusion criteria. The number of publications showed an upward trend over the past decade, with a significant peak in 2021. China (583 papers) and the United States (454 papers) were the major contributing countries. Imperial College London emerged as the leading institution. Key research frontiers identified include the roles of NF kappa B and air pollution in AR. Keyword burst analysis revealed emerging topics such as respiratory allergy and personalized treatment strategies. Notable limitations include the exclusive use of the WoSCC database and the restriction to English-language publications. Conclusion The field of immunoregulatory mechanisms in allergic rhinitis has seen significant growth, with China and the United States leading the research. Future research should focus on developing personalized treatment plans and understanding the comprehensive impact of environmental factors. Continued interdisciplinary collaboration and international cooperation will be essential for advancing therapeutic strategies in AR.
Collapse
Affiliation(s)
- Yandan Wang
- Department of Otolaryngology, Huaihe Hospital, Henan University, Kaifeng, China
| | - Liangran Zhang
- Department of Otolaryngology, Huaihe Hospital, Henan University, Kaifeng, China
| | - Baoyuan Shi
- Department of Otolaryngology, Huaihe Hospital, Henan University, Kaifeng, China
| | - Junpeng Luo
- Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, China
| |
Collapse
|
41
|
Wang X, Cao L, Liu S, Zhou Y, Zhou J, Zhao W, Gao S, Liu R, Shi Y, Shao C, Fang J. The critical roles of IGFs in immune modulation and inflammation. Cytokine 2024; 183:156750. [PMID: 39243567 DOI: 10.1016/j.cyto.2024.156750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/31/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Insulin-like growth factors (IGFs) are crucial for embryonic and postnatal growth and development, influencing cell survival, metabolism, myogenesis, and cancer progression. Many studies have demonstrated that IGFs also play prominent roles in the modulation of both innate and adaptive immune systems during inflammation. Strikingly, IGFs dictate the phenotype and functional properties of macrophages and T cells. Furthermore, the interplay between IGFs and inflammatory cytokines may generate tissue-protective properties during inflammation. Herein, we review the recent advances on the dialogue between immune cells and IGFs, especially zooming in on the significance of immunomodulatory properties in inflammatory conditions, cancer and autoimmune diseases. The investigation of IGFs may have broad clinical implications.
Collapse
Affiliation(s)
- Xin Wang
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Lijuan Cao
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China; Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Shisong Liu
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yipeng Zhou
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jiarui Zhou
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Wenxuan Zhao
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Shengqi Gao
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Rui Liu
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China; Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Yufang Shi
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China; Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Changshun Shao
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China.
| | - Jiankai Fang
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
42
|
Vásquez-Pérez JM, González-Guevara E, Gutiérrez-Buenabad D, Martínez-Gopar PE, Martinez-Lazcano JC, Cárdenas G. Is Nasal Dysbiosis a Required Component for Neuroinflammation in Major Depressive Disorder? Mol Neurobiol 2024:10.1007/s12035-024-04375-2. [PMID: 39120823 DOI: 10.1007/s12035-024-04375-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024]
Abstract
Human microbiota is known to influence immune and cerebral responses by direct and/or indirect mechanisms, including hypothalamic-pituitary-adrenal axis signaling, activation of neural afferent circuits to the brain, and by altering the peripheral immune responses (cellular and humoral immune function, circulatory inflammatory cells, and the production of several inflammatory mediators, such as cytokines, chemokines, and reactive oxygen species). The inflammatory responses in the nasal mucosa (rhinitis) or paranasal sinuses (chronic rhinosinusitis) are dual conditions related with a greater risk for developing depression. In the nasal cavity, anatomic components of the olfactive function are in direct contact with the CNS through the olfactory receptors, neurons, and axons that end in the olfactory bulb and the entorhinal cortex. Local microbiome alterations (dysbiosis) are linked to transepithelial translocation of microorganisms and their metabolites, which disrupts the epithelial barrier and favors vascular permeability, increasing the levels of several inflammatory molecules (both cytokines and non-cytokine mediators: extracellular vesicles (exosomes) and neuropeptides), triggering local inflammation (rhinitis) and the spread of these components into the central nervous system (neuroinflammation). In this review, we discuss the role of microbiota-related immunity in conditions affecting the nasal mucosa (chronic rhinosinusitis and allergic rhinitis) and their relevance in major depressive disorders, focusing on the few mechanisms known to be involved and providing some hypothetical proposals on the pathophysiology of depression.
Collapse
Affiliation(s)
- Jorge Manuel Vásquez-Pérez
- Laboratorio de Neurogénesis, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, 14370, Ciudad de México, Mexico
- Programa de Posgrado Doctorado en Ciencias Biomédicas, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Ciudad Universitaria, Coyoacán, Ciudad de Mexico, Mexico
| | - Edith González-Guevara
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, 14269, Ciudad de México, Mexico
| | - Diana Gutiérrez-Buenabad
- Laboratorio de Neurogénesis, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, 14370, Ciudad de México, Mexico
- Programa de Posgrado Doctorado en Ciencias Biomédicas, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Ciudad Universitaria, Coyoacán, Ciudad de Mexico, Mexico
| | - Pablo Eliasib Martínez-Gopar
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, 14269, Ciudad de México, Mexico
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del IPN, Unidad Sede Sur, 14330, Ciudad de México, Mexico
| | - Juan Carlos Martinez-Lazcano
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, 14269, Ciudad de México, Mexico
| | - Graciela Cárdenas
- Departamento de Neurología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Insurgentes Sur 3877, La Fama, Tlalpan, 14269, Ciudad de Mexico, Mexico.
| |
Collapse
|
43
|
Delgado Dolset MI, Pablo-Torres C, Contreras N, Couto-Rodríguez A, Escolar-Peña A, Graña-Castro O, Izquierdo E, López-Rodríguez JC, Macías-Camero A, Pérez-Gordo M, Villaseñor A, Zubeldia-Varela E, Barber D, Escribese MM. Severe Allergy as a Chronic Inflammatory Condition From a Systems Biology Perspective. Clin Exp Allergy 2024; 54:550-584. [PMID: 38938054 DOI: 10.1111/cea.14517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/14/2024] [Accepted: 05/26/2024] [Indexed: 06/29/2024]
Abstract
Persistent and unresolved inflammation is a common underlying factor observed in several and seemingly unrelated human diseases, including cardiovascular and neurodegenerative diseases. Particularly, in atopic conditions, acute inflammatory responses such as those triggered by insect venom, food or drug allergies possess also a life-threatening potential. However, respiratory allergies predominantly exhibit late immune responses associated with chronic inflammation, that can eventually progress into a severe phenotype displaying similar features as those observed in other chronic inflammatory diseases, as is the case of uncontrolled severe asthma. This review aims to explore the different facets and systems involved in chronic allergic inflammation, including processes such as tissue remodelling and immune cell dysregulation, as well as genetic, metabolic and microbiota alterations, which are common to other inflammatory conditions. Our goal here was to deepen on the understanding of an entangled disease as is chronic allergic inflammation and expose potential avenues for the development of better diagnostic and intervention strategies.
Collapse
Affiliation(s)
- M I Delgado Dolset
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - C Pablo-Torres
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - N Contreras
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Couto-Rodríguez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Escolar-Peña
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - O Graña-Castro
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - E Izquierdo
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - J C López-Rodríguez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Macías-Camero
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - M Pérez-Gordo
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Villaseñor
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - E Zubeldia-Varela
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - D Barber
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - M M Escribese
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| |
Collapse
|
44
|
Wiciński M, Fajkiel-Madajczyk A, Kurant Z, Liss S, Szyperski P, Szambelan M, Gromadzki B, Rupniak I, Słupski M, Sadowska-Krawczenko I. Ashwagandha's Multifaceted Effects on Human Health: Impact on Vascular Endothelium, Inflammation, Lipid Metabolism, and Cardiovascular Outcomes-A Review. Nutrients 2024; 16:2481. [PMID: 39125360 PMCID: PMC11314093 DOI: 10.3390/nu16152481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Withania somnifera, commonly known as Ashwagandha, has been popular for many years. Numerous studies have shown that the extract of this plant, due to its wealth of active substances, can induce anti-inflammatory, neuroprotective, immunomodulatory, hepatoprotective, cardioprotective, anti-diabetic, adaptogenic, anti-arthritic, anti-stress, and antimicrobial effects. This review examines the impact of Ashwagandha extract on the vascular endothelium, inflammation, lipid metabolism, and cardiovascular outcomes. Studies have shown that Ashwagandha extracts exhibit an anti-angiogenic effect by inhibiting vascular endothelial growth factor (VEGF)-induced capillary sprouting and formation by lowering the mean density of microvessels. Furthermore, the results of numerous studies highlight the anti-inflammatory role of Ashwagandha extract, as the action of this plant causes a decrease in the expression of pro-inflammatory cytokines. Interestingly, withanolides, present in Ashwagandha root, have shown the ability to inhibit the differentiation of preadipocytes into adipocytes. Research results have also proved that W. somnifera demonstrates cardioprotective effects due to its antioxidant properties and reduces ischemia/reperfusion-induced apoptosis. It seems that this plant can be successfully used as a potential treatment for several conditions, mainly those with increased inflammation. More research is needed to elucidate the exact mechanisms by which the substances contained in W. somnifera extracts can act in the human body.
Collapse
Affiliation(s)
- Michał Wiciński
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie Skłodowskiej 9, 85-094 Bydgoszcz, Poland; (M.W.); (Z.K.); (S.L.); (P.S.); (M.S.); (B.G.)
| | - Anna Fajkiel-Madajczyk
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie Skłodowskiej 9, 85-094 Bydgoszcz, Poland; (M.W.); (Z.K.); (S.L.); (P.S.); (M.S.); (B.G.)
| | - Zuzanna Kurant
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie Skłodowskiej 9, 85-094 Bydgoszcz, Poland; (M.W.); (Z.K.); (S.L.); (P.S.); (M.S.); (B.G.)
| | - Sara Liss
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie Skłodowskiej 9, 85-094 Bydgoszcz, Poland; (M.W.); (Z.K.); (S.L.); (P.S.); (M.S.); (B.G.)
| | - Paweł Szyperski
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie Skłodowskiej 9, 85-094 Bydgoszcz, Poland; (M.W.); (Z.K.); (S.L.); (P.S.); (M.S.); (B.G.)
| | - Monika Szambelan
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie Skłodowskiej 9, 85-094 Bydgoszcz, Poland; (M.W.); (Z.K.); (S.L.); (P.S.); (M.S.); (B.G.)
| | - Bartłomiej Gromadzki
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie Skłodowskiej 9, 85-094 Bydgoszcz, Poland; (M.W.); (Z.K.); (S.L.); (P.S.); (M.S.); (B.G.)
| | - Iga Rupniak
- Department of Neonatology, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, ul. Ujejskiego 75, 85-168 Bydgoszcz, Poland; (I.R.); (I.S.-K.)
| | - Maciej Słupski
- Department of Hepatobiliary and General Surgery, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie Skłodowskiej 9, 85-094 Bydgoszcz, Poland;
| | - Iwona Sadowska-Krawczenko
- Department of Neonatology, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, ul. Ujejskiego 75, 85-168 Bydgoszcz, Poland; (I.R.); (I.S.-K.)
| |
Collapse
|
45
|
Cheng YH, Wu HI, Chen YY, Lee YH, Wang BJ, Wang YJ. Adverse outcome pathway-based approach to reveal the mechanisms of skin sensitization and long-term aging effects of chlorothalonil. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135176. [PMID: 39029193 DOI: 10.1016/j.jhazmat.2024.135176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/21/2024]
Abstract
Chlorothalonil (CHT) is a widely used antifungal agent and is reported to be a sensitizer that can cause allergic contact dermatitis (ACD). ACD initiation is associated with various innate immune cell contributions and is usually accompanied by persistent inflammation, which is a potential contributing factor to skin damage. However, detailed information on the mechanisms by which CHT induces skin sensitization and damage is still insufficient. This study focused on investigating the possible sensitization process and mechanism of CHT and the adverse effects of repeated CHT exposure. CHT activates dendritic cells and promotes the proliferation of lymph cells in the skin sensitization phase, causing severe inflammation. Keratinocytes activate the NLRP3 inflammasome pathway to cause inflammation during CHT treatment, and macrophages also secrete inflammatory cytokines. In addition, CHT-induced inflammation triggered skin wrinkles, decreased epidermal thickness and decreased collagen. Cell experiments also showed that repeated exposure to CHT led to cell proliferation inhibition and senescence, and CHT-induced autophagy dysfunction was not only the reason for inflammation but also for senescence. This study defined the possible process through which CHT is involved in the skin sensitization phase and elucidated the mechanism of CHT-induced inflammation in innate immune responses. We also determined that repeated CHT exposure caused persistent inflammation, ultimately leading to skin aging.
Collapse
Affiliation(s)
- Yung-Hsuan Cheng
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 70428, Taiwan, ROC
| | - Hsuan-I Wu
- Department of Food Safety, Hygiene and Risk Management, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 70428, Taiwan, ROC
| | - Yu-Ying Chen
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 70428, Taiwan, ROC
| | - Yu-Hsuan Lee
- Department of Cosmeceutics, China Medical University, Taichung, Taiwan, ROC
| | - Bour-Jr Wang
- Department of Cosmetic Science and Institute of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan, ROC; Department of Occupational and Environmental Medicine, National Cheng Kung University Hospital, Tainan 70403, Taiwan, ROC.
| | - Ying-Jan Wang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 70428, Taiwan, ROC; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC.
| |
Collapse
|
46
|
Ho G, Lam L, Tran T, Wei J, Hashimoto M. Innate neuroimmunity across aging and neurodegeneration: a perspective from amyloidogenic evolvability. Front Cell Dev Biol 2024; 12:1430593. [PMID: 39071802 PMCID: PMC11272618 DOI: 10.3389/fcell.2024.1430593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/17/2024] [Indexed: 07/30/2024] Open
Abstract
In Alzheimer's Disease (AD), amyloidogenic proteins (APs), such as β-amyloid (Aβ) and tau, may act as alarmins/damage-associated molecular patterns (DAMPs) to stimulate neuroinflammation and cell death. Indeed, recent evidence suggests that brain-specific type 2 immune networks may be important in modulating amyloidogenicity and brain homeostasis. Central to this, components of innate neuroimmune signaling, particularly type 2 components, assume distinctly specialized roles in regulating immune homeostasis and brain function. Whereas balanced immune surveillance stems from normal type 2 brain immune function, appropriate microglial clearance of aggregated misfolded proteins and neurotrophic and synaptotrophic signaling, aberrant pro-inflammatory activity triggered by alarmins might disrupt this normal immune homeostasis with reduced microglial amyloid clearance, synaptic loss, and ultimately neurodegeneration. Furthermore, since increased inflammation may in turn cause neurodegeneration, it is predicted that AP aggregation and neuroinflammation could synergistically promote even more damage. The reasons for maintaining such adverse biological conditions which have not been weeded out during evolution remain unclear. Here, we discuss these issues from a viewpoint of amyloidogenic evolvability, namely, aEVO, a hypothetic view of an adaptation to environmental stress by AP aggregates. Speculatively, the interaction of AP aggregation and neuroinflammation for aEVO in reproduction, which is evolutionally beneficial, might become a co-activating relationship which promotes AD pathogenesis through antagonistic pleiotropy. If validated, simultaneously suppressing both AP aggregation and specific innate neuroinflammation could greatly increase therapeutic efficacy in AD. Overall, combining a better understanding of innate neuroimmunity in aging and disease with the aEVO hypothesis may help uncover novel mechanism of pathogenesis of AD, leading to improved diagnostics and treatments.
Collapse
Affiliation(s)
- Gilbert Ho
- PCND Neuroscience Research Institute, Poway, CA, United States
| | - Linh Lam
- PCND Neuroscience Research Institute, Poway, CA, United States
| | - Tony Tran
- PCND Neuroscience Research Institute, Poway, CA, United States
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, China
| | | |
Collapse
|
47
|
Shen Q, Cao M, Yu C, Tang J, Song L, Ding Y, Ju L, Wei JF, Li L, Huang W. Biodegradable Mesoporous Organosilica-Based Nanostabilizer Targeting Mast Cells for Long-Term Treatment of Allergic Diseases. ACS NANO 2024; 18:16934-16946. [PMID: 38907988 DOI: 10.1021/acsnano.4c03069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Allergic diseases are immune system dysfunctions mediated by mast cell (MC) activation stimulated by specific allergens. However, current small molecular MC stabilizers for allergic disease prevention often require multiple doses over a long period of time and are associated with serious side effects. Herein, we develop a diselenide-bridged mesoporous silica nanostabilizer, proving that it could specifically target sensitized MCs via the recognition of IgE aptamer and IgE. Meantime, the IgE aptamer can also mitigate allergic reactions by preventing re-exposure of allergens from the surface of sensitized MCs. Furthermore, the diselenide-bridged scaffold can be reduced by the intracellular excessive ROS, subsequently achieving redox homeostasis via ROS depletion. Finally, the precise release of small molecular MC stabilizers along with the biodegradation of nanocarrier can stabilize the membranes of MCs. In vivo assays in passive cutaneous anaphylactic (PCA) and allergic rhinitis (AR) mice indicated that our current strategy further endowed it with a high efficacy, long-term therapeutic time window, as well as negligible inflammatory side effects for allergic diseases, offering a promising therapeutic strategy for the clinical generalization of allergic diseases.
Collapse
Affiliation(s)
- Qian Shen
- Key Laboratory of Flexible Electronics (KLOFE), School of Flexible Electronics (Future Technologies) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Mengda Cao
- Department of Pharmacy, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing 210009, China
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210044, China
| | - Changmin Yu
- Key Laboratory of Flexible Electronics (KLOFE), School of Flexible Electronics (Future Technologies) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Jian Tang
- Department of Pharmacy, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Lebin Song
- Department of Pharmacy, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Yanan Ding
- Key Laboratory of Flexible Electronics (KLOFE), School of Flexible Electronics (Future Technologies) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Linjie Ju
- Department of Pharmacy, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Ji-Fu Wei
- Department of Pharmacy, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Lin Li
- Key Laboratory of Flexible Electronics (KLOFE), School of Flexible Electronics (Future Technologies) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing 211816, China
- The Institute of Flexible Electronics, Xiamen University, Xiamen 361005, China
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE), School of Flexible Electronics (Future Technologies) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing 211816, China
- The Institute of Flexible Electronics, Xiamen University, Xiamen 361005, China
| |
Collapse
|
48
|
Basith S, Pham NT, Manavalan B, Lee G. SEP-AlgPro: An efficient allergen prediction tool utilizing traditional machine learning and deep learning techniques with protein language model features. Int J Biol Macromol 2024; 273:133085. [PMID: 38871100 DOI: 10.1016/j.ijbiomac.2024.133085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 05/20/2024] [Accepted: 06/09/2024] [Indexed: 06/15/2024]
Abstract
Allergy is a hypersensitive condition in which individuals develop objective symptoms when exposed to harmless substances at a dose that would cause no harm to a "normal" person. Most current computational methods for allergen identification rely on homology or conventional machine learning using limited set of feature descriptors or validation on specific datasets, making them inefficient and inaccurate. Here, we propose SEP-AlgPro for the accurate identification of allergen protein from sequence information. We analyzed 10 conventional protein-based features and 14 different features derived from protein language models to gauge their effectiveness in differentiating allergens from non-allergens using 15 different classifiers. However, the final optimized model employs top 10 feature descriptors with top seven machine learning classifiers. Results show that the features derived from protein language models exhibit superior discriminative capabilities compared to traditional feature sets. This enabled us to select the most discriminatory baseline models, whose predicted outputs were aggregated and used as input to a deep neural network for the final allergen prediction. Extensive case studies showed that SEP-AlgPro outperforms state-of-the-art predictors in accurately identifying allergens. A user-friendly web server was developed and made freely available at https://balalab-skku.org/SEP-AlgPro/, making it a powerful tool for identifying potential allergens.
Collapse
Affiliation(s)
- Shaherin Basith
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea.
| | - Nhat Truong Pham
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Balachandran Manavalan
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Gwang Lee
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea.
| |
Collapse
|
49
|
Khan M, Banerjee S, Muskawad S, Maity R, Chowdhury SR, Ejaz R, Kuuzie E, Satnarine T. The Impact of Artificial Intelligence on Allergy Diagnosis and Treatment. Curr Allergy Asthma Rep 2024; 24:361-372. [PMID: 38954325 DOI: 10.1007/s11882-024-01152-y] [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] [Accepted: 05/19/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE OF REVIEW Artificial intelligence (AI), be it neuronal networks, machine learning or deep learning, has numerous beneficial effects on healthcare systems; however, its potential applications and diagnostic capabilities for immunologic diseases have yet to be explored. Understanding AI systems can help healthcare workers better assimilate artificial intelligence into their practice and unravel its potential in diagnostics, clinical research, and disease management. RECENT FINDINGS We reviewed recent advancements in AI systems and their integration in healthcare systems, along with their potential benefits in the diagnosis and management of diseases. We explored machine learning as employed in allergy diagnosis and its learning patterns from patient datasets, as well as the possible advantages of using AI in the field of research related to allergic reactions and even remote monitoring. Considering the ethical challenges and privacy concerns raised by clinicians and patients with regard to integrating AI in healthcare, we explored the new guidelines adapted by regulatory bodies. Despite these challenges, AI appears to have been successfully incorporated into various healthcare systems and is providing patient-centered solutions while simultaneously assisting healthcare workers. Artificial intelligence offers new hope in the field of immunologic disease diagnosis, monitoring, and management and thus has the potential to revolutionize healthcare systems.
Collapse
Affiliation(s)
- Maham Khan
- Fatima Jinnah Medical University, Lahore, Pakistan.
| | | | | | - Rick Maity
- Institute of Post Graduate Medical Education and Research, Kolkata, West Bengal, India
| | | | - Rida Ejaz
- Shifa College of Medicine, Islamabad, Pakistan
| | | | | |
Collapse
|
50
|
Li Z, Tian J, Yang F. Tyrosine nitration enhances the allergenic potential of house dust mite allergen Der p 2. ENVIRONMENTAL RESEARCH 2024; 252:118826. [PMID: 38579999 DOI: 10.1016/j.envres.2024.118826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/07/2024]
Abstract
Nitration of allergenic proteins caused by atmospheric pollutants O3 and NO2 may enhance their allergenic potential. In the study, the influence of nitration was investigated on the allergenicity of Der p 2, which is a main allergen from house dust mites and plays an important role in allergenic rhinitis and asthma. The results reveal that nitrated Der p 2 enhanced the IgE-binding capacity, upregulated the mRNA expression and release of IL-6 and IL-8 from bronchial epithelial cells, and induced higher levels of specific-IgE, TH2 cytokines and white blood cells in mice. Besides, nitrated Der p 2 caused more severe oxidative stress and allergenic symptoms in mice. It is concluded that nitration enhanced the allergenicity of Der p 2 through not only directly inducing higher amount of specific-IgE and stronger responses of TH2 cytokines, but also indirectly aggravating allergic symptoms by oxidative stress and adjuvant-like activation airway epithelial cells. The study suggests that the contribution of nitration to the promotion in allergenicity should not be ignored when precisely assessing the risk of house dust mite allergens in real environment.
Collapse
Affiliation(s)
- Zhiqi Li
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Natural Resources and Environmental Science, Zhejiang University, 310058, Hangzhou, China
| | - Jingyi Tian
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Natural Resources and Environmental Science, Zhejiang University, 310058, Hangzhou, China
| | - Fangxing Yang
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Natural Resources and Environmental Science, Zhejiang University, 310058, Hangzhou, China; Innovation Center of Yangtze River Delta, Zhejiang University, 314100, Jiashan, China.
| |
Collapse
|