1
|
Singh G, Thakur N, Kumar R. Nanoparticles in drinking water: Assessing health risks and regulatory challenges. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 949:174940. [PMID: 39047836 DOI: 10.1016/j.scitotenv.2024.174940] [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: 05/19/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
Nanoparticles (NPs) pose a significant concern in drinking water due to their potential health risks and environmental impact. This review provides a comprehensive analysis of the current understanding of NP sources and contamination in drinking water, focusing on health concerns, mitigation strategies, regulatory frameworks, and future perspectives. This review highlights the importance of nano-specific pathways, fate processes, health risks & toxicity, and the need for realistic toxicity assessments. Different NPs like titanium dioxide, silver, nanoplastics, nanoscale liquid crystal monomers, copper oxide, and others pose potential health risks through ingestion, inhalation, or dermal exposure, impacting organs and potentially leading to oxidative stress, inflammatory responses, DNA damage, cytotoxicity, disrupt intracellular energetic mechanisms, reactive oxygen species generation, respiratory and immune toxicity, and genotoxicity in humans. Utilizing case studies and literature reviews, we investigate the health risks associated with NPs in freshwater environments, emphasizing their relevance to drinking water quality. Various mitigation and treatment strategies, including filtration systems (e.g., reverse osmosis, and ultra/nano-filtration), adsorption processes, coagulation/flocculation, electrocoagulation, advanced oxidation processes, membrane distillation, and ultraviolet treatment, all of which demonstrate high removal efficiencies for NPs from drinking water. Regulatory frameworks and challenges for the production, applications, and disposal of NPs at both national and international levels are discussed, emphasizing the need for tailored regulations to address NP contamination and standardize safety testing and risk assessment practices. Looking ahead, this review underscores the necessity of advancing detection methods and nanomaterial-based treatment technologies while stressing the pivotal role of public awareness and tailored regulatory guidelines in upholding drinking water quality standards. This review emphasizes the urgency of addressing NP contamination in drinking water and provides insights into potential solutions and future research directions. Lastly, this review worth concluded with future recommendations on advanced analytical techniques and sensitive sensors for NP detection for safeguarding public health and policy implementations.
Collapse
Affiliation(s)
- Gagandeep Singh
- Department of Biosciences (UIBT), Chandigarh University, Ludhiana, Punjab 140413, India
| | - Neelam Thakur
- Department of Zoology, Sardar Patel University, Vallabh Government College, Campus, Mandi, Himachal Pradesh 175001, India.
| | - Rakesh Kumar
- Department of Biosystems Engineering, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
2
|
Lu J, Zhao XJ, Ruan Y, Liu XJ, Di X, Xu R, Wang JY, Qian MY, Jin HM, Li WJ, Shen X. Desloratadine ameliorates paclitaxel-induced peripheral neuropathy and hypersensitivity reactions in mice. Acta Pharmacol Sin 2024; 45:2061-2076. [PMID: 38789495 PMCID: PMC11420356 DOI: 10.1038/s41401-024-01301-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Paclitaxel (PTX) serves as a primary chemotherapy agent against diverse solid tumors including breast cancer, lung cancer, head and neck cancer and ovarian cancer, having severe adverse effects including PTX-induced peripheral neuropathy (PIPN) and hypersensitivity reactions (HSR). A recommended anti-allergic agent diphenhydramine (DIP) has been used to alleviate PTX-induced HSR. Desloratadine (DLT) is a third generation of histamine H1 receptor antagonist, but also acted as a selective antagonist of 5HTR2A. In this study we investigated whether DLT ameliorated PIPN-like symptoms in mice and the underlying mechanisms. PIPN was induced in male mice by injection of PTX (4 mg/kg, i.p.) every other day for 4 times. The mice exhibited 50% reduction in mechanical threshold, paw thermal response latency and paw cold response latency compared with control mice. PIPN mice were treated with DLT (10, 20 mg/kg, i.p.) 30 min before each PTX administration in the phase of establishing PIPN mice model and then administered daily for 4 weeks after the model was established. We showed that DLT administration dose-dependently elevated the mechanical, thermal and cold pain thresholds in PIPN mice, whereas administration of DIP (10 mg/kg, i.p.) had no ameliorative effects on PIPN-like symptoms. We found that the expression of 5HTR2A was selectively elevated in the activated spinal astrocytes of PIPN mice. Spinal cord-specific 5HTR2A knockdown by intrathecal injection of AAV9-5Htr2a-shRNA significantly alleviated the mechanical hyperalgesia, thermal and cold hypersensitivity in PIPN mice, while administration of DLT (20 mg/kg) did not further ameliorate PIPN-like symptoms. We demonstrated that DLT administration alleviated dorsal root ganglion neuronal damage and suppressed sciatic nerve destruction, spinal neuron apoptosis and neuroinflammation in the spinal cord of PIPN mice. Furthermore, we revealed that DLT administration suppressed astrocytic neuroinflammation via the 5HTR2A/c-Fos/NLRP3 pathway and blocked astrocyte-neuron crosstalk by targeting 5HTR2A. We conclude that spinal 5HTR2A inhibition holds promise as a therapeutic approach for PIPN and we emphasize the potential of DLT as a dual-functional agent in ameliorating PTX-induced both PIPN and HSR in chemotherapy. In summary, we determined that spinal 5HTR2A was selectively activated in PIPN mice and DLT could ameliorate the PTX-induced both PIPN- and HSR-like pathologies in mice. DLT alleviated the damages of DRG neurons and sciatic nerves, while restrained spinal neuronal apoptosis and CGRP release in PIPN mice. The underlying mechanisms were intensively investigated by assay against the PIPN mice with 5HTR2A-specific knockdown in the spinal cord by injection of adeno-associated virus 9 (AAV9)-5Htr2a-shRNA. DLT inhibited astrocytic NLRP3 inflammasome activation-mediated spinal neuronal damage through 5HTR2A/c-FOS pathway. Our findings have supported that spinal 5HTR2A inhibition shows promise as a therapeutic strategy for PIPN and highlighted the potential advantage of DLT as a dual-functional agent in preventing against PTX-induced both PIPN and HSR effects in anticancer chemotherapy.
Collapse
Affiliation(s)
- Jian Lu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xue-Jian Zhao
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuan Ruan
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiao-Jing Liu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xuan Di
- School of Pharmacy, Experiment Center for Science and Technology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rui Xu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jia-Ying Wang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Min-Yi Qian
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hong-Ming Jin
- School of Pharmacy, Experiment Center for Science and Technology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wen-Jun Li
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xu Shen
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
3
|
Nesovic LD, Gonzalez Cruz PE, Rychener N, Wilks LR, Gill HS. Standardizing the skin tape stripping method for sensitization and using it to create a mouse model of peanut allergy. Int J Pharm 2024; 662:124479. [PMID: 39019298 DOI: 10.1016/j.ijpharm.2024.124479] [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: 03/30/2024] [Revised: 07/11/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND Animal models for food allergies serve as crucial tools in understanding allergy mechanisms and assessing the efficacy of potential desensitization methods. The effectiveness of inducing allergies in mice through intragastric lavage sensitization varies. The intraperitoneal method can trigger systemic anaphylaxis, however it lacks anatomical relevance. Hence, a uniform and reliable allergy induction method in mice is required. Tape -stripping can mimic atopic dermatitis (AD), a precursor to lifelong peanut allergies in humans. Furthermore, skin damage triggers the upregulation of skin alarmins and the expansion of small-intestinal mast cells, both implicated in allergy development. METHODS We standardized a skin-based sensitization method in a mouse model of peanut allergy using skin tape-stripping followed by allergen application. We compared this method with intragastric sensitization. RESULTS Skin-based sensitization led to increased mast cells, goblet cells, and eosinophils in the small intestine, elevated systemic IgE levels, murine mast cell protease-1 (mMCP-1), histamine, and eosinophilic activity in peripheral blood. Moreover, it resulted in a significant hypothermic response, with nearly 30% mortality following an oral challenge one-month post-sensitization. CONCLUSION Our research offers a standardized and readily reproducible method for inducing peanut allergy in mice, which could also be adapted for other food allergens.
Collapse
Affiliation(s)
- Lazar D Nesovic
- Department of Chemical Engineering, Texas Tech University, 8th and Canton, Lubbock, TX 79409, USA
| | - Pedro E Gonzalez Cruz
- Department of Chemical Engineering, Texas Tech University, 8th and Canton, Lubbock, TX 79409, USA
| | - Natalie Rychener
- Department of Chemical Engineering, Texas Tech University, 8th and Canton, Lubbock, TX 79409, USA
| | - Logan R Wilks
- Department of Chemical Engineering, Texas Tech University, 8th and Canton, Lubbock, TX 79409, USA
| | - Harvinder S Gill
- Department of Chemical Engineering, Texas Tech University, 8th and Canton, Lubbock, TX 79409, USA; Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, USA.
| |
Collapse
|
4
|
Krempski J, Yamani A, Thota LNR, Marella S, Ganesan V, Sharma A, Kaneshige A, Bai L, Zhou H, Foster PS, Wang S, Obi AT, Hogan SP. IL-4-STAT6 axis amplifies histamine-induced vascular endothelial dysfunction and hypovolemic shock. J Allergy Clin Immunol 2024; 154:719-734. [PMID: 38777155 DOI: 10.1016/j.jaci.2024.05.009] [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: 07/13/2023] [Revised: 04/22/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Mast cell-derived mediators induce vasodilatation and fluid extravasation, leading to cardiovascular failure in severe anaphylaxis. We previously revealed a synergistic interaction between the cytokine IL-4 and the mast cell-derived mediator histamine in modulating vascular endothelial (VE) dysfunction and severe anaphylaxis. The mechanism by which IL-4 exacerbates histamine-induced VE dysfunction and severe anaphylaxis is unknown. OBJECTIVE We sought to identify the IL-4-induced molecular processes regulating the amplification of histamine-induced VE barrier dysfunction and the severity of IgE-mediated anaphylactic reactions. METHODS RNA sequencing, Western blot, Ca2+ imaging, and barrier functional analyses were performed on the VE cell line (EA.hy926). Pharmacologic degraders (selective proteolysis-targeting chimera) and genetic (lentiviral short hairpin RNA) inhibitors were used to determine the roles of signal transducer and activator of transcription 3 (STAT3) and STAT6 in conjunction with in vivo model systems of histamine-induced hypovolemic shock. RESULTS IL-4 enhancement of histamine-induced VE barrier dysfunction was associated with increased VE-cadherin degradation, intracellular calcium flux, and phosphorylated Src levels and required transcription and de novo protein synthesis. RNA sequencing analyses of IL-4-stimulated VE cells identified dysregulation of genes involved in cell proliferation, cell development, and cell growth, and transcription factor motif analyses revealed a significant enrichment of differential expressed genes with putative STAT3 and STAT6 motif. IL-4 stimulation in EA.hy926 cells induced both serine residue 727 and tyrosine residue 705 phosphorylation of STAT3. Genetic and pharmacologic ablation of VE STAT3 activity revealed a role for STAT3 in basal VE barrier function; however, IL-4 enhancement and histamine-induced VE barrier dysfunction was predominantly STAT3 independent. In contrast, IL-4 enhancement and histamine-induced VE barrier dysfunction was STAT6 dependent. Consistent with this finding, pharmacologic knockdown of STAT6 abrogated IL-4-mediated amplification of histamine-induced hypovolemia. CONCLUSIONS These studies unveil a novel role of the IL-4/STAT6 signaling axis in the priming of VE cells predisposing to exacerbation of histamine-induced anaphylaxis.
Collapse
Affiliation(s)
- James Krempski
- Mary H. Weiser Food Allergy Center, Michigan Medicine, University of Michigan, Ann Arbor, Mich
| | - Amnah Yamani
- Mary H. Weiser Food Allergy Center, Michigan Medicine, University of Michigan, Ann Arbor, Mich; Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Sahiti Marella
- Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Mich
| | - Varsha Ganesan
- Mary H. Weiser Food Allergy Center, Michigan Medicine, University of Michigan, Ann Arbor, Mich; Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Mich
| | - Ankit Sharma
- Mary H. Weiser Food Allergy Center, Michigan Medicine, University of Michigan, Ann Arbor, Mich
| | - Atsunori Kaneshige
- Department of Internal Medicine, University of Michigan, Ann Arbor, Mich; Department of Pharmacology, University of Michigan, Ann Arbor, Mich; Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Mich
| | - Longchuan Bai
- Department of Internal Medicine, University of Michigan, Ann Arbor, Mich; Department of Pharmacology, University of Michigan, Ann Arbor, Mich; Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Mich
| | - Haibin Zhou
- Department of Internal Medicine, University of Michigan, Ann Arbor, Mich; Department of Pharmacology, University of Michigan, Ann Arbor, Mich; Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Mich
| | - Paul S Foster
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Shaomeng Wang
- Department of Internal Medicine, University of Michigan, Ann Arbor, Mich; Department of Pharmacology, University of Michigan, Ann Arbor, Mich; Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Mich
| | - Andrea T Obi
- Conrad Jobst Vascular Research Laboratories, University of Michigan Medical School, Ann Arbor, Mich
| | - Simon P Hogan
- Mary H. Weiser Food Allergy Center, Michigan Medicine, University of Michigan, Ann Arbor, Mich; Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Mich.
| |
Collapse
|
5
|
Piotin A, Oulehri W, Charles AL, Tacquard C, Collange O, Mertes PM, Geny B. Oxidative Stress and Mitochondria Are Involved in Anaphylaxis and Mast Cell Degranulation: A Systematic Review. Antioxidants (Basel) 2024; 13:920. [PMID: 39199166 PMCID: PMC11352116 DOI: 10.3390/antiox13080920] [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: 06/14/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
Anaphylaxis, an allergic reaction caused by the massive release of active mediators, can lead to anaphylactic shock (AS), the most severe and potentially life-threatening form of anaphylactic reaction. Nevertheless, understanding of its pathophysiology to support new therapies still needs to be improved. We performed a systematic review, assessing the role and the complex cellular interplay of mitochondria and oxidative stress during anaphylaxis, mast cell metabolism and degranulation. After presenting the main characteristics of anaphylaxis, the oxidant/antioxidant balance and mitochondrial functions, we focused this review on the involvement of mitochondria and oxidative stress in anaphylaxis. Then, we discussed the role of oxidative stress and mitochondria following mast cell stimulation by allergens, leading to degranulation, in order to further elucidate mechanistic pathways. Finally, we considered potential therapeutic interventions implementing these findings for the treatment of anaphylaxis. Experimental studies evaluated mainly cardiomyocyte metabolism during AS. Cardiac dysfunction was associated with left ventricle mitochondrial impairment and lipid peroxidation. Studies evaluating in vitro mast cell degranulation, following Immunoglobulin E (IgE) or non-IgE stimulation, revealed that mitochondrial respiratory complex integrity and membrane potential are crucial for mast cell degranulation. Antigen stimulation raises reactive oxygen species (ROS) production from nicotinamide adenine dinucleotide phosphate (NADPH) oxidases and mitochondria, leading to mast cell degranulation. Moreover, mast cell activation involved mitochondrial morphological changes and mitochondrial translocation to the cell surface near exocytosis sites. Interestingly, antioxidant administration reduced degranulation by lowering ROS levels. Altogether, these results highlight the crucial role of oxidative stress and mitochondria during anaphylaxis and mast cell degranulation. New therapeutics against anaphylaxis should probably target oxidative stress and mitochondria, in order to decrease anaphylaxis-induced systemic and major organ deleterious effects.
Collapse
Affiliation(s)
- Anays Piotin
- Physiology and Functional Exploration Service, Strasbourg University Hospital, 67000 Strasbourg, France;
- Division of Asthma and Allergy, Chest Diseases Department, Strasbourg University Hospital, 67000 Strasbourg, France
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
| | - Walid Oulehri
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
- Department of Anesthesia and Intensive Care, Strasbourg University Hospital, 67000 Strasbourg, France;
| | - Anne-Laure Charles
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
| | - Charles Tacquard
- Department of Anesthesia and Intensive Care, Strasbourg University Hospital, 67000 Strasbourg, France;
- Établissement Français du Sang (EFS) Grand Est, French National Institute of Health and Medical Research), (INSERM) BPPS UMR_S1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), University of Strasbourg, 67000 Strasbourg, France
| | - Olivier Collange
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
- Department of Anesthesia and Intensive Care, Strasbourg University Hospital, 67000 Strasbourg, France;
| | - Paul-Michel Mertes
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
- Department of Anesthesia and Intensive Care, Strasbourg University Hospital, 67000 Strasbourg, France;
| | - Bernard Geny
- Physiology and Functional Exploration Service, Strasbourg University Hospital, 67000 Strasbourg, France;
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
| |
Collapse
|
6
|
Hjálmsdóttir Á, Hasler F, Waeckerle-Men Y, Duda A, López-Deber MP, Pihlgren M, Vukicevic M, Kündig TM, Johansen P. T cell independent antibody responses with class switch and memory using peptides anchored on liposomes. NPJ Vaccines 2024; 9:115. [PMID: 38909055 PMCID: PMC11193769 DOI: 10.1038/s41541-024-00902-3] [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: 06/05/2023] [Accepted: 05/23/2024] [Indexed: 06/24/2024] Open
Abstract
Vaccines generally require T lymphocytes for B-cell activation and immunoglobulin class switching in response to peptide or protein antigens. In the absence of T cells, limited IgG class switch takes place, germinal centers are short-lived, and the B cells lack memory. Here, immunization of mice with liposomes containing 15mer peptides and monophosphoryl lipid A (MPLA) as adjuvant, induced T-cell independent (TI) IgG class switch within three days, as well as germinal center formation. The antibody responses were long-lived, strictly dependent on Toll-like receptor 4 (TLR4) signaling, partly dependent on Bruton's tyrosine kinase (BTK) signal transmission, and independent of signaling through T-cell receptors, MHC class II and inflammasome. The antibody response showed characteristics of both TI type 1 and TI type 2. All IgG subclasses could be boosted months after primary immunization, and the biological function of the secreted antibodies was demonstrated in murine models of allergic anaphylaxis and of bacterial infection. Moreover, antibody responses after immunization with peptide- and MPLA-loaded liposomes could be triggered in neonatal mice and in mice receiving immune-suppressants. This study demonstrates T-cell independent endogenous B-cell memory and recall responses in vivo using a peptide antigen. The stimulation of these antibody responses required a correct and dense assembly and administration of peptide and adjuvant on the surface of liposomes. In the future, TI vaccines may prove beneficial in pathological conditions in which T-cell immunity is compromised through disease or medicines or when rapid, antibody-mediated immune protection is needed.
Collapse
Affiliation(s)
| | - Fabio Hasler
- Department of Dermatology, University of Zurich, Zurich, Switzerland
| | | | - Agathe Duda
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | | | - Maria Pihlgren
- AC Immune SA, EPFL Innovation Park EPFL, Lausanne, Switzerland
| | | | - Thomas M Kündig
- Department of Dermatology, University of Zurich, Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Pål Johansen
- Department of Dermatology, University of Zurich, Zurich, Switzerland.
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
7
|
Rathod S, Hoshitsuki K, Zhu Y, Ramsey M, Fernandez CA. Asparaginase-specific basophil recognition and activation predict Asparaginase hypersensitivity in mice. Front Immunol 2024; 15:1392099. [PMID: 38686384 PMCID: PMC11057047 DOI: 10.3389/fimmu.2024.1392099] [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: 02/26/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Background Asparaginase (ASNase) is a crucial part of acute leukemia treatment, but immune responses to the agent can reduce its effectiveness and increase the risk of relapse. Currently, no reliable and validated biomarker predicts ASNase-induced hypersensitivity reactions during therapy. We aimed to identify predictive biomarkers and determine immune cells responsible for anaphylaxis using a murine model of ASNase hypersensitivity. Methods Our preclinical study uses a murine model to investigate predictive biomarkers of ASNase anaphylaxis, including anti-ASNase antibody responses, immune complex (IC) levels, ASNase-specific binding to leukocytes or basophils, and basophil activation. Results Our results indicate that mice immunized to ASNase exhibited dynamic IgM, IgG, and IgE antibody responses. The severity of ASNase-induced anaphylaxis was found to be correlated with levels of IgG and IgE, but not IgM. Basophils from immunized mice were able to recognize and activate in response to ASNase ex vivo, and the extent of recognition and activation also correlated with the severity of anaphylaxis observed. Using a multivariable model that included all biomarkers significantly associated with anaphylaxis, independent predictors of ASNase-induced hypersensitivity reactions were found to be ASNase IC levels and ASNase-specific binding to leukocytes or basophils. Consistent with our multivariable analysis, we found that basophil depletion significantly protected mice from ASNase-induced hypersensitivity reactions, supporting that basophils are essential and can be used as a predictive marker of ASNase-induced anaphylaxis. Conclusions Our study demonstrates the need for using tools that can detect both IC- and IgE-mediated hypersensitivity reactions to mitigate the risk of ASNase-induced hypersensitivity reactions during treatment.
Collapse
Affiliation(s)
| | | | | | | | - Christian A. Fernandez
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
8
|
Park JY, Kim MJ, Choi YA, Lee SW, Lee S, Jang YH, Kim SH. Ethanol Extract of Ampelopsis brevipedunculata Rhizomes Suppresses IgE-Mediated Mast Cell Activation and Anaphylaxis. Adv Pharmacol Pharm Sci 2024; 2024:5083956. [PMID: 38605816 PMCID: PMC11008974 DOI: 10.1155/2024/5083956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/12/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024] Open
Abstract
More than 20% of the world's population suffers from allergic diseases, including allergic asthma, rhinitis, and atopic dermatitis that severely reduce the patient's quality of life. The treatment of allergy has been developed, but there are still unmet needs. Ampelopsis brevipedunculata (Maxim.) Trautv. is a traditional medicinal herb with beneficial bioactivities, such as antioxidant, anti-hypertension, anti-viral, anti-mutagenic, and skin and liver (anti-hepatotoxic) protective actions. However, its anti-allergic effect has not been addressed. This study designed to investigate the pharmacological effect of an ethanol extract of A. brevipedunculata rhizomes (ABE) on mast cell and anaphylaxis models. For in vivo studies, we used ovalbumin-induced active systemic anaphylaxis (ASA) and immunoglobulin (Ig) E-mediated passive cutaneous anaphylaxis (PCA) models. In ASA model, oral administration of ABE (1, 10, and 100 mg/kg) attenuated the anaphylactic responses, such as hypothermia, serum histamine, and IgE productions. In PCA model, ABE also suppressed the plasma extravasation and swelling. The underlying mechanisms of action were identified in various mast cell types. In vitro, ABE (10, 30, and 60 µg/mL) inhibited the release of essential allergic mediators, such as histamine and β-hexosaminidase, in a concentration-dependent manner. ABE prevented the rapid increase in intracellular calcium levels induced by the DNP-HSA challenge. In addition, ABE downregulated the tumor necrosis factor-α and interleukin-4 by suppressing the activation of nuclear factor-κB. Collectively, this study is the first to identify the anti-allergic effect of ABE, suggesting that ABE is a promising candidate for treating allergic diseases.
Collapse
Affiliation(s)
- Ji-Yeong Park
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Min-Jong Kim
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Young-Ae Choi
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Seung Woong Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Soyoung Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Yong Hyun Jang
- Department of Dermatology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Sang-Hyun Kim
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
9
|
Bao C, Abraham SN. Mast cell-sensory neuron crosstalk in allergic diseases. J Allergy Clin Immunol 2024; 153:939-953. [PMID: 38373476 PMCID: PMC10999357 DOI: 10.1016/j.jaci.2024.02.005] [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: 06/15/2023] [Revised: 01/12/2024] [Accepted: 02/14/2024] [Indexed: 02/21/2024]
Abstract
Mast cells (MCs) are tissue-resident immune cells, well-positioned at the host-environment interface for detecting external antigens and playing a critical role in mobilizing innate and adaptive immune responses. Sensory neurons are afferent neurons innervating most areas of the body but especially in the periphery, where they sense external and internal signals and relay information to the brain. The significance of MC-sensory neuron communication is now increasingly becoming recognized, especially because both cell types are in close physical proximity at the host-environment interface and around major organs of the body and produce specific mediators that can activate each other. In this review, we explore the roles of MC-sensory neuron crosstalk in allergic diseases, shedding light on how activated MCs trigger sensory neurons to initiate signaling in pruritus, shock, and potentially abdominal pain in allergy, and how activated sensory neurons regulate MCs in homeostasis and atopic dermatitis associated with contact hypersensitivity and type 2 inflammation. Throughout the review, we also discuss how these 2 sentinel cell types signal each other, potentially resulting in a positive feedback loop that can sustain inflammation. Unraveling the mysteries of MC-sensory neuron crosstalk is likely to unveil their critical roles in various disease conditions and enable the development of new therapeutic approaches to combat these maladies.
Collapse
Affiliation(s)
- Chunjing Bao
- Department of Pathology, Duke University Medical Center, Durham, NC
| | - Soman N Abraham
- Department of Pathology, Duke University Medical Center, Durham, NC; Department of Immunology, Duke University Medical Center, Durham, NC; Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC; Department of Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore, Singapore.
| |
Collapse
|
10
|
Mayorga C, Ariza A, Muñoz-Cano R, Sabato V, Doña I, Torres MJ. Biomarkers of immediate drug hypersensitivity. Allergy 2024; 79:601-612. [PMID: 37947156 DOI: 10.1111/all.15933] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/29/2023] [Accepted: 10/17/2023] [Indexed: 11/12/2023]
Abstract
Immediate drug hypersensitivity reactions (IDHRs) are a burden for patients and the health systems. This problem increases when taking into account that only a small proportion of patients initially labelled as allergic are finally confirmed after an allergological workup. The diverse nature of drugs involved will imply different interactions with the immunological system. Therefore, IDHRs can be produced by a wide array of mechanisms mediated by the drug interaction with specific antibodies or directly on effector target cells. These heterogeneous mechanisms imply an enhanced complexity for an accurate diagnosis and the identification of the phenotype and endotype at early stages of the reaction is of vital importance. Currently, several endophenotypic categories (type I IgE/non-IgE, cytokine release, Mast-related G-protein coupled receptor X2 (MRGPRX2) or Cyclooxygenase-1 (COX-1) inhibition and their associated biomarkers have been proposed. A precise knowledge of endotypes will permit to discriminate patients within the same phenotype, which is crucial in order to personalise diagnosis, future treatment and prevention to improve the patient's quality of life.
Collapse
Affiliation(s)
- Cristobalina Mayorga
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina - IBIMA Plataforma BIONAND, Málaga, Spain
- Allergy Unit, Hospital Regional Universitario de Málaga-HRUM, Málaga, Spain
| | - Adriana Ariza
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina - IBIMA Plataforma BIONAND, Málaga, Spain
| | - Rosa Muñoz-Cano
- Allergy Department, Hospital Clinic, Institut d'Investigacions Biomediques August Pi i Sunyer - IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Vito Sabato
- Department of Immunology, Allergology, Rheumatology, Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Inmaculada Doña
- Allergy Unit, Hospital Regional Universitario de Málaga-HRUM, Málaga, Spain
| | - Maria J Torres
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina - IBIMA Plataforma BIONAND, Málaga, Spain
- Allergy Unit, Hospital Regional Universitario de Málaga-HRUM, Málaga, Spain
- Medicine Department, Universidad de Málaga-UMA, Málaga, Spain
| |
Collapse
|
11
|
Sharma SR, Choudhary SK, Vorobiov J, Commins SP, Karim S. Tick bite-induced alpha-gal syndrome and immunologic responses in an alpha-gal deficient murine model. Front Immunol 2024; 14:1336883. [PMID: 38390396 PMCID: PMC10882631 DOI: 10.3389/fimmu.2023.1336883] [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: 11/11/2023] [Accepted: 12/26/2023] [Indexed: 02/24/2024] Open
Abstract
Introduction Alpha-Gal Syndrome (AGS) is a delayed allergic reaction due to specific IgE antibodies targeting galactose-α-1,3-galactose (α-gal), a carbohydrate found in red meat. This condition has gained significant attention globally due to its increasing prevalence, with more than 450,000 cases estimated just in the United States alone. Previous research has established a connection between AGS and tick bites, which sensitize individuals to α-gal antigens and elevate the levels of specific IgE. However, the precise mechanism by which tick bites influence the host's immune system and contribute to the development of AGS remains poorly understood. This study investigates various factors related to ticks and the host associated with the development of AGS following a tick bite, using mice with a targeted disruption of alpha-1,3-galactosyltransferase (AGKO) as a model organism. Methods Lone-star tick (Amblyomma americanum) and gulf-coast tick (Amblyomma maculatum) nymphs were used to sensitize AGKO mice, followed by pork meat challenge. Tick bite site biopsies from sensitized and non-sensitized mice were subjected to mRNA gene expression analysis to assess the host immune response. Antibody responses in sensitized mice were also determined. Results Our results showed a significant increase in the total IgE, IgG1, and α-gal IgG1 antibodies titers in the lone-star tick-sensitized AGKO mice compared to the gulf-coast tick-sensitized mice. Pork challenge in Am. americanum -sensitized mice led to a decline in body temperature after the meat challenge. Gene expression analysis revealed that Am. americanum bites direct mouse immunity toward Th2 and facilitate host sensitization to the α-gal antigen. Conclusion This study supports the hypothesis that specific tick species may increase the risk of developing α-gal-specific IgE and hypersensitivity reactions or AGS, thereby providing opportunities for future research on the mechanistic role of tick and host-related factors in AGS development.
Collapse
Affiliation(s)
- Surendra Raj Sharma
- School of Biological, Environment and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Shailesh K. Choudhary
- Department of Medicine and Pediatrics, University of North Carolina, Chapel Hill, NC, United States
| | - Julia Vorobiov
- Department of Medicine and Pediatrics, University of North Carolina, Chapel Hill, NC, United States
| | - Scott P. Commins
- Department of Medicine and Pediatrics, University of North Carolina, Chapel Hill, NC, United States
| | - Shahid Karim
- School of Biological, Environment and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| |
Collapse
|
12
|
Gallizzi AA, Heinken A, Guéant-Rodriguez RM, Guéant JL, Safar R. A systematic review and meta-analysis of proteomic and metabolomic alterations in anaphylaxis reactions. Front Immunol 2024; 15:1328212. [PMID: 38384462 PMCID: PMC10879545 DOI: 10.3389/fimmu.2024.1328212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/11/2024] [Indexed: 02/23/2024] Open
Abstract
Background Anaphylaxis manifests as a severe immediate-type hypersensitivity reaction initiated through the immunological activation of target B-cells by allergens, leading to the release of mediators. However, the well-known underlying pathological mechanisms do not fully explain the whole variety of clinical and immunological presentations. We performed a systemic review of proteomic and metabolomic studies and analyzed the extracted data to improve our understanding and identify potential new biomarkers of anaphylaxis. Methods Proteomic and metabolomic studies in both human subjects and experimental models were extracted and selected through a systematic search conducted on databases such as PubMed, Scopus, and Web of Science, up to May 2023. Results Of 137 retrieved publications, we considered 12 for further analysis, including seven on proteome analysis and five on metabolome analysis. A meta-analysis of the four human studies identified 118 proteins with varying expression levels in at least two studies. Beside established pathways of mast cells and basophil activation, functional analysis of proteomic data revealed a significant enrichment of biological processes related to neutrophil activation and platelet degranulation and metabolic pathways of arachidonic acid and icosatetraenoic acid. The pathway analysis highlighted also the involvement of neutrophil degranulation, and platelet activation. Metabolome analysis across different models showed 13 common metabolites, including arachidonic acid, tryptophan and lysoPC(18:0) lysophosphatidylcholines. Conclusion Our review highlights the underestimated role of neutrophils and platelets in the pathological mechanisms of anaphylactic reactions. These findings, derived from a limited number of publications, necessitate confirmation through human studies with larger sample sizes and could contribute to the development of new biomarkers for anaphylaxis. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42024506246.
Collapse
Affiliation(s)
- Adrienne Astrid Gallizzi
- INSERM, UMR_S1256, NGERE – Nutrition, Genetics, and Environmental Risk Exposure, Faculty of Medicine of Nancy, University of Lorraine, Vandoeuvre-lès-Nancy, France
| | - Almut Heinken
- INSERM, UMR_S1256, NGERE – Nutrition, Genetics, and Environmental Risk Exposure, Faculty of Medicine of Nancy, University of Lorraine, Vandoeuvre-lès-Nancy, France
| | - Rosa-Maria Guéant-Rodriguez
- INSERM, UMR_S1256, NGERE – Nutrition, Genetics, and Environmental Risk Exposure, Faculty of Medicine of Nancy, University of Lorraine, Vandoeuvre-lès-Nancy, France
- Department of Molecular Medicine and Personalized Therapeutics, Department of Biochemistry, Molecular Biology, Nutrition, and Metabolism, University Hospital of Nancy, Vandoeuvre-lès-Nancy, France
| | - Jean-Louis Guéant
- INSERM, UMR_S1256, NGERE – Nutrition, Genetics, and Environmental Risk Exposure, Faculty of Medicine of Nancy, University of Lorraine, Vandoeuvre-lès-Nancy, France
- Department of Molecular Medicine and Personalized Therapeutics, Department of Biochemistry, Molecular Biology, Nutrition, and Metabolism, University Hospital of Nancy, Vandoeuvre-lès-Nancy, France
| | - Ramia Safar
- INSERM, UMR_S1256, NGERE – Nutrition, Genetics, and Environmental Risk Exposure, Faculty of Medicine of Nancy, University of Lorraine, Vandoeuvre-lès-Nancy, France
| |
Collapse
|
13
|
Huang M, Shao H, Zhang X, Yang F, Wang J, Tan S, Chen H, Li X. Comparison of cow's milk allergy models highlighted higher humoral and Th2 immune responses in BALB/c than C3H/HeNCrl mice. Food Chem Toxicol 2024; 184:114315. [PMID: 38081529 DOI: 10.1016/j.fct.2023.114315] [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: 09/10/2023] [Revised: 11/09/2023] [Accepted: 11/28/2023] [Indexed: 12/25/2023]
Abstract
Cow's milk allergy (CMA) is common in early childhood and the incidence is increasing. However, its mechanisms of action are still not fully understood due to the range of different clinical symptoms. So far, the development of different mouse models has been the best choice to study the molecular mechanisms triggering allergy. However, the selection of suitable strains for the establishment of animal models truly representative of associated human pathologies is still a challenge. Hence, we focused on both C3H/HeNCrl and BALB/c mice to characterize their susceptibility to CMA. After intraperitoneal sensitization, BALB/c and C3H/HeNCrl strains were challenged with β-lactoglobulin (BLG), and compared in allergic symptoms and active immune response, which assessed by specific antibody production and cytokine release. At first, both groups exhibited anaphylaxis, showed specific BLG-related IgE, Th2 response and seemed both suitable for the development of CMA models. However, a detailed analysis revealed that BALB/c had both stronger humoral and Th2 immune responses, producing more antibodies (IgE and IgG/IgG1/IgG2a), and releasing higher levels of Th2-associated cytokines (IL-4, IL-5, IL-13) compared to C3H/HeNCrl mice. Therefore, BALB/c strain would represent a preferential choice in the establishment of CMA models. This study highlights the subtle differences and major outcomes in the selection of mouse strains for the development of suitable food allergy models.
Collapse
Affiliation(s)
- Meijia Huang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, PR China; School of Food Science, Henan Institute of Science and Technology, Xinxiang, 453003, PR China; School of Food Science and Technology, Nanchang University, Nanchang, 330031, Jiangxi, PR China
| | - Huming Shao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, PR China; School of Food Science and Technology, Nanchang University, Nanchang, 330031, Jiangxi, PR China
| | - Xing Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, PR China; School of Food Science and Technology, Nanchang University, Nanchang, 330031, Jiangxi, PR China
| | - Fan Yang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, PR China; School of Food Science and Technology, Nanchang University, Nanchang, 330031, Jiangxi, PR China
| | - Jingshu Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, PR China; School of Food Science and Technology, Nanchang University, Nanchang, 330031, Jiangxi, PR China
| | - Shuijie Tan
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, PR China; School of Food Science and Technology, Nanchang University, Nanchang, 330031, Jiangxi, PR China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, PR China; Sino-German Joint Research Institute, Nanchang University, Nanchang, 330047, Jiangxi, PR China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, 330047, Jiangxi, PR China
| | - Xin Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, PR China; School of Food Science and Technology, Nanchang University, Nanchang, 330031, Jiangxi, PR China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, 330047, Jiangxi, PR China.
| |
Collapse
|
14
|
Xu Y, Zhang F, Mu G, Zhu X. Effect of lactic acid bacteria fermentation on cow milk allergenicity and antigenicity: A review. Compr Rev Food Sci Food Saf 2024; 23:e13257. [PMID: 38284611 DOI: 10.1111/1541-4337.13257] [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: 03/02/2023] [Revised: 09/22/2023] [Accepted: 10/02/2023] [Indexed: 01/30/2024]
Abstract
Cow milk is a major allergenic food. The potential prevention and treatment effects of lactic acid bacteria (LAB)-fermented dairy products on allergic symptoms have garnered considerable attention. Cow milk allergy (CMA) is mainly attributed to extracellular and/or cell envelope proteolytic enzymes with hydrolysis specificity. Numerous studies have demonstrated that LAB prevents the risk of allergies by modulating the development and regulation of the host immune system. Specifically, LAB and its effectors can enhance intestinal barrier function and affect immune cells by interfering with humoral and cellular immunity. Fermentation hydrolysis of allergenic epitopes is considered the main mechanism of reducing CMA. This article reviews the linear epitopes of allergens in cow milk and the effect of LAB on these allergens and provides insight into the means of predicting allergenic epitopes by conventional laboratory analysis methods combined with molecular simulation. Although LAB can reduce CMA in several ways, the mechanism of action remains partially clarified. Therefore, this review additionally attempts to summarize the main mechanism of LAB fermentation to provide guidance for establishing an effective preventive and treatment method for CMA and serve as a reference for the screening, research, and application of LAB-based intervention.
Collapse
Affiliation(s)
- Yunpeng Xu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, P. R. China
| | - Feifei Zhang
- Liaoning Ocean and Fisheries Science Research Institute, Dalian, Liaoning, P. R. China
| | - Guangqing Mu
- Dalian Key Laboratory of Functional Probiotics, Dalian, Liaoning, P. R. China
| | - Xuemei Zhu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, P. R. China
| |
Collapse
|
15
|
Waritani T, Lomax S, Cutler D, Chang J. Development and evaluation of mouse anti-Ara h 1 and Ara h 3 IgE monoclonal antibodies for advancing peanut allergy research. MethodsX 2023; 11:102470. [PMID: 38034322 PMCID: PMC10681920 DOI: 10.1016/j.mex.2023.102470] [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: 08/08/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Immediate hypersensitivity reactions to peanuts are a considerable public health concern due to the acute and severe IgE mediated reactions. To conduct research on the pathogenesis and therapeutics of peanut allergies, it is imperative to have mouse anti-crude peanut extract (CPE) IgE monoclonal antibodies (mAbs) for both in-vitro and in-vivo assays. Without these tools, it is difficult to advance research in this field. In this study, four hybridomas producing anti-CPE IgE mAbs were developed and the IgE mAbs were validated using immune-blot analysis, Sandwich ELISA, Indirect ELISA, a cell-based assay using RBL-2H3 cells, and footpad type I hypersensitivity reaction studies in mice. The results indicate that two of the four mAbs can be effectively used for both in-vitro and in-vivo peanut allergy studies, as they induce allergic reactions with sensitization alone in mice. These novel anti-Ara h1 and Ara h 3 IgE mAbs, in combination with the detailed protocols outlined in this article, offer valuable guidance for studying acute allergic reactions involving mast cells across various platforms. With some considerations, the IgE mAbs can significantly advance peanut allergy research.
Collapse
Affiliation(s)
- Takaki Waritani
- Chondrex, Inc., 16928 Woodinville-Redmond Rd NE STE B101, Woodinville, WA 98072, USA
| | - Sidney Lomax
- Chondrex, Inc., 16928 Woodinville-Redmond Rd NE STE B101, Woodinville, WA 98072, USA
| | - Dawn Cutler
- Chondrex, Inc., 16928 Woodinville-Redmond Rd NE STE B101, Woodinville, WA 98072, USA
| | - Jessica Chang
- Chondrex, Inc., 16928 Woodinville-Redmond Rd NE STE B101, Woodinville, WA 98072, USA
| |
Collapse
|
16
|
Sharma SR, Choudhary SK, Vorobiov J, Commins SP, Karim S. Tick bite-induced Alpha-Gal Syndrome and Immunologic Responses in an Alpha-Gal Deficient Murine Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.09.566281. [PMID: 38014105 PMCID: PMC10680608 DOI: 10.1101/2023.11.09.566281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Introduction Alpha-Gal Syndrome (AGS) is a delayed allergic reaction due to specific IgE antibodies targeting galactose-α-1,3-galactose (α-gal), a carbohydrate found in red meat. This condition has gained significant attention globally due to its increasing prevalence, with more than 450,000 cases estimated in the United States alone. Previous research has established a connection between AGS and tick bites, which sensitize individuals to α-gal antigens and elevate the levels of α-gal specific IgE. However, the precise mechanism by which tick bites influence the hosťs immune system and contribute to the development of AGS remains poorly understood. This study investigates various factors related to ticks and the host associated with the development of AGS following a tick bite, using mice with a targeted disruption of alpha-1,3-galactosyltransferase (AGKO) as a model organism. Methods Lone-star tick (Amblyomma americanum) and gulf-coast tick (Amblyomma maculatum) nymphs were used to sensitize AGKO mice, followed by pork meat challenge. Tick bite site biopsies from sensitized and non-sensitized mice were subjected to mRNA gene expression analysis to assess the host immune response. Antibody responses in sensitized mice were also determined. Results Our results showed a significant increase in the titer of total IgE, IgG1, and α-gal IgG1 antibodies in the lone-star tick-sensitized AGKO mice compared to the gulf-coast tick-sensitized mice. Pork challenge in Am. americanum -sensitized mice led to a decline in body temperature after the meat challenge. Gene expression analysis revealed that Am. americanum bites direct mouse immunity toward Th2 and facilitate host sensitization to the α-gal antigen, while Am. maculatum did not. Conclusion This study supports the hypothesis that specific tick species may increase the risk of developing α-gal-specific IgE and hypersensitivity reactions or AGS, thereby providing opportunities for future research on the mechanistic role of tick and host-related factors in AGS development.
Collapse
Affiliation(s)
- Surendra Raj Sharma
- School of Biological, Environment and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Shailesh K Choudhary
- Department of Medicine & Pediatrics, University of North Carolina, Chapel Hill, NC 27599-7280, USA
| | - Julia Vorobiov
- Department of Medicine & Pediatrics, University of North Carolina, Chapel Hill, NC 27599-7280, USA
| | - Scott P Commins
- Department of Medicine & Pediatrics, University of North Carolina, Chapel Hill, NC 27599-7280, USA
| | - Shahid Karim
- School of Biological, Environment and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS 39406, USA
| |
Collapse
|
17
|
Nuñez-Borque E, Fernandez-Bravo S, Rodríguez Del Rio P, Palacio-García L, Di Giannatale A, Di Paolo V, Galardi A, Colletti M, Pascucci L, Tome-Amat J, Cuesta-Herranz J, Ibañez-Sandin MD, Laguna JJ, Benito-Martin A, Esteban V. Novel mediator in anaphylaxis: decreased levels of miR-375-3p in serum and within extracellular vesicles of patients. Front Immunol 2023; 14:1209874. [PMID: 37965316 PMCID: PMC10642912 DOI: 10.3389/fimmu.2023.1209874] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 10/13/2023] [Indexed: 11/16/2023] Open
Abstract
Introduction Anaphylaxis is among the most severe manifestations of allergic disorders, but its molecular basis remains largely unknown and reliable diagnostic markers are not currently available. MicroRNAs (miRNAs) regulate several pathophysiological processes and have been proposed as non-invasive biomarkers. Therefore, this study aims to evaluate their involvement in anaphylactic reaction and their value as biomarkers. Methods Acute (anaphylaxis) and baseline (control) serum samples from 67 patients with anaphylaxis were studied. Among them, 35 were adults with drug-induced anaphylaxis, 13 adults with food-induced anaphylaxis and 19 children with food-induced anaphylaxis. The circulating serum miRNAs profile was characterized by next-generation sequencing (NGS). For this purpose, acute and baseline samples from 5 adults with drug-induced anaphylaxis were used. RNA was extracted, retrotranscribed, sequenced and the readings obtained were mapped to the human database miRBase_20. In addition, a system biology analysis (SBA) was performed with its target genes and revealed pathways related to anaphylactic mediators signaling. Moreover, functional and molecular endothelial permeability assays were conducted with miR-375-3p-transfected cells in response to cAMP. Results A total of 334 miRNAs were identified, of which 21 were significant differentially expressed between both phases. Extracellular vesicles (EVs) were characterized by Western blot, electron microscopy and NanoSight. A decrease of miR-375-3p levels was determined by qPCR in both serum and EVs of patients with anaphylaxis (****p<.0001). Precisely, the decrease of miR-375-3p correlated with the increase of two inflammatory cytokines: monocyte chemoattractant protein-1 (MCP-1) and granulocyte macrophage colony-stimulating factor (GM-CSF). On the other hand, functional and molecular data obtained showed that miR-375-3p partially blocked the endothelial barrier maintenance and stabilization by disassembly of cell-cell junctions exhibiting low Rac1-Cdc42 levels. Discussion These findings demonstrate a differential serum profile of circulating miRNAs in patients with anaphylaxis and exhibit the miR-375-3p modulation in serum and EVs during drug- and food-mediated anaphylactic reactions. Furthermore, the in silico and in vitro studies show a negative role for miR-375-3p/Rac1-Cdc42 in the endothelial barrier stability.
Collapse
Affiliation(s)
- Emilio Nuñez-Borque
- Department of Allergy and Immunology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Sergio Fernandez-Bravo
- Department of Allergy and Immunology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Pablo Rodríguez Del Rio
- Allergy Department, Hospital Infantil Universitario Niño Jesús, Fundación Hospital Niño Jesús (HNJ), Instituto de Investigación del Hospital de La Princesa (IIS-P), Madrid, Spain
| | - Lucia Palacio-García
- Department of Allergy and Immunology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Angela Di Giannatale
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children’s Hospital, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Virginia Di Paolo
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children’s Hospital, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Angela Galardi
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children’s Hospital, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Marta Colletti
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children’s Hospital, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Luisa Pascucci
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Jaime Tome-Amat
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (UPM-INIA), Universidad Politécnica de Madrid, Madrid, Spain
| | - Javier Cuesta-Herranz
- Department of Allergy. Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - María Dolores Ibañez-Sandin
- Allergy Department, Hospital Infantil Universitario Niño Jesús, Fundación Hospital Niño Jesús (HNJ), Instituto de Investigación del Hospital de La Princesa (IIS-P), Madrid, Spain
| | - José Julio Laguna
- Allergy Unit, Allergo-Anaesthesia Unit, Cruz Roja Central Hospital, Villanueva de la Cañada, Madrid, Spain
- Faculty of Medicine and Biomedicine, Universidad Alfonso X el Sabio (UAX), Madrid, Spain
| | - Alberto Benito-Martin
- Faculty of Medicine and Biomedicine, Universidad Alfonso X el Sabio (UAX), Madrid, Spain
| | - Vanesa Esteban
- Department of Allergy and Immunology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Faculty of Medicine and Biomedicine, Universidad Alfonso X el Sabio (UAX), Madrid, Spain
| |
Collapse
|
18
|
Okojie AK, Uweru JO, Coburn MA, Li S, Cao-Dao VD, Eyo UB. Distinguishing the effects of systemic CSF1R inhibition by PLX3397 on microglia and peripheral immune cells. J Neuroinflammation 2023; 20:242. [PMID: 37865779 PMCID: PMC10590528 DOI: 10.1186/s12974-023-02924-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/10/2023] [Indexed: 10/23/2023] Open
Abstract
Microglia, the primary immune cells of the central nervous system (CNS), are derived from the yolk sac and populate the brain during development. Once microglia migrate to the CNS, they are self-renewing and require CSF1R signaling for their maintenance. Pexidartinib (PLX3397, PLX), a small molecule inhibitor of the CSF1R, has been shown to effectively deplete microglia since microglial maintenance is CSF1R-dependent. There have, however, been several conflicting reports that have shown the potential off-target effects of PLX on peripheral immune cells particularly those of lymphoid origin. Given this controversy in the use of the PLX family of drugs, it has become important to ascertain to what extent PLX affects the peripheral immune profile in lymphoid (spleen, and bone marrow) and non-lymphoid (kidney, lungs, and heart) organs. PLX3397 chow treatment at 660 mg/kg for 7 days significantly reduced CD45+ macrophages, CX3CR1-GFP cells, CD11b+CD45intermediate cells, and P2RY12 expression in the brain. However, there were minimal effects on peripheral immune cells from both lymphoid and non-lymphoid organs except in the heart where there was a significant decrease in CD3+ cells, inflammatory and patrolling monocytes, and CD11b+Ly6G+ neutrophils. We then stimulated the immune system with 1 mg/kg of LPS which resulted in a significant reduction in the number of innate immune cells. In this context, PLX did not alter the cytokine profile in the serum and the brain of naïve mice but did so in the LPS-stimulated group resulting in a significant reduction in TNFα, IL-1α, IFN-γ and IL-1β. Furthermore, PLX did not alter locomotor activity in the open field test suggesting that microglia do not contribute to LPS-induced sickness behavior. Our results provide an assessment of immune cell populations with PLX3397 treatment on brain, lymphoid and non-lymphoid organs without and during LPS treatment that can serve as a resource for understanding consequences of such approaches.
Collapse
Affiliation(s)
- Akhabue K Okojie
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA.
| | - Joseph O Uweru
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Morgan A Coburn
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Sihan Li
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Vivian D Cao-Dao
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Ukpong B Eyo
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
19
|
Stackowicz J, Gillis CM, Godon O, Iannascoli B, Conde E, Leveque E, Worrall WPM, Galli SJ, Bruhns P, Reber LL, Jönsson F. Conditional neutrophil depletion challenges their contribution to mouse models of anaphylaxis. Allergy 2023; 78:2767-2770. [PMID: 37022292 DOI: 10.1111/all.15738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/19/2023] [Accepted: 04/03/2023] [Indexed: 04/07/2023]
Affiliation(s)
- Julien Stackowicz
- Unit of Antibodies in Therapy and Pathology, INSERM UMR1222, Institut Pasteur, Université de Paris Cité, Paris
- College Doctoral, Sorbonne Université, Paris, France
| | - Caitlin M Gillis
- Unit of Antibodies in Therapy and Pathology, INSERM UMR1222, Institut Pasteur, Université de Paris Cité, Paris
| | - Ophélie Godon
- Unit of Antibodies in Therapy and Pathology, INSERM UMR1222, Institut Pasteur, Université de Paris Cité, Paris
| | - Bruno Iannascoli
- Unit of Antibodies in Therapy and Pathology, INSERM UMR1222, Institut Pasteur, Université de Paris Cité, Paris
| | - Eva Conde
- Unit of Antibodies in Therapy and Pathology, INSERM UMR1222, Institut Pasteur, Université de Paris Cité, Paris
- College Doctoral, Sorbonne Université, Paris, France
| | - Edouard Leveque
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse, France
| | - William P M Worrall
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse, France
| | - Stephen J Galli
- Departments of Pathology and of Microbiology and Immunology, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, California, USA
| | - Pierre Bruhns
- Unit of Antibodies in Therapy and Pathology, INSERM UMR1222, Institut Pasteur, Université de Paris Cité, Paris
| | - Laurent L Reber
- Unit of Antibodies in Therapy and Pathology, INSERM UMR1222, Institut Pasteur, Université de Paris Cité, Paris
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse, France
| | - Friederike Jönsson
- Unit of Antibodies in Therapy and Pathology, INSERM UMR1222, Institut Pasteur, Université de Paris Cité, Paris
- CNRS, Paris, France
| |
Collapse
|
20
|
Liu J, Li J, Yin J. Changes of allergic inflammation and immunological parameters after Alt a 1 and A. alternata immunotherapy in mice. World Allergy Organ J 2023; 16:100807. [PMID: 37638361 PMCID: PMC10457585 DOI: 10.1016/j.waojou.2023.100807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Background The efficacy of allergen-specific subcutaneousimmunotherapy (SCIT) with Alt a 1 of the fungus A. alternata is still unknown. Yet, few studies compare the therapeutic effects and immunological mechanisms of Alt a 1 and A. alternata extracts. We aim to explore and compare the changes in allergic inflammation and immunological mechanisms of Alt a 1 and A. alternata in mice. Methods Female BALB/c mice administrated recombinant Alt a 1 (rAlt a 1), native Alt a 1 (nAlt a 1), and A. alternata. Lung histology, airway hyper-reactivity (AHR), bronchoalveolar lavage fluid (BALF) cytokine levels, serum immunoglobulin responses, the expression of Bcl-6, the percentages of T follicular helper cells (Tfh), cytokine-related Tfh subtypes, regulatory B cells (Breg), and IL-10+ Breg cells were detected. Results High-purity nAlt 1 protein was obtained. SCIT with Alt a 1 and Alternaria decreased airway and lung inflammation, including improvement of lung pathology, lower levels of AHR, reduction of total cell numbers, and IL-4 and IL-13 levels in BALF. Furthermore, Alt a 1-SCIT effectively suppressed the IgE responses, elevated IgG titers, and was superior in decreasing the expression of Bcl-6. Additionally, Alternaria-SCIT significantly decreased the expression of Tfh cells, L-4+ Tfh, and IL-5+ Tfh cells in the spleen, whereas Alt a 1 showed superior therapeutic effects in the lymph node. IL-13+ Tfh cells in these two treatment groups not being significant. IL-17A+ Tfh cells were alleviated most effectively after A. alternata-SCIT in both the spleen and lymph node. Intriguingly, IL-10+ Breg cells decreased remarkably in response to SCIT with rAlt a 1. Conclusions Treatments with Alt a 1 and A. alternata extracts had beneficial effects on allergic inflammation. Alt a 1-SCIT resulted in prominent improvement in the immunoglobulin responses, Bcl-6, and IL-10+ Breg cells. Alternaria-SCIT was more likely to suppress the expression of Tfh and cytokine-related Tfh subtypes.
Collapse
Affiliation(s)
- Juan Liu
- Department of Allergy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment on Allergic Diseases, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Disease (NCRC-DID), Beijing, China
| | - Junda Li
- Department of Allergy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment on Allergic Diseases, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Disease (NCRC-DID), Beijing, China
| | - Jia Yin
- Department of Allergy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment on Allergic Diseases, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Disease (NCRC-DID), Beijing, China
| |
Collapse
|
21
|
Gouel-Chéron A, Dejoux A, Lamanna E, Bruhns P. Animal Models of IgE Anaphylaxis. BIOLOGY 2023; 12:931. [PMID: 37508362 PMCID: PMC10376466 DOI: 10.3390/biology12070931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023]
Abstract
Allergies and atopy have emerged as significant public health concerns, with a progressively increasing incidence over the last two decades. Anaphylaxis is the most severe form of allergic reactions, characterized by a rapid onset and potentially fatal outcome, even in healthy individuals. Due to the unpredictable nature and potential lethality of anaphylaxis and the wide range of allergens involved, clinical studies in human patients have proven to be challenging. Diagnosis is further complicated by the lack of reliable laboratory biomarkers to confirm clinical suspicion. Thus, animal models have been developed to replicate human anaphylaxis and explore its pathophysiology. Whereas results obtained from animal models may not always be directly translatable to humans, they serve as a foundation for understanding the underlying mechanisms. Animal models are an essential tool for investigating new biomarkers that could be incorporated into the allergy workup for patients, as well as for the development of novel treatments. Two primary pathways have been described in animals and humans: classic, predominantly involving IgE and histamine, and alternative, reliant on IgG and the platelet-activating factor. This review will focus essentially on the former and aims to describe the most utilized IgE-mediated anaphylaxis animal models, including their respective advantages and limitations.
Collapse
Affiliation(s)
- Aurélie Gouel-Chéron
- Université Paris Cité, 75010 Paris, France
- Anaesthesiology and Critical Care Medicine Department, DMU Parabol, Bichat-Claude Bernard Hospital, AP-HP, 75018 Paris, France
- Institut Pasteur, Université de Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| | - Alice Dejoux
- Institut Pasteur, Université de Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- Sorbonne Université, Collège Doctoral, 75005 Paris, France
| | - Emma Lamanna
- Institut Pasteur, Université de Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- Neovacs SA, 92150 Suresnes, France
| | - Pierre Bruhns
- Institut Pasteur, Université de Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| |
Collapse
|
22
|
Ma RX, Hu JQ, Fu W, Zhong J, Cao C, Wang CC, Qi SQ, Zhang XL, Liu GH, Gao YD. Intermittent fasting protects against food allergy in a murine model via regulating gut microbiota. Front Immunol 2023; 14:1167562. [PMID: 37228621 PMCID: PMC10205017 DOI: 10.3389/fimmu.2023.1167562] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/14/2023] [Indexed: 05/27/2023] Open
Abstract
Background The prevalence of food allergy (FA) is increasing. Decreases in the diversity of gut microbiota may contribute to the pathogenesis of FA by regulating IgE production of B cells. Intermittent fasting (IF) is a popular diet with the potential to regulate glucose metabolism, boosting immune memory and optimizing gut microbiota. The potential effect of long-term IF on the prevention and treatment of FA is still unknown. Methods Two IF protocols (16 h fasting/8 h feeding and 24 h fasting/24 h feeding) were conducted on mice for 56 days, while the control mice were free to intake food (free diet group, FrD). To construct the FA model, all mice were sensitized and intragastrical challenged with ovalbumin (OVA) during the second half of IF (day 28 to day 56). Rectal temperature reduction and diarrhea were recorded to evaluate the symptoms of FA. Levels of serum IgE, IgG1, Th1/Th2 cytokines, mRNA expression of spleen T cell related transcriptional factors, and cytokines were examined. H&E, immunofluorescence, and toluidine blue staining were used to assess the structural changes of ileum villi. The composition and abundance of gut microbiota were analyzed by 16srRNA sequencing in cecum feces. Results The diarrhea score and rectal temperature reduction were lower in the two fasting groups compared to the FrD groups. Fasting was associated with lower levels of serum OVA-sIgE, OVA-sIgG1, interleukin (IL)-4 and IL-5, and mRNA expression of IL-4, IL-5, and IL-10 in the spleen. While no significant association was observed in interferon (IFN)-γ, tumor necrosis factor (TNF)-α, IL-6, IL-2 levels. Less mast cell infiltration in ileum was observed in the 16h/8h fasting group compared to the FrD group. ZO-1 expression in the ileum of the two fasting groups was higher in IF mice. The 24h/24h fasting reshaped the gut microbiota, with a higher abundance of Alistipes and Rikenellaceae strains compared to the other groups. Conclusion In an OVA-induced mice FA model, long-term IF may attenuate FA by reducing Th2 inflammation, maintaining the integrity of the intestinal epithelial barrier, and preventing gut dysbiosis.
Collapse
Affiliation(s)
- Ru-xue Ma
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jia-qian Hu
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wei Fu
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jian Zhong
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Can Cao
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chang-chang Wang
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shi-quan Qi
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiao-Lian Zhang
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, China
| | - Guang-hui Liu
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ya-dong Gao
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, China
| |
Collapse
|
23
|
Chen WA, Chang DY, Chen BM, Lin YC, Barenholz Y, Roffler SR. Antibodies against Poly(ethylene glycol) Activate Innate Immune Cells and Induce Hypersensitivity Reactions to PEGylated Nanomedicines. ACS NANO 2023; 17:5757-5772. [PMID: 36926834 PMCID: PMC10062034 DOI: 10.1021/acsnano.2c12193] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/03/2023] [Indexed: 06/09/2023]
Abstract
Nanomedicines and macromolecular drugs can induce hypersensitivity reactions (HSRs) with symptoms ranging from flushing and breathing difficulties to hypothermia, hypotension, and death in the most severe cases. Because many normal individuals have pre-existing antibodies that bind to poly(ethylene glycol) (PEG), which is often present on the surface of nanomedicines and macromolecular drugs, we examined if and how anti-PEG antibodies induce HSRs to PEGylated liposomal doxorubicin (PLD). Anti-PEG IgG but not anti-PEG IgM induced symptoms of HSRs including hypothermia, altered lung function, and hypotension after PLD administration in C57BL/6 and nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice. Hypothermia was significantly reduced by blocking FcγRII/III, by depleting basophils, monocytes, neutrophils, or mast cells, and by inhibiting secretion of histamine and platelet-activating factor. Anti-PEG IgG also induced hypothermia in mice after administration of other PEGylated liposomes, nanoparticles, or proteins. Humanized anti-PEG IgG promoted binding of PEGylated nanoparticles to human immune cells and induced secretion of histamine from human basophils in the presence of PLD. Anti-PEG IgE could also induce hypersensitivity reactions in mice after administration of PLD. Our results demonstrate an important role for IgG antibodies in induction of HSRs to PEGylated nanomedicines through interaction with Fcγ receptors on innate immune cells and provide a deeper understanding of HSRs to PEGylated nanoparticles and macromolecular drugs that may facilitate development of safer nanomedicines.
Collapse
Affiliation(s)
- Wei-An Chen
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Deng-Yuan Chang
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Bing-Mae Chen
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Chen Lin
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
- Graduate
Institute of Life Sciences, National Defense
Medical Center, Taipei 11529, Taiwan
| | - Yechezekel Barenholz
- Department
of Biochemistry, Faculty of Medicine, The
Hebrew University, Jerusalem 91120, Israel
| | - Steve R. Roffler
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
- Graduate
Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
24
|
Bao C, Chen O, Sheng H, Zhang J, Luo Y, Hayes BW, Liang H, Liedtke W, Ji RR, Abraham SN. A mast cell-thermoregulatory neuron circuit axis regulates hypothermia in anaphylaxis. Sci Immunol 2023; 8:eadc9417. [PMID: 36930731 PMCID: PMC10331449 DOI: 10.1126/sciimmunol.adc9417] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 02/23/2023] [Indexed: 03/19/2023]
Abstract
IgE-mediated anaphylaxis is an acute life-threatening systemic reaction to allergens, including certain foods and venoms. Anaphylaxis is triggered when blood-borne allergens activate IgE-bound perivascular mast cells (MCs) throughout the body, causing an extensive systemic release of MC mediators. Through precipitating vasodilatation and vascular leakage, these mediators are believed to trigger a sharp drop in blood pressure in humans and in core body temperature in animals. We report that the IgE/MC-mediated drop in body temperature in mice associated with anaphylaxis also requires the body's thermoregulatory neural circuit. This circuit is activated when granule-borne chymase from MCs is deposited on proximal TRPV1+ sensory neurons and stimulates them via protease-activated receptor-1. This triggers the activation of the body's thermoregulatory neural network, which rapidly attenuates brown adipose tissue thermogenesis to cause hypothermia. Mice deficient in either chymase or TRPV1 exhibited limited IgE-mediated anaphylaxis, and, in wild-type mice, anaphylaxis could be recapitulated simply by systemically activating TRPV1+ sensory neurons. Thus, in addition to their well-known effects on the vasculature, MC products, especially chymase, promote IgE-mediated anaphylaxis by activating the thermoregulatory neural circuit.
Collapse
Affiliation(s)
- Chunjing Bao
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ouyang Chen
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Huaxin Sheng
- Multidisciplinary Neuroprotection Laboratories, Center of Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jeffrey Zhang
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yikai Luo
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Byron W. Hayes
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Han Liang
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wolfgang Liedtke
- Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York NY 10010
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Soman N. Abraham
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham NC 27710, USA
- Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore 169857, Singapore
| |
Collapse
|
25
|
Kow ASF, Khoo LW, Tan JW, Abas F, Lee MT, Israf DA, Shaari K, Tham CL. Clinacanthus nutans aqueous leaves extract exerts anti-allergic activity in preclinical anaphylactic models via alternative IgG pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 303:116003. [PMID: 36464074 DOI: 10.1016/j.jep.2022.116003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 10/31/2022] [Accepted: 11/28/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Allergy is mediated by the crosslinking of immunoglobulins (Ig) -E or -G to their respective receptors, which degranulates mast cells, macrophages, basophils, or neutrophils, releasing allergy-causing mediators. The removal of these mediators such as histamine, platelet-activating factor (PAF) and interleukins (ILs) released by effector cells will alleviate allergy. Clinacanthus nutans (C. nutans), an herbal plant in Southeast Asia, is used traditionally to treat skin rash, an allergic symptom. Previously, we have reported that C. nutans aqueous leaves extract (CNAE) was able to suppress the release of β-hexosaminidase and histamine but not interleukin-4 (IL-4) and tumor necrosis factor-alpha (TNF-α) in the IgE-induced mast cell degranulation model at 5 mg/mL and above. We also found that CNAE could protect rats against ovalbumin-challenged active systemic anaphylaxis (OVA-ASA) through the downregulation and upregulation of certain metabolites using proton nuclear magnetic resonance (1H-NMR) metabolomics approach. AIM OF THE STUDY As allergy could be mediated by both IgE and IgG, we further evaluated the anti-allergy potential of CNAE in both in vitro model of IgG-induced macrophage activation and in vivo anaphylaxis models to further dissect the mechanism of action underlying the anti-allergic properties of CNAE. MATERIAL & METHODS The anti-allergy potential of CNAE was evaluated in in vivo anaphylaxis models of ovalbumin-challenged active systemic anaphylaxis (OVA-ASA) and IgE-challenged passive systemic anaphylaxis (PSA) using Sprague Dawley rats as well as IgG-challenged passive systemic anaphylaxis (IgG-PSA) using C57BL/6 mice. Meanwhile, in vitro model of IgG-induced macrophage activation model was performed using IC-21 macrophages. The release of soluble mediators from both IgE and IgG-mediated pathways were measured using enzyme-linked immunosorbent assay (ELISA). The signaling molecules targeted by CNAE were identified by performing Western blot. RESULTS IgG, platelet-activating factor (PAF) and IL-6 was suppressed by CNAE in OVA-ASA, but not IgE. In addition, CNAE significantly suppressed PAF and IL-6 in IgG-PSA but did not suppress histamine, IL-4 and leukotrienes C4 (LTC4) in IgE-PSA. CNAE also inhibited IL-6 and TNF-α by inhibiting the phosphorylation of ERK1/2 in the IgG-induced macrophage activation model. CONCLUSION Overall, our findings supported that CNAE exerts its anti-allergic properties by suppressing the IgG pathway and its mediators by inhibiting ERK1/2 phosphorylation, thus providing scientific evidence supporting its traditional use in managing allergy.
Collapse
Affiliation(s)
- Audrey Siew Foong Kow
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia; Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, 56000, Malaysia.
| | - Leng Wei Khoo
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, 43400, Malaysia.
| | - Ji Wei Tan
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia; School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Malaysia.
| | - Faridah Abas
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, 43400, Malaysia; Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, Serdang, 43400, Malaysia.
| | - Ming-Tatt Lee
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, 56000, Malaysia; Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan; Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan.
| | - Daud Ahmad Israf
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia.
| | - Khozirah Shaari
- Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, Serdang, 43400, Malaysia.
| | - Chau Ling Tham
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia.
| |
Collapse
|
26
|
Hu S, Zhang Y, Dang B, Wang Y, Zheng G, Zhang T, An H. Myricetin alleviated immunologic contact urticaria and mast cell degranulation via the PI3K/Akt/NF-κB pathway. Phytother Res 2023; 37:2024-2035. [PMID: 36649930 DOI: 10.1002/ptr.7726] [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: 10/26/2022] [Revised: 12/13/2022] [Accepted: 12/27/2022] [Indexed: 01/19/2023]
Abstract
Immunologic contact urticaria (ICU) is characterized by the wheal and flare reaction from direct contact with a chemical or protein agent, which involves a type I hypersensitivity mediated by allergen-specific immunoglobulin E (sIgE). Myricetin (Myr), a bioactive flavonoid, exhibits antiinflammatory activities. Our results showed that treatment with Myr could alleviate ICU symptoms, including a decrease in the number of wheals and scratching, and inhibit ear swelling in the IgE/DNFB-induced mice. The serum level of IgE, histamine, interleukin (IL)-4, TNF-α, and MCP-1 were reduced in Myr-treated mice. Myr also attenuated mast cells (MCs) degranulation and H-PGDS, TSLP, IL-33, PI3K, Akt, and NF-κB mRNA levels in ICU model. The IgE-mediated anaphylaxis mouse models demonstrated anti-allergic effects of Myr. In vitro analysis showed that Myr reduced IgE-induced calcium (Ca2+ ) influx, suppressed degranulation, and chemokine release in LAD2 cells (human primary mast cells). Myr can significantly inhibited PLCγ1, Akt, NF-κB, and p38 phosphorylation. In conclusion, the study demonstrated that Myr alleviate ICU symptoms and inhibit mast cell activation via PI3K/Akt/NF-κB signal pathway.
Collapse
Affiliation(s)
- Shiting Hu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yonghui Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Baowen Dang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yuejing Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Guodong Zheng
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Tao Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Hongli An
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| |
Collapse
|
27
|
Baldo BA. Allergic and other adverse reactions to drugs used in anesthesia and surgery. ANESTHESIOLOGY AND PERIOPERATIVE SCIENCE 2023; 1:16. [PMCID: PMC10264870 DOI: 10.1007/s44254-023-00018-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/02/2023] [Accepted: 04/11/2023] [Indexed: 11/13/2023]
Abstract
The list of drugs patients may be exposed to during the perioperative and postoperative periods is potentially extensive. It includes induction agents, neuromuscular blocking drugs (NMBDs), opioids, antibiotics, sugammadex, colloids, local anesthetics, polypeptides, antifibrinolytic agents, heparin and related anticoagulants, blue dyes, chlorhexidine, and a range of other agents depending on several factors related to individual patients’ clinical condition and progress in the postoperative recovery period. To avoid poor or ultrarapid metabolizers to a particular drug (for example tramadol and codeine) or possible adverse drug reactions (ADRs), some drugs may need to be avoided during or after surgery. This will be the case for patients with a history of anaphylaxis or other adverse events/intolerances to a known drug. Other drugs may be ceased for a period before surgery, e.g., anticoagulants that increase the chance of bleeding; diuretics for patients with acute renal failure; antihypertensives relative to kidney injury after major vascular surgery; and serotonergic drugs that together with some opioids may rarely induce serotonin toxicity. Studies of germline variations shown by genotyping and phenotyping to identify a predisposition of genetic factors to ADRs offer an increasingly important approach to individualize drug therapy. Studies of associations of human leukocyte antigen (HLA) genes with some serious delayed immune-mediated reactions are ongoing and variations of drug-metabolizing cytochrome CYP450 enzymes, P-glycoprotein, and catechol-O -methyltransferase show promise for the assessment of ADRs and non-responses to drugs, particularly opioids and other analgesics. Surveys of ADRs from an increasing number of institutions often cover small numbers of patients, are retrospective in nature, fail to clearly identify culprit drugs, and do not adequately distinguish immune-mediated from non-immune-mediated anaphylactoid reactions. From the many surveys undertaken, the large list of agents identified during and after anesthesia and surgery are examined for their ADR involvement. Drugs are classified into those most often involved, (NMBD and antibiotics); drugs that are becoming more frequently implicated, namely antibiotics (particularly teicoplanin), and blue dyes; those becoming less frequently involved; and drugs more rarely involved in perioperative, and postoperative adverse reactions but still important and necessary to keep in mind for the occasional potential sensitive patient. Clinicians should be aware of the similarities between drug-induced true allergic type I IgE/FcεRI- and pseudoallergic MRGPRX2-mediated ADRs, the clinical features of each, and their distinguishing characteristics. Procedures for identifying MRGPRX2 agonists and diagnosing and distinguishing pseudoallergic from allergic reaction mechanisms are discussed.
Collapse
Affiliation(s)
- Brian A. Baldo
- Molecular Immunology Unit, Kolling Institute of Medical Research, Royal North Shore Hospital of Sydney, St Leonards, Australia
- Department of Medicine, University of Sydney, Sydney, NSW Australia
- Lindfield, Australia
| |
Collapse
|
28
|
Terashi M, Yamaki K, Koyama Y. Development of a Novel IgG 1 Anaphylaxis Mouse Model with Uniquely Characteristic Skin Manifestations Induced Through the FcγRIII-Histamine Pathway. Immunol Invest 2023; 52:83-103. [PMID: 36201173 DOI: 10.1080/08820139.2022.2130799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Studies of passive anaphylaxis, in which mouse immunoglobulin G (IgG) and its antigens are administered to mice, believe that platelet-activating factor (PAF) is more important than histamine and that basophils or macrophages are primarily involved. However, the full extent of IgG-dependent anaphylaxis is still unclear; that is, little agreement has been reached about the mechanism. METHODS First, we established the novel model of IgG1 anaphylaxis induced by the intravenous administration of two types of IgG1 and a fluorescent dye-labeled antigen, as IgG1 immune complex in HR-1 hairless mice. Subsequently, pharmacological analysis was used to investigate the underlying mechanisms of IgG1 anaphylaxis in this established model. RESULTS The novel IgG1 anaphylaxis model can induce the IgG-induced Anaphylaxis-dependent Spotted Distribution of fluorescently labeled Immune complexes in the Skin, named "G-ASDIS". Moreover, this model was triggered primarily by the FcγRIII-dependent histamine release, which is different from the conventional model in which PAF was involved in the development of IgG1 anaphylaxis. Basophils in the circulation and mast cells in the skin may participate in the development of IgG1 anaphylaxis and increased G-ASDIS. CONCLUSION Our results propose that the novel axis, namely the FcγRIII-basophils and/or mast cell-histamine pathway, is important for IgG1 anaphylaxis. Further analysis of our model in addition to other models will lead to a broader analysis and understanding of the IgG1 anaphylaxis mechanism.
Collapse
Affiliation(s)
- Masato Terashi
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Hyogo, Japan
| | - Kouya Yamaki
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Hyogo, Japan
| | - Yutaka Koyama
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Hyogo, Japan
| |
Collapse
|
29
|
Arai T, Kokubo T, Tang R, Abo H, Terui A, Hirakawa J, Akita H, Kawashima H, Hisaka A, Hatakeyama H. Tumor-associated neutrophils and macrophages exacerbate antidrug IgG-mediated anaphylactic reaction against an immune checkpoint inhibitor. J Immunother Cancer 2022; 10:jitc-2022-005657. [PMID: 36543377 PMCID: PMC9772690 DOI: 10.1136/jitc-2022-005657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND With the increased use of immune checkpoint inhibitors (ICIs), side effects and toxicity are a great concern. Anaphylaxis has been identified as a potential adverse event induced by ICIs. Anaphylaxis is a life-threatening medical emergency. However, the mechanisms and factors that can potentially influence the incidence and severity of anaphylaxis in patients with cancer remain unclear. METHODS Healthy, murine colon 26, CT26, breast 4T1, EMT6, and renal RENCA tumor-bearing mice were treated with an anti-PD-L1 antibody (clone 10F.9G2). Symptoms of anaphylaxis were evaluated along with body temperature and mortality. The amounts of antidrug antibody and platelet-activating factor (PAF) in the blood were quantified via ELISA and liquid chromatography-mass spectrometry (LC-MS/MS). Immune cells were analyzed and isolated using a flow cytometer and magnetic-activated cell sorting, respectively. RESULTS Repeated administration of the anti-PD-L1 antibody 10F.9G2 to tumor-bearing mice caused fatal anaphylaxis, depending on the type of tumor model. After administration, antidrug immunoglobulin G (IgG), but not IgE antibodies, were produced, and PAF was released as a chemical mediator during anaphylaxis, indicating that anaphylaxis was caused by an IgG-dependent pathway. Anaphylaxis induced by 10F.9G2 was treated with a PAF receptor antagonist. We identified that neutrophils and macrophages were PAF-producing effector cells during anaphylaxis, and the tumor-bearing models with increased numbers of neutrophils and macrophages showed lethal anaphylaxis after treatment with 10F.9G2. Depletion of both neutrophils and macrophages using clodronate liposomes prevented anaphylaxis in tumor-bearing mice. CONCLUSIONS Thus, increased numbers of neutrophils and macrophages associated with cancer progression may be risk factors for anaphylaxis. These findings may provide useful insights into the mechanism of anaphylaxis following the administration of immune checkpoint inhibitors in human subjects.
Collapse
Affiliation(s)
- Takahiro Arai
- Lratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Tomomi Kokubo
- Lratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Ruiheng Tang
- Lratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan,Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Hirohito Abo
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Ayu Terui
- Lratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Jotaro Hirakawa
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Hidetaka Akita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hiroto Kawashima
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Akihiro Hisaka
- Lratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Hiroto Hatakeyama
- Lratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan,Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| |
Collapse
|
30
|
Bohn A, Blumenstiel J. Update Anaphylaxie – wenig Neues, aber weiter wichtig. NOTARZT 2022. [DOI: 10.1055/a-1961-9717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Affiliation(s)
- Andreas Bohn
- Berufsfeuerwehr, Stadt Münster, Munster, Deutschland
| | - Jonas Blumenstiel
- Abteilung Anästhesie, Schüchtermann-Klinik Bad Rothenfelde, Bad Rothenfelde, Deutschland
| |
Collapse
|
31
|
Carpio-Escalona LV, González-de-Olano D. Immunological and Non-Immunological Risk Factors in Anaphylaxis. CURRENT TREATMENT OPTIONS IN ALLERGY 2022. [DOI: 10.1007/s40521-022-00319-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
32
|
The Effects of Honey Sulfonamides on Immunological and Hematological Parameters in Wistar Rats. Medicina (B Aires) 2022; 58:medicina58111558. [PMID: 36363515 PMCID: PMC9697197 DOI: 10.3390/medicina58111558] [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: 09/20/2022] [Revised: 10/19/2022] [Accepted: 10/27/2022] [Indexed: 01/25/2023] Open
Abstract
Sulfonamides are among the most used drugs in beekeeping due to their effectiveness, despite their long-term persistence in tissues. Bee honey containing such residues poses numerous risks to human health. The aim of the study was to evaluate the effects on immunological and hematological parameters of Wistar rats produced by sulfonamide residues in bee honey, through the evaluation of various blood parameters such as triiodothyronine and thyroxine levels, hematocrit, hemoglobin, red blood cell count and mean corpuscular hemoglobin concentration in a given volume of erythrocytes following administration of sulfonamide-containing honey. The hematological and immunological parameters showed significant variations in the group of rats that had been fed with honey spiked with sulfonamides compared to the control group. Changes in hematological indices were demonstrated in terms of a significant reduction in the number of erythrocytes, the amount of hemoglobin, and the value of hematocrit, thus confirming the induction of anemia in the tested group. Investigation of thyroid function through the analysis of triiodothyronine (T3) and thyroxine (T4) and their ratio showed a very significant decrease in plasma thyroxine levels in laboratory rats that were fed sulfonamide-spiked honey compared to the control group. The mean T3 concentration decreased from 0.70 ± 0.14 ng/dL to 0.34 ± 0.03 ng/dL, while the mean T4 concentration was reduced from 4.50 ± 0.30 μg/dL to 3.32 ± 0.21 μg/dL, thus demonstrating toxic effects on thyroid function. In sum, the presence of sulfonamides induced significant changes in the evaluated parameters indicating that the consumption of contaminated honey samples represents a high risk factor for thyroid dysfunction with potentially serious health impacts.
Collapse
|
33
|
Combined IgE neutralization and Bifidobacterium longum supplementation reduces the allergic response in models of food allergy. Nat Commun 2022; 13:5669. [PMID: 36167830 PMCID: PMC9515155 DOI: 10.1038/s41467-022-33176-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/06/2022] [Indexed: 11/09/2022] Open
Abstract
IgE is central to the development of allergic diseases, and its neutralization alleviates allergic symptoms. However, most of these antibodies are based on IgG1, which is associated with an increased risk of fragment crystallizable-mediated side effects. Moreover, omalizumab, an anti-IgE antibody approved for therapeutic use, has limited benefits for patients with high IgE levels. Here, we assess a fusion protein with extracellular domain of high affinity IgE receptor, FcεRIα, linked to a IgD/IgG4 hybrid Fc domain we term IgETRAP, to reduce the risk of IgG1 Fc-mediated side effects. IgETRAP shows enhanced IgE binding affinity compared to omalizumab. We also see an enhanced therapeutic effect of IgETRAP in food allergy models when combined with Bifidobacterium longum, which results in mast cell number and free IgE levels. The combination of IgETRAP and B. longum may therefore represent a potent treatment for allergic patients with high IgE levels. IgE is a critical component of the allergic response and therapeutic targeting can alleviate symptomology. Here the authors propose the combined use of Bifidobacterium longum and a FcεRIα extracellular domain linked to a IgD/IgG4 hybrid Fc domain fusion protein called IgETRAP and show reduction of mast cell and IgE levels in models of food allergy.
Collapse
|
34
|
Modulatory effects of Porphyra-derived polysaccharides, oligosaccharides and their mixture on antigen-specific immune responses in ovalbumin-sensitized mice. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105209] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
35
|
Turner K, Boyd C, Rossi G, Sharp CR, Claus MA, Francis A, Smart L. Allergy, inflammation, hepatopathy and coagulation biomarkers in dogs with suspected anaphylaxis due to insect envenomation. Front Vet Sci 2022; 9:875339. [PMID: 36003410 PMCID: PMC9393546 DOI: 10.3389/fvets.2022.875339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives To compare concentrations of biomarkers of; allergy [mast cell tryptase (MCT) and histamine], inflammation [interleukin (IL)-6,-10, and-18, CXCL8, CCL2, keratinocyte chemoattractant (KC), C-reactive protein (CRP)], endothelial glycocalyx shedding (hyaluronan), coagulation [prothrombin time, activated partial thromboplastin time, fibrinogen concentration, and von Willebrand Factor antigen, protein C (PC) and antithrombin (AT) activity], and hepatopathy [alanine transaminase (ALT), aspartate transaminase (AST), alkaline phosphatase (ALP), and total bilirubin] between dogs with anaphylaxis after suspected insect exposure, dogs with critical illness, and healthy dogs. Design This was a single center prospective clinical observational comparative biomarker study that included 25 dogs with anaphylaxis (evidence of insect exposure, acute dermatological signs, and other organ involvement), 30 dogs with other critical illness, and 20 healthy dogs. Differences across groups in biomarker concentrations were tested using one-way ANOVA or Kruskal-Wallis test, with significant P values (<0.05) reported for pairwise differences detected by post-hoc tests. Logistic regression models were used to calculate the area under the receiver operator characteristic curve (AUROC) for discrimination between anaphylaxis and non-anaphylactic illness. Results Histamine concentration was significantly higher in the anaphylaxis group than the healthy (P < 0.001) and critically ill groups (P < 0.001), whereas no differences in MCT were detected amongst groups. Biomarker concentrations that were increased relative to healthy dogs in both the anaphylaxis and critically ill groups included IL-10 (P < 0.001 and P = 0.007, respectively), CCL2 (P = 0.007 and P < 0.001, respectively) and AST (both P < 0.001), whereas only the critically ill group had significantly increased CRP (P < 0.001), IL-6 (P < 0.001), KC (P < 0.001), ALP (P < 0.001), and fibrinogen (P = 0.016) concentrations, compared to the healthy group. Only dogs with anaphylaxis had significantly higher hyaluronan (P = 0.021) and ALT (P = 0.021) concentrations, and lower PC (P = 0.030) and AT (P = 0.032) activities, compared to healthy dogs. Both CRP and histamine concentration showed good discrimination between anaphylaxis and other critical illness, with an AUROC of 0.96 (95% CI 0.91-1) and 0.81 (95% CI 0.69-0.93), respectively. Conclusions This preliminary study in dogs with anaphylaxis after suspected insect exposure, found evidence of an early innate immune response, glycocalyx shedding and anticoagulant consumption. Both CRP and histamine showed potential clinical utility for differentiation between anaphylaxis and other critical illness.
Collapse
Affiliation(s)
- Kate Turner
- Emergency and Critical Care Department, School of Veterinary Medicine, Murdoch University, Perth, WA, Australia
| | - Corrin Boyd
- Emergency and Critical Care Department, School of Veterinary Medicine, Murdoch University, Perth, WA, Australia
| | - Gabriele Rossi
- Veterinary Pathology Department, School of Veterinary Medicine, Murdoch University, Perth, WA, Australia
| | - Claire R. Sharp
- Emergency and Critical Care Department, School of Veterinary Medicine, Murdoch University, Perth, WA, Australia
- Harry Butler Institute, Murdoch University, Perth, WA, Australia
| | - Melissa A. Claus
- Emergency and Critical Care Department, School of Veterinary Medicine, Murdoch University, Perth, WA, Australia
| | - Abbie Francis
- Telethon Kids Cancer Centre, Telethon Kids Institute, Nedlands, WA, Australia
- Discipline of Pediatrics, Medical School, The University of Western Australia, Nedlands, WA, Australia
| | - Lisa Smart
- Emergency and Critical Care Department, School of Veterinary Medicine, Murdoch University, Perth, WA, Australia
- Emergency and Critical Care Department, Small Animal Specialist Hospital, North Ryde, NSW, Australia
| |
Collapse
|
36
|
Chudakov DB, Kotsareva OD, Konovalova MV, Tsaregorodtseva DS, Shevchenko MA, Sergeev AA, Fattakhova GV. Early IgE Production Is Linked with Extrafollicular B- and T-Cell Activation in Low-Dose Allergy Model. Vaccines (Basel) 2022; 10:vaccines10060969. [PMID: 35746576 PMCID: PMC9231339 DOI: 10.3390/vaccines10060969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 06/01/2022] [Accepted: 06/14/2022] [Indexed: 12/23/2022] Open
Abstract
Despite its paramount importance, the predominant association of early IgE production with harmless antigens, via germinal-center B- and T-cell subpopulations or extrafollicular activation, remains unresolved. The aim of this work was to clarify whether the reinforced IgE production following the subcutaneous immunization of BALB/c mice with low antigen doses in withers adipose tissue might be linked with intensified extrafollicular or germinal-center responses. The mice were immunized three times a week for 4 weeks in the withers region, which is enriched in subcutaneous fat and tissue-associated B cells, with high and low OVA doses and via the intraperitoneal route for comparison. During long-term immunization with both low and high antigen doses in the withers region, but not via the intraperitoneal route, we observed a significant accumulation of B220-CD1d-CD5-CD19+ B-2 extrafollicular plasmablasts in the subcutaneous fat and regional lymph nodes but not in the intraperitoneal fat. Only low antigen doses induced a significant accumulation of CXCR4+ CXCR5- CD4+ extrafollicular T helpers in the withers adipose tissue but not in the regional lymph nodes or abdominal fat. Only in subcutaneous fat was there a combination of extrafollicular helper accumulation. In conclusion, extrafollicular B- and T-cell activation are necessary for early IgE class switching.
Collapse
Affiliation(s)
- Dmitrii Borisovich Chudakov
- Laboratory of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 16/10 Miklukho-Maklaya St., 117997 Moscow, Russia; (O.D.K.); (M.V.K.); (M.A.S.); (A.A.S.); (G.V.F.)
- Correspondence: ; Tel.: +7-495-330-4011
| | - Olga Dmitrievna Kotsareva
- Laboratory of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 16/10 Miklukho-Maklaya St., 117997 Moscow, Russia; (O.D.K.); (M.V.K.); (M.A.S.); (A.A.S.); (G.V.F.)
| | - Maryia Vladimirovna Konovalova
- Laboratory of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 16/10 Miklukho-Maklaya St., 117997 Moscow, Russia; (O.D.K.); (M.V.K.); (M.A.S.); (A.A.S.); (G.V.F.)
| | - Daria Sergeevna Tsaregorodtseva
- Faculty of Medical Biology, Sechenov First Moscow State Medical University, 2 Bolshaya Pirogovskaya St., 1194535 Moscow, Russia;
| | - Marina Alexandrovna Shevchenko
- Laboratory of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 16/10 Miklukho-Maklaya St., 117997 Moscow, Russia; (O.D.K.); (M.V.K.); (M.A.S.); (A.A.S.); (G.V.F.)
| | - Anton Andreevich Sergeev
- Laboratory of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 16/10 Miklukho-Maklaya St., 117997 Moscow, Russia; (O.D.K.); (M.V.K.); (M.A.S.); (A.A.S.); (G.V.F.)
| | - Gulnar Vaisovna Fattakhova
- Laboratory of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 16/10 Miklukho-Maklaya St., 117997 Moscow, Russia; (O.D.K.); (M.V.K.); (M.A.S.); (A.A.S.); (G.V.F.)
| |
Collapse
|
37
|
Allantoin Inhibits Compound 48/80-Induced Pseudoallergic Reactions In Vitro and In Vivo. Molecules 2022; 27:molecules27113473. [PMID: 35684410 PMCID: PMC9182162 DOI: 10.3390/molecules27113473] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 11/26/2022] Open
Abstract
Pseudoallergic reactions are hypersensitivity reactions mediated by an IgE-independent mechanism. Since allantoin (AT)-mediated pseudoallergy has not been studied, in this study, our objective is to investigate the anti-pseudoallergy effect of AT and its underlying mechanism. In vitro, β-hexosaminidase (β-Hex) and histamine (HIS) release assays, inflammatory cytokine assays, toluidine blue staining, and F-actin microfilament staining were used to evaluate the inhibitory effect of AT in RBL-2H3 cells stimulated with Compound 48/80 (C48/80). Western blot analysis is further performed to investigate intracellular calcium fluctuation-related signaling pathways. In vivo, Evans Blue extraction, paw swelling, and the diameter of Evans Blue extravasation were evaluated, and skin tissues are examined for histopathological examination in mice with passive cutaneous anaphylaxis (PCA) induced by C48/80. Body temperature is measured, and the levels of cytokines are further determined by ELISA kits in mice with active systemic anaphylaxis (ASA) induced by C48/80. The results show that AT dose-dependently inhibited degranulation in C48/80-stimulated RBL-2H3 cells by inhibiting β-Hex and HIS release, reducing the levels of TNF-α, IL-8, and MCP-1, inhibiting shape changes due to degranulation and disassembling the F-actin cytoskeleton. Furthermore, AT dose-dependently inhibits the phosphorylation of PLCγ and IP3R. In vivo, AT decreased Evans Blue extravasation, paw swelling, and the diameter of Evans Blue extravasation and significantly ameliorate pathological changes and mast cell degranulation in C48/80-induced PCA. Furthermore, AT help the mice recover from the C48/80-induced decrease in body temperature and decreased the levels of cytokines in C48/80-treated ASA mice. Our results indicate that allantoin inhibits compound 48/80-induced pseudoallergic reactions. AT has the potential to be used in IgE-independent anti-allergic and anti-inflammatory therapies.
Collapse
|
38
|
Landers JJ, Janczak KW, Shakya AK, Zarnitsyn V, Patel SR, Baker JR, Gill HS, O'Konek JJ. Targeted allergen-specific immunotherapy within the skin improves allergen delivery to induce desensitization to peanut. Immunotherapy 2022; 14:539-552. [PMID: 35196877 PMCID: PMC9043875 DOI: 10.2217/imt-2021-0206] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 01/26/2022] [Indexed: 11/21/2022] Open
Abstract
Aim: Epicutaneous immunotherapy (EPIT) with peanut has been demonstrated to be safe but efficacy may be limited by allergen uptake through the skin barrier. To enhance allergen uptake into the skin, the authors used peanut-coated microneedles and compared them with EPIT in a peanut allergy mouse model. Methods: Sensitized mice were treated with peanut-coated microneedles or peanut-EPIT and then challenged with peanut to determine protection. Results: Treatment with peanut-coated microneedles was safe and showed enhanced desensitization to peanut compared with peanut-EPIT administered via a similar schedule. Protection was associated with reduced Th2 immune responses and mast cell accumulation in the intestine. Conclusion: Peanut-coated microneedles have the potential to present a safe method of improving allergen delivery for cutaneous immunotherapy.
Collapse
Affiliation(s)
- Jeffrey J Landers
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Katarzyna W Janczak
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | - James R Baker
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Jessica J O'Konek
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
39
|
Kong YW, Dreaden EC. PEG: Will It Come Back to You? Polyethelyne Glycol Immunogenicity, COVID Vaccines, and the Case for New PEG Derivatives and Alternatives. Front Bioeng Biotechnol 2022; 10:879988. [PMID: 35573237 PMCID: PMC9092184 DOI: 10.3389/fbioe.2022.879988] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 04/11/2022] [Indexed: 11/21/2022] Open
Affiliation(s)
- Yi Wen Kong
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States
- *Correspondence: Yi Wen Kong, ; Erik C Dreaden, ,
| | - Erik C Dreaden
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA, United States
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
- *Correspondence: Yi Wen Kong, ; Erik C Dreaden, ,
| |
Collapse
|
40
|
Nuñez-Borque E, Fernandez-Bravo S, Yuste-Montalvo A, Esteban V. Pathophysiological, Cellular, and Molecular Events of the Vascular System in Anaphylaxis. Front Immunol 2022; 13:836222. [PMID: 35371072 PMCID: PMC8965328 DOI: 10.3389/fimmu.2022.836222] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/07/2022] [Indexed: 01/10/2023] Open
Abstract
Anaphylaxis is a systemic hypersensitivity reaction that can be life threatening. Mechanistically, it results from the immune activation and release of a variety of mediators that give rise to the signs and symptoms of this pathological event. For years, most of the research in anaphylaxis has focused on the contribution of the immune component. However, approaches that shed light on the participation of other cellular and molecular agents are necessary. Among them, the vascular niche receives the various signals (e.g., histamine) that elicit the range of anaphylactic events. Cardiovascular manifestations such as increased vascular permeability, vasodilation, hypotension, vasoconstriction, and cardiac alterations are crucial in the pathophysiology of anaphylaxis and are highly involved to the development of the most severe cases. Specifically, the endothelium, vascular smooth muscle cells, and their molecular signaling outcomes play an essential role downstream of the immune reaction. Therefore, in this review, we synthesized the vascular changes observed during anaphylaxis as well as its cellular and molecular components. As the risk of anaphylaxis exists both in clinical procedures and in routine life, increasing our knowledge of the vascular physiology and their molecular mechanism will enable us to improve the clinical management and how to treat or prevent anaphylaxis. Key Message Anaphylaxis, the most severe allergic reaction, involves a variety of immune and non-immune molecular signals that give rise to its pathophysiological manifestations. Importantly, the vascular system is engaged in processes relevant to anaphylactic events such as increased vascular permeability, vasodilation, hypotension, vasoconstriction, and decreased cardiac output. The novelty of this review focuses on the fact that new studies will greatly improve the understanding of anaphylaxis when viewed from a vascular molecular angle and specifically from the endothelium. This knowledge will improve therapeutic options to treat or prevent anaphylaxis.
Collapse
Affiliation(s)
- Emilio Nuñez-Borque
- Department of Allergy and Immunology, Instituto en Investigación Sanitaria - Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Sergio Fernandez-Bravo
- Department of Allergy and Immunology, Instituto en Investigación Sanitaria - Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Alma Yuste-Montalvo
- Department of Allergy and Immunology, Instituto en Investigación Sanitaria - Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Vanesa Esteban
- Department of Allergy and Immunology, Instituto en Investigación Sanitaria - Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Faculty of Medicine and Biomedicine, Alfonso X El Sabio University, Madrid, Spain
| |
Collapse
|
41
|
Fereydouni M, Motaghed M, Ahani E, Kafri T, Dellinger K, Metcalfe DD, Kepley CL. Harnessing the Anti-Tumor Mediators in Mast Cells as a New Strategy for Adoptive Cell Transfer for Cancer. Front Oncol 2022; 12:830199. [PMID: 35433433 PMCID: PMC9009255 DOI: 10.3389/fonc.2022.830199] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/28/2022] [Indexed: 12/12/2022] Open
Abstract
The emergence of cancer immunotherapies utilizing adoptive cell transfer (ACT) continues to be one of the most promising strategies for cancer treatment. Mast cells (MCs) which occur throughout vascularized tissues, are most commonly associated with Type I hypersensitivity, bind immunoglobin E (IgE) with high affinity, produce anti-cancer mediators such as tumor necrosis factor alpha (TNF-α) and granulocyte macrophage colony-stimulating factor (GM-CSF), and generally populate the tumor microenvironments. Yet, the role of MCs in cancer pathologies remains controversial with evidence for both anti-tumor and pro-tumor effects. Here, we review the studies examining the role of MCs in multiple forms of cancer, provide an alternative, MC-based hypothesis underlying the mechanism of therapeutic tumor IgE efficacy in clinical trials, and propose a novel strategy for using tumor-targeted, IgE-sensitized MCs as a platform for developing new cellular cancer immunotherapies. This autologous MC cancer immunotherapy could have several advantages over current cell-based cancer immunotherapies and provide new mechanistic strategies for cancer therapeutics alone or in combination with current approaches.
Collapse
Affiliation(s)
- Mohammad Fereydouni
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina Greensboro (UNCG), Greensboro, NC, United States
| | - Mona Motaghed
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, Greensboro, NC, United States
| | - Elnaz Ahani
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, Greensboro, NC, United States
| | - Tal Kafri
- Gene Therapy Center and Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kristen Dellinger
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, Greensboro, NC, United States
| | - Dean D. Metcalfe
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Christopher L. Kepley
- Department of Molecular and Cellular Sciences, Liberty University College of Osteopathic Medicine, Lynchburg, VA, United States
- *Correspondence: Christopher L. Kepley,
| |
Collapse
|
42
|
Homeostatic serum IgE is secreted by plasma cells in the thymus and enhances mast cell survival. Nat Commun 2022; 13:1418. [PMID: 35301301 PMCID: PMC8930980 DOI: 10.1038/s41467-022-29032-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 02/23/2022] [Indexed: 12/29/2022] Open
Abstract
Increased serum levels of immunoglobulin E (IgE) is a risk factor for various diseases, including allergy and anaphylaxis. However, the source and ontogeny of B cells producing IgE under steady state conditions are not well defined. Here, we show plasma cells that develop in the thymus and potently secrete IgE and other immunoglobulins, including IgM, IgA, and IgG. The development of these IgE-secreting plasma cells are induced by IL-4 produced by invariant Natural Killer T cells, independent of CD1d-mediated interaction. Single-cell transcriptomics suggest the developmental landscape of thymic B cells, and the thymus supports development of transitional, mature, and memory B cells in addition to plasma cells. Furthermore, thymic plasma cells produce polyclonal antibodies without somatic hypermutation, indicating they develop via the extra-follicular pathway. Physiologically, thymic-derived IgEs increase the number of mast cells in the gut and skin, which correlates with the severity of anaphylaxis. Collectively, we define the ontogeny of thymic plasma cells and show that steady state thymus-derived IgEs regulate mast cell homeostasis, opening up new avenues for studying the genetic causes of allergic disorders. Elevated levels of IgE is associated with a range of allergic pathology but the source of such IgE producing B cells during the steady state is poorly understood. Here, Kwon et al. show that homeostatic IgE is secreted by plasma cells in the thymus and link this to mast cell survival.
Collapse
|
43
|
Udoye CC, Rau CN, Freye SM, Almeida LN, Vera-Cruz S, Othmer K, Korkmaz RÜ, Clauder AK, Lindemann T, Niebuhr M, Ott F, Kalies K, Recke A, Busch H, Fähnrich A, Finkelman FD, Manz RA. B-cell receptor physical properties affect relative IgG1 and IgE responses in mouse egg allergy. Mucosal Immunol 2022; 15:1375-1388. [PMID: 36114245 PMCID: PMC9705252 DOI: 10.1038/s41385-022-00567-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 08/10/2022] [Accepted: 09/01/2022] [Indexed: 02/04/2023]
Abstract
Mutated and unmutated IgE and IgG play different and partly opposing roles in allergy development, but the mechanisms controlling their relative production are incompletely understood. Here, we analyzed the IgE-response in murine food allergy. Deep sequencing of the complementary-determining region (CDR) repertoires indicated that an ongoing unmutated extrafollicular IgE response coexists with a germinal center response, even after long-lasting allergen challenges. Despite overall IgG1-dominance, a significant proportion of clonotypes contained several-fold more IgE than IgG1. Clonotypes with differential bias to either IgE or IgG1 showed distinct hypermutation and clonal expansion. Hypermutation rates were associated with different physiochemical binding properties of individual B-cell receptors (BCR). Increasing BCR signaling strength inhibited class switching from IgG1 to IgE in vitro, preferentially constraining IgE formation. These data indicate that antigen-binding properties of individual BCRs determine differential IgE hypermutation and IgE versus IgG1 production on the level of single B-cell clones.
Collapse
Affiliation(s)
- Christopher C. Udoye
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Christina N. Rau
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Sarah M. Freye
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Larissa N. Almeida
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Sarah Vera-Cruz
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Kai Othmer
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Rabia Ü. Korkmaz
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Ann-Katrin Clauder
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Timo Lindemann
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Markus Niebuhr
- grid.4562.50000 0001 0057 2672Institute for Anatomy, University of Lübeck, Lübeck, Germany
| | - Fabian Ott
- grid.4562.50000 0001 0057 2672Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology and Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Kathrin Kalies
- grid.4562.50000 0001 0057 2672Institute for Anatomy, University of Lübeck, Lübeck, Germany
| | - Andreas Recke
- Department of Dermatology, Allergology and Venereology, University off Lübeck, Lübeck, Germany
| | - Hauke Busch
- grid.4562.50000 0001 0057 2672Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology and Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Anke Fähnrich
- grid.4562.50000 0001 0057 2672Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology and Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Fred D. Finkelman
- grid.239573.90000 0000 9025 8099Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, University of Cincinnati College of Medicine and the Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Rudolf A. Manz
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| |
Collapse
|
44
|
Bernardo LR, Ferreira LKDP, Ferreira LAMP, Vieira CID, Alves AF, Figueiredo PTR, Piuvezam MR. 4-Carvomenthenol, a monoterpene of essential oils, and its underlying effects on anti-inflammatory activity and immediate hypersensitivity reaction. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e20780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
45
|
Brock I, Eng N, Maitland A. Adult-onset mast cell activation syndrome following scombroid poisoning: a case report and review of the literature. J Med Case Rep 2021; 15:620. [PMID: 34920756 PMCID: PMC8684076 DOI: 10.1186/s13256-021-03190-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 11/12/2021] [Indexed: 11/18/2022] Open
Abstract
Background Mast cells are closely associated with epithelium, serving as sentinels responsible for the recognition of tissue injury and coordination of the initial inflammatory response. Upon detection of the injured cell content, mast cells then tailor the release of preformed and newly produced chemical mediators to the detected challenge, via an array of pathogen receptors. In addition to immunoglobulin E receptor-triggered mast cell activation, commonly referred to as allergic or atopic disorders, non-immunoglobulin E receptor mediated mast cell activation follows engagement of toll-like receptors, immunoglobulin G receptors, and complement receptors. Upon containment of the extrinsic challenge, acute inflammation is downregulated, and repair of the injured tissue ensues. The mast cell compartments must return to a baseline steady state to remain tolerant towards self-antigens and harmless entities, including environmental conditions, to prevent unnecessary immune activation and chronic hypersensitivity disorders. Over the past 50 years, an increasing number of patients are experiencing episodes of aberrant mast cell activation, not associated with allergen-specific mast cell disease or systemic mastocytosis. This led to proposed diagnostic criteria of mast cell activation syndrome. Mast cell activation syndrome is a heterogeneous disorder, defined by a combination of (1) recurrent symptoms typical of mast cell activation, (2) an increase of validated mast cell derived mediators, and (3) response to treatment with mast cell stabilizing or mast cell mediator-targeted therapies. Onset of mast cell activation syndrome ostensibly reflects the loss of tolerance in the mast cell compartment to nonthreatening entities and nonhazardous environmental conditions. The etiology of chronic mast cell dysregulation and associated intolerance to self-antigens or harmless entities is not well understood, but a growing number of studies point to exposure of the epithelial borders, which leads to inappropriate or excessive mast cell activation or impaired resolution of acute inflammation following neutralization of the identified pathogen. Case presentation Here we present a case of adult onset mast cell activation syndrome following scombroid poisoning. Scombroid toxicity is usually a self-limited illness, but there are individuals who have been shown to have severe symptoms or persistent illness following histamine fish poisoning. We describe a 74-year-old Caucasian woman, with a history of drug-induced urticaria, who developed a constellation of hypersensitivity illnesses consistent with the diagnosis of mast cell activation syndrome after ingestion of tainted fish. Conclusion Mast cell activation disease causes problems of increased complexity in children and adults. The increased prevalence and severity of mast cell activation disease has been attributed to dramatic changes in our lifestyles and modern living environments. These changes likely impact the integrity of the epithelial barriers, leading to loss of tolerance in the mast cell compartment. Here, we present a case of a nonatopic, 74-year-old female who developed mast cell activation disease after exposure to a potent environmental toxin. Mast cell activation disease commonly involves several organ systems, with patients often referred to a succession of different specialists. This results in delayed diagnosis and suboptimal care. Instead, early recognition of mast cell activation disease would lead to better outcomes. We review the literature, describing the diagnostic criteria for mast cell activation disorders that can improve recognition of this multiorgan system syndrome. Further research is needed into the interaction of epithelial barrier disruption and the dysregulation of the immune system.
Collapse
Affiliation(s)
- Isabelle Brock
- Comprehensive Asthma and Allergy, 200 S Broadway Suite 104, Tarrytown, NY, 10591, USA. .,Qolify, 99 Wall St Suite 1147, New York, NY, 10005, USA. .,Department of Medical and Molecular Genetics, Indiana University School of Medicine, 975 W. Walnut Street, IB 130, Indianapolis, IN, 46202, USA.
| | - Nicole Eng
- Nicole Eng, BS, Biomedical Studies, University of Pittsburgh, Pittsburgh, USA
| | - Anne Maitland
- Comprehensive Asthma and Allergy, 200 S Broadway Suite 104, Tarrytown, NY, 10591, USA.,Department of Neurology, Mount Sinai- South Nassau Hospital, The Chiari-EDS Center, 1420 Broadway, Hewlett, NY, 11557, USA
| |
Collapse
|
46
|
Smeekens JM, Kulis MD. Mouse Models of Food Allergy in the Pursuit of Novel Treatment Modalities. FRONTIERS IN ALLERGY 2021; 2:810067. [PMID: 35387036 PMCID: PMC8974753 DOI: 10.3389/falgy.2021.810067] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/23/2021] [Indexed: 11/13/2022] Open
Abstract
The prevalence of IgE-mediated food allergies has increased dramatically in the past three decades, now affecting up to 10% of the US population. IgE-mediated food allergy is an immunologic disease, involving a variety of cells, including B and T cells, mast cells, basophils, ILC2s, and epithelial cells. Mouse models of food allergy mimic the overall immunologic processes known to exist in humans. Due to the limitations of invasive sampling of human tissue and the similarities of the human and mouse immune systems, comprehensive pathogenesis studies of food allergy have been performed in mouse models. Mouse models have been effective in elucidating the roles of non-oral routes of sensitization and identifying key cells and molecules involved in allergic sensitization. Furthermore, the development of novel therapeutic approaches for food allergy has been accelerated through the use of pre-clinical mouse models. Despite the groundbreaking findings stemming from research in mice, there are continued efforts to improve the translational utility of these models. Here, we highlight the achievements in understanding food allergy development and efforts to bring novel treatment approaches into clinical trials.
Collapse
Affiliation(s)
- Johanna M. Smeekens
- Division of Allergy and Immunology, Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC, United States
- University of North Carolina Food Allergy Initiative, Chapel Hill, NC, United States
- *Correspondence: Johanna M. Smeekens
| | - Michael D. Kulis
- Division of Allergy and Immunology, Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC, United States
- University of North Carolina Food Allergy Initiative, Chapel Hill, NC, United States
| |
Collapse
|
47
|
Liu J, Yin J. Immunotherapy With Recombinant Alt a 1 Suppresses Allergic Asthma and Influences T Follicular Cells and Regulatory B Cells in Mice. Front Immunol 2021; 12:747730. [PMID: 34804031 PMCID: PMC8602824 DOI: 10.3389/fimmu.2021.747730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/18/2021] [Indexed: 01/07/2023] Open
Abstract
Background Alternaria is a major source of asthma-inducing allergens. Allergen-specific immunotherapy improves the progression of allergic asthma. The current treatment is based on crude Alternaria extracts. Alt a 1 is the predominant allergen in Alternaria. However, the treatment efficacy of recombinant Alt a 1 (rAlt a 1) in an asthmatic animal model and its influence on Tfh and Breg cells are unknown. Objective To explore the therapeutic treatment effects of rAlt a 1 on the progress of an asthmatic mouse model and its effect on Tfh and Breg cells. Methods We synthesized and purified rAlt a 1. Alternaria-sensitized and challenged mice received subcutaneous immunotherapy (SCIT) with four different rAlt a 1 dosages (5, 50, 100, and 150 µg) or PBS only. Finally, lung and airway inflammation, mouse mast cell protease 1 (MMCP-1), serum immunoglobulin responses, Tfh and Breg cell levels, and the correlation between asthmatic features (inflammation grades and IL-4 and IL-10 levels) and these two cell types were measured after Alternaria rechallenge. Results High purity and allergenic potency of rAlt a 1 protein were obtained. Following treatment with four different rAlt a 1 dosages, both lung and airway inflammation ameliorated, including lung pathology, serum MMCP-1 levels, inflammatory cell numbers, and cytokine levels in bronchoalveolar lavage fluid (BALF). Additionally, rAlt a 1-SCIT increased the expression of Alternaria-sIgG1, rAlt a 1-sIgG1, rAlt a 1-sIgG2a, and rAlt a 1-sIgG2b in serum. Moreover, the number and percentage of CXCR5+PD-1+Tfh cells were increased in the PC control, while they decreased in the rAlt a 1-SCIT groups. Meanwhile, the absolute numbers and proportions of Breg cells were evaluated after administration of rAlt a 1. A positive correlation was observed between CXCR5+PD-1+Tfh cells and inflammation grades (r = 0.50, p = 0.01), as well as a slightly strong positive relationship with IL-4 (r = 0.55, p = 0.005) and IL-10 (r = 0.58, p = 0.003) levels; Breg cells showed an opposite correlation with the grades of inflammation (r = -0.68, p = 0.0003), along with a negative correlation to IL-4 (r = -0.61, p = 0.001) and IL-10 (r = -0.53, p = 0.008) levels. Conclusions We verified that treatment with rAlt a 1 can alleviate asthma progression and further have a regulatory effect on Tfh and Breg cells in an Alternaria-induced asthmatic mouse model.
Collapse
Affiliation(s)
- Juan Liu
- Department of Allergy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Department of Allergy, Peking Union Medical College Hospital, Beijing Key Laboratory of Precision Medicine For Diagnosis and Treatment on Allergic Diseases, Beijing, China.,Department of Allergy, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Disease, Beijing, China
| | - Jia Yin
- Department of Allergy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Department of Allergy, Peking Union Medical College Hospital, Beijing Key Laboratory of Precision Medicine For Diagnosis and Treatment on Allergic Diseases, Beijing, China.,Department of Allergy, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Disease, Beijing, China
| |
Collapse
|
48
|
Vultaggio A, Perlato M, Nencini F, Vivarelli E, Maggi E, Matucci A. How to Prevent and Mitigate Hypersensitivity Reactions to Biologicals Induced by Anti-Drug Antibodies? Front Immunol 2021; 12:765747. [PMID: 34790200 PMCID: PMC8591239 DOI: 10.3389/fimmu.2021.765747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/13/2021] [Indexed: 01/06/2023] Open
Abstract
Biologicals are widely used therapeutic agents for rheumatologic diseases, cancers, and other chronic inflammatory diseases. They are characterized by complex structures and content of variable amounts of foreign regions, which may lead to anti-drug antibodies (ADA) development. ADA onset may limit the clinical usage of biologicals because they may decrease their safety. In fact they are mainly associated with immediate hypersensitivity reactions (HSRs). Development of ADAs is reduced by concomitant immunosuppressive treatment, while it is increased by longer intervals between drug administrations; thus, regular infusion regimens should be preferred to reduce HSRs. Once ADAs have formed, some procedures can be implemented to reduce the risk of HSRs. ADAs may belong to different isotype; the detection of IgE ADA is advisable to be assessed when high and early ADAs are detected, in order to reduce the risk of severe HRs. In patients who need to reintroduce the biological culprit, as alternative therapies are not available, drug desensitization (DD) may be applied. Desensitization should be conceptually dedicated to patients with an IgE-mediated HSR; however, it can be performed also in patients who had developed non-IgE-mediated HSRs. Although the underlying mechanisms behind successful DD has not been fully clarified, the DD procedure is associated with the inhibition of mast cell degranulation and cytokine production. Additionally, some data are emerging about the inhibition of drug-specific immune responses during DD.
Collapse
Affiliation(s)
| | - Margherita Perlato
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Francesca Nencini
- Immunoallergology Unit, Careggi University Hospital, Florence, Italy
| | | | - Enrico Maggi
- Translational Immunology Unit, Immunology Area, Pediatric Hospital Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS), Rome, Italy
| | - Andrea Matucci
- Immunoallergology Unit, Careggi University Hospital, Florence, Italy
| |
Collapse
|
49
|
Stackowicz J, Gaudenzio N, Serhan N, Conde E, Godon O, Marichal T, Starkl P, Balbino B, Roers A, Bruhns P, Jönsson F, Moguelet P, Georgin-Lavialle S, Broderick L, Hoffman HM, Galli SJ, Reber LL. Neutrophil-specific gain-of-function mutations in Nlrp3 promote development of cryopyrin-associated periodic syndrome. J Exp Med 2021; 218:212620. [PMID: 34477811 PMCID: PMC8421266 DOI: 10.1084/jem.20201466] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 06/10/2021] [Accepted: 07/30/2021] [Indexed: 12/14/2022] Open
Abstract
Gain-of-function mutations in NLRP3 are responsible for a spectrum of autoinflammatory diseases collectively referred to as “cryopyrin-associated periodic syndromes” (CAPS). Treatment of CAPS patients with IL-1–targeted therapies is effective, confirming a central pathogenic role for IL-1β. However, the specific myeloid cell population(s) exhibiting inflammasome activity and sustained IL-1β production in CAPS remains elusive. Previous reports suggested an important role for mast cells (MCs) in this process. Here, we report that, in mice, gain-of-function mutations in Nlrp3 restricted to neutrophils, and to a lesser extent macrophages/dendritic cells, but not MCs, are sufficient to trigger severe CAPS. Furthermore, in patients with clinically established CAPS, we show that skin-infiltrating neutrophils represent a substantial biological source of IL-1β. Together, our data indicate that neutrophils, rather than MCs, can represent the main cellular drivers of CAPS pathology.
Collapse
Affiliation(s)
- Julien Stackowicz
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR 1222, Institut national de la santé et de la recherche médicale, Paris, France.,Sorbonne Université, Paris, France
| | - Nicolas Gaudenzio
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Institut national de la santé et de la recherche médicale, UMR 1291, Centre National de la Recherche Scientifique, UMR 5051, University of Toulouse III, Toulouse, France
| | - Nadine Serhan
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Institut national de la santé et de la recherche médicale, UMR 1291, Centre National de la Recherche Scientifique, UMR 5051, University of Toulouse III, Toulouse, France
| | - Eva Conde
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR 1222, Institut national de la santé et de la recherche médicale, Paris, France
| | - Ophélie Godon
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR 1222, Institut national de la santé et de la recherche médicale, Paris, France
| | - Thomas Marichal
- GIGA-Research and Faculty of Veterinary Medicine, Liège University, Liège, Belgium
| | - Philipp Starkl
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Research Laboratory of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Bianca Balbino
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR 1222, Institut national de la santé et de la recherche médicale, Paris, France.,Sorbonne Université, Paris, France
| | - Axel Roers
- Institute for Immunology, Medical Faculty Carl Gustav Carus, University of Technology Dresden, Dresden, Germany
| | - Pierre Bruhns
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR 1222, Institut national de la santé et de la recherche médicale, Paris, France
| | - Friederike Jönsson
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR 1222, Institut national de la santé et de la recherche médicale, Paris, France
| | - Philippe Moguelet
- Faculty of Medicine, Sorbonne University, Tenon Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Sophie Georgin-Lavialle
- Internal Medicine Department, Tenon Hospital, Assistance Publique - Hôpitaux de Paris, Sorbonne University, Paris, France
| | - Lori Broderick
- Division of Pediatric Allergy, Immunology and Rheumatology, University of California, San Diego, and Rady Children's Hospital, San Diego, CA
| | - Hal M Hoffman
- Division of Pediatric Allergy, Immunology and Rheumatology, University of California, San Diego, and Rady Children's Hospital, San Diego, CA
| | - Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, CA.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA.,Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA
| | - Laurent L Reber
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR 1222, Institut national de la santé et de la recherche médicale, Paris, France.,Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Institut national de la santé et de la recherche médicale, UMR 1291, Centre National de la Recherche Scientifique, UMR 5051, University of Toulouse III, Toulouse, France
| |
Collapse
|
50
|
Pennington LF, Gasser P, Brigger D, Guntern P, Eggel A, Jardetzky TS. Structure-guided design of ultrapotent disruptive IgE inhibitors to rapidly terminate acute allergic reactions. J Allergy Clin Immunol 2021; 148:1049-1060. [PMID: 33991582 PMCID: PMC8502201 DOI: 10.1016/j.jaci.2021.03.050] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 02/24/2021] [Accepted: 03/26/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Anaphylaxis represents one of the most severe and fatal forms of allergic reactions. Like most other allergies, it is caused by activation of basophils and mast cells by allergen-mediated cross-linking of IgE bound to its high-affinity receptor, FcεRI, on the cell surface. The systemic release of soluble mediators induces an inflammatory cascade, rapidly causing symptoms with peak severity in minutes to hours after allergen exposure. Primary treatment for anaphylaxis consists of immediate intramuscular administration of adrenaline. OBJECTIVE While adrenaline alleviates life-threatening symptoms of an anaphylactic reaction, there are currently no disease-modifying interventions available. We sought to develop potent and fast-acting IgE inhibitors with the potential to rapidly terminate acute allergic reactions. METHODS Using affinity maturation by yeast display and structure-guided molecular engineering, we generated 3 optimized disruptive IgE inhibitors based on designed ankyrin repeat proteins and assessed their ability to actively remove IgE from allergic effector cells in vitro as well as in vivo in mice. RESULTS The engineered IgE inhibitors rapidly dissociate preformed IgE:FcεRI complexes, terminate IgE-mediated signaling in preactivated human blood basophils in vitro, and shut down preinitiated allergic reactions and anaphylaxis in mice in vivo. CONCLUSIONS Fast-acting disruptive IgE inhibitors demonstrate the feasibility of developing kinetically optimized inhibitors for the treatment of anaphylaxis and the rapid desensitization of allergic individuals.
Collapse
Affiliation(s)
- Luke F Pennington
- Department of Structural Biology, Stanford University School of Medicine, Stanford, Calif; Program in Immunology, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy Research at Stanford University, Stanford, Calif
| | - Pascal Gasser
- Department of Rheumatology and Immunology, Bern University Hospital, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Daniel Brigger
- Department of Rheumatology and Immunology, Bern University Hospital, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Pascal Guntern
- Department of Rheumatology and Immunology, Bern University Hospital, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Alexander Eggel
- Department of Rheumatology and Immunology, Bern University Hospital, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland.
| | - Theodore S Jardetzky
- Department of Structural Biology, Stanford University School of Medicine, Stanford, Calif; Program in Immunology, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy Research at Stanford University, Stanford, Calif.
| |
Collapse
|