1
|
Pahima HT, Dwyer DF. Update on mast cell biology. J Allergy Clin Immunol 2025; 155:1115-1123. [PMID: 39800266 DOI: 10.1016/j.jaci.2024.12.1092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/15/2025]
Abstract
Mast cells (MCs) are heterogeneous tissue-resident effector cells that are thought to play central roles in allergic inflammatory disease, yet the degree of heterogeneity and nature of these roles has remained elusive. In recent years, advances in tissue culture systems, preclinical mouse models, and the continued spread of single-cell RNA sequencing have greatly advanced our understanding of MC phenotypes in health and disease. These approaches have identified novel interactions of MC subsets with immune cells, neurons, and tissue structural cells, changing our understanding of how MCs both drive and help resolve tissue inflammation, reshape tissue microenvironments, and influence host behavior. This review addresses key studies from 2022 to 2024 that have advanced our understanding of MC biology in mice and humans.
Collapse
Affiliation(s)
- Hadas Tamar Pahima
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Daniel F Dwyer
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass.
| |
Collapse
|
2
|
Duque-Wilckens N, Joseph D, Syed M, Smith B, Maradiaga N, Yeh SY, Srinivasan V, Sotomayor F, Durga K, Nestler E, Moesers AJ, Robison AJ. FosB/ΔFosB activation in mast cells regulates gene expression to modulate allergic inflammation in male mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.06.592755. [PMID: 38766119 PMCID: PMC11100602 DOI: 10.1101/2024.05.06.592755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Mast cells are innate immune cells that regulate physiological processes by releasing pre-stored and newly synthesized mediators in response to allergens, infection, and other stimuli. Dysregulated mast cell activity can lead to multisystemic pathologies, but the underlying regulatory mechanisms remain poorly understood. We found that FOSB and ΔFOSB, transcription factors encoded by the FosB gene, are robustly expressed in mast cells following IgE-antigen stimulation, suggesting a role in modulating stimulus-induced mast cell functions. Using phenotypic, gene binding, and gene expression analyses in wild-type and mast cell-specific FosB knockout male mice, we demonstrate that FOSB/ΔFOSB modulates mast cell functions by limiting reactivity to allergen-like stimuli both in vitro and in vivo . These effects seem to be mediated, at least in part, by FOSB/ΔFOSB-driven enhanced expression of DUSP4, a dual-specificity phosphatase that attenuates MAPK signaling. These findings highlight FOSB/ΔFOSB as critical regulators of mast cell activity and potential targets for therapeutic intervention.
Collapse
|
3
|
Murphy CT, Bachelder EM, Ainslie KM. Mast cell activators as adjuvants for intranasal mucosal vaccines. Int J Pharm 2025; 672:125300. [PMID: 39914508 DOI: 10.1016/j.ijpharm.2025.125300] [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: 12/18/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/10/2025]
Abstract
Mast cells have roles in immune regulation, allergy, and host response to pathogens. Compounds that activate mast cells (MCAs) can serve as vaccine adjuvants, potentially outperforming current FDA-approved options, especially for mucosal vaccines. While most vaccines are administered intramuscularly, intranasal and needle-free formulations offer benefits like improved compliance and accessibility. However, the lack of effective adjuvants limits mucosal vaccine development. This review explores MCAs as promising alternatives to traditional adjuvants, aiming to enhance mucosal vaccine efficacy. We summarize the nascent work of formulating MCAs like compound 48/80 into nanoparticles, with excipients such as chitosan and chitosan/alginate. Other MCAs like the peptide mastoparan 7 complexed with CpG have formed nanoparticle complexes that illustrate protective mucosal immunity in a model of influenza. The small molecule MCA ST101036, when encapsulated in acetalated dextran particles, has demonstrated enhanced immune responses and protection in a West Nile Virus model of infection. This review highlights the potential of MCAs as potent vaccine adjuvants, particularly for mucosal vaccines, and summarizes, recent advancements in formulating these activators into nanoparticles to enhance immune responses and protection.
Collapse
Affiliation(s)
- Connor T Murphy
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC Chapel Hill NC USA
| | - Eric M Bachelder
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC Chapel Hill NC USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC Chapel Hill NC USA; Department of Biomedical Engineering NC State/UNC Chapel Hill NC USA; Department of Microbiology and Immunology, School of Medicine, UNC Chapel Hill NC USA.
| |
Collapse
|
4
|
Arul Arasan TS, Jorgensen R, Van Antwerp C, Ng PKW, Gangur V. Advances in Mechanisms of Anaphylaxis in Wheat Allergy: Utility of Rodent Models. Foods 2025; 14:883. [PMID: 40077585 PMCID: PMC11899146 DOI: 10.3390/foods14050883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/20/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Wheat is a staple and nutritious food that is consumed globally. However, it is identified as a major allergenic food because of its capacity to trigger life-threatening systemic anaphylaxis. The specific mechanisms that underlie this systemic anaphylaxis in wheat allergy are incompletely understood. As a result, several rodent models have been developed to study anaphylaxis in wheat allergies. In this paper, we have conducted a comprehensive review of wheat-induced anaphylaxis using Google Scholar and PubMed databases with relevant keywords. The following objectives were addressed: (1) to determine the complexity of wheat-induced anaphylaxis; (2) to summarize the role of genetic susceptibility in wheat anaphylaxis; (3) to identify the environmental factors involved in the development of wheat anaphylaxis; (4) to map the current status of mechanisms involved in wheat anaphylaxis; (5) to identify the approaches, strengths, and limitations of rodent models of wheat anaphylaxis; and (6) to identify challenges and opportunities in this area of science. Our findings provide a comprehensive updated critical resource for the future research agenda in wheat allergy-associated anaphylaxis, particularly using rodent models as attractive pre-clinical tools.
Collapse
Affiliation(s)
- Tamil Selvan Arul Arasan
- Food Allergy and Immunology Laboratory, Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA; (T.S.A.A.); (R.J.); (C.V.A.)
| | - Rick Jorgensen
- Food Allergy and Immunology Laboratory, Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA; (T.S.A.A.); (R.J.); (C.V.A.)
| | - Chris Van Antwerp
- Food Allergy and Immunology Laboratory, Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA; (T.S.A.A.); (R.J.); (C.V.A.)
| | - Perry K. W. Ng
- Cereal Science Laboratory, Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48823, USA;
| | - Venu Gangur
- Food Allergy and Immunology Laboratory, Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA; (T.S.A.A.); (R.J.); (C.V.A.)
| |
Collapse
|
5
|
Globig P, Morakabati P, Höfer V, Willmes DM, Babina M, Worm M. Acetylsalicylic acid aggravates anaphylaxis in a PGE2-dependent manner. J Clin Invest 2025; 135:e175397. [PMID: 40026245 DOI: 10.1172/jci175397] [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: 09/11/2023] [Accepted: 01/08/2025] [Indexed: 03/05/2025] Open
Abstract
Acetylsalicylic acid (ASA) can exert proanaphylactic effects, but the extent of this phenomenon and its underlying mechanisms are undefined. Yet, low homeostatic prostaglandin E2 (PGE2) levels have been associated with anaphylaxis. In this study, we investigated whether the proanaphylactic effect of ASA is PGE2 dependent. We assessed the effect of ASA in experimental anaphylaxis models, analyzed a large dataset of patients with anaphylaxis, and performed titrated allergen challenges in ASA-treated allergic individuals. Registry data indicated an increased risk for severe anaphylaxis in patients with ASA comedication. ASA pretreatment aggravated allergen-dependent anaphylaxis in mice, whereas histamine-induced anaphylaxis remained unaffected. Exacerbation was due to reduced PGE2, as its stabilization or the use of prostanoid E receptor (EP) agonists reversed the proanaphylactic effects of ASA. EP2-, EP3-, and EP4 receptor-deficient mice revealed that each receptor individually contributed to ASA susceptibility. In patients with allergy, prior ASA intake increased skin responsiveness to allergen but not to histamine. Conversely, the responses of basophils to ex vivo FcεRI aggregation remained unaltered, indicating that ASA operated by enhancing the stimulability of mast cells in a PGE2-dependent manner. Collectively, our data reveal a central role of the PGE2 network in ASA-aggravated anaphylaxis. EP receptors could be potential targets to prevent or alter the outcome of anaphylaxis.
Collapse
Affiliation(s)
- Philipp Globig
- Division of Allergy and Immunology, Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Payam Morakabati
- Division of Allergy and Immunology, Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Veronika Höfer
- Division of Allergy and Immunology, Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Diana M Willmes
- Division of Allergy and Immunology, Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Magda Babina
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Berlin, Germany
| | - Margitta Worm
- Division of Allergy and Immunology, Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
6
|
Plum T, Feyerabend TB, Rodewald HR. Beyond classical immunity: Mast cells as signal converters between tissues and neurons. Immunity 2024; 57:2723-2736. [PMID: 39662090 DOI: 10.1016/j.immuni.2024.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/15/2024] [Accepted: 11/12/2024] [Indexed: 12/13/2024]
Abstract
Mast cells are regarded as effectors in immune defense against parasites and venoms and play an essential role in the pathology of allergic diseases. More recently, mast cells have been shown to receive stimuli derived from type 2 immunity, tissue damage, stress, and inflammation. Mast cells then rapidly convert these diverse signals into appropriate, organ-specific protective reflexes that can limit inflammation or reduce tissue damage. In this review, we consider functions of mast cells in sensations-such as pain, itch, and nausea-arising from tissue insults and inflammation and the ensuing protective responses. In light of emerging data highlighting the involvement of mast cells in neuroimmune communication, we also propose that mast cells are "signal converters" linking immunological and tissue states with nervous system responses.
Collapse
Affiliation(s)
- Thomas Plum
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany.
| | - Thorsten B Feyerabend
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany.
| |
Collapse
|
7
|
Lu S, Jiang Q, Zhou P, Yin L, Wang N, Xu J, Qian Q, Tao M, Yin H, Han L, Gu Y, Gao F, Liu J, Chen S. Targeting Dlat-Trpv3 pathway by hyperforin elicits non-canonical promotion of adipose thermogenesis as an effective anti-obesity strategy. J Adv Res 2024:S2090-1232(24)00555-1. [PMID: 39631519 DOI: 10.1016/j.jare.2024.11.035] [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: 06/27/2024] [Revised: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024] Open
Abstract
INTRODUCTION Promoting adipose thermogenesis is considered as a promising therapeutic intervention in obesity. However, endeavors to develop anti-obesity medications by targeting the canonical thermogenesis regulatory pathway, particularly β3-adrenergic receptor (β3-AR)-dependent mechanism, have failed due to the off-target effects of β3-AR agonists, exacerbating the risk of cardiovascular disease. Hyperforin (HPF), a natural compound extracted from the traditional herbal St. John's Wort, binds to Dihydrolipoamide s-acetyltransferase (Dlat) and exerts effective anti-obesity properties through promoting adipose thermogenesis. OBJECTIVES The objective of this study was to investigate the oral efficacy and pharmacokinetics profile of HPF, and explore the detailed mechanism by which Dlat modulates HPF-mediated adipose thermogenesis. METHODS To assess the anti-obesity efficacy of orally administered HPF in vivo, Dlat heterozygous knockout (Dlat+/-) mice and wild-type (WT) mice, both fed a high-fat diet (HFD), underwent a validation process that involved the use of metabolic cages, NMR analysis, and infrared imaging. Sprague Dawley rats were employed to determine the pharmacokinetic parameters of HPF. Seahorse assays, JC-1 staining, qPCR, and immunoblotting were performed to evaluate cellular thermogenic efficacy of HPF and Dlat in vitro. RESULTS Our study uncovered a non-canonical thermogenesis pathway involving Dlat, transient receptor potential vanilloid 3 (Trpv3, a calcium channel) and AMPK. Dlat interacted with Trpv3 to activate it, resulting in an increase in intracellular calcium (Ca2+) and the activation of Camkkβ. Camkkβ then stimulated AMPK, leading to elevated Ucp1 expression and initiating adipose thermogenesis. HPF promoted thermogenesis in adipose tissues through enhancing the Dlat-Trpv3 interaction independently of β3-AR, causing minimal cardiac side effects. Notably, HPF's thermogenic effects were reduced in Dlat+/- mice. Moreover, HPF exerted favorable oral bioavailability, a relatively long half-life, and extensive distribution within adipose tissues. CONCLUSION In summary, our study demonstrates that HPF targets a novel mechanism for promoting adipose thermogenesis and exhibits potent and safe anti-obesity efficacy.
Collapse
Affiliation(s)
- Sijia Lu
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Quanxin Jiang
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Peihui Zhou
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Limin Yin
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Ning Wang
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Junting Xu
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Qiqi Qian
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Mijia Tao
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hanrui Yin
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Liu Han
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yunqing Gu
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Fei Gao
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Junli Liu
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Suzhen Chen
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
8
|
Houghton V, Eiwegger T, Florsheim EB, Knibb RC, Thuret S, Santos AF. From bite to brain: Neuro-immune interactions in food allergy. Allergy 2024; 79:3326-3340. [PMID: 39462229 DOI: 10.1111/all.16366] [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: 06/25/2024] [Revised: 09/17/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024]
Abstract
Immunoglobulin E (IgE)-mediated food allergies are reported to affect around 3.5% of children and 2.4% of adults, with symptoms varying in range and severity. While being the gold standard for diagnosis, oral food challenges are burdensome, and diagnostic tools based on specific IgE can be flawed. Furthering our understanding of the mechanisms behind food allergy onset, severity and persistence could help reveal immune profiles associated with the disease, to ultimately aid in diagnosis. Alterations to cytokine levels and immune cell ratios have been identified, though further research is needed to fully capture the heterogenous nature of food allergy. Moreover, the existence of such immune alterations also raises the question of potential wider systemic effects. For example, recent research has emphasised the existence and impact of neuro-immune interactions and implicated behavioural and neurological changes associated with food allergy. This review will provide an overview of such food allergy-driven neuro-immune interactions, with the aim of emphasising the importance of furthering our understanding of the immune mechanisms underlying IgE-mediated food allergy.
Collapse
Affiliation(s)
- Vikki Houghton
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Thomas Eiwegger
- Department of Pediatric and Adolescent Medicine, University Hospital St. Pölten, St. Pölten, Austria
- Translational Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Canada
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Esther Borges Florsheim
- Center for Health Through Microbiomes, Biodesign Institute Arizona State University Tempe, Arizona, USA
- School of Life Sciences, Arizona State University Tempe, Arizona, USA
| | - Rebecca C Knibb
- Institute of Health and Neurodevelopment, Aston University, Birmingham, UK
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Alexandra F Santos
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
- Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
- Children's Allergy Service, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
9
|
Wang Z, Song K, Kim BS, Manion J. Sensory neuroimmune interactions at the barrier. Mucosal Immunol 2024; 17:1151-1160. [PMID: 39374664 DOI: 10.1016/j.mucimm.2024.10.001] [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: 07/30/2024] [Revised: 09/24/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
Epithelial barriers such as the skin, lung, and gut, in addition to having unique physiologic functions, are designed to preserve tissue homeostasis upon challenge with a variety of allergens, irritants, or pathogens. Both the innate and adaptive immune systems play a critical role in responding to epithelial cues triggered by environmental stimuli. However, the mechanisms by which organs sense and coordinate complex epithelial, stromal, and immune responses have remained a mystery. Our increasing understanding of the anatomic and functional characteristics of the sensory nervous system is greatly advancing a new field of peripheral neuroimmunology and subsequently changing our understanding of mucosal immunology. Herein, we detail how sensory biology is informing mucosal neuroimmunology, even beyond neuroimmune interactions seen within the central and autonomic nervous systems.
Collapse
Affiliation(s)
- Zhen Wang
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Keaton Song
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brian S Kim
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA.
| | - John Manion
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Department of Urology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Surgery, Harvard Medical School, Boston, MA 02115, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
10
|
Nagamine M, Kaitani A, Izawa K, Ando T, Yoshikawa A, Nakamura M, Maehara A, Yamamoto R, Okamoto Y, Wang H, Yamada H, Maeda K, Nakano N, Shimizu T, Ogawa H, Okumura K, Kitaura J. Neuronal substance P-driven MRGPRX2-dependent mast cell degranulation products differentially promote vascular permeability. Front Immunol 2024; 15:1477072. [PMID: 39640264 PMCID: PMC11617324 DOI: 10.3389/fimmu.2024.1477072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/30/2024] [Indexed: 12/07/2024] Open
Abstract
Mas-related G protein-coupled receptor b2 (Mrgprb2) binding to its cationic endogenous and exogenous ligands induces mast cell degranulation and promotes inflammation in mice. However, the physiological roles of its human homologue MRGPRX2 remain unclear. Here we aimed to elucidate the mechanisms by which MRGPRX2 regulates vascular permeability, and generated MRGPRX2 knock-in (MRGPRX2-KI) and Mrgprb2 knockout (Mrgprb2-KO) mice. Substance P (SP) and ciprofloxacin strongly degranulated MRGPRX2-KI peritoneal mast cells (PMCs) better than WT PMCs, whereas Dermatophagoides pteronyssinus (Der p) extract and phenol-soluble modulin α3 (PSMα3) did not degranulate PMCs. SP-stimulated MRGPRX2-KI PMCs released large amounts of histamine and mast cell protease 4 (MCPT4) chymase. Der p extract, PSMα3, and MCPT4, but not histamine, induced SP release from dorsal root ganglion (DRG) cells. However, this effect of Der p extract/PSMα3 was suppressed by a transient receptor potential vanilloid 1 (TRPV1) antagonist. SP-, ciprofloxacin-, Der p extract-, PSMα3-, and MCPT4-induced vascular permeability was highest in MRGPRX2-KI mice, which depended on SP. In addition, SP-, ciprofloxacin- and PSMα3-induced MRGPRX2-dependent vascular hyperpermeability was suppressed by antihistamine and chymase inhibitor. TRPV1 antagonist also inhibited PSMα3-induced MRGPRX2-dependent vascular hyperpermeability. Both Mrgprb2-KO and MRGPRX2-KI did not influence the histamine-induced murine vascular hyperpermeability. Overall, our results suggest that neuronal SP induces MRGPRX2-dependent mast cell degranulation, releasing histamine and chymase, which promote vascular hyperpermeability directly or indirectly via DRG cell activation. Importantly, the worsening cycle (MRGPRX2 → mast cell degranulation → chymase → DRG activation → SP → MRGPRX2) seems to play an important role in human MRGPRX2-depdendent inflammation.
Collapse
Affiliation(s)
- Masakazu Nagamine
- Department of Science of Allergy and Inflammation, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ayako Kaitani
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kumi Izawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tomoaki Ando
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akihisa Yoshikawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Otorhinolaryngology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masahiro Nakamura
- Department of Otorhinolaryngology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akie Maehara
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Risa Yamamoto
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoko Okamoto
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hexing Wang
- Department of Science of Allergy and Inflammation, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiromichi Yamada
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Keiko Maeda
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Immunological Diagnosis, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nobuhiro Nakano
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Toshiaki Shimizu
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hideoki Ogawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ko Okumura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Jiro Kitaura
- Department of Science of Allergy and Inflammation, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
11
|
Argueta DA, Tran H, Goel Y, Nguyen A, Nguyen J, Kiven SB, Chen C, Abdulla F, Vercellotti GM, Belcher JD, Gupta K. Mast cell extracellular trap formation underlies vascular and neural injury and hyperalgesia in sickle cell disease. Life Sci Alliance 2024; 7:e202402788. [PMID: 39242155 PMCID: PMC11381676 DOI: 10.26508/lsa.202402788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/09/2024] Open
Abstract
Sickle cell disease (SCD) is the most common inherited monogenetic disorder. Chronic and acute pain are hallmark features of SCD involving neural and vascular injury and inflammation. Mast cells reside in the vicinity of nerve fibers and vasculature, but how they influence these structures remains unknown. We therefore examined the mechanism of mast cell activation in a sickle microenvironment replete with cell-free heme and inflammation. Mast cells exposed to this environment showed an explosion of nuclear contents with the release of citrullinated histones, suggestive of mast cell extracellular trap (MCET) release. MCETs interacted directly with the vasculature and nerve fibers, a cause of vascular and neural injury in sickle cell mice. MCET formation was dependent upon peptidylarginine deiminase 4 (PAD4). Inhibition of PAD4 ameliorated vasoocclusion, chronic and acute hyperalgesia, and inflammation in sickle mice. PAD4 activation may also underlie neutrophil trap formation in SCD, thus providing a novel target to treat the sequelae of vascular and neural injury in SCD.
Collapse
Affiliation(s)
- Donovan A Argueta
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Huy Tran
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Yugal Goel
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Aithanh Nguyen
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Julia Nguyen
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Stacy B Kiven
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Chunsheng Chen
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Fuad Abdulla
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Gregory M Vercellotti
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - John D Belcher
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Kalpna Gupta
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, USA
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| |
Collapse
|
12
|
Chiu IM, Sokol CL. Neuroimmune recognition of allergens. Curr Opin Immunol 2024; 90:102458. [PMID: 39213825 PMCID: PMC11423315 DOI: 10.1016/j.coi.2024.102458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/07/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Interactions between the nervous system and the immune system play crucial roles in initiating and directing the type 2 immune response. Sensory neurons can initiate innate and adaptive type 2 immunity through their ability to detect allergens and promote dendritic cell and mast cell responses. Neurons also indirectly promote type 2 inflammation through suppression of type 1 immune responses. Type 2 cytokines promote neuronal function by directly activating or sensitizing neurons. This positive neuroimmune feedback loop may not only enhance allergic inflammation but also promote the system-wide responses of aversion, anaphylaxis, and allergen polysensitization that are characteristic of allergic immunity.
Collapse
Affiliation(s)
- Isaac M Chiu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| | - Caroline L Sokol
- Center for Immunology & Inflammatory Diseases, Division of Rheumatology, Allergy & Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
13
|
Bai T, Shao H, Yang F, Zhang X, Tong P, Meng X, Wu Y, Chen H, Li X. Maternal High-Fat Diet Exacerbates Epicutaneous Sensitization and Oral Challenge-Induced Food Allergy to Ovalbumin in Offspring Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:21240-21253. [PMID: 39261017 DOI: 10.1021/acs.jafc.4c05373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Dietary factors have been associated with an increased prevalence of food allergy (FA). However, little is known about how an unhealthy diet in early life affects FA reactions in offspring. The objective of this study is to provide a scientific foundation for developing and promoting healthy dietary patterns in early life. In this study, we found that maternal high-fat diet (HFD) during pregnancy and lactation exacerbates FA (HFD-FA) in offspring mice, leading to increased serum levels of mast cell protease 1. First, we studied the systemic immunity of the HFD-FA mice and observed elevated levels of proinflammatory cytokines (IL-4, IL-6, and IL-1β) and a reduced frequency of Treg cells in splenocytes. Additionally, the HFD-FA mice showed increased gut permeability, accumulation of intestinal mast cells, and a decrease in the Treg cell frequency in the mesenteric lymph nodes. Furthermore, our findings also indicated a reduction in gut microbial diversity and abundance in HFD-FA mice. Importantly, lipid metabolism profiling revealed unique lipid profiles in the HFD-FA mice, with significant upregulation of triglycerides and downregulation of sphingolipids. Taken together, our results suggest that maternal HFD alters intestinal homeostasis and increases FA susceptibility in offspring mice.
Collapse
Affiliation(s)
- Tianliang Bai
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, Jiangxi, China
- School of Food Science and Technology, Nanchang University, Nanchang 330031, Jiangxi, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, Jiangxi, China
| | - Huming Shao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, Jiangxi, China
- School of Food Science and Technology, Nanchang University, Nanchang 330031, Jiangxi, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, Jiangxi, China
| | - Fan Yang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, Jiangxi, China
- School of Food Science and Technology, Nanchang University, Nanchang 330031, Jiangxi, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, Jiangxi, China
| | - Xing Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, Jiangxi, China
- School of Food Science and Technology, Nanchang University, Nanchang 330031, Jiangxi, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, Jiangxi, China
| | - Ping Tong
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, Jiangxi, China
- School of Food Science and Technology, Nanchang University, Nanchang 330031, Jiangxi, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, Jiangxi, China
| | - Xuanyi Meng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, Jiangxi, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, Jiangxi, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang 330000, Jiangxi, China
| | - Yong Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, Jiangxi, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, Jiangxi, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang 330000, Jiangxi, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, Jiangxi, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, Jiangxi, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang 330000, Jiangxi, China
| | - Xin Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, Jiangxi, China
- School of Food Science and Technology, Nanchang University, Nanchang 330031, Jiangxi, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, Jiangxi, China
| |
Collapse
|
14
|
Matatia PR, Christian E, Sokol CL. Sensory sentinels: Neuroimmune detection and food allergy. Immunol Rev 2024; 326:83-101. [PMID: 39092839 PMCID: PMC11436315 DOI: 10.1111/imr.13375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Food allergy is classically characterized by an inappropriate type-2 immune response to allergenic food antigens. However, how allergens are detected and how that detection leads to the initiation of allergic immunity is poorly understood. In addition to the gastrointestinal tract, the barrier epithelium of the skin may also act as a site of food allergen sensitization. These barrier epithelia are densely innervated by sensory neurons, which respond to diverse physical environmental stimuli. Recent findings suggest that sensory neurons can directly detect a broad array of immunogens, including allergens, triggering sensory responses and the release of neuropeptides that influence immune cell function. Reciprocally, immune mediators modulate the activation or responsiveness of sensory neurons, forming neuroimmune feedback loops that may impact allergic immune responses. By utilizing cutaneous allergen exposure as a model, this review explores the pivotal role of sensory neurons in allergen detection and their dynamic bidirectional communication with the immune system, which ultimately orchestrates the type-2 immune response. Furthermore, it sheds light on how peripheral signals are integrated within the central nervous system to coordinate hallmark features of allergic reactions. Drawing from this emerging evidence, we propose that atopy arises from a dysregulated neuroimmune circuit.
Collapse
Affiliation(s)
- Peri R. Matatia
- Center for Immunology & Inflammatory Diseases, Division of Rheumatology, Allergy & Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA
| | - Elena Christian
- Center for Immunology & Inflammatory Diseases, Division of Rheumatology, Allergy & Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA
| | - Caroline L. Sokol
- Center for Immunology & Inflammatory Diseases, Division of Rheumatology, Allergy & Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
15
|
Wang Y, Zhang X, Hung I, Liu C, Ren W, Ge L, Wang H. Melatonergic Signaling Sustains Food Allergy Through FcεRI Recycling. RESEARCH (WASHINGTON, D.C.) 2024; 7:0418. [PMID: 39040920 PMCID: PMC11260513 DOI: 10.34133/research.0418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 06/07/2024] [Indexed: 07/24/2024]
Abstract
The prevalence of food allergies is increasing dramatically and causing serious public health concerns. Notably, melatonin metabolism imbalance in patients with food allergies; however, the role of melatonin in food allergies remains unclear. Here, we demonstrated that melatonin suppresses food allergy responses and reprograms the gut microbiota of food-allergic mice, while melatonin aggravates food allergy during gut microbiota depletion. Mechanistically, melatonin boosts the degranulation of mast cells by up-regulating the expression of membrane high-affinity immunoglobulin E (IgE) receptor (FcεRI). Melatonin increases the mRNA expression of Rabenosyn-5 (a component of factors for endosome recycling and Rab interactions) through melatonin receptor 2 (MT2)-extracellular signal-regulated kinase (ERK) signaling, thereby driving the recycling of FcεRI and elevating the abundance of membrane FcεRI. Likewise, the inhibition of MT2 attenuates melatonin-induced food allergy in mice with gut microbiota depletion. Collectively, our finding provides insights into the pathogenesis of food allergies and provides a potential therapeutic target for the prevention and treatment of food allergies.
Collapse
Affiliation(s)
- Youxia Wang
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science,
South China Agricultural University, Guangzhou, China
| | - Xinmei Zhang
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science,
South China Agricultural University, Guangzhou, China
| | - Ifen Hung
- Anyou Biotechnology Group Co. Ltd., Taicang, China
- Joint Laboratory of Functional Nutrition and Animal Health, Centree Bio-tech (Wuhan) Co. Ltd., Wuhan, China
| | - Chunxue Liu
- Anyou Biotechnology Group Co. Ltd., Taicang, China
| | - Wenkai Ren
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science,
South China Agricultural University, Guangzhou, China
| | - Liangpeng Ge
- National Center of Technology Innovation for Pigs; Chongqing Academy of Animal Sciences; Key Laboratory of Pig Industry Science, Ministry of Agriculture, Chongqing, China
| | - Hao Wang
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science,
South China Agricultural University, Guangzhou, China
- College of Animal Science and Veterinary Medicine,
Henan Institute of Science and Technology, Xinxiang, Henan, China
| |
Collapse
|
16
|
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
|
17
|
Huang YZ, Ma JX, Bian YJ, Bai QR, Gao YH, Di SK, Lei YT, Yang H, Yang XN, Shao CY, Wang WH, Cao P, Li CZ, Zhu MX, Sun MY, Yu Y. TRPV1 analgesics disturb core body temperature via a biased allosteric mechanism involving conformations distinct from that for nociception. Neuron 2024; 112:1815-1831.e4. [PMID: 38492574 DOI: 10.1016/j.neuron.2024.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/30/2024] [Accepted: 02/21/2024] [Indexed: 03/18/2024]
Abstract
Efforts on developing transient receptor potential vanilloid 1 (TRPV1) drugs for pain management have been hampered by deleterious hypo- or hyperthermia caused by TRPV1 agonists/antagonists. Here, we compared the effects of four antagonists on TRPV1 polymodal gating and core body temperature (CBT) in Trpv1+/+, Trpv1-/-, and Trpv1T634A/T634A. Neither the effect on proton gating nor drug administration route, hair coverage, CBT rhythmic fluctuations, or inflammation had any influence on the differential actions of TRPV1 drugs on CBT. We identified the S4-S5 linker region exposed to the vanilloid pocket of TRPV1 to be critical for hyperthermia associated with certain TRPV1 antagonists. PSFL2874, a TRPV1 antagonist we discovered, is effective against inflammatory pain but devoid of binding to the S4-S5 linker and inducing CBT changes. These findings implicate that biased allosteric mechanisms exist for TRPV1 coupling to nociception and CBT regulation, opening avenues for the development of non-opioid analgesics without affecting CBT.
Collapse
Affiliation(s)
- Yi-Zhe Huang
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jing-Xian Ma
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yu-Jing Bian
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Qin-Ru Bai
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yu-Hao Gao
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Shu-Ke Di
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yun-Tao Lei
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Hui Yang
- Tea Research Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, Hunan, China
| | - Xiao-Na Yang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Chang-Yan Shao
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Wen-Hui Wang
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Peng Cao
- Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chang-Zhu Li
- State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Changsha 410004, Hunan, China
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Meng-Yang Sun
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Ye Yu
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
18
|
Du F, Rische CH, Li Y, Vincent MP, Krier-Burris RA, Qian Y, Yuk SA, Almunif S, Bochner BS, Qiao B, Scott EA. Controlled adsorption of multiple bioactive proteins enables targeted mast cell nanotherapy. NATURE NANOTECHNOLOGY 2024; 19:698-704. [PMID: 38228804 PMCID: PMC11105988 DOI: 10.1038/s41565-023-01584-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 11/24/2023] [Indexed: 01/18/2024]
Abstract
Protein adsorption onto nanomaterials often results in denaturation and loss of bioactivity. Controlling the adsorption process to maintain the protein structure and function has potential for a range of applications. Here we report that self-assembled poly(propylene sulfone) (PPSU) nanoparticles support the controlled formation of multicomponent enzyme and antibody coatings and maintain their bioactivity. Simulations indicate that hydrophobic patches on protein surfaces induce a site-specific dipole relaxation of PPSU assemblies to non-covalently anchor the proteins without disrupting the protein hydrogen bonding or structure. As a proof of concept, a nanotherapy employing multiple mast-cell-targeted antibodies for preventing anaphylaxis is demonstrated in a humanized mouse model. PPSU nanoparticles displaying an optimized ratio of co-adsorbed anti-Siglec-6 and anti-FcεRIα antibodies effectively inhibit mast cell activation and degranulation, preventing anaphylaxis. Protein immobilization on PPSU surfaces provides a simple and rapid platform for the development of targeted protein nanomedicines.
Collapse
Affiliation(s)
- Fanfan Du
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Clayton H Rische
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Department of Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yang Li
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, USA
| | - Michael P Vincent
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Rebecca A Krier-Burris
- Department of Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yuan Qian
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Simseok A Yuk
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Sultan Almunif
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Bruce S Bochner
- Department of Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Baofu Qiao
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- Department of Natural Sciences, Baruch College, City University of New York, New York, NY, USA
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA.
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA.
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
19
|
Culhuac EB, Bello M. Evaluation of Urtica dioica Phytochemicals against Therapeutic Targets of Allergic Rhinitis Using Computational Studies. Molecules 2024; 29:1765. [PMID: 38675586 PMCID: PMC11052477 DOI: 10.3390/molecules29081765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/06/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Allergic rhinitis (AR) is a prevalent inflammatory condition affecting millions globally, with current treatments often associated with significant side effects. To seek safer and more effective alternatives, natural sources like Urtica dioica (UD) are being explored. However, UD's mechanism of action remains unknown. Therefore, to elucidate it, we conducted an in silico evaluation of UD phytochemicals' effects on known therapeutic targets of allergic rhinitis: histamine receptor 1 (HR1), neurokinin 1 receptor (NK1R), cysteinyl leukotriene receptor 1 (CLR1), chemoattractant receptor-homologous molecule expressed on type 2 helper T cells (CRTH2), and bradykinin receptor type 2 (BK2R). The docking analysis identified amentoflavone, alpha-tocotrienol, neoxanthin, and isorhamnetin 3-O-rutinoside as possessing a high affinity for all the receptors. Subsequently, molecular dynamics (MD) simulations were used to analyze the key interactions; the free energy of binding was calculated through Generalized Born and Surface Area Solvation (MMGBSA), and the conformational changes were evaluated. Alpha-tocotrienol exhibited a high affinity while also inducing positive conformational changes across all targets. Amentoflavone primarily affected CRTH2, neoxanthin targeted NK1R, CRTH2, and BK2R, and isorhamnetin-3-O-rutinoside acted on NK1R. These findings suggest UD's potential to treat AR symptoms by inhibiting these targets. Notably, alpha-tocotrienol emerges as a promising multi-target inhibitor. Further in vivo and in vitro studies are needed for validation.
Collapse
Affiliation(s)
- Erick Bahena Culhuac
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico;
- Facultad de Ciencias, Universidad Autónoma del Estado de México, Toluca 50000, Mexico
| | - Martiniano Bello
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico;
| |
Collapse
|
20
|
Granton E, Brown L, Defaye M, Moazen P, Almblad H, Randall TE, Rich JD, Geppert A, Abdullah NS, Hassanabad MF, Hiroki CH, Farias R, Nguyen AP, Schubert C, Lou Y, Andonegui G, Iftinca M, Raju D, Vargas MA, Howell PL, Füzesi T, Bains J, Kurrasch D, Harrison JJ, Altier C, Yipp BG. Biofilm exopolysaccharides alter sensory-neuron-mediated sickness during lung infection. Cell 2024; 187:1874-1888.e14. [PMID: 38518773 DOI: 10.1016/j.cell.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 01/04/2024] [Accepted: 03/01/2024] [Indexed: 03/24/2024]
Abstract
Infections of the lung cause observable sickness thought to be secondary to inflammation. Signs of sickness are crucial to alert others via behavioral-immune responses to limit contact with contagious individuals. Gram-negative bacteria produce exopolysaccharide (EPS) that provides microbial protection; however, the impact of EPS on sickness remains uncertain. Using genome-engineered Pseudomonas aeruginosa (P. aeruginosa) strains, we compared EPS-producers versus non-producers and a virulent Escherichia coli (E. coli) lung infection model in male and female mice. EPS-negative P. aeruginosa and virulent E. coli infection caused severe sickness, behavioral alterations, inflammation, and hypothermia mediated by TLR4 detection of the exposed lipopolysaccharide (LPS) in lung TRPV1+ sensory neurons. However, inflammation did not account for sickness. Stimulation of lung nociceptors induced acute stress responses in the paraventricular hypothalamic nuclei by activating corticotropin-releasing hormone neurons responsible for sickness behavior and hypothermia. Thus, EPS-producing biofilm pathogens evade initiating a lung-brain sensory neuronal response that results in sickness.
Collapse
Affiliation(s)
- Elise Granton
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Luke Brown
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Manon Defaye
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Inflammation Research Network, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Parisa Moazen
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Henrik Almblad
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Trevor E Randall
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Jacquelyn D Rich
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Andrew Geppert
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Nasser S Abdullah
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Inflammation Research Network, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Mortaza F Hassanabad
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Carlos H Hiroki
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Raquel Farias
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Angela P Nguyen
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Courtney Schubert
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Yuefei Lou
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Graciela Andonegui
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mircea Iftinca
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Inflammation Research Network, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Deepa Raju
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Mario A Vargas
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - P Lynne Howell
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Tamás Füzesi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Cumming School of Medicine Optogenetics Core Facility, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jaideep Bains
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Krembil Research Institute, University Health Network, Toronto, ON, Canada.
| | - Deborah Kurrasch
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Joe Jonathan Harrison
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Biological Sciences, University of Calgary, Calgary, AB, Canada.
| | - Christophe Altier
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Inflammation Research Network, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.
| | - Bryan G Yipp
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
21
|
Zhao Y, Wang H, Wu Z, Zhu Y, Wang J. Case Report: A Rare Case of Iodixanol-Induced Anaphylactic Shock in Cerebral Angiography. J Asthma Allergy 2024; 17:361-367. [PMID: 38623449 PMCID: PMC11018123 DOI: 10.2147/jaa.s460263] [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: 01/18/2024] [Accepted: 04/01/2024] [Indexed: 04/17/2024] Open
Abstract
Background Adverse reactions induced by isoosmolar contrast medium (iodixanol) are mostly mild, with rashes and headaches being the most common. Although anaphylactic shock has been reported, no related incidents have been documented on cerebral angiography. Objective This article reports a serious case of anaphylactic shock possibly induced by iodixanol and provides an overview of the case report. Case Summary A 65-year-old female with persistent headaches for nearly six months and CTA examination revealed multiple intracranial aneurysms. After two treatments, she returned to the hospital for aneurysm of reexamination a month ago. Following a preoperative assessment, cerebral angiography was performed. Three minutes after the procedure, the patient experienced dizziness, increased heart rate, followed by hypotension (BP 90/43 mm Hg), a sudden drop-in heart rate (HR 68 bpm), and a drop in SpO2 to 92%. Intravenous dexamethasone for anti-allergic were administered immediately, along with therapy through oxygen-inhalation. However, the patient then developed limb convulsions, unresponsiveness, and was urgently given diazepam for sedation and sputum aspiration to maintain airway patency. Blood pressure decrease to 53/29 mm Hg, and SpO2 readings were unavailable. Intravenous dopamine to elevates blood pressure, and assists breathing by intubating in the endotracheal. After 3 minutes, as the blood pressure remained undetectable, intermittent intravenous epinephrine 1mg was administered to raise the blood pressure, gradually restoring it to 126/90 mm Hg, and SpO2 increased to 95%. The patient was diagnosed with iodixanol-induced anaphylactic shock and urgently transferred to the NICU for monitoring and treatment. The patient died despite immediate treatment. Conclusion A 65-year-old female developed serious anaphylactic shock during cerebral angiography after receiving iodixanol. Although iodixanol is considered one of the safest iodinated contrast mediums (ICM), clinicians should be aware of its the potential for serious hypersensitivity reactions that can lead to fatal and life-threatening events.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, People’s Republic of China
| | - Hua Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Zhengjun Wu
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, People’s Republic of China
| | - Yunxiang Zhu
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, People’s Republic of China
| | - Jingsong Wang
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, People’s Republic of China
| |
Collapse
|
22
|
Theoharides TC, Twahir A, Kempuraj D. Mast cells in the autonomic nervous system and potential role in disorders with dysautonomia and neuroinflammation. Ann Allergy Asthma Immunol 2024; 132:440-454. [PMID: 37951572 DOI: 10.1016/j.anai.2023.10.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/16/2023] [Accepted: 10/06/2023] [Indexed: 11/14/2023]
Abstract
Mast cells (MC) are ubiquitous in the body, and they are critical for not only in allergic diseases but also in immunity and inflammation, including having potential involvement in the pathophysiology of dysautonomias and neuroinflammatory disorders. MC are located perivascularly close to nerve endings and sites such as the carotid bodies, heart, hypothalamus, the pineal gland, and the adrenal gland that would allow them not only to regulate but also to be affected by the autonomic nervous system (ANS). MC are stimulated not only by allergens but also many other triggers including some from the ANS that can affect MC release of neurosensitizing, proinflammatory, and vasoactive mediators. Hence, MC may be able to regulate homeostatic functions that seem to be dysfunctional in many conditions, such as postural orthostatic tachycardia syndrome, autism spectrum disorder, myalgic encephalomyelitis/chronic fatigue syndrome, and Long-COVID syndrome. The evidence indicates that there is a possible association between these conditions and diseases associated with MC activation. There is no effective treatment for any form of these conditions other than minimizing symptoms. Given the many ways MC could be activated and the numerous mediators released, it would be important to develop ways to inhibit stimulation of MC and the release of ANS-relevant mediators.
Collapse
Affiliation(s)
- Theoharis C Theoharides
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, Florida; Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts.
| | - Assma Twahir
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, Florida
| | - Duraisamy Kempuraj
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, Florida
| |
Collapse
|
23
|
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
|
24
|
Smith J, Tan JKH, Short C, O'Neill H, Moro C. The effect of myeloablative radiation on urinary bladder mast cells. Sci Rep 2024; 14:6219. [PMID: 38485999 PMCID: PMC10940702 DOI: 10.1038/s41598-024-56655-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 03/08/2024] [Indexed: 03/18/2024] Open
Abstract
Radiation-induced cystitis is an inflammatory condition affecting the urinary bladder, which can develop as a side effect of abdominopelvic radiotherapy, specifically external-beam radiation therapy or myeloablative radiotherapy. A possible involvement of mast cells in the pathophysiology of radiation-induced cystitis has been indicated in cases of external-beam radiation therapy; however, there is no evidence that these findings apply to the myeloablative aetiology. As such, this study investigated potential changes to urinary bladder mast cell prevalence when exposed to myeloablative radiation. Lethally irradiated C57BL/6J mice that received donor rescue bone marrow cells exhibited an increased mast cell frequency amongst host leukocytes 1 week following irradiation. By 4 weeks, no significant difference in either frequency or cell density was observed. However mast cell diameter was smaller, and a significant increase in mast cell number in the adventitia was observed. This study highlights that mast cells constitute a significant portion of the remaining host leukocyte population following radiation exposure, with changes to mast cell distribution and decreased cell diameter four weeks following radiation-induced injury.
Collapse
Affiliation(s)
- Jessica Smith
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, 4226, Australia
| | - Jonathan Kah Huat Tan
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QLD, 4226, Australia
| | - Christie Short
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QLD, 4226, Australia
| | - Helen O'Neill
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QLD, 4226, Australia
| | - Christian Moro
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, 4226, Australia.
| |
Collapse
|
25
|
Deng L, Costa F, Blake KJ, Choi S, Chandrabalan A, Yousuf MS, Shiers S, Dubreuil D, Vega-Mendoza D, Rolland C, Deraison C, Voisin T, Bagood MD, Wesemann L, Frey AM, Palumbo JS, Wainger BJ, Gallo RL, Leyva-Castillo JM, Vergnolle N, Price TJ, Ramachandran R, Horswill AR, Chiu IM. S. aureus drives itch and scratch-induced skin damage through a V8 protease-PAR1 axis. Cell 2023; 186:5375-5393.e25. [PMID: 37995657 PMCID: PMC10669764 DOI: 10.1016/j.cell.2023.10.019] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 08/20/2023] [Accepted: 10/18/2023] [Indexed: 11/25/2023]
Abstract
Itch is an unpleasant sensation that evokes a desire to scratch. The skin barrier is constantly exposed to microbes and their products. However, the role of microbes in itch generation is unknown. Here, we show that Staphylococcus aureus, a bacterial pathogen associated with itchy skin diseases, directly activates pruriceptor sensory neurons to drive itch. Epicutaneous S. aureus exposure causes robust itch and scratch-induced damage. By testing multiple isogenic bacterial mutants for virulence factors, we identify the S. aureus serine protease V8 as a critical mediator in evoking spontaneous itch and alloknesis. V8 cleaves proteinase-activated receptor 1 (PAR1) on mouse and human sensory neurons. Targeting PAR1 through genetic deficiency, small interfering RNA (siRNA) knockdown, or pharmacological blockade decreases itch and skin damage caused by V8 and S. aureus exposure. Thus, we identify a mechanism of action for a pruritogenic bacterial factor and demonstrate the potential of inhibiting V8-PAR1 signaling to treat itch.
Collapse
Affiliation(s)
- Liwen Deng
- Department of Immunology, Harvard Medical School, Boston, MA 02215, USA
| | - Flavia Costa
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kimbria J Blake
- Department of Immunology, Harvard Medical School, Boston, MA 02215, USA
| | - Samantha Choi
- Department of Immunology, Harvard Medical School, Boston, MA 02215, USA
| | - Arundhasa Chandrabalan
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Muhammad Saad Yousuf
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Daniel Dubreuil
- Departments of Neurology and Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Daniela Vega-Mendoza
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Corinne Rolland
- IRSD, Université de Toulouse, INSERM, INRAe, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Celine Deraison
- IRSD, Université de Toulouse, INSERM, INRAe, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Tiphaine Voisin
- Department of Immunology, Harvard Medical School, Boston, MA 02215, USA
| | - Michelle D Bagood
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lucia Wesemann
- Department of Immunology, Harvard Medical School, Boston, MA 02215, USA
| | - Abigail M Frey
- Department of Immunology, Harvard Medical School, Boston, MA 02215, USA
| | - Joseph S Palumbo
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Brian J Wainger
- Departments of Neurology and Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | | | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRAe, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
26
|
Yang D, Almanzar N, Chiu IM. The role of cellular and molecular neuroimmune crosstalk in gut immunity. Cell Mol Immunol 2023; 20:1259-1269. [PMID: 37336989 PMCID: PMC10616093 DOI: 10.1038/s41423-023-01054-5] [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: 03/31/2023] [Accepted: 06/01/2023] [Indexed: 06/21/2023] Open
Abstract
The gastrointestinal tract is densely innervated by the peripheral nervous system and populated by the immune system. These two systems critically coordinate the sensations of and adaptations to dietary, microbial, and damaging stimuli from the external and internal microenvironment during tissue homeostasis and inflammation. The brain receives and integrates ascending sensory signals from the gut and transduces descending signals back to the gut via autonomic neurons. Neurons regulate intestinal immune responses through the action of local axon reflexes or through neuronal circuits via the gut-brain axis. This neuroimmune crosstalk is critical for gut homeostatic maintenance and disease resolution. In this review, we discuss the roles of distinct types of gut-innervating neurons in the modulation of intestinal mucosal immunity. We will focus on the molecular mechanisms governing how different immune cells respond to neural signals in host defense and inflammation. We also discuss the therapeutic potential of strategies targeting neuroimmune crosstalk for intestinal diseases.
Collapse
Affiliation(s)
- Daping Yang
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA
| | - Nicole Almanzar
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
27
|
Tauber M, Basso L, Martin J, Bostan L, Pinto MM, Thierry GR, Houmadi R, Serhan N, Loste A, Blériot C, Kamphuis JB, Grujic M, Kjellén L, Pejler G, Paul C, Dong X, Galli SJ, Reber LL, Ginhoux F, Bajenoff M, Gentek R, Gaudenzio N. Landscape of mast cell populations across organs in mice and humans. J Exp Med 2023; 220:e20230570. [PMID: 37462672 PMCID: PMC10354537 DOI: 10.1084/jem.20230570] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/21/2023] Open
Abstract
Mast cells (MCs) are tissue-resident immune cells that exhibit homeostatic and neuron-associated functions. Here, we combined whole-tissue imaging and single-cell RNA sequencing datasets to generate a pan-organ analysis of MCs in mice and humans at steady state. In mice, we identify two mutually exclusive MC populations, MrgprB2+ connective tissue-type MCs and MrgprB2neg mucosal-type MCs, with specific transcriptomic core signatures. While MrgprB2+ MCs develop in utero independently of the bone marrow, MrgprB2neg MCs develop after birth and are renewed by bone marrow progenitors. In humans, we unbiasedly identify seven MC subsets (MC1-7) distributed across 12 organs with different transcriptomic core signatures. MC1 are preferentially enriched in the bladder, MC2 in the lungs, and MC4, MC6, and MC7 in the skin. Conversely, MC3 and MC5 are shared by most organs but not skin. This comprehensive analysis offers valuable insights into the natural diversity of MC subtypes in both mice and humans.
Collapse
Affiliation(s)
- Marie Tauber
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Lilian Basso
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Jeremy Martin
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Luciana Bostan
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Marlene Magalhaes Pinto
- Centre for Inflammation Research and Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Guilhem R. Thierry
- Aix Marseille University, CNRS, INSERM, Centre d'immunologie de Marseille-Luminy, Marseille, France
| | - Raïssa Houmadi
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Nadine Serhan
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Alexia Loste
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Camille Blériot
- Institut Necker des Enfants Malades, CNRS UMR8253, Paris, France
| | - Jasper B.J. Kamphuis
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Mirjana Grujic
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Lena Kjellén
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Carle Paul
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
- Toulouse University and Centre Hospitalier Universitaire, Toulouse, France
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, School of Medicine, Center for Sensory Biology, Johns Hopkins University, Baltimore, MD, USA
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephen J. Galli
- Departments of Pathology and Microbiology and Immunology, Stanford University, Stanford, CA, USA
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA
| | - Laurent L. Reber
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Florent Ginhoux
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM U1015, Gustave Roussy, Villejuif, France
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Marc Bajenoff
- Aix Marseille University, CNRS, INSERM, Centre d'immunologie de Marseille-Luminy, Marseille, France
| | - Rebecca Gentek
- Centre for Inflammation Research and Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Nicolas Gaudenzio
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
- Genoskin SAS, Toulouse, France
| |
Collapse
|
28
|
Chen O, Luo X, Ji RR. Macrophages and microglia in inflammation and neuroinflammation underlying different pain states. MEDICAL REVIEW (2021) 2023; 3:381-407. [PMID: 38283253 PMCID: PMC10811354 DOI: 10.1515/mr-2023-0034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/26/2023] [Indexed: 01/30/2024]
Abstract
Pain is a main symptom in inflammation, and inflammation induces pain via inflammatory mediators acting on nociceptive neurons. Macrophages and microglia are distinct cell types, representing immune cells and glial cells, respectively, but they share similar roles in pain regulation. Macrophages are key regulators of inflammation and pain. Macrophage polarization plays different roles in inducing and resolving pain. Notably, macrophage polarization and phagocytosis can be induced by specialized pro-resolution mediators (SPMs). SPMs also potently inhibit inflammatory and neuropathic pain via immunomodulation and neuromodulation. In this review, we discuss macrophage signaling involved in pain induction and resolution, as well as in maintaining physiological pain. Microglia are macrophage-like cells in the central nervous system (CNS) and drive neuroinflammation and pathological pain in various inflammatory and neurological disorders. Microglia-produced inflammatory cytokines can potently regulate excitatory and inhibitory synaptic transmission as neuromodulators. We also highlight sex differences in macrophage and microglial signaling in inflammatory and neuropathic pain. Thus, targeting macrophage and microglial signaling in distinct locations via pharmacological approaches, including immunotherapies, and non-pharmacological approaches will help to control chronic inflammation and chronic pain.
Collapse
Affiliation(s)
- Ouyang Chen
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University Medical Center, Durham, NC, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Xin Luo
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University Medical Center, Durham, NC, USA
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ru-Rong Ji
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University Medical Center, Durham, NC, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
29
|
Bordon Y. Mast cells chill in anaphylaxis. Nat Rev Immunol 2023; 23:270. [PMID: 37024577 DOI: 10.1038/s41577-023-00875-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|