1
|
Srisomboon Y, Tojima I, Iijima K, Kita H, O'Grady SM. Allergen-induced activation of epithelial P2Y 2 receptors promotes adenosine triphosphate exocytosis and type 2 immunity in airways. J Allergy Clin Immunol 2025; 155:1607-1622. [PMID: 39863058 DOI: 10.1016/j.jaci.2025.01.019] [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: 08/23/2024] [Revised: 12/29/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Environmental allergens induce the release of danger signals from the airway epithelium that trigger type 2 immune responses and promote airway inflammation. OBJECTIVE We investigated the role of allergen-stimulated P2Y2 receptor activation in regulating adenosine triphosphate (ATP), IL-33, and DNA release by human bronchial epithelial (hBE) cells and mouse airways. METHODS The hBE cells were exposed to Alternaria alternata extract and secretion of ATP, IL-33, and DNA were studied in vitro. Molecular and cellular mechanisms were examined by biochemical and genetic approaches. Mice were treated intranasally with pharmacologic agents and exposed to Alternaria extract. RESULTS Exposure of hBE cells to Alternaria extract stimulated P2Y2 receptors coupled to phospholipase C β3, leading to activation of multiple protein kinase C (PKC) isoforms and an increase in intracellular Ca2+ concentration. Small interfering RNAs targeting PKC δ or inhibiting PKC δ activity with delcasertib blocked exocytosis of ATP and reduced IL-33 and DNA secretion. Moreover, a peptide antagonist for myristoylated alanine-rich C-kinase substrate (MARCKS) reduced vesicular ATP release. A proximity ligand assay showed that Alternaria extract stimulated MARCKS desorption from the plasma membrane and delcasertib prevented the response. Finally, the P2Y2 receptor antagonist AR-C118925XX and delcasertib blocked IL-33, DNA, and type 2 cytokine secretion in vivo in mice exposed to Alternaria. CONCLUSION P2Y2 receptor stimulation after allergen exposure promoted activation of PLC β3, PKC δ, and MARCKS protein desorption from the apical membrane, which facilitated ATP exocytosis and subsequent secretion of IL-33 and DNA. Epithelial P2Y2 receptors serve as primary sensors for aeroallergen-induced alarmin release by airway epithelial cells.
Collapse
Affiliation(s)
- Yotesawee Srisomboon
- Departments of Animal Science, Integrative Biology, and Physiology, University of Minnesota, St Paul, Minn
| | - Ichiro Tojima
- Division of Allergy, Asthma and Clinical Immunology, Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Koji Iijima
- Division of Allergy, Asthma and Clinical Immunology, Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Hirohito Kita
- Division of Allergy, Asthma and Clinical Immunology, Department of Medicine, Mayo Clinic, Scottsdale, Ariz.
| | - Scott M O'Grady
- Departments of Animal Science, Integrative Biology, and Physiology, University of Minnesota, St Paul, Minn.
| |
Collapse
|
2
|
Metwally H. STAT Signature Dish: Serving Immunity with a Side of Dietary Control. Biomolecules 2025; 15:487. [PMID: 40305224 PMCID: PMC12024614 DOI: 10.3390/biom15040487] [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: 02/17/2025] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
Immunity is a fundamental aspect of animal biology, defined as the host's ability to detect and defend against harmful pathogens and toxic substances to preserve homeostasis. However, immune defenses are metabolically demanding, requiring the efficient allocation of limited resources to balance immune function with other physiological and developmental needs. To achieve this balance, organisms have evolved sophisticated signaling networks that enable precise, context-specific responses to internal and external cues. These networks are essential for survival and adaptation in multicellular systems. Central to this regulatory architecture is the STAT (signal transducer and activator of Transcription) family, a group of versatile signaling molecules that govern a wide array of biological processes across eukaryotes. STAT signaling demonstrates remarkable plasticity, from orchestrating host defense mechanisms to regulating dietary metabolism. Despite its critical role, the cell-specific and context-dependent nuances of STAT signaling remain incompletely understood, highlighting a significant gap in our understanding. This review delves into emerging perspectives on immunity, presenting dynamic frameworks to explore the complexity and adaptability of STAT signaling and the underlying logic driving cellular decision-making. It emphasizes how STAT pathways integrate diverse physiological processes, from immune responses to dietary regulation, ultimately supporting organismal balance and homeostasis.
Collapse
Affiliation(s)
- Hozaifa Metwally
- Laboratory of Immune Regulation, The World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
3
|
Edwards DL, Huang M, Wang TT. Soluble Factors and Mechanisms Regulated by Sialylated IgG Signaling. Immunol Rev 2025; 330:e70021. [PMID: 40084926 PMCID: PMC12042769 DOI: 10.1111/imr.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Inflammation is a complex biological response that can be both induced and actively suppressed by IgG-Fc gamma receptor (FcγR) interactions. This review explores the role of IgG sialylation in reducing or blocking inflammatory responses. We first revisit foundational studies that established the anti-inflammatory properties of sialylated IgG1 Fc. These early investigations revealed that the sialylated fraction is crucial for intravenous immunoglobulin's (IVIg's) ability to reduce inflammation in many autoinflammatory diseases and defined a paracrine signaling mechanism underlying this activity. Next, we discuss a recently identified mechanism whereby sialylated IgG directly induces RE1-Silencing Transcription Factor (REST) which functions as a transcriptional repressor of NF-κB1. This mechanism suggests a very broad role for sialylated IgG signaling in inflammation control since NF-κB is a central mediator of responses downstream of diverse activating receptors on both adaptive and innate immune cells. Finally, we review a set of soluble factors that are suppressed by sialylated IgG signaling in the murine airway and in purified human macrophages, providing additional insight into mechanisms by which sialylated IgG contributes to broad inflammatory control.
Collapse
Affiliation(s)
- Desmond L. Edwards
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305
| | - Min Huang
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305
| | - Taia T. Wang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305
- Department of Medicine, Division of Infectious Diseases, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
4
|
Olsthoorn SEM, van Krimpen A, Hendriks RW, Stadhouders R. Chronic Inflammation in Asthma: Looking Beyond the Th2 Cell. Immunol Rev 2025; 330:e70010. [PMID: 40016948 PMCID: PMC11868696 DOI: 10.1111/imr.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 02/11/2025] [Indexed: 03/01/2025]
Abstract
Asthma is a common chronic inflammatory disease of the airways. A substantial number of patients present with severe and therapy-resistant asthma, for which the underlying biological mechanisms remain poorly understood. In most asthma patients, airway inflammation is characterized by chronic activation of type 2 immunity. CD4+ T helper 2 (Th2) cells are the canonical producers of the cytokines that fuel type 2 inflammation: interleukin (IL)-4, IL-5, IL-9, and IL-13. However, more recent findings have shown that other lymphocyte subsets, in particular group 2 innate lymphoid cells (ILC2s) and type 2 CD8+ cytotoxic T (Tc2) cells, can also produce large amounts of type 2 cytokines. Importantly, a substantial number of severe therapy-resistant asthma patients present with chronic type 2 inflammation, despite the high sensitivity of Th2 cells for suppression by corticosteroids-the mainstay drugs for asthma. Emerging evidence indicates that ILC2s and Tc2 cells are more abundant in severe asthma patients and can adopt corticosteroid-resistance states. Moreover, many severe asthma patients do not present with overt type 2 airway inflammation, implicating non-type 2 immunity as a driver of disease. In this review, we will discuss asthma pathophysiology and focus on the roles played by ILC2s, Tc2 cells, and non-type 2 lymphocytes, placing special emphasis on severe disease forms.
Collapse
Affiliation(s)
- Simone E. M. Olsthoorn
- Department of Pulmonary MedicineErasmus MC University Medical CenterRotterdamthe Netherlands
| | - Anneloes van Krimpen
- Department of Pulmonary MedicineErasmus MC University Medical CenterRotterdamthe Netherlands
| | - Rudi W. Hendriks
- Department of Pulmonary MedicineErasmus MC University Medical CenterRotterdamthe Netherlands
| | - Ralph Stadhouders
- Department of Pulmonary MedicineErasmus MC University Medical CenterRotterdamthe Netherlands
| |
Collapse
|
5
|
Mannion JM, Rahimi RA. Tissue-Resident Th2 Cells in Type 2 Immunity and Allergic Diseases. Immunol Rev 2025; 330:e70006. [PMID: 39981858 PMCID: PMC11897987 DOI: 10.1111/imr.70006] [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: 08/23/2024] [Accepted: 02/06/2025] [Indexed: 02/22/2025]
Abstract
Type 2 immunity represents a unique immune module that provides host protection against macro-parasites and noxious agents such as venoms and toxins. In contrast, maladaptive type 2 immune responses cause allergic diseases. While multiple cell types play important roles in type 2 immunity, recent studies in humans and murine models of chronic allergic diseases have shown that a distinct population of tissue-resident, CD4+ T helper type 2 (Th2) cells play a critical role in chronic allergic inflammation. The rules regulating Th2 cell differentiation have remained less well defined than other T cell subsets, but recent studies have shed new light into the specific mechanisms controlling Th2 cell biology in vivo. Here, we review our current understanding of the checkpoints regulating the development and function of tissue-resident Th2 cells with a focus on chronic allergic diseases. We discuss evidence for a barrier tissue checkpoint in initial Th2 cell priming, including the role of neuropeptides, damage-associated molecular patterns, and dendritic cell macro-clusters. Furthermore, we review the evidence for a second barrier tissue checkpoint that instructs the development of multi-cytokine producing, tissue-resident Th2 cells that orchestrate allergic inflammation. Lastly, we discuss potential approaches to therapeutically target tissue-resident Th2 cells in chronic allergic diseases.
Collapse
Affiliation(s)
- Jenny M Mannion
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rod A Rahimi
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Tajima A, Yamazaki F, Kishimoto I, Ma N, Kume N, Walls AF, Kambe N, Tanizaki H. Basophil-Derived IL-4 Production and Its Potential Pro-Tumoural Role in Th2-Polarisation Within Sentinel Lymph Nodes of Primary Cutaneous Melanoma. Exp Dermatol 2025; 34:e70028. [PMID: 39865442 DOI: 10.1111/exd.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 12/01/2024] [Accepted: 12/21/2024] [Indexed: 01/28/2025]
Abstract
Chronic inflammation in the tumour microenvironment (TME) via Th2-polarisation promotes melanoma progression and metastasis, making it a target for immunotherapy. Interleukin (IL)-4 is considered essential for Th2-polarisation in the TME; however, its source remains unknown. Basophils have been postulated as one of its sources. Basophil-derived IL-4 contributes to Th2-polarisation in parasitic infections and allergic diseases and has been implicated in tumour immunity. To identify basophil infiltration into the TME of human melanoma skin lesions and sentinel lymph nodes (SLNs) and demonstrate that basophils produce IL-4. Immunohistochemistry (IHC) with a basophil-specific BB1 antibody and in situ hybridisation. Basophils tended to infiltrate skin lesions at Stage II or later. Higher numbers of infiltrating basophils correlated with the Breslow depth and a shorter progression-free survival, indicating an association with poor prognosis. In SLNs, basophils infiltrated at early stages without metastasis (Stages I and II), with the number of infiltrating basophils being significantly higher in Stage II than in Stage I. IHC revealed that IL-4 levels were also significantly elevated in Stage II SLNs compared to Stage I SLNs. Furthermore, a positive correlation was observed between the number of basophils infiltrating SLNs and IL-4 expression. In situ hybridisation confirmed that basophils expressed IL4. These findings are consistent with previous reports of early-stage melanoma SLNs having a Th2-environment and suggest that basophil-derived IL-4 may contribute to a metastasis-promoting environment in SLNs through Th2-polarisation. Basophils may represent potential immunotherapeutic targets for pro-tumour changes that occur in SLNs in early-stage melanoma.
Collapse
Affiliation(s)
- Aki Tajima
- Department of Dermatology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Fumikazu Yamazaki
- Department of Dermatology, Kansai Medical University, Hirakata, Osaka, Japan
- Department of Dermatology, Tokai University, Isehara, Kanagawa, Japan
| | - Izumi Kishimoto
- Department of Dermatology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Ni Ma
- Department of Dermatology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Noriko Kume
- Department of Dermatology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Andrew F Walls
- Immunopharmacology Group, Clinical and Experimental Sciences, University of Southampton, Southampton, UK
| | - Naotomo Kambe
- Department of Dermatology, Kansai Medical University, Hirakata, Osaka, Japan
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hideaki Tanizaki
- Department of Dermatology, Kansai Medical University, Hirakata, Osaka, Japan
| |
Collapse
|
7
|
Santacroce G, Rossi CM, Lenti MV, Ghosh S, Iacucci M, Di Sabatino A. Understanding tissue injury and remodelling in eosinophilic oesophagitis: development towards personalised medicine. Gut 2024:gutjnl-2024-333994. [PMID: 39658262 DOI: 10.1136/gutjnl-2024-333994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 11/15/2024] [Indexed: 12/12/2024]
Abstract
Eosinophilic oesophagitis (EoE) is a chronic, immune-mediated condition characterised by eosinophilic infiltration of the oesophagus, leading to significant morbidity due to oesophageal dysfunction. The pathogenic course of EoE begins with tissue injury, marked by the intricate interplay of oesophageal barrier dysfunction and T helper 2-mediated inflammation. In response to tissue damage, a subsequent phase of tissue remodelling features a complex interaction between epithelial cells and stromal cells, aimed at tissue repair. The persistence of inflammation drives these mechanisms towards oesophageal fibrostenosis, mainly through the transforming growth factor-dependent, myofibroblast-driven accumulation of the extracellular matrix. Currently, treatment options for EoE are limited, with dietary intervention, proton pump inhibitors and oral steroids serving as first-line therapies. Dupilumab, an antiinterleukin (IL) 4/IL-13 agent, is the only biologic that has been approved by European and American regulatory authorities. However, emerging OMIC technologies significantly advance our understanding of EoE pathogenesis, revealing novel cellular and molecular mechanisms driving the disease. This progress has accelerated the identification of new therapeutic targets and agents, some already under clinical investigation, potentially expanding our therapeutic arsenal and paving the way for more personalised approaches. In this evolving landscape, artificial intelligence (AI) has shown great potential to further elaborate on the complexities of EoE heterogeneity, offering standardised tools for diagnosis, disease phenotyping, and prediction of treatment response. Though still in their early stages, integrating OMICs and AI marks a pivotal step towards precision medicine in EoE.
Collapse
Affiliation(s)
- Giovanni Santacroce
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Lombardia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Lombardia, Italy
| | - Carlo Maria Rossi
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Lombardia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Lombardia, Italy
| | - Marco Vincenzo Lenti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Lombardia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Lombardia, Italy
| | - Subrata Ghosh
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Marietta Iacucci
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Antonio Di Sabatino
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Lombardia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Lombardia, Italy
| |
Collapse
|
8
|
Leyva-Castillo JM, Das M, Strakosha M, McGurk A, Artru E, Kam C, Alasharee M, Wesemann DR, Tomura M, Karasuyama H, Brombacher F, Geha RS. IL-4 acts on skin-derived dendritic cells to promote the T H2 response to cutaneous sensitization and the development of allergic skin inflammation. J Allergy Clin Immunol 2024; 154:1462-1471.e3. [PMID: 38996877 PMCID: PMC11625010 DOI: 10.1016/j.jaci.2024.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 06/06/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024]
Abstract
BACKGROUND Atopic dermatitis is characterized by scratching and a TH2-dominated local and systemic response to cutaneously encountered antigens. Dendritic cells (DCs) capture antigens in the skin and rapidly migrate to draining lymph nodes (dLNs) where they drive the differentiation of antigen-specific naive T cells. OBJECTIVE We sought to determine whether non-T-cell-derived IL-4 acts on skin-derived DCs to promote the TH2 response to cutaneously encountered antigen and allergic skin inflammation. METHODS DCs from dLNs of ovalbumin (OVA)-exposed skin were analyzed by flow cytometry and for their ability to polarize OVA-specific naive CD4+ T cells. Skin inflammation following epicutaneous sensitization of tape-stripped skin was assessed by flow cytometry of skin cells and real-time quantitative PCR of cytokines. Cytokine secretion and antibody levels were evaluated by ELISA. RESULTS Scratching upregulated IL4 expression in human skin. Similarly, tape stripping caused rapid basophil-dependent upregulation of cutaneous Il4 expression in mouse skin. In vitro treatment of DCs from skin dLNs with IL-4 promoted their capacity to drive TH2 differentiation. DCs from dLNs of OVA-sensitized skin of Il4-/- mice and CD11c-CreIl4rflox/- mice, which lack IL-4Rα expression in DCs (DCΔ/Δll4ra mice), were impaired in their capacity to drive TH2 polarization compared with DCs from controls. Importantly, OVA-sensitized DCΔ/Δll4ra mice demonstrated impaired allergic skin inflammation and OVA-specific systemic TH2 response evidenced by reduced TH2 cytokine secretion by OVA-stimulated splenocytes and lower levels of OVA-specific IgE and IgG1 antibodies, compared with controls. CONCLUSIONS Mechanical skin injury causes basophil-dependent upregulation of cutaneous IL-4. IL-4 acts on skin DCs that capture antigen and migrate to dLNs to promote their capacity for TH2 polarization and drive allergic skin inflammation.
Collapse
Affiliation(s)
| | - Mrinmoy Das
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Boston, Mass
| | - Maria Strakosha
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Boston, Mass
| | - Alex McGurk
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Boston, Mass
| | - Emilie Artru
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Boston, Mass
| | - Christy Kam
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Boston, Mass
| | - Mohammed Alasharee
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Boston, Mass
| | - Duane R Wesemann
- Division of Allergy and Clinical Immunology, Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass; Broad Institute, Cambridge, Mass; Ragon Institute, Cambridge, Mass
| | - Michio Tomura
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Japan
| | - Hajime Karasuyama
- Inflammation, Infection and Immunity Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Frank Brombacher
- International Center for Genetic Engineering and Biotechnology & University of Cape Town & South Africa Medical Research Council, Cape Town, South Africa
| | - Raif S Geha
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Boston, Mass.
| |
Collapse
|
9
|
Szeto AC, Ferreira AC, McKenzie AN. Molecular mechanisms regulating T helper 2 cell differentiation and function. Curr Opin Immunol 2024; 91:102483. [PMID: 39357077 PMCID: PMC7617428 DOI: 10.1016/j.coi.2024.102483] [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: 07/12/2024] [Revised: 08/29/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024]
Abstract
T helper 2 (TH2) cells orchestrate type 2 immunity during protective antihelminth immunity and help restore tissue homoeostasis. Their misdirected activities against innocuous substances also underlie atopic diseases, such as asthma and allergy. Recent technological advances are uncovering novel insights into the molecular mechanisms governing TH2 cell differentiation and function.
Collapse
Affiliation(s)
- Aydan Ch Szeto
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Ana Cf Ferreira
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | | |
Collapse
|
10
|
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
|
11
|
Huang R, Zhang B, Ye W, Tang Z, Zheng Q. IL-4 Downregulates PD-L1 Level Via SOCS1 Upregulation-Induced JNK Deactivation to Enhance Antitumor Immunity in In Vitro Colorectal Cancer. J Interferon Cytokine Res 2024; 44:486-495. [PMID: 39364618 DOI: 10.1089/jir.2024.0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024] Open
Abstract
Interleukin-4 (IL-4) controls cell growth and immune system regulation in tumorigenesis and can inhibit the growth of colon cancer cell lines, but the possible mechanism is unclear. In this study, we investigated the possible mechanism of IL-4 in colorectal cancer (CRC) through in vitro experiments. CRC cells received treatment with IL-4 (50 ng/mL), investigating the suppressor of cytokine signaling 1 (SOCS1)-related mechanism underlying the role of IL-4 in the progression and immunosuppression of CRC. The malignant processes of CRC cells and CD8+T cell-mediated immune response in CRC cells were determined by CCK-8, Transwell, wound healing, and flow cytometry assays. Programmed death ligand 1 (PD-L1), SOCS1 expressions, and c-Jun N-terminal kinase (JNK) activation in CRC cells were analyzed by quantitative reverse transcription polymerase chain reaction and/or Western blot. IL-4 repressed the malignant processes, yet promoted the apoptosis of CRC cells. Besides, IL-4 downregulated PD-L1 level, upregulated SOCS1 level, and restrained JNK activation in CRC cells, while enhancing CRC cell-killing effect of CD8+T cells. IL-4-induced effects on the aforementioned malignant processes of CRC cells and the killing effect of CD8+T cells toward CRC cells were all reversed when SOCS1 was knocked down in the CRC cells. IL-4 downregulates PD-L1 level via SOCS1 upregulation-induced JNK deactivation to enhance antitumor immunity in in vitro CRC. The study provides a theoretical basis for the clinical application of IL-4 in antitumor immunity in CRC.
Collapse
Affiliation(s)
- Ruiyan Huang
- The Second Department of Oncology, Wenzhou Central Hospital, Wenzhou, China
| | - Baofan Zhang
- The Second Department of Oncology, Wenzhou Central Hospital, Wenzhou, China
| | - Wanchun Ye
- The Second Department of Oncology, Wenzhou Central Hospital, Wenzhou, China
| | - Zhongjie Tang
- The Second Department of Oncology, Wenzhou Central Hospital, Wenzhou, China
| | - Qingsong Zheng
- The Second Department of Oncology, Wenzhou Central Hospital, Wenzhou, China
| |
Collapse
|
12
|
Hao Y, Zhang L, Meng Q, Jia Q, Ma J, Zhang X. Development and validation of an enzyme-linked immunosorbent assay for the quantification of a recombinant humanized anti-IL-4Rα monoclonal antibody CM310 in serum and its application to pharmacokinetic study in Sprague-Dawley Rats. Anal Biochem 2024; 694:115623. [PMID: 39059567 DOI: 10.1016/j.ab.2024.115623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
CM310 is a recombinant humanized monoclonal antibody targeting Interleukin (IL)-4 receptor alpha (IL-4Rα). IL-4Rα blockade prevents IL-4 and IL-13 from binding to their receptor, thereby inhibiting downstream signaling pathways that drive Type 2 helper T-cell (Th2) inflammation. CM310 holds potential for treating Th2-related inflammatory diseases, such as asthma, atopic dermatitis and chronic sinusitis with nasal polyposis. In this study, a direct enzyme-linked immunosorbent assay (ELISA) was developed to measure the concentrations of CM310 in rat serum. Seven calibration standards (ranging from 25 to 1600 ng/mL) and three quality controls (70, 500 and 1250 ng/mL) were defined. The limit of detection (LOD), lower limit of quantification (LLOQ) and upper limit of quantification (ULOQ) were 13, 25 and 1600 ng/mL, respectively. The method exhibited excellent precision and accuracy and successfully applied to in vitro serum stability and pharmacokinetic (PK) studies. In conclusion, we have developed and validated a highly sensitive and selective method for measuring CM310 in Sprague-Dawley rats. The development and validation ELISA method met the acceptable criteria, which suggested that these can be applied to quantify CM310, as well as in PK studies.
Collapse
Affiliation(s)
- Yimeng Hao
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Libo Zhang
- Keymed Biosciences Inc., Chengdu, Sichuan Province, China
| | - Qinghe Meng
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, China
| | - Qian Jia
- Keymed Biosciences Inc., Chengdu, Sichuan Province, China
| | - Jing Ma
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.
| |
Collapse
|
13
|
He K, Xiao H, MacDonald WA, Mehta I, Kishore A, Vincent A, Xu Z, Ray A, Chen W, Weaver CT, Lambrecht BN, Das J, Poholek AC. Spatial microniches of IL-2 combine with IL-10 to drive lung migratory T H2 cells in response to inhaled allergen. Nat Immunol 2024; 25:2124-2139. [PMID: 39394532 PMCID: PMC11934206 DOI: 10.1038/s41590-024-01986-8] [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: 10/16/2023] [Accepted: 09/12/2024] [Indexed: 10/13/2024]
Abstract
The mechanisms that guide T helper 2 (TH2) cell differentiation in barrier tissues are unclear. Here we describe the molecular pathways driving allergen-specific TH2 cells using temporal, spatial and single-cell transcriptomic tracking of house dust mite-specific T cells in mice. Differentiation and migration of lung allergen-specific TH2 cells requires early expression of the transcriptional repressor Blimp-1. Loss of Blimp-1 during priming in the lymph node ablated the formation of TH2 cells in the lung, indicating early Blimp-1 promotes TH2 cells with migratory capability. IL-2/STAT5 signals and autocrine/paracrine IL-10 from house dust mite-specific T cells were essential for Blimp-1 and subsequent GATA3 upregulation through repression of Bcl6 and Bach2. Spatial microniches of IL-2 in the lymph node supported the earliest Blimp-1+TH2 cells, demonstrating lymph node localization is a driver of TH2 initiation. Our findings identify an early requirement for IL-2-mediated spatial microniches that integrate with allergen-driven IL-10 from responding T cells to drive allergic asthma.
Collapse
Affiliation(s)
- Kun He
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hanxi Xiao
- Center for Systems Immunology, Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Joint CMU-Pitt PhD Program in Computational Biology, Pittsburgh, PA, USA
| | - William A MacDonald
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Health Sciences Sequencing Core, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Isha Mehta
- Center for Systems Immunology, Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Akash Kishore
- Center for Systems Immunology, Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Augusta Vincent
- Center for Systems Immunology, Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zhongli Xu
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- School of Medicine, Tsinghua University, Beijing, China
| | - Anuradha Ray
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Wei Chen
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Biostatistics, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Casey T Weaver
- Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Bart N Lambrecht
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Jishnu Das
- Center for Systems Immunology, Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amanda C Poholek
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Center for Systems Immunology, Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
14
|
Anvar MT, Rashidan K, Arsam N, Rasouli-Saravani A, Yadegari H, Ahmadi A, Asgari Z, Vanan AG, Ghorbaninezhad F, Tahmasebi S. Th17 cell function in cancers: immunosuppressive agents or anti-tumor allies? Cancer Cell Int 2024; 24:355. [PMID: 39465401 PMCID: PMC11514949 DOI: 10.1186/s12935-024-03525-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/08/2024] [Indexed: 10/29/2024] Open
Abstract
T helper (Th) 17 cells, a distinct subset of Th lymphocytes, are known for their prominent interleukin (IL)-17 production and other pro-inflammatory cytokines. These cells exhibit remarkable plasticity, allowing them to exhibit different phenotypes in the cancer microenvironment. This adaptability enables Th17 cells to promote tumor progression by immunosuppressive activities and angiogenesis, but also mediate anti-tumor immune responses through employing immune cells in tumor setting or even by directly converting toward Th1 phenotype and producing interferon-gamma (IFN-γ). This dual role of Th17 cells in cancer makes it a double-edged sword in encountering cancer. In this review, we aim to elucidate the complexities of Th17 cell function in cancer by summarizing recent studies and, ultimately, to design novel therapeutic strategies, especially targeting Th17 cells in the tumor milieu, which could pave the way for more effective cancer treatments.
Collapse
Affiliation(s)
- Milad Taghizadeh Anvar
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimiya Rashidan
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nima Arsam
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ashkan Rasouli-Saravani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamidreza Yadegari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Ahmadi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeynab Asgari
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Ghorbani Vanan
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Farid Ghorbaninezhad
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Safa Tahmasebi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Lee SH, Sacks DL. Resilience of dermis resident macrophages to inflammatory challenges. Exp Mol Med 2024; 56:2105-2112. [PMID: 39349826 PMCID: PMC11542019 DOI: 10.1038/s12276-024-01313-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/24/2024] [Accepted: 07/04/2024] [Indexed: 10/03/2024] Open
Abstract
The skin serves as a complex barrier organ populated by tissue-resident macrophages (TRMs), which play critical roles in defense, homeostasis, and tissue repair. This review examines the functions of dermis resident TRMs in different inflammatory settings, their embryonic origins, and their long-term self-renewal capabilities. We highlight the M2-like phenotype of dermal TRMs and their specialized functions in perivascular and perineuronal niches. Their interactions with type 2 immune cells, autocrine cytokines such as IL-10, and their phagocytic clearance of apoptotic cells have been explored as mechanisms for M2-like dermal TRM self-maintenance and function. In conclusion, we address the need to bridge murine models with human studies, with the possibility of targeting TRMs to promote skin immunity or restrain cutaneous pathology.
Collapse
Affiliation(s)
- Sang Hun Lee
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David L Sacks
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
16
|
Wang R, Lan C, Benlagha K, Camara NOS, Miller H, Kubo M, Heegaard S, Lee P, Yang L, Forsman H, Li X, Zhai Z, Liu C. The interaction of innate immune and adaptive immune system. MedComm (Beijing) 2024; 5:e714. [PMID: 39286776 PMCID: PMC11401974 DOI: 10.1002/mco2.714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 08/11/2024] [Accepted: 08/11/2024] [Indexed: 09/19/2024] Open
Abstract
The innate immune system serves as the body's first line of defense, utilizing pattern recognition receptors like Toll-like receptors to detect pathogens and initiate rapid response mechanisms. Following this initial response, adaptive immunity provides highly specific and sustained killing of pathogens via B cells, T cells, and antibodies. Traditionally, it has been assumed that innate immunity activates adaptive immunity; however, recent studies have revealed more complex interactions. This review provides a detailed dissection of the composition and function of the innate and adaptive immune systems, emphasizing their synergistic roles in physiological and pathological contexts, providing new insights into the link between these two forms of immunity. Precise regulation of both immune systems at the same time is more beneficial in the fight against immune-related diseases, for example, the cGAS-STING pathway has been found to play an important role in infections and cancers. In addition, this paper summarizes the challenges and future directions in the field of immunity, including the latest single-cell sequencing technologies, CAR-T cell therapy, and immune checkpoint inhibitors. By summarizing these developments, this review aims to enhance our understanding of the complexity interactions between innate and adaptive immunity and provides new perspectives in understanding the immune system.
Collapse
Affiliation(s)
- Ruyuan Wang
- Department of Thyroid and Breast Surgery Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Caini Lan
- Cancer Center Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Kamel Benlagha
- Alloimmunity, Autoimmunity and Transplantation Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160 Paris France
| | - Niels Olsen Saraiva Camara
- Department of Immunology Institute of Biomedical Sciences University of São Paulo (USP) São Paulo São Paulo Brazil
| | - Heather Miller
- Coxiella Pathogenesis Section, Laboratory of Bacteriology Rocky Mountain Laboratories National Institute of Allergy and Infectious Diseases, National Institutes of Health Hamilton Montana USA
| | - Masato Kubo
- Division of Molecular Pathology Research Institute for Biomedical Sciences (RIBS) Tokyo University of Science Noda Chiba Japan
| | - Steffen Heegaard
- Department of Ophthalmology Rigshospitalet Hospital Copenhagen University Copenhagen Denmark
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong China
| | - Lu Yang
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology Wuhan Hubei China
| | - Huamei Forsman
- Department of Laboratory Medicine Institute of Biomedicine, University of Gothenburg Gothenburg Sweden
| | - Xingrui Li
- Department of Thyroid and Breast Surgery Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Zhimin Zhai
- Department of Hematology The Second Hospital of Anhui Medical University Hefei China
| | - Chaohong Liu
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology Wuhan Hubei China
| |
Collapse
|
17
|
Wang S, Guo L, Gu F, Bao J, Guo Y, Zhang Y, Wang Z, Li R, Wu Z, Li J. Quercetin restores respiratory mucosal barrier dysfunction in Mycoplasma gallisepticum-infected chicks by enhancing Th2 immune response. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155953. [PMID: 39154527 DOI: 10.1016/j.phymed.2024.155953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/31/2024] [Accepted: 08/10/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Mycoplasma gallisepticum (MG) has long been a pathogenic microorganism threatening the global poultry industry. Previous studies have demonstrated that the mechanism by which quercetin (QUE) inhibits the colonization of MG in chicks differs from that of antibiotics. However, the molecular mechanism by which QUE facilitates the clearance of MG remains unclear. PURPOSE The aim of this study was to investigate the molecular mechanism of MG clearance by QUE, with the expectation of providing new options for the treatment of MG. METHODS A model of MG infection in chicks and MG-induced M1 polarization in HD-11 cells were established. The mechanism of QUE clearance of MG was investigated by evaluating the relationship between tracheal mucosal barrier integrity, antibody levels, Th1/Th2 immune balance and macrophage metabolism and M1/M2 polarization balance. Furthermore, network pharmacology and molecular docking techniques were employed to explore the potential molecular pathways connecting QUE, M2 polarization, and fatty acid oxidation (FAO). RESULTS The findings indicate that QUE remodels tracheal mucosal barrier function by regulating tight junctions and secretory immunoglobulin A (sIgA) expression levels. This process entails the regulatory function of QUE on the Th1/Th2 immune imbalance that is induced by MG infection in the tracheal mucosa. Moreover, QUE intervention impeded the M1 polarization of HD-11 cells induced by MG infection, while simultaneously promoting M2 polarization through the induction of FAO. Conversely, inhibitors of the FAO pathway impede this effect. The results of computer network analysis suggest that QUE may induce FAO via the PI3K/AKT pathway to promote M2 polarization. Notably, inhibition of the PI3K/AKT pathway was found to effectively inhibit M2 polarization in HD-11 cells, while having a limited effect on FAO. CONCLUSIONS QUE promotes M2 polarization of HD-11 cells to enhance Th2 immune response through FAO and PI3K/AKT pathways, thereby restoring tracheal mucosal barrier function and ultimately inhibiting MG colonization.
Collapse
Affiliation(s)
- Shun Wang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China
| | - Liyang Guo
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China
| | - Fuhua Gu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China
| | - Jiaxin Bao
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China
| | - Yuquan Guo
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China
| | - Yongjie Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China
| | - Ze Wang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China
| | - Rui Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China
| | - Zhiyong Wu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China.
| | - Jichang Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China.
| |
Collapse
|
18
|
Cheng Y, Zhao M, Zhu C, Tang X, Wang W, Tang H, Zheng X, Zhu Z, Sheng Y, Wang Z, Zhou F, Gao J. Proteomic Analysis Reveals Oxidative Phosphorylation and JAK-STAT Pathways Mediated Pathogenesis of Pemphigus Vulgaris. Exp Dermatol 2024; 33:e15184. [PMID: 39373252 DOI: 10.1111/exd.15184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/14/2024] [Accepted: 09/17/2024] [Indexed: 10/08/2024]
Abstract
Pemphigus vulgaris (PV) stands as a rare autoimmune bullous disease, while the precise underlying mechanism remains incompletely elucidated. High-throughput proteomic methodologies, such as LC-MS/MS, have facilitated the quantification and characterisation of proteomes from clinical skin samples, enhancing our comprehension of PV pathogenesis. The objective of this study is to elucidate the signalling mechanisms underlying PV through proteomic analysis. Proteins and cell suspension were extracted from skin biopsies obtained from both PV patients and healthy volunteers and subsequently analysed using LC-MS/MS and scRNA-seq. Cultured keratinocytes were treated with PV serum, followed by an assessment of protein expression levels using immunofluorescence and western blotting. A total of 880, 605, and 586 differentially expressed proteins (DEPs) were identified between the lesion vs. control, non-lesion vs. control, and lesion vs. non-lesion groups, respectively. The oxidative phosphorylation (OXPHOS) pathway showed activation in PV. Keratinocytes are the major cell population in the epidermis and highly expressed ATP5PF, ATP6V1G1, COX6B1, COX6A1, and NDUFA9. In the cellular model, there was a notable increase in the expression levels of OXPHOS-related proteins (V-ATP5A, III-UQCRC2, II-SDHB, I-NDUFB8), along with STAT1, p-STAT1, and p-JAK1. Furthermore, both the OXPHOS inhibitor metformin and the JAK1 inhibitor tofacitinib demonstrated therapeutic effects on PV serum-induced cell separation, attenuating cell detachment. Metformin notably reduced the expression of V-ATP5A, III-UQCRC2, II-SDHB, I-NDUFB8, p-STAT1, p-JAK1, whereas tofacitinib decreased the expression of p-STAT1 and p-JAK1, with minimal impact on the expression of V-ATP5A, III-UQCRC2, II-SDHB, and I-NDUFB8. Our results indicate a potential involvement of the OXPHOS and JAK-STAT1 pathways in the pathogenesis of PV.
Collapse
Affiliation(s)
- Yuqi Cheng
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Mingming Zhao
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - CaiHong Zhu
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Xianfa Tang
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Wenjun Wang
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Huayang Tang
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Xiaodong Zheng
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Zhengwei Zhu
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Yujun Sheng
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Zaixing Wang
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Fusheng Zhou
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Jinping Gao
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| |
Collapse
|
19
|
高 雅, 龙 波, 余 少. [Research progress of memory T cells in the pathogenesis of allergic rhinitis]. LIN CHUANG ER BI YAN HOU TOU JING WAI KE ZA ZHI = JOURNAL OF CLINICAL OTORHINOLARYNGOLOGY HEAD AND NECK SURGERY 2024; 38:975-978. [PMID: 39390941 PMCID: PMC11839565 DOI: 10.13201/j.issn.2096-7993.2024.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Indexed: 10/12/2024]
Abstract
Allergic rhinitis(AR) is a non-infectious chronic inflammatory disease of the nasal mucosa mainly mediated by immunoglobulin E(IgE) in atopic individuals after exposure to allergens. T cells are the core cell population. In recent years, studies have shown that memory T cells play an important role in the development of allergic rhinitis. This article reviews the pathogenesis of memory T cells in allergic rhinitis, in order to further improve the pathogenesis of allergic rhinitis and provide theoretical basis and reference for subsequent clinical drug treatment.
Collapse
Affiliation(s)
- 雅玟 高
- 同济大学附属同济医院耳鼻咽喉头颈外科(上海,200065)Department of Otorhinolaryngology Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - 波锦 龙
- 同济大学附属同济医院耳鼻咽喉头颈外科(上海,200065)Department of Otorhinolaryngology Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - 少卿 余
- 同济大学附属同济医院耳鼻咽喉头颈外科(上海,200065)Department of Otorhinolaryngology Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| |
Collapse
|
20
|
Dash MK, Samal S, Rout S, Behera CK, Sahu MC, Das B. Immunomodulation in dengue: towards deciphering dengue severity markers. Cell Commun Signal 2024; 22:451. [PMID: 39327552 PMCID: PMC11425918 DOI: 10.1186/s12964-024-01779-4] [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: 05/06/2024] [Accepted: 08/06/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Dengue is a vector-borne debilitating disease that is manifested as mild dengue fever, dengue with warning signs, and severe dengue. Dengue infection provokes a collective immune response; in particular, the innate immune response plays a key role in primary infection and adaptive immunity during secondary infection. In this review, we comprehensively walk through the various markers of immune response against dengue pathogenesis and outcome. MAIN BODY Innate immune response against dengue involves a collective response through the expression of proinflammatory cytokines, such as tumor necrosis factors (TNFs), interferons (IFNs), and interleukins (ILs), in addition to anti-inflammatory cytokines and toll-like receptors (TLRs) in modulating viral pathogenesis. Monocytes, dendritic cells (DCs), and mast cells are the primary innate immune cells initially infected by DENV. Such immune cells modulate the expression of various markers, which can influence disease severity by aiding virus entry and proinflammatory responses. Adaptive immune response is mainly aided by B and T lymphocytes, which stimulate the formation of germinal centers for plasmablast development and antibody production. Such antibodies are serotype-dependent and can aid in virus entry during secondary infection, mediated through a different serotype, such as in antibody-dependent enhancement (ADE), leading to DENV severity. The entire immunological repertoire is exhibited differently depending on the immune status of the individual. SHORT CONCLUSION Dengue fever through severe dengue proceeds along with the modulated expression of several immune markers. In particular, TLR2, TNF-α, IFN-I, IL-6, IL-8, IL-17 and IL-10, in addition to intermediate monocytes (CD14+CD16+) and Th17 (CD4+IL-17+) cells are highly expressed during severe dengue. Such markers could assist greatly in severity assessment, prompt diagnosis, and treatment.
Collapse
Affiliation(s)
- Manoj Kumar Dash
- School of Biotechnology, Kalinga Institute of Industrial Technology, Deemed to Be University, Bhubaneswar, Odisha, 751024, India
| | - Sagnika Samal
- School of Biotechnology, Kalinga Institute of Industrial Technology, Deemed to Be University, Bhubaneswar, Odisha, 751024, India
| | - Shailesh Rout
- School of Biotechnology, Kalinga Institute of Industrial Technology, Deemed to Be University, Bhubaneswar, Odisha, 751024, India
| | - Chinmay Kumar Behera
- Department of Pediatrics, Kalinga Institute of Medical Sciences, Deemed to Be University, Bhubaneswar, Odisha, 751024, India
| | | | - Biswadeep Das
- School of Biotechnology, Kalinga Institute of Industrial Technology, Deemed to Be University, Bhubaneswar, Odisha, 751024, India.
| |
Collapse
|
21
|
Mertelsmann AM, Bowers SF, Wright D, Maganga JK, Mazigo HD, Ndhlovu LC, Changalucha JM, Downs JA. Effects of Schistosoma haematobium infection and treatment on the systemic and mucosal immune phenotype, gene expression and microbiome: A systematic review. PLoS Negl Trop Dis 2024; 18:e0012456. [PMID: 39250522 PMCID: PMC11412685 DOI: 10.1371/journal.pntd.0012456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 09/19/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Urogenital schistosomiasis caused by Schistosoma haematobium affects approximately 110 million people globally, with the majority of cases in low- and middle-income countries. Schistosome infections have been shown to impact the host immune system, gene expression, and microbiome composition. Studies have demonstrated variations in pathology between schistosome subspecies. In the case of S. haematobium, infection has been associated with HIV acquisition and bladder cancer. However, the underlying pathophysiology has been understudied compared to other schistosome species. This systematic review comprehensively investigates and assimilates the effects of S. haematobium infection on systemic and local host mucosal immunity, cellular gene expression and microbiome. METHODS We conducted a systematic review assessing the reported effects of S. haematobium infections and anthelmintic treatment on the immune system, gene expression and microbiome in humans and animal models. This review followed PRISMA guidelines and was registered prospectively in PROSPERO (CRD42022372607). Randomized clinical trials, cohort, cross-sectional, case-control, experimental ex vivo, and animal studies were included. Two reviewers performed screening independently. RESULTS We screened 3,177 studies and included 94. S. haematobium was reported to lead to: (i) a mixed immune response with a predominant type 2 immune phenotype, increased T and B regulatory cells, and select pro-inflammatory cytokines; (ii) distinct molecular alterations that would compromise epithelial integrity, such as increased metalloproteinase expression, and promote immunological changes and cellular transformation, specifically upregulation of genes p53 and Bcl-2; and (iii) microbiome dysbiosis in the urinary, intestinal, and genital tracts. CONCLUSION S. haematobium induces distinct alterations in the host's immune system, molecular profile, and microbiome. This leads to a diverse range of inflammatory and anti-inflammatory responses and impaired integrity of the local mucosal epithelial barrier, elevating the risks of secondary infections. Further, S. haematobium promotes cellular transformation with oncogenic potential and disrupts the microbiome, further influencing the immune system and genetic makeup. Understanding the pathophysiology of these interactions can improve outcomes for the sequelae of this devastating parasitic infection.
Collapse
Affiliation(s)
- Anna M Mertelsmann
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
- Center for Global Health, Weill Cornell Medicine, New York, New York, United States of America
| | - Sheridan F Bowers
- Center for Global Health, Weill Cornell Medicine, New York, New York, United States of America
| | - Drew Wright
- Samuel J. Wood Library & C.V. Starr Biomedical Information Center, Weill Cornell Medical College, New York, New York, United States of America
| | - Jane K Maganga
- Mwanza Intervention Trials Unit/National Institute for Medical Research, Mwanza, Tanzania
| | - Humphrey D Mazigo
- Department of Parasitology and Entomology, Catholic University of Health and Allied Sciences, Mwanza, Tanzania
| | - Lishomwa C Ndhlovu
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - John M Changalucha
- Mwanza Intervention Trials Unit/National Institute for Medical Research, Mwanza, Tanzania
| | - Jennifer A Downs
- Center for Global Health, Weill Cornell Medicine, New York, New York, United States of America
- Mwanza Intervention Trials Unit/National Institute for Medical Research, Mwanza, Tanzania
- Weill Bugando School of Medicine, Mwanza, Tanzania
| |
Collapse
|
22
|
Sánchez-Menéndez C, de la Calle-Jiménez O, Mateos E, Vigón L, Fuertes D, Murciano Antón MA, San José E, García-Gutiérrez V, Cervero M, Torres M, Coiras M. Different polarization and functionality of CD4+ T helper subsets in people with post-COVID condition. Front Immunol 2024; 15:1431411. [PMID: 39257580 PMCID: PMC11385313 DOI: 10.3389/fimmu.2024.1431411] [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: 05/11/2024] [Accepted: 08/09/2024] [Indexed: 09/12/2024] Open
Abstract
Introduction After mild COVID-19 that does not require hospitalization, some individuals develop persistent symptoms that may worsen over time, producing a multisystemic condition termed Post-COVID condition (PCC). Among other disorders, PCC is characterized by persistent changes in the immune system that may not be solved several months after COVID-19 diagnosis. Methods People with PCC were recruited to determine the distribution and functionality of CD4+ T helper (Th) subsets in comparison with individuals with mild, severe, and critical presentations of acute COVID-19 to evaluate their contribution as risk or protective factors for PCC. Results People with PCC showed low levels of Th1 cells, similar to individuals with severe and critical COVID-19, although these cells presented a higher capacity to express IFNγ in response to stimulation. Th2/Th1 correlation was negative in individuals with acute forms of COVID-19, but there was no significant Th2/Th1 correlation in people with PCC. Th2 cells from people with PCC presented high capacity to express IL-4 and IL-13, which are related to low ventilation and death associated with COVID-19. Levels of proinflammatory Th9 and Th17 subsets were significantly higher in people with PCC in comparison with acute COVID-19, being Th1/Th9 correlation negative in these individuals, which probably contributed to a more pro-inflammatory than antiviral scenario. Th17 cells from approximately 50% of individuals with PCC had no capacity to express IL-17A and IL-22, similar to individuals with critical COVID-19, which would prevent clearing extracellular pathogens. Th2/Th17 correlation was positive in people with PCC, which in the absence of negative Th1/Th2 correlation could also contribute to the proinflammatory state. Finally, Th22 cells from most individuals with PCC had no capacity to express IL-13 or IL-22, which could increase tendency to reinfections due to impaired epithelial regeneration. Discussion People with PCC showed skewed polarization of CD4+ Th subsets with altered functionality that was more similar to individuals with severe and critical presentations of acute COVID-19 than to people who fully recovered from mild disease. New strategies aimed at reprogramming the immune response and redirecting CD4+ Th cell polarization may be necessary to reduce the proinflammatory environment characteristic of PCC.
Collapse
Affiliation(s)
- Clara Sánchez-Menéndez
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- PhD Program in Biomedical Sciences and Public Health, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Olivia de la Calle-Jiménez
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Internal Medicine Service, Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - Elena Mateos
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Lorena Vigón
- AIDS Immunopathology, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Fuertes
- School of Telecommunications Engineering, Universidad Politécnica de Madrid, Madrid, Spain
| | - María Aranzazu Murciano Antón
- Family Medicine, Centro de Salud Doctor Pedro Laín Entralgo, Alcorcón, Madrid, Spain
- International PhD School, Universidad Rey Juan Carlos, Alcorcón, Madrid, Spain
| | - Esther San José
- Immunomodulation Unit, Department of Health Sciences, Faculty of Biomedical and Health Sciences, European University of Madrid, Madrid, Spain
| | - Valentín García-Gutiérrez
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Miguel Cervero
- School of Medicine, Universidad Alfonso X El Sabio, Madrid, Spain
| | - Montserrat Torres
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Mayte Coiras
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
23
|
Sun X, Gu R, Bai J. Differentiation and regulation of CD4 + T cell subsets in Parkinson's disease. Cell Mol Life Sci 2024; 81:352. [PMID: 39153043 PMCID: PMC11335276 DOI: 10.1007/s00018-024-05402-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, and its hallmark pathological features are the loss of dopaminergic (DA) neurons in the midbrain substantia nigra pars compacta (SNpc) and the accumulation of alpha-synuclein (α-syn). It has been shown that the integrity of the blood-brain barrier (BBB) is damaged in PD patients, and a large number of infiltrating T cells and inflammatory cytokines have been detected in the cerebrospinal fluid (CSF) and brain parenchyma of PD patients and PD animal models, including significant change in the number and proportion of different CD4+ T cell subsets. This suggests that the neuroinflammatory response caused by CD4+ T cells is an important risk factor for the development of PD. Here, we systematically review the differentiation of CD4+ T cell subsets, and focus on describing the functions and mechanisms of different CD4+ T cell subsets and their secreted cytokines in PD. We also summarize the current immunotherapy targeting CD4+ T cells with a view to providing assistance in the diagnosis and treatment of PD.
Collapse
Affiliation(s)
- Xiaowei Sun
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
- Southwest United Graduate School, Kunming, 650500, China
| | - Rou Gu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Jie Bai
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China.
- Southwest United Graduate School, Kunming, 650500, China.
| |
Collapse
|
24
|
Baral B, Saini V, Kandpal M, Kundu P, Dixit AK, Parmar HS, Meena AK, Trivedi P, Jha HC. The interplay of co-infections in shaping COVID-19 severity: Expanding the scope beyond SARS-CoV-2. J Infect Public Health 2024; 17:102486. [PMID: 39002466 DOI: 10.1016/j.jiph.2024.102486] [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: 12/10/2023] [Revised: 06/17/2024] [Accepted: 06/23/2024] [Indexed: 07/15/2024] Open
Abstract
High mortality has been reported in severe cases of COVID-19. Emerging reports suggested that the severity is not only due to SARS-CoV-2 infection, but also due to coinfections by other pathogens exhibiting symptoms like COVID-19. During the COVID-19 pandemic, simultaneous respiratory coinfections with various viral (Retroviridae, Flaviviridae, Orthomyxoviridae, and Picoviridae) and bacterial (Mycobacteriaceae, Mycoplasmataceae, Enterobacteriaceae and Helicobacteraceae) families have been observed. These pathogens intensify disease severity by potentially augmenting SARSCoV-2 replication, inflammation, and modulation of signaling pathways. Coinfection emerges as a critical determinant of COVID-19 severity, principally instigated by heightened pro-inflammatory cytokine levels, as cytokine storm. Thereby, in co-infection scenario, the severity is also driven by the modulation of inflammatory signaling pathways by both pathogens possibly associated with interleukin, interferon, and cell death exacerbating the severity. In the current review, we attempt to understand the role of co- infections by other pathogens and their involvement in the severity of COVID-19.
Collapse
Affiliation(s)
- Budhadev Baral
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India
| | - Vaishali Saini
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India
| | - Meenakshi Kandpal
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India
| | - Pratik Kundu
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India
| | - Amit Kumar Dixit
- Central Ayurveda Research Institute, 4-CN Block, Sector -V, Bidhannagar, Kolkata 700 091, India
| | - Hamendra Singh Parmar
- School of Biotechnology, Devi Ahilya Vishwavidyalaya, Takshashila Campus, Indore, Madhya Pradesh 452001, India
| | - Ajay Kumar Meena
- Regional Ayurveda Research Institute, Gwalior, Amkhoh, Gwalior, Madhya Pradesh 474001, India
| | - Pankaj Trivedi
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Hem Chandra Jha
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India; Centre for Rural Development and Technology, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India.
| |
Collapse
|
25
|
Zheng W, Wai CYY, Sit JKC, Cheng NS, Leung CWM, Leung TF. Routinely Used and Emerging Diagnostic and Immunotherapeutic Approaches for Wheat Allergy. Biomedicines 2024; 12:1549. [PMID: 39062122 PMCID: PMC11275021 DOI: 10.3390/biomedicines12071549] [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: 05/11/2024] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Wheat, a component of the staple diet globally, is a common food allergen in children. The symptoms of wheat allergy (WA) range from skin rash to shortness of breath, significantly impairing quality of life. Following initial clinical suspicion, individuals may undergo routinely used allergy tests such as a wheat allergen-specific skin prick test (SPT), a blood test for specific immunoglobulin E (sIgE) levels, or oral food challenge. Conventional management of WA lies in wheat avoidance, yet accidental consumption may be inevitable owing to the ubiquity of wheat in various food products. This article aims to provide an overview of the immunologic pathway of WA, followed by its emerging diagnostic methods, namely alcohol-soluble SPT extracts, component-resolved diagnosis, and the basophil activation test (BAT). The mechanisms underlying wheat allergen-specific oral immunotherapy (OIT) as well as a summary of the efficacy, tolerability, and safety of related clinical trials will then be discussed.
Collapse
Affiliation(s)
- Wanqi Zheng
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong; (W.Z.); (C.Y.Y.W.); (J.K.C.S.); (N.S.C.)
| | - Christine Yee Yan Wai
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong; (W.Z.); (C.Y.Y.W.); (J.K.C.S.); (N.S.C.)
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jason Ka Chun Sit
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong; (W.Z.); (C.Y.Y.W.); (J.K.C.S.); (N.S.C.)
| | - Nam Sze Cheng
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong; (W.Z.); (C.Y.Y.W.); (J.K.C.S.); (N.S.C.)
| | | | - Ting Fan Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong; (W.Z.); (C.Y.Y.W.); (J.K.C.S.); (N.S.C.)
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
26
|
Szeto AC, Clark PA, Ferreira AC, Heycock M, Griffiths EL, Jou E, Mannion J, Luan SL, Storrar S, Knolle MD, Kozik P, Jolin HE, Fallon PG, McKenzie AN. Mef2d potentiates type-2 immune responses and allergic lung inflammation. Science 2024; 384:eadl0370. [PMID: 38935708 PMCID: PMC7616247 DOI: 10.1126/science.adl0370] [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: 09/25/2023] [Accepted: 05/02/2024] [Indexed: 06/29/2024]
Abstract
Innate lymphoid cells (ILCs) and adaptive T lymphocytes promote tissue homeostasis and protective immune responses. Their production depends on the transcription factor GATA3, which is further elevated specifically in ILC2s and T helper 2 cells to drive type-2 immunity during tissue repair, allergic disorders, and anti-helminth immunity. The control of this crucial up-regulation is poorly understood. Using CRISPR screens in ILCs we identified previously unappreciated myocyte-specific enhancer factor 2d (Mef2d)-mediated regulation of GATA3-dependent type-2 lymphocyte differentiation. Mef2d-deletion from ILC2s and/or T cells specifically protected against an allergen lung challenge. Mef2d repressed Regnase-1 endonuclease expression to enhance IL-33 receptor production and IL-33 signaling and acted downstream of calcium-mediated signaling to translocate NFAT1 to the nucleus to promote type-2 cytokine-mediated immunity.
Collapse
Affiliation(s)
- Aydan C.H. Szeto
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Paula A. Clark
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Ana C.F. Ferreira
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Morgan Heycock
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Emma L. Griffiths
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Eric Jou
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Jonathan Mannion
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
- Cambridge
University Hospitals,
Cambridge, CB2 0QQ, United Kingdom
| | - Shi-Lu Luan
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Sophie Storrar
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Martin D. Knolle
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
- Cambridge
University Hospitals,
Cambridge, CB2 0QQ, United Kingdom
| | - Patrycja Kozik
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Helen E. Jolin
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | | | | |
Collapse
|
27
|
Aimulajiang K, Chu W, Liao S, Wen Z, He R, Lu M, Xu L, Song X, Li X, Yan R. Succinate coenzyme A ligase β-like protein from Trichinella spiralis is a potential therapeutic molecule for allergic asthma. Immun Inflamm Dis 2024; 12:e1321. [PMID: 38888451 PMCID: PMC11184933 DOI: 10.1002/iid3.1321] [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: 07/25/2023] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND For decades, studies have demonstrated the anti-inflammatory potential of proteins secreted by helminths in allergies and asthma. Previous studies have demonstrated the immunomodulatory capabilities of Succinate Coenzyme A ligase beta-like protein (SUCLA-β) derived from Trichinella spiralis, a crucial excretory product of this parasite. OBJECTIVE To explore the therapeutic potential of SUCLA-β in alleviating and controlling ovalbumin (OVA)-induced allergic asthma, as well as its influence on host immune modulation. METHODS In this research, we utilized the rTs-SUCLA-β protein derived from T. spiralis to investigate its potential in mitigating airway inflammation in a murine model of asthma induced by OVA sensitization/stimulation, both pre- and post-challenge. The treatment's efficacy was assessed by quantifying the extent of inflammation in the lungs. RESULTS Treatment with rTs-SUCLA-β demonstrated efficacy in ameliorating OVA-induced airway inflammation, as evidenced by a reduction in eosinophil infiltration, levels of OVA-specific Immunoglobulin E, interferon-γ, interleukin (IL)-9, and IL-17A, along with an elevation in IL-10. The equilibrium between Th17 and Treg cells plays a pivotal role in modulating the abundance of inflammatory cells within the organism, thereby ameliorating inflammation and alleviating symptoms associated with allergic asthma. CONCLUSIONS AND CLINICAL RELEVANCE Our data revealed that T. spiralis-derived Ts-SUCLA-β protein may inhibit the allergic airway inflammation by regulating host immune responses.
Collapse
Affiliation(s)
- Kalibixiati Aimulajiang
- Laboratory of Animal Health and Food Safety, MOE Joint International Research, College of Veterinary MedicineNanjing Agricultural UniversityNanjingJiangsuP. R. China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine InstituteThe First Affiliated Hospital of Xinjiang Medical UniversityUrumqiXinjiangP. R. China
| | - Wen Chu
- Laboratory of Animal Health and Food Safety, MOE Joint International Research, College of Veterinary MedicineNanjing Agricultural UniversityNanjingJiangsuP. R. China
| | - Shuyi Liao
- Laboratory of Animal Health and Food Safety, MOE Joint International Research, College of Veterinary MedicineNanjing Agricultural UniversityNanjingJiangsuP. R. China
- Changsha Animal and Plant Disease Control CenterChangshaHunanP. R. China
| | - Zhaohai Wen
- Laboratory of Animal Health and Food Safety, MOE Joint International Research, College of Veterinary MedicineNanjing Agricultural UniversityNanjingJiangsuP. R. China
- Department of Veterinary Medicine, College of Coastal Agricultural SciencesGuangdong Ocean UniversityZhanjiangP. R. China
| | - Rongdong He
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine InstituteThe First Affiliated Hospital of Xinjiang Medical UniversityUrumqiXinjiangP. R. China
| | - Mingmin Lu
- Laboratory of Animal Health and Food Safety, MOE Joint International Research, College of Veterinary MedicineNanjing Agricultural UniversityNanjingJiangsuP. R. China
| | - Lixin Xu
- Laboratory of Animal Health and Food Safety, MOE Joint International Research, College of Veterinary MedicineNanjing Agricultural UniversityNanjingJiangsuP. R. China
| | - Xiaokai Song
- Laboratory of Animal Health and Food Safety, MOE Joint International Research, College of Veterinary MedicineNanjing Agricultural UniversityNanjingJiangsuP. R. China
| | - Xiangrui Li
- Laboratory of Animal Health and Food Safety, MOE Joint International Research, College of Veterinary MedicineNanjing Agricultural UniversityNanjingJiangsuP. R. China
| | - Ruofeng Yan
- Laboratory of Animal Health and Food Safety, MOE Joint International Research, College of Veterinary MedicineNanjing Agricultural UniversityNanjingJiangsuP. R. China
| |
Collapse
|
28
|
Tchen J, Simon Q, Chapart L, Thaminy MK, Vibhushan S, Saveanu L, Lamri Y, Saidoune F, Pacreau E, Pellefigues C, Bex-Coudrat J, Karasuyama H, Miyake K, Hidalgo J, Fallon PG, Papo T, Blank U, Benhamou M, Hanouna G, Sacre K, Daugas E, Charles N. PD-L1- and IL-4-expressing basophils promote pathogenic accumulation of T follicular helper cells in lupus. Nat Commun 2024; 15:3389. [PMID: 38649353 PMCID: PMC11035650 DOI: 10.1038/s41467-024-47691-w] [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: 08/30/2023] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by anti-nuclear autoantibodies whose production is promoted by autoreactive T follicular helper (TFH) cells. During SLE pathogenesis, basophils accumulate in secondary lymphoid organs (SLO), amplify autoantibody production and disease progression through mechanisms that remain to be defined. Here, we provide evidence for a direct functional relationship between TFH cells and basophils during lupus pathogenesis, both in humans and mice. PD-L1 upregulation on basophils and IL-4 production are associated with TFH and TFH2 cell expansions and with disease activity. Pathogenic TFH cell accumulation, maintenance, and function in SLO were dependent on PD-L1 and IL-4 in basophils, which induced a transcriptional program allowing TFH2 cell differentiation and function. Our study establishes a direct mechanistic link between basophils and TFH cells in SLE that promotes autoantibody production and lupus nephritis.
Collapse
Affiliation(s)
- John Tchen
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Quentin Simon
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
- Inovarion, 75005, Paris, France
| | - Léa Chapart
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Morgane K Thaminy
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Shamila Vibhushan
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Loredana Saveanu
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Yasmine Lamri
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Fanny Saidoune
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Emeline Pacreau
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Christophe Pellefigues
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Julie Bex-Coudrat
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Hajime Karasuyama
- Inflammation, Infection and Immunity Laboratory, TMDU Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kensuke Miyake
- Inflammation, Infection and Immunity Laboratory, TMDU Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Juan Hidalgo
- Universidad Autonoma de Barcelona, Facultad de Biociencias, Unidad de Fisiologia Animal Bellaterra, Bellaterra Campus, 08193, Barcelona, Spain
| | | | - Thomas Papo
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
- Service de Médecine Interne, Hôpital Bichat, Assistance Publique - Hôpitaux de Paris, 75018, Paris, France
| | - Ulrich Blank
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Marc Benhamou
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Guillaume Hanouna
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
- Service de Néphrologie, Hôpital Bichat, Assistance Publique - Hôpitaux de Paris, 75018, Paris, France
| | - Karim Sacre
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
- Service de Médecine Interne, Hôpital Bichat, Assistance Publique - Hôpitaux de Paris, 75018, Paris, France
| | - Eric Daugas
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
- Service de Néphrologie, Hôpital Bichat, Assistance Publique - Hôpitaux de Paris, 75018, Paris, France
| | - Nicolas Charles
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France.
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France.
| |
Collapse
|
29
|
Lyons-Cohen MR, Shamskhou EA, Gerner MY. Site-specific regulation of Th2 differentiation within lymph node microenvironments. J Exp Med 2024; 221:e20231282. [PMID: 38442268 PMCID: PMC10912907 DOI: 10.1084/jem.20231282] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/13/2023] [Accepted: 02/08/2024] [Indexed: 03/07/2024] Open
Abstract
T helper 2 (Th2) responses protect against pathogens while also driving allergic inflammation, yet how large-scale Th2 responses are generated in tissue context remains unclear. Here, we used quantitative imaging to investigate early Th2 differentiation within lymph nodes (LNs) following cutaneous allergen administration. Contrary to current models, we observed extensive activation and "macro-clustering" of early Th2 cells with migratory type-2 dendritic cells (cDC2s), generating specialized Th2-promoting microenvironments. Macro-clustering was integrin-mediated and promoted localized cytokine exchange among T cells to reinforce differentiation, which contrasted the behavior during Th1 responses. Unexpectedly, formation of Th2 macro-clusters was dependent on the site of skin sensitization. Differences between sites were driven by divergent activation states of migratory cDC2 from different dermal tissues, with enhanced costimulatory molecule expression by cDC2 in Th2-generating LNs promoting prolonged T cell activation, macro-clustering, and cytokine sensing. Thus, the generation of dedicated Th2 priming microenvironments through enhanced costimulatory molecule signaling initiates Th2 responses in vivo and occurs in a skin site-specific manner.
Collapse
Affiliation(s)
- Miranda R. Lyons-Cohen
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA, USA
| | - Elya A. Shamskhou
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA, USA
| | - Michael Y. Gerner
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
30
|
Acar Eser N, Dikmetas O, Kocabeyoglu S, Tan C, Irkec M. Evaluation of Keratoconus Disease with Tear Cytokine and Chemokine Levels Before and After Corneal Cross-Linking Treatment. Ocul Immunol Inflamm 2024; 32:269-275. [PMID: 36745704 DOI: 10.1080/09273948.2023.2165950] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/06/2022] [Accepted: 01/03/2023] [Indexed: 02/08/2023]
Abstract
OBJECTIVES To compare tear cytokine and chemokine levels of keratoconus (KC) patients with controls to perceive etiology distinctly and to clarify the molecular changes after cross-linking (CXL). METHODS Tear samples were gathered from 34 participants in this prospective study. Participants underwent anterior and posterior segment examinations with slit-lamp biomicroscopy. Patients were assessed by corneal topography before and 3 months after CXL. Flat (K1), steep (K2), and average keratometry (Kmean), cylinder (CYL), and central corneal thickness (CCT) values were evaluated. After 3 months from CXL, samples were re-collected, and comparisons were made with preoperative values. RESULTS Levels of IFN-gamma, IL-8, IL-12, IL-17, TNF-α, IL-4 and IL-13 were detected higher in KC patients (p= 0.008, p= 0.047, p= 0.001, p= 0.001, p= 0.001, p= 0.001, p= 0.027, respectively). After CXL IL-4, IL-5, IL-6, IL-7, IL-8, TNF-α levels showed significant decrease (p= 0.005, p= 0.045, p= 0.010, p= 0.022, p= 0.001, p=0.002, respectively). As for the topographic measurements, postoperative CCT values were increased whereas Kmean reduced after CXL (p < 0.001, p = 0.015, respectively). (p= 0.001, p= 0.027, respectively). CONCLUSION Our findings imply that inflammation plays a key role in the development of KC and that this link is influenced by CXL therapy.
Collapse
Affiliation(s)
- Nazan Acar Eser
- Department of Ophthalmology, Ulucanlar Eye Training and Research Hospital, University of Health Science, Ankara, Turkey
| | - Ozlem Dikmetas
- Department of Ophthalmology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Sibel Kocabeyoglu
- Department of Ophthalmology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Cagman Tan
- Department of Pediatrics Immunology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Murat Irkec
- Department of Ophthalmology, Hacettepe University School of Medicine, Ankara, Turkey
| |
Collapse
|
31
|
Hayashi R, Srisomboon Y, Iijima K, Maniak PJ, Tei R, Kobayashi T, Matsunaga M, Luo H, Masuda MY, O'Grady SM, Kita H. Cholinergic sensing of allergen exposure by airway epithelium promotes type 2 immunity in the lungs. J Allergy Clin Immunol 2024; 153:793-808.e2. [PMID: 38000698 PMCID: PMC10939907 DOI: 10.1016/j.jaci.2023.10.031] [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: 05/02/2023] [Revised: 09/26/2023] [Accepted: 10/09/2023] [Indexed: 11/26/2023]
Abstract
BACKGROUND Nonneuronal cells, including epithelial cells, can produce acetylcholine (ACh). Muscarinic ACh receptor antagonists are used clinically to treat asthma and other medical conditions; however, knowledge regarding the roles of ACh in type 2 immunity is limited. OBJECTIVE Our aim was to investigate the roles of epithelial ACh in allergic immune responses. METHODS Human bronchial epithelial (HBE) cells were cultured with allergen extracts, and their ACh production and IL-33 secretion were studied in vitro. To investigate immune responses in vivo, naive BALB/c mice were treated intranasally with different muscarinic ACh receptor antagonists and then exposed intranasally to allergens. RESULTS At steady state, HBE cells expressed cellular components necessary for ACh production, including choline acetyltransferase and organic cation transporters. Exposure to allergens caused HBE cells to rapidly release ACh into the extracellular medium. Pharmacologic or small-interfering RNA-based blocking of ACh production or autocrine action through the M3 muscarinic ACh receptors in HBE cells suppressed allergen-induced ATP release, calcium mobilization, and extracellular secretion of IL-33. When naive mice were exposed to allergens, ACh was quickly released into the airway lumen. A series of clinical M3 muscarinic ACh receptor antagonists inhibited allergen-induced IL-33 secretion and innate type 2 immune response in the mouse airways. In a preclinical murine model of asthma, an ACh receptor antagonist suppressed allergen-induced airway inflammation and airway hyperreactivity. CONCLUSIONS ACh is released quickly by airway epithelial cells on allergen exposure, and it plays an important role in type 2 immunity. The epithelial ACh system can be considered a therapeutic target in allergic airway diseases.
Collapse
Affiliation(s)
- Ryusuke Hayashi
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic, Scottsdale, Ariz; Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Yotesawee Srisomboon
- Department of Animal Science, University of Minnesota, St Paul, Minn; Department of Integrative Biology and Physiology, University of Minnesota, St Paul, Minn
| | - Koji Iijima
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic, Scottsdale, Ariz; Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Peter J Maniak
- Department of Animal Science, University of Minnesota, St Paul, Minn; Department of Integrative Biology and Physiology, University of Minnesota, St Paul, Minn
| | - Rinna Tei
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic, Scottsdale, Ariz; Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Takao Kobayashi
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic, Scottsdale, Ariz; Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Mayumi Matsunaga
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic, Scottsdale, Ariz; Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Huijun Luo
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic, Scottsdale, Ariz; Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Mia Y Masuda
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minn; Mayo Clinic Graduate School of Biomedical Sciences, Scottsdale, Ariz
| | - Scott M O'Grady
- Department of Animal Science, University of Minnesota, St Paul, Minn; Department of Integrative Biology and Physiology, University of Minnesota, St Paul, Minn
| | - Hirohito Kita
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic, Scottsdale, Ariz; Department of Medicine, Mayo Clinic, Scottsdale, Ariz; Department of Immunology, Mayo Clinic Rochester, Rochester, Minn; Department of Immunology, Mayo Clinic Arizona, Scottsdale, Ariz.
| |
Collapse
|
32
|
Yang F, Zhou L, Shen Y, Wang X, Fan X, Yang L. Multi-omics approaches for drug-response characterization in primary biliary cholangitis and autoimmune hepatitis variant syndrome. J Transl Med 2024; 22:214. [PMID: 38424613 PMCID: PMC10902991 DOI: 10.1186/s12967-024-05029-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/24/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Primary biliary cholangitis (PBC) and autoimmune hepatitis (AIH) variant syndrome (VS) exhibit a complex overlap of AIH features with PBC, leading to poorer prognoses than those with PBC or AIH alone. The biomarkers associated with drug response and potential molecular mechanisms in this syndrome have not been fully elucidated. METHODS Whole-transcriptome sequencing was employed to discern differentially expressed (DE) RNAs within good responders (GR) and poor responders (PR) among patients with PBC/AIH VS. Subsequent gene ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were conducted for the identified DE RNAs. Plasma metabolomics was employed to delineate the metabolic profiles distinguishing PR and GR groups. The quantification of immune cell profiles and associated cytokines was achieved through flow cytometry and immunoassay technology. Uni- and multivariable logistic regression analyses were conducted to construct a predictive model for insufficient biochemical response. The performance of the model was assessed by computing the area under the receiver operating characteristic (AUC) curve, sensitivity, and specificity. FINDINGS The analysis identified 224 differentially expressed (DE) mRNAs, 189 DE long non-coding RNAs, 39 DE circular RNAs, and 63 DE microRNAs. Functional pathway analysis revealed enrichment in lipid metabolic pathways and immune response. Metabolomics disclosed dysregulated lipid metabolism and identified PC (18:2/18:2) and PC (16:0/20:3) as predictors. CD4+ T helper (Th) cells, including Th2 cells and regulatory T cells (Tregs), were upregulated in the GR group. Pro-inflammatory cytokines (IFN-γ, TNF-α, IL-9, and IL-17) were downregulated in the GR group, while anti-inflammatory cytokines (IL-10, IL-4, IL-5, and IL-22) were elevated. Regulatory networks were constructed, identifying CACNA1H and ACAA1 as target genes. A predictive model based on these indicators demonstrated an AUC of 0.986 in the primary cohort and an AUC of 0.940 in the validation cohort for predicting complete biochemical response. CONCLUSION A combined model integrating genomic, metabolic, and cytokinomic features demonstrated high accuracy in predicting insufficient biochemical response in patients with PBC/AIH VS. Early recognition of individuals at elevated risk for insufficient response allows for the prompt initiation of additional treatments.
Collapse
Affiliation(s)
- Fan Yang
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, #37 Guoxue Road, Chengdu, 610041, Sichuan, China
| | - Leyu Zhou
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, #37 Guoxue Road, Chengdu, 610041, Sichuan, China
| | - Yi Shen
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, #37 Guoxue Road, Chengdu, 610041, Sichuan, China
| | - Xianglin Wang
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, #37 Guoxue Road, Chengdu, 610041, Sichuan, China
| | - Xiaoli Fan
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, #37 Guoxue Road, Chengdu, 610041, Sichuan, China.
| | - Li Yang
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, #37 Guoxue Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
33
|
Jacques C, Marchand F, Chatelais M, Floris I. Actives from the Micro-Immunotherapy Medicine 2LMIREG ® Reduce the Expression of Cytokines and Immune-Related Markers Including Interleukin-2 and HLA-II While Modulating Oxidative Stress and Mitochondrial Function. J Inflamm Res 2024; 17:1161-1181. [PMID: 38406323 PMCID: PMC10894519 DOI: 10.2147/jir.s445053] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/13/2024] [Indexed: 02/27/2024] Open
Abstract
Introduction Micro-immunotherapy (MI) is a therapeutic option employing low doses (LD) and ultra-low doses (ULD) of cytokines and immune factors to help the organism at modulating the immune responses. In an overpowering inflammatory context, this strategy may support the restoration of the body's homeostasis, as the active ingredients of MI medicines' (MIM) could boost or slow down the physiological functions of the immune cells. The aim of the study is to evaluate for the first time the in vitro anti-inflammatory properties of some actives employed by the MIM of interest in several human immune cell models. Methods In the first part of the study, the effects of the actives from the MIM of interest were assessed from a molecular standpoint: the expression of HLA-II, interleukin (IL)-2, and the secretion of several other cytokines were evaluated. In addition, as mitochondrial metabolism is also involved in the inflammatory processes, the second part of the study aimed at assessing the effects of these actives on the mitochondrial reactive oxygen species (ROS) production and on the mitochondrial membrane potential. Results We showed that the tested actives decreased the expression of HLA-DR and HLA-DP in IFN-γ-stimulated endothelial cells and in LPS-treated-M1-macrophages. The tested MIM slightly reduced the intracellular expression of IL-2 in CD4+ and CD8+ T-cells isolated from PMA/Iono-stimulated human PBMCs. Additionally, while the secretion of IL-2, IL-10, and IFN-γ was diminished, the treatment increased IL-6, IL-9, and IL-17A, which may correspond to a "Th17-like" secretory pattern. Interestingly, in PMA/Iono-treated PBMCs, we reported that the treatment reduced the ROS production in B-cells. Finally, in PMA/Iono-treated human macrophages, we showed that the treatment slightly protected the cells from early cell death/apoptosis. Discussion Overall, these results provide data about the molecular and functional anti-inflammatory effects of several actives contained in the tested MIM in immune-related cells, and their impact on two mitochondria-related processes.
Collapse
Affiliation(s)
- Camille Jacques
- Preclinical Research Department, Labo’Life France, Pescalis-Les Magnys, Moncoutant-sur-Sevre, 79320, France
| | | | | | - Ilaria Floris
- Preclinical Research Department, Labo’Life France, Pescalis-Les Magnys, Moncoutant-sur-Sevre, 79320, France
| |
Collapse
|
34
|
Simões R, Ribeiro AC, Dias R, Freitas V, Soares S, Pérez-Gregorio R. Unveiling the Immunomodulatory Potential of Phenolic Compounds in Food Allergies. Nutrients 2024; 16:551. [PMID: 38398875 PMCID: PMC10891931 DOI: 10.3390/nu16040551] [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: 01/08/2024] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Food allergies are becoming ever more prevalent around the world. This pathology is characterized by the breakdown of oral tolerance to ingested food allergens, resulting in allergic reactions in subsequent exposures. Due to the possible severity of the symptoms associated with this pathology, new approaches to prevent it and reduce associated symptoms are of utmost importance. In this framework, dietary phenolic compounds appear as a tool with a not fully explored potential. Some phenolic compounds have been pointed to with the ability to modulate food allergies and possibly reduce their symptoms. These compounds can modulate food allergies through many different mechanisms, such as altering the bioaccessibility and bioavailability of potentially immunogenic peptides, by modulating the human immune system and by modulating the composition of the human microbiome that resides in the oral cavity and the gastrointestinal tract. This review deepens the state-of-the-art of the modulation of these mechanisms by phenolic compounds. While this review shows clear evidence that dietary supplementation with foods rich in phenolic compounds might constitute a new approach to the management of food allergies, it also highlights the need for further research to delve into the mechanisms of action of these compounds and decipher systematic structure/activity relationships.
Collapse
Affiliation(s)
- Rodolfo Simões
- REQUIMTE/LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua Campo Alegre 687, s/n, 4169-007 Porto, Portugal
- Food and Health Omics Group, Food and Agroecology Institute, University of Vigo, Campus As Lagoas, s/n, 32004 Ourense, Spain
- Food and Health Omics Group, Department of Chemistry and Biochemistry, Galicia Sur Health Research Institute (IISGS), SERGAS-UVIGO, 32002 Ourense, Spain
| | - Ana Catarina Ribeiro
- REQUIMTE/LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua Campo Alegre 687, s/n, 4169-007 Porto, Portugal
| | - Ricardo Dias
- REQUIMTE/LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua Campo Alegre 687, s/n, 4169-007 Porto, Portugal
| | - Victor Freitas
- REQUIMTE/LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua Campo Alegre 687, s/n, 4169-007 Porto, Portugal
| | - Susana Soares
- REQUIMTE/LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua Campo Alegre 687, s/n, 4169-007 Porto, Portugal
| | - Rosa Pérez-Gregorio
- REQUIMTE/LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua Campo Alegre 687, s/n, 4169-007 Porto, Portugal
- Food and Health Omics Group, Food and Agroecology Institute, University of Vigo, Campus As Lagoas, s/n, 32004 Ourense, Spain
- Food and Health Omics Group, Department of Chemistry and Biochemistry, Galicia Sur Health Research Institute (IISGS), SERGAS-UVIGO, 32002 Ourense, Spain
| |
Collapse
|
35
|
Miron RJ, Estrin NE, Sculean A, Zhang Y. Understanding exosomes: Part 2-Emerging leaders in regenerative medicine. Periodontol 2000 2024; 94:257-414. [PMID: 38591622 DOI: 10.1111/prd.12561] [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: 02/04/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
Exosomes are the smallest subset of extracellular signaling vesicles secreted by most cells with the ability to communicate with other tissues and cell types over long distances. Their use in regenerative medicine has gained tremendous momentum recently due to their ability to be utilized as therapeutic options for a wide array of diseases/conditions. Over 5000 publications are currently being published yearly on this topic, and this number is only expected to dramatically increase as novel therapeutic strategies continue to be developed. Today exosomes have been applied in numerous contexts including neurodegenerative disorders (Alzheimer's disease, central nervous system, depression, multiple sclerosis, Parkinson's disease, post-traumatic stress disorders, traumatic brain injury, peripheral nerve injury), damaged organs (heart, kidney, liver, stroke, myocardial infarctions, myocardial infarctions, ovaries), degenerative processes (atherosclerosis, diabetes, hematology disorders, musculoskeletal degeneration, osteoradionecrosis, respiratory disease), infectious diseases (COVID-19, hepatitis), regenerative procedures (antiaging, bone regeneration, cartilage/joint regeneration, osteoarthritis, cutaneous wounds, dental regeneration, dermatology/skin regeneration, erectile dysfunction, hair regrowth, intervertebral disc repair, spinal cord injury, vascular regeneration), and cancer therapy (breast, colorectal, gastric cancer and osteosarcomas), immune function (allergy, autoimmune disorders, immune regulation, inflammatory diseases, lupus, rheumatoid arthritis). This scoping review is a first of its kind aimed at summarizing the extensive regenerative potential of exosomes over a broad range of diseases and disorders.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Nathan E Estrin
- Advanced PRF Education, Venice, Florida, USA
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Anton Sculean
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- Department of Oral Implantology, University of Wuhan, Wuhan, China
| |
Collapse
|
36
|
Kim HY, Jeong D, Kim JH, Chung DH. Innate Type-2 Cytokines: From Immune Regulation to Therapeutic Targets. Immune Netw 2024; 24:e6. [PMID: 38455467 PMCID: PMC10917574 DOI: 10.4110/in.2024.24.e6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 03/09/2024] Open
Abstract
The intricate role of innate type-2 cytokines in immune responses is increasingly acknowledged for its dual nature, encompassing both protective and pathogenic dimensions. Ranging from defense against parasitic infections to contributing to inflammatory diseases like asthma, fibrosis, and obesity, these cytokines intricately engage with various innate immune cells. This review meticulously explores the cellular origins of innate type-2 cytokines and their intricate interactions, shedding light on factors that amplify the innate type-2 response, including TSLP, IL-25, and IL-33. Recent advancements in therapeutic strategies, specifically the utilization of biologics targeting pivotal cytokines (IL-4, IL-5, and IL-13), are discussed, offering insights into both challenges and opportunities. Acknowledging the pivotal role of innate type-2 cytokines in orchestrating immune responses positions them as promising therapeutic targets. The evolving landscape of research and development in this field not only propels immunological knowledge forward but also holds the promise of more effective treatments in the future.
Collapse
Affiliation(s)
- Hye Young Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul 03760, Korea
| | - Dongjin Jeong
- Laboratory of Immune Regulation, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University Medical Research Center, Seoul 03080, Korea
| | - Ji Hyung Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Doo Hyun Chung
- Laboratory of Immune Regulation, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University Medical Research Center, Seoul 03080, Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
37
|
Nakano K, Whitehead GS, Lyons-Cohen MR, Grimm SA, Wilkinson CL, Izumi G, Livraghi-Butrico A, Cook DN, Nakano H. Chemokine CCL19 promotes type 2 T-cell differentiation and allergic airway inflammation. J Allergy Clin Immunol 2024; 153:487-502.e9. [PMID: 37956733 PMCID: PMC10922373 DOI: 10.1016/j.jaci.2023.10.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 10/06/2023] [Accepted: 10/12/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND Allergic asthma is driven largely by allergen-specific TH2 cells, which develop in regional lymph nodes on the interaction of naive CD4+ T cells with allergen-bearing dendritic cells that migrate from the lung. This migration event is dependent on CCR7 and its chemokine ligand, CCL21. However, is has been unclear whether the other CCR7 ligand, CCL19, has a role in allergic airway disease. OBJECTIVE This study sought to define the role of CCL19 in TH2 differentiation and allergic airway disease. METHODS Ccl19-deficient mice were studied in an animal model of allergic asthma. Dendritic cells or fibroblastic reticular cells from wild-type and Ccl19-deficient mice were cultured with naive CD4+ T cells, and cytokine production was measured by ELISA. Recombinant CCL19 was added to CD4+ T-cell cultures, and gene expression was assessed by RNA-sequencing and quantitative PCR. Transcription factor activation was assessed by flow cytometry. RESULTS Lungs of Ccl19-deficient mice had less allergic airway inflammation, reduced airway hyperresponsiveness, and less IL-4 and IL-13 production compared with lungs of Ccl19-sufficient animals. Naive CD4+ T cells cocultured with Ccl19-deficient dendritic cells or fibroblastic reticular cells produced lower amounts of type 2 cytokines than did T cells cocultured with their wild-type counterparts. Recombinant CCL19 increased phosphorylation of STAT5 and induced expression of genes associated with TH2 cell and IL-2 signaling pathways. CONCLUSIONS These results reveal a novel, TH2 cell-inducing function of CCL19 in allergic airway disease and suggest that strategies to block this pathway might help to reduce the incidence or severity of allergic asthma.
Collapse
Affiliation(s)
- Keiko Nakano
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Gregory S Whitehead
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Miranda R Lyons-Cohen
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Sara A Grimm
- Integrative Bioinformatics Support Group, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Christina L Wilkinson
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Gentaro Izumi
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | | | - Donald N Cook
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC.
| | - Hideki Nakano
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC.
| |
Collapse
|
38
|
Liu Q, Wang Y, Harpaz N. Coexisting Th1 and Th2 cytokines in patients with collagenous gastritis and implications for its pathogenesis. J Pediatr Gastroenterol Nutr 2024; 78:231-240. [PMID: 38374564 DOI: 10.1002/jpn3.12109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/28/2023] [Accepted: 12/02/2023] [Indexed: 02/21/2024]
Abstract
OBJECTIVES Collagenous gastritis (CG) is a rare cause of refractory dyspepsia and anemia that frequently affects children and young adults and whose histological hallmark is chronic mucosal inflammation with a subepithelial collagen band. The etiology remains obscure, and no established treatments exist. We investigated the pathogenesis of CG by determining the expression profiles of genes related to immunity and inflammation in index biopsies. METHODS Gastric biopsies from 10 newly diagnosed patients with CG were evaluated using the NanoString nCounter assay. Gastric biopsies from 14 normal individuals served as controls. The gene expression ratios for CG versus controls were determined in pooled samples and confirmed in individual samples by quantitative reverse transcription polymerase chain reaction. The results were compared with previously reported expression data from a cohort of patients with collagenous colitis, a colonic disorder with similar morphology, including subepithelial collagen band. RESULTS CG biopsies featured enhanced expression of key genes encoding both Th1 (IFNγ, TNF-α, IL-2, IL-10, IL-12A, IL-12B, and IL-18) and Th2 cytokines (IL-3, IL-4, IL-5, IL-6, and IL-13). In contrast, biopsies from patients with CC exhibited upregulated Th1 cytokines only. CONCLUSIONS We show in this first published gene expression profiling study that CG involves simultaneous upregulation of Th1 and Th2 cytokines. This finding is unique, contrasting with other types of chronic gastritis as well as with collagenous colitis, which shares the presence of a collagen band. Involvement of Th2 immunity in CG would support further investigation of potential dietary, environmental, or allergic factors to guide future therapeutic trials.
Collapse
Affiliation(s)
- Qingqing Liu
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yanping Wang
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology and Laboratory Medicine, Loyola University Health System, Maywood, Illinois, USA
| | - Noam Harpaz
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
39
|
Rafaqat S, Patoulias D, Behnoush AH, Sharif S, Klisic A. Interleukins: pathophysiological role in acute pancreatitis. Arch Med Sci 2024; 20:138-156. [PMID: 38414463 PMCID: PMC10895951 DOI: 10.5114/aoms/178183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/02/2024] [Indexed: 02/29/2024] Open
Abstract
Acute pancreatitis (AP) is a common inflammatory state characterized by a clinical course that can lead to serious local and extrapancreatic organ malfunction and failure. Interleukins (ILs) are biologically active glycoproteins primarily produced by macrophages and lymphocytes. According to the literature, there are many ILs. However, this article represents a summary of the role of ILs in AP, such as IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, and IL-20. The ways to modulate IL activity to reduce inflammation and improve outcomes in individuals with this condition are under investigation. Drugs that target specific ILs might be developed to mitigate the effects of AP.
Collapse
Affiliation(s)
- Saira Rafaqat
- Department of Zoology, Lahore College for Women University, Lahore, Punjab, Pakistan
| | - Dimitrios Patoulias
- Outpatient Department of Cardiometabolic Medicine, Second Department of Cardiology, Aristotle University of Thessaloniki, General Hospital “Hippokration”, Thessaloniki, Greece
| | | | - Saima Sharif
- Department of Zoology, Lahore College for Women University, Lahore, Punjab, Pakistan
| | - Aleksandra Klisic
- University of Montenegro-Faculty of Medicine, Podgorica, Montenegro
- Center for Laboratory Diagnostics, Primary Health Care Center, Podgorica, Montenegro
| |
Collapse
|
40
|
Sulaiman I, Okwuofu EO, Mohtarrudin N, Lim JCW, Stanslas J. An Andrographis paniculata Burm. Nees extract standardized for three main Andrographolides prevents house dust mite-induced airway inflammation, remodeling, and hyperreactivity by regulating Th1/Th2 gene expression in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117082. [PMID: 37652197 DOI: 10.1016/j.jep.2023.117082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Andrographis paniculata Burm. Nees (AP) is an herb used traditionally in Indian and Chinese traditional medicine for the treatment of various inflammatory and respiratory tract diseases. However, the anti-inflammatory potential of standardized Andrographis paniculata 50% ethanol extract (APEE50) in the murine model of asthma has not been investigated. AIM OF THE STUDY This study aimed to evaluate the protective anti-inflammatory potential and better understand the underlying mechanism of action of APEE50 in a clinically-relevant mouse asthma model. Thereafter, develop the ethanolic extract of AP as a supplement for asthma prophylaxis. MATERIALS AND METHOD APEE50 was prepared and standardized for AGP, NAG, and DDAG using a high-performance liquid chromatography system. Asthma was induced according to a 14-day house dust mite (HDM) induction protocol. The prophylactic potential of APEE50 (50 mg/kg - 200 mg/kg) was determined by assessing cardinal asthma features, which included BALF leukocyte and differential cell count, BALF cytokine assay, histology, gene expression, and airway hyperreactivity study. RESULTS APEE50 significantly inhibited HDM-induced airway eosinophilia and neutrophilia. In addition to decreased levels of IL-4, IL-5, IL-13, and eotaxin in bronchoalveolar fluid, APEE50 abrogated HDM-induced airway mucus over-secretion and airway hyper-responsiveness. Administration of APEE50 downregulated HDM-induced upregulation of the oxidative stress enzyme Duox1 (dual oxidase 1) and marginally induced Nfe2l2 (nuclear factor erythroid 2-related factor 2) gene expressions. Similarly, Th2-related (Serpinb2, Clca3a1, Il4 and Il13) and Muc5ac gene expression were significantly downregulated. CONCLUSION Prophylactic administration of APEE50 prevented the progression of HDM-induced asthmatic responses by down-regulating Th2 cytokine gene expression and oxidative stress level.
Collapse
Affiliation(s)
- Ibrahim Sulaiman
- Pharmacotherapeutic Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Emmanuel Oshiogwe Okwuofu
- Pharmacotherapeutic Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Norhafizah Mohtarrudin
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Jonathan Chee Woei Lim
- Pharmacotherapeutic Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Johnson Stanslas
- Pharmacotherapeutic Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.
| |
Collapse
|
41
|
Amoriello R, Memo C, Ballerini L, Ballerini C. The brain cytokine orchestra in multiple sclerosis: from neuroinflammation to synaptopathology. Mol Brain 2024; 17:4. [PMID: 38263055 PMCID: PMC10807071 DOI: 10.1186/s13041-024-01077-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/18/2024] [Indexed: 01/25/2024] Open
Abstract
The central nervous system (CNS) is finely protected by the blood-brain barrier (BBB). Immune soluble factors such as cytokines (CKs) are normally produced in the CNS, contributing to physiological immunosurveillance and homeostatic synaptic scaling. CKs are peptide, pleiotropic molecules involved in a broad range of cellular functions, with a pivotal role in resolving the inflammation and promoting tissue healing. However, pro-inflammatory CKs can exert a detrimental effect in pathological conditions, spreading the damage. In the inflamed CNS, CKs recruit immune cells, stimulate the local production of other inflammatory mediators, and promote synaptic dysfunction. Our understanding of neuroinflammation in humans owes much to the study of multiple sclerosis (MS), the most common autoimmune and demyelinating disease, in which autoreactive T cells migrate from the periphery to the CNS after the encounter with a still unknown antigen. CNS-infiltrating T cells produce pro-inflammatory CKs that aggravate local demyelination and neurodegeneration. This review aims to recapitulate the state of the art about CKs role in the healthy and inflamed CNS, with focus on recent advances bridging the study of adaptive immune system and neurophysiology.
Collapse
Affiliation(s)
- Roberta Amoriello
- International School for Advanced Studies (SISSA/ISAS), 34136, Trieste, Italy.
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, 50139, Florence, Italy.
| | - Christian Memo
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, 50139, Florence, Italy
| | - Laura Ballerini
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, 50139, Florence, Italy
| | - Clara Ballerini
- International School for Advanced Studies (SISSA/ISAS), 34136, Trieste, Italy.
| |
Collapse
|
42
|
Skacel J, Melichar B, Mohelnikova-Duchonova B, Lemstrova R. Hyperprogression on anti-PD-1 treatment. Is subsequent therapy feasible? A case report and review of the literature. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2023; 167:376-384. [PMID: 35703362 DOI: 10.5507/bp.2022.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/18/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Hyperprogressive disease (HPD) is a new phenomenon that has emerged in the immunotherapy era. HPD is defined as a rapid tumour growth with detrimental effect on the patient condition and disease course. The management and treatment following HPD is not defined. We present here the case report of patient with HPD and review of the literature on putative mechanisms of HPD and following disease management. METHODS AND RESULTS A 60-year old male patient with metastatic melanoma was indicated for systemic treatment with anti-programmed cell death (PD)-1 antibody. Rapid tumour growth and detrimental effect on the patient general condition after administration of a single dose of anti-PD-1 antibody met the criteria of HPD. The patient underwent the second line taxane-based chemotherapy with good tolerance and disease stabilization. The third line treatment with anti- cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) antibody ipilimumab was well tolerated and resulted in partial response. Re-challenge with anti-CTLA-4 antibody was feasible, but only with a modest clinical effect. CONCLUSION Prompt recognition of HPD and administration of salvage chemotherapy with taxane-based regimens may be crucial. HPD is rarely observed with ipilimumab treatment. Administration of ipilimumab as well as an ipilimumab re-challenge are feasible after HPD on anti-PD-1 antibodies. Investigation of new predictive biomarkers of HPD is warranted as well as new agents that potentiate the immune response in patients affected with this insidious complication.
Collapse
Affiliation(s)
- Jan Skacel
- Department of Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Zdravotniku 7, 779 00 Olomouc, Czech Republic
| | - Bohuslav Melichar
- Department of Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Zdravotniku 7, 779 00 Olomouc, Czech Republic
| | - Beatrice Mohelnikova-Duchonova
- Department of Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Zdravotniku 7, 779 00 Olomouc, Czech Republic
| | - Radmila Lemstrova
- Department of Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Zdravotniku 7, 779 00 Olomouc, Czech Republic
| |
Collapse
|
43
|
Agha-Hosseini F, Moosavi MS, Bahrami H. A Systematic Review of Interleukin-17 in Oral Lichen Planus: From Etiopathogenesis to Treatment. Clin Med Res 2023; 21:201-215. [PMID: 38296640 PMCID: PMC11149959 DOI: 10.3121/cmr.2023.1822] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/18/2023] [Accepted: 10/03/2023] [Indexed: 02/02/2024]
Abstract
Lichen planus (LP) is a chronic autoimmune disease of skin and mucous membranes. World Health Organization has announced oral lichen planus (OLP) as a premalignant lesion. The exact etiology of OLP remains unknown; however, different mechanisms may be involved in its immunopathogenesis. The upregulation of cytokines, chemokines, and adhesion molecules is consistent with a persistent and erratic immunological response to OLP-mediated antigens generated by oral keratinocytes and innate immune cells. These molecules attract T cells, and mast cells to the disease site and regulate complex interactions among cells that lead to death of keratinocytes, degradation of basement membrane, and chronicity of the disease. It is believed that CD8+ and CD4+ T helper 1 (Th1) cells are the main lymphocytes involved in this process, although recent evidence suggests implication of other T helper subgroups, such as Th23, Th17, and regulatory T cells (Tregs), proposing a more complex cellular immunity process to be involved in its pathogenesis. The emphasis of this research review is on the function of IL-17 in the pathophysiology of OLP and how current discoveries may point to future treatment strategies. This research protocol will follow Preferred Reporting Items for Systematic Reviews (PRISMA 2020) checklist. An electronic search was conducted in PubMed, Scopus, Google Scholar, Embase, and Cochrane databases for articles published from 1960 to June 2022. Based on the eligibility criteria, 21 articles were enrolled. In comparison to healthy controls, the findings of this review demonstrated greater expression of IL-17 and Th-17 in the blood, saliva, and tissues of OLP and LP patients. Additionally, there was a strong link between the relative levels of IL-17 and IL-23 expression. Treatment with monoclonal antibodies against Th-17/Tc-17, IL-12/IL-23, and IL-23 would result in significant long-term improvement of LP symptoms.
Collapse
Affiliation(s)
- Farzaneh Agha-Hosseini
- Professor, Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences and Department of Oral Medicine, Faculty of Dentistry, Tehran University of Medical Sciences, Tehran, Iran, The Academy of Medical Sciences Tehran, Iran, DDS, MSc, Fellowship of Research Biology. E-mail:
| | - Mahdieh-Sadat Moosavi
- Associate Professor, Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences and Department of Oral Medicine, Faculty of Dentistry, Tehran University of Medical Sciences, Tehran, Iran. E-mail:
| | - Hosna Bahrami
- Postgraduate student, Department of Oral & Maxillofacial Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Deng R, Li J, Wu H, Wang M. Mechanistic insight into the adjuvant effect of co-exposure to ultrafine carbon black and high humidity on allergic asthma. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2023; 45:9653-9667. [PMID: 37794280 DOI: 10.1007/s10653-023-01764-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/18/2023] [Indexed: 10/06/2023]
Abstract
Respiratory diseases continue to be a major global concern, with allergies and asthma often discussed as critical areas of study. While the role of environmental risk factors, such as non-allergenic pollutants and high humidity, in asthma induction is often mentioned, there is still a lack of thorough research on their co-exposure. This study aims to investigate the adjuvant effect of ultrafine carbon black (30-50 nm) and high humidity (70% relative humidity) on the induction of allergic asthma. A mouse model of asthma was established using ovalbumin, and airway hyperresponsiveness, remodeling, and inflammation were measured as the endpoint effects of asthma. The mediating role of the oxidative stress pathway and the transient receptor potential vanilloid 1 pathway in asthma induction was validated using pathway inhibitors vitamin E and capsaicin, respectively. Co-exposure to ultrafine carbon black and high humidity had a significant impact on metabolic pathways in the lung, including aminoacyl-tRNA biosynthesis, glycerophospholipid metabolism, and ATP-binding cassette transporters. However, administering vitamin E and capsaicin altered the effects of co-exposure on the lung metabolome. These results offer new insights into the health risk assessment of co-exposure to environmental risk factors and provide an important reference point for the prevention and treatment of allergic asthma.
Collapse
Affiliation(s)
- Rui Deng
- Joint International Research Laboratory of Green Buildings and Built Environments (Ministry of Education), School of Civil Engineering, Chongqing University, Chongqing, 400045, China.
| | - Jia Li
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Haiping Wu
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Mingpu Wang
- Joint International Research Laboratory of Green Buildings and Built Environments (Ministry of Education), School of Civil Engineering, Chongqing University, Chongqing, 400045, China
| |
Collapse
|
45
|
Huang D, Zhang Y, Yu Y, Jiang Y, Kong L, Ding Y, Shi Y, Gao Y. Long-term efficacy and safety of dupilumab for severe bullous pemphigoid: A prospective cohort study. Int Immunopharmacol 2023; 125:111157. [PMID: 37925949 DOI: 10.1016/j.intimp.2023.111157] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Bullous pemphigoid (BP) is a common subepidermal bullous disease. Dupilumab is a novel treatment for BP. However, its long-term efficacy and safety have not been demonstrated in prospective studies. OBJECTIVE Evaluate the long-term efficacy and safety of dupilumab in treating severe BP. METHODS Patients were divided into two groups: the methylprednisolone monotherapy group (M), and the methylprednisolone and dupilumab combination therapy group (D + M). This study consisted of two stages. The first stage focused on the initial treatment phase, where the early efficacy and safety was evaluated. The study then entered the 12-month maintenance treatment stage, where we assessed recurrence in both groups. Additionally, we evaluated the rate of healing of skin lesions, glucocorticoids burden and length of hospital stay and various laboratory test indicators. RESULTS After four weeks of treatment, the Bullous Pemphigoid Disease Area Index (BPDAI) and pruritus Numerical Rating Scale scores of the D + M group decreased significantly more than those of the M group. The median BPDAI at week 4 was 0 (range: 0.0-3.0) in the D + M group and 10.0 (5.0-12.0) in the M group (P < 0.001). Patients treated with dupilumab experienced a faster cessation of new blisters, quicker glucocorticoid reduction, shorter healing times, and shorter hospital stays (P < 0.001). Additionally, after two weeks of treatment, the levels of eosinophils and immunoglobulin E also decreased (P < 0.001). Follow-up studies further demonstrated that dupilumab monotherapy was associated with a lower recurrence rate. Notably, no serious adverse effects were observed in the study. CONCLUSIONS Our study provides evidence for the efficacy of dupilumab in the treatment of BP based on prospective studies. Additionally, our findings suggest that dupilumab can be considered a reliable single-agent maintenance treatment due to its good safety profile and lower relapse.
Collapse
Affiliation(s)
- Dawei Huang
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200443, China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai 200443, China
| | - Yuexin Zhang
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200443, China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai 200443, China
| | - Yingyuan Yu
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200443, China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai 200443, China
| | - Yuxiong Jiang
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200443, China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai 200443, China
| | - Luyang Kong
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200443, China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai 200443, China
| | - Yangfeng Ding
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200443, China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai 200443, China.
| | - Yuling Shi
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200443, China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai 200443, China.
| | - Yunlu Gao
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200443, China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai 200443, China.
| |
Collapse
|
46
|
Bensemmane L, Milliat F, Treton X, Linard C. Systemically delivered adipose stromal vascular fraction mitigates radiation-induced gastrointestinal syndrome by immunomodulating the inflammatory response through a CD11b + cell-dependent mechanism. Stem Cell Res Ther 2023; 14:325. [PMID: 37953266 PMCID: PMC10641938 DOI: 10.1186/s13287-023-03562-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND Stromal vascular fraction (SVF) treatment promoted the regeneration of the intestinal epithelium, limiting lethality in a mouse model of radiation-induced gastrointestinal syndrome (GIS). The SVF has a heterogeneous cell composition; the effects between SVF and the host intestinal immunity are still unknown. The specific role of the different cells contained in the SVF needs to be clarified. Monocytes-macrophages have a crucial role in repair and monocyte recruitment and activation are orchestrated by the chemokine receptors CX3CR1 and CCR2. METHODS Mice exposed to abdominal radiation (18 Gy) received a single intravenous injection of SVF (2.5 × 106 cells), obtained by enzymatic digestion of inguinal fat tissue, on the day of irradiation. Intestinal immunity and regeneration were evaluated by flow cytometry, RT-PCR and histological analyses. RESULTS Using flow cytometry, we showed that SVF treatment modulated intestinal monocyte differentiation at 7 days post-irradiation by very early increasing the CD11b+Ly6C+CCR2+ population in the intestine ileal mucosa and accelerating the phenotype modification to acquire CX3CR1 in order to finally restore the F4/80+CX3CR1+ macrophage population. In CX3CR1-depleted mice, SVF treatment fails to mature the Ly6C-MCHII+CX3CR1+ population, leading to a macrophage population deficit associated with proinflammatory environment maintenance and defective intestinal repair; this impaired SVF efficiency on survival. Consistent with a CD11b+ being involved in SVF-induced intestinal repair, we showed that SVF-depleted CD11b+ treatment impaired F4/80+CX3CR1+macrophage pool restoration and caused loss of anti-inflammatory properties, abrogating stem cell compartment repair and survival. CONCLUSIONS These data showed that SVF treatment mitigates the GIS-involving immunomodulatory effect. Cooperation between the monocyte in SVF and the host monocyte defining the therapeutic properties of the SVF is necessary to guarantee the effective action of the SVF on the GIS.
Collapse
Affiliation(s)
- Lydia Bensemmane
- PSE-SANTE/SERAMED/LRMed, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), 92260, Fontenay-Aux-Roses, France
| | - Fabien Milliat
- PSE-SANTE/SERAMED/LRMed, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), 92260, Fontenay-Aux-Roses, France
| | | | - Christine Linard
- PSE-SANTE/SERAMED/LRMed, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), 92260, Fontenay-Aux-Roses, France.
| |
Collapse
|
47
|
Bernstein ZJ, Shenoy A, Chen A, Heller NM, Spangler JB. Engineering the IL-4/IL-13 axis for targeted immune modulation. Immunol Rev 2023; 320:29-57. [PMID: 37283511 DOI: 10.1111/imr.13230] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 05/19/2023] [Indexed: 06/08/2023]
Abstract
The structurally and functionally related interleukin-4 (IL-4) and IL-13 cytokines play pivotal roles in shaping immune activity. The IL-4/IL-13 axis is best known for its critical role in T helper 2 (Th2) cell-mediated Type 2 inflammation, which protects the host from large multicellular pathogens, such as parasitic helminth worms, and regulates immune responses to allergens. In addition, IL-4 and IL-13 stimulate a wide range of innate and adaptive immune cells, as well as non-hematopoietic cells, to coordinate various functions, including immune regulation, antibody production, and fibrosis. Due to its importance for a broad spectrum of physiological activities, the IL-4/IL-13 network has been targeted through a variety of molecular engineering and synthetic biology approaches to modulate immune behavior and develop novel therapeutics. Here, we review ongoing efforts to manipulate the IL-4/IL-13 axis, including cytokine engineering strategies, formulation of fusion proteins, antagonist development, cell engineering approaches, and biosensor design. We discuss how these strategies have been employed to dissect IL-4 and IL-13 pathways, as well as to discover new immunotherapies targeting allergy, autoimmune diseases, and cancer. Looking ahead, emerging bioengineering tools promise to continue advancing fundamental understanding of IL-4/IL-13 biology and enabling researchers to exploit these insights to develop effective interventions.
Collapse
Affiliation(s)
- Zachary J Bernstein
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anjali Shenoy
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amy Chen
- Department of Molecular and Cellular Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nicola M Heller
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
- Division of Allergy and Clinical Immunology, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jamie B Spangler
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Sidney Kimmel Cancer Center, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
48
|
Yuan Q, Yang W, Zhang X. Immune cells in pemphigus vulgaris and bullous Pemphigoid: From pathogenic roles to targeting therapies. Int Immunopharmacol 2023; 123:110694. [PMID: 37523970 DOI: 10.1016/j.intimp.2023.110694] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/27/2023] [Accepted: 07/19/2023] [Indexed: 08/02/2023]
Abstract
Pemphigus vulgaris (PV) and bullous pemphigoid (BP) are two major subtypes of autoimmune bullous diseases (AIBD), characterized by blisters and erosions of skin and/or mucous membranes with dysregulated immune activity. Current literature established that T and B cells are the main executors of PV and BP. Emerging evidence revealed that macrophages and related cytokines also contribute to these diseases. While the role of lymphocytes on PV and BP is well established, the definitive functions of macrophages in disease progression are not fully understood. Furthermore, current status of clinical trials targeting immune cells is poorly recapitulated in PV and BP. In this review, we summarized current knowledge in this rapidly advancing field, with emphasis on the individual functions of immune cells and their interactions, as well as ongoing clinical trials targeting immune cells, to provide novel insights in mechanistic understanding and clinical management of PV and BP.
Collapse
Affiliation(s)
- Qiuyun Yuan
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Wanchun Yang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Xuefeng Zhang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan Province, China.
| |
Collapse
|
49
|
Minskaia E, Maimaris J, Jenkins P, Albuquerque AS, Hong Y, Eleftheriou D, Gilmour KC, Grace R, Moreira F, Grimbacher B, Morris EC, Burns SO. Autosomal Dominant STAT6 Gain of Function Causes Severe Atopy Associated with Lymphoma. J Clin Immunol 2023; 43:1611-1622. [PMID: 37316763 PMCID: PMC10499697 DOI: 10.1007/s10875-023-01530-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 05/29/2023] [Indexed: 06/16/2023]
Abstract
The transcription factor STAT6 (Signal Transducer and Activator of Transcription 6) is a key regulator of Th2 (T-helper 2) mediated allergic inflammation via the IL-4 (interleukin-4) JAK (Janus kinase)/STAT signalling pathway. We identified a novel heterozygous germline mutation STAT6 c.1255G > C, p.D419H leading to overactivity of IL-4 JAK/STAT signalling pathway, in a kindred affected by early-onset atopic dermatitis, food allergy, eosinophilic asthma, anaphylaxis and follicular lymphoma. STAT6 D419H expression and functional activity were compared with wild type STAT6 in transduced HEK293T cells and to healthy control primary skin fibroblasts and peripheral blood mononuclear cells (PBMC). We observed consistently higher STAT6 levels at baseline and higher STAT6 and phosphorylated STAT6 following IL-4 stimulation in D419H cell lines and primary cells compared to wild type controls. The pSTAT6/STAT6 ratios were unchanged between D419H and control cells suggesting that elevated pSTAT6 levels resulted from higher total basal STAT6 expression. The selective JAK1/JAK2 inhibitor ruxolitinib reduced pSTAT6 levels in D419H HEK293T cells and patient PBMC. Nuclear staining demonstrated increased STAT6 in patient fibroblasts at baseline and both STAT6 and pSTAT6 after IL-4 stimulation. We also observed higher transcriptional upregulation of downstream genes (XBP1 and EPAS1) in patient PBMC. Our study confirms STAT6 gain of function (GOF) as a novel monogenetic cause of early onset atopic disease. The clinical association of lymphoma in our kindred, along with previous data linking somatic STAT6 D419H mutations to follicular lymphoma suggest that patients with STAT6 GOF disease may be at higher risk of lymphomagenesis.245 words.
Collapse
Affiliation(s)
- Ekaterina Minskaia
- University College London Institute of Immunity and Transplantation, London, UK
| | - Jesmeen Maimaris
- University College London Institute of Immunity and Transplantation, London, UK.
- Department of Immunology, Royal Free London NHS Foundation Trust, London, UK.
| | - Persephone Jenkins
- University College London Institute of Immunity and Transplantation, London, UK
| | | | - Ying Hong
- Inflammation and Rheumatology Section, University College London Institute of Child Health, London, UK
| | - Despina Eleftheriou
- Inflammation and Rheumatology Section, University College London Institute of Child Health, London, UK
- Rheumatology Department, Great Ormond Street Hospital National Health Service (NHS) Foundation Trust, London, UK
| | - Kimberly C Gilmour
- Clinical Immunology Laboratory, Great Ormond Street Hospital of Children NHS Foundation Trust and NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Richard Grace
- Department of Haematology, East Sussex Healthcare NHS Trust, Saint Leonards-on-sea, UK
| | - Fernando Moreira
- Department of Immunology, Royal Free London NHS Foundation Trust, London, UK
| | - Bodo Grimbacher
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Emma C Morris
- University College London Institute of Immunity and Transplantation, London, UK
- Department of Immunology, Royal Free London NHS Foundation Trust, London, UK
| | - Siobhan O Burns
- University College London Institute of Immunity and Transplantation, London, UK
- Department of Immunology, Royal Free London NHS Foundation Trust, London, UK
| |
Collapse
|
50
|
Rochman Y, Kotliar M, Ben-Baruch Morgenstern N, Barski A, Wen T, Rothenberg ME. TSLP shapes the pathogenic responses of memory CD4 + T cells in eosinophilic esophagitis. Sci Signal 2023; 16:eadg6360. [PMID: 37699081 PMCID: PMC10602003 DOI: 10.1126/scisignal.adg6360] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 08/23/2023] [Indexed: 09/14/2023]
Abstract
The cytokine thymic stromal lymphopoietin (TSLP) mediates type 2 immune responses, and treatments that interfere with TSLP activity are in clinical use for asthma. Here, we investigated whether TSLP contributes to allergic inflammation by directly stimulating human CD4+ T cells and whether this process is operational in eosinophilic esophagitis (EoE), a disease linked to variants in TSLP. We showed that about 10% of esophageal-derived memory CD4+ T cells from individuals with EoE and less than 3% of cells from control individuals expressed the receptor for TSLP and directly responded to TSLP, as determined by measuring the phosphorylation of STAT5, a transcription factor activated downstream of TSLP stimulation. Accordingly, increased numbers of TSLP-responsive memory CD4+ T cells were present in the circulation of individuals with EoE. TSLP increased the proliferation of CD4+ T cells, enhanced type 2 cytokine production, induced the increased abundance of its own receptor, and modified the expression of 212 genes. The epigenetic response to TSLP was associated with an enrichment in BATF and IRF4 chromatin-binding sites, and these transcription factors were induced by TSLP, providing a feed-forward loop. The numbers of circulating and esophageal CD4+ T cells responsive to TSLP correlated with the numbers of esophageal eosinophils, supporting a potential functional role for TSLP in driving the pathogenesis of EoE and providing the basis for a blood-based diagnostic test based on the extent of TSLP-induced STAT5 phosphorylation in circulating CD4+ T cells. These findings highlight the potential therapeutic value of TSLP inhibitors for the treatment of EoE.
Collapse
Affiliation(s)
- Yrina Rochman
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Michael Kotliar
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Netali Ben-Baruch Morgenstern
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Artem Barski
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
- Division of Human Genetics, Department of Pediatrics Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Ting Wen
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Marc E. Rothenberg
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|