1
|
Vinh DC. Human immunity to fungal infections. J Exp Med 2025; 222:e20241215. [PMID: 40232283 PMCID: PMC11998751 DOI: 10.1084/jem.20241215] [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: 12/02/2024] [Revised: 01/07/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
Fungi increasingly threaten health globally. Mycoses range from life-threatening, often iatrogenic conditions, to enigmatic syndromes occurring without apparent immunosuppression. Despite some recent advances in antifungal drug development, complementary therapeutic strategies are essential for addressing these opportunistic pathogens. One promising avenue is leveraging host immunity to combat fungal infections; this necessitates deeper understanding of the molecular immunology of human fungal susceptibility to differentiate beneficial versus harmful immunopathological responses. Investigating human models of fungal diseases in natural settings, particularly through genetic immunodeficiencies and ethnographic-specific genetic vulnerabilities, reveals crucial immune pathways essential for fighting various yeasts and molds. This review highlights the diversity in intrinsic fungal susceptibility across individuals and populations, through genetic- and autoantibody-mediated processes, complementing previous principles learned from animal studies and iatrogenic contexts. Improved understanding of human immunity to fungal diseases will facilitate the development of host-directed immunotherapies and targeted public health interventions, paving the way for precision medicine in fungal disease management.
Collapse
Affiliation(s)
- Donald C. Vinh
- Department of Medicine (Division of Infectious Diseases), McGill University Health Center, Montreal, Canada
- Department of OptiLab (Division of Medical Microbiology, Division of Molecular Genetics-Immunology), McGill University Health Center, Montreal, Canada
- Department of Human Genetics, McGill University, Montreal, Canada
- Center of Reference for Genetic Research in Infection and Immunity, McGill University Health Center Research Institute, Montreal, Canada
| |
Collapse
|
2
|
Wang R, Wu D, Wang C, Livingston A, Wu X, Liu M, Yang XO. Platelet-Sourced TGF-β Promotes Th17 Responses and Enhances Airway Neutrophilia. Biomolecules 2025; 15:482. [PMID: 40305199 PMCID: PMC12024734 DOI: 10.3390/biom15040482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 05/02/2025] Open
Abstract
Microbial, especially fungal, sensitization has been associated with the development and exacerbation of treatment-refractory neutrophilic asthma. Among the airway-inhabiting fungi, Aspergillus fumigatus and Candida albicans are the dominant species that elicit protective T helper (Th) 17 and other T cell responses, contributing to airway neutrophilia and steroid resistance. However, it is not fully understood how fungal airway colonization impacts the immunopathogenesis of asthma. Here, we used a neutrophilic asthma model induced by C. albicans to study the immune regulation of this disease. We found that intranasal administration of C. albicans induced platelet infiltration into the lung. Platelet-expressed latent TGF-β could be activated specifically by Th17 cells and drive the commitment, maintenance, and expansion of Th17 cells. In Candida-induced asthma, an adoptive transfer of platelets enhanced Th17 responses, increasing airway neutrophil influx. Thus, managing airway mycobiota and reducing platelet intrapulmonary infiltration may serve as a promising interventional approach.
Collapse
Affiliation(s)
- Ruoning Wang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (R.W.); (D.W.); (A.L.); (X.W.)
| | - Dandan Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (R.W.); (D.W.); (A.L.); (X.W.)
| | - Chunqing Wang
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (C.W.); (M.L.)
| | - Amanda Livingston
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (R.W.); (D.W.); (A.L.); (X.W.)
| | - Xiang Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (R.W.); (D.W.); (A.L.); (X.W.)
- Department of Parasitology, School of Basic Medical Sciences, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (C.W.); (M.L.)
| | - Xuexian O. Yang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (R.W.); (D.W.); (A.L.); (X.W.)
| |
Collapse
|
3
|
Vuscan P, Röring RJ, Kischkel B, Tintoré M, Cuñé J, de Lecea C, Joosten LAB, Netea MG. Effect of Saccharomyces cerevisiae β-glucan on the T helper cytokine profile. Cytokine 2025; 187:156871. [PMID: 39889564 DOI: 10.1016/j.cyto.2025.156871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/27/2024] [Accepted: 01/13/2025] [Indexed: 02/03/2025]
Abstract
ABBi16 is a high-complexity blend of β-1,3/1,6-glucans from Saccharomyces cerevisiae with strong immunomodulatory activities, that have been recently shown to support anti-tumoral immune responses through the induction of trained immunity. Whether ABBi16 also modulates the balance between the various T helper (Th) lymphocyte responses is not known. Here, we show that ABBi16 induces Th1 responses, as indicated by stimulation of IFNγ and TNF production by human peripheral blood mononuclear cells (PBMCs). Moreover, the elevated secretion of IL-10 and IL-22 suggests a potential regulatory response of the Th1/Th2/Th17 balance. Co-stimulating PBMCs with ABBi16 alongside Bacille Calmette-Guerin (BCG), IL-1beta + IL-23, and IL-12 + IL-18 cytokine combinations further enhanced Th1 polarization and IL-22 induction, hinting at an additive effect of β-glucan on both Th1 and regulatory Th17 immune responses. ABBi16 did not induce IL-17 production, the prototype pro-inflammatory product of Th17 responses, suggesting that it can be safely used as an oral supplement in patients with autoimmune conditions. These results highlight the potential of ABBi16 to regulate the Th1/Th2/Th17 balance toward antimicrobial and regulatory effects.
Collapse
Affiliation(s)
- Patricia Vuscan
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rutger J Röring
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands; Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Brenda Kischkel
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Maria Tintoré
- AB Biotek Human Nutrition and Health, Barcelona, Spain
| | - Jordi Cuñé
- AB Biotek Human Nutrition and Health, Barcelona, Spain
| | | | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands; Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands; Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Germany.
| |
Collapse
|
4
|
Begagic E, Vranic S, Sominanda A. The role of interleukin 17 in cancer: a systematic review. Carcinogenesis 2025; 46:bgae079. [PMID: 39673782 DOI: 10.1093/carcin/bgae079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/08/2024] [Accepted: 12/13/2024] [Indexed: 12/16/2024] Open
Abstract
Interleukin 17 (IL17) is a cytokine involved in immune regulation and has been increasingly recognized for its role in cancer progression. This systematic review aims to integrate data on IL17's role in various tumors to better understand its implications for cancer prognosis and treatment. The review included 105 studies (27.6% experimental and 72.4% clinical). Clinical studies involved 9266 patients: 31.2% males, 60.0% females, and 8.8% with undefined gender. IL17A and IL17 were the most studied subtypes (36.2% and 33.3%, respectively). Breast cancer (26.7%), colorectal carcinoma (13.3%), and hematologic malignancies (10.5%) were the most researched neoplasms. IL17A promoted tumor growth in breast cancer and correlated with poor outcomes in colorectal, breast, and lung cancers. IL17 also played a significant role in immune modulation in gliomas and other tumors. IL17A significantly influences tumor growth and prognosis across various cancers, with notable roles in immune modulation and poor outcomes in multiple cancer types.
Collapse
Affiliation(s)
- Emir Begagic
- Department of Neurosurgery, Cantonal Hospital Zenica, Crkvice 67, 72000 Zenica, Bosnia and Herzegovina
| | - Semir Vranic
- Department of Pathology, College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Ajith Sominanda
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| |
Collapse
|
5
|
Palacios A, Kumar A, Caliwag FMC, Becerril-Garcia MA. Neonatal Immunity to Candida: Current Understanding and Contributions of Murine Models. Crit Rev Immunol 2025; 45:63-76. [PMID: 39976518 DOI: 10.1615/critrevimmunol.2024055053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Neonatal candidiasis poses significant clinical challenges due to its potential for severe morbidity and mortality in vulnerable infants. Due to their underdeveloped immune system, neonates are at a higher risk for infections caused by Candida species. They can vary from mild to severe, including penetrating deep tissues, bloodstream spread, and dissemination to organs. The immune system of newborns is marked by a limited innate immune response, with lower levels of pro-inflammatory cytokines. Adaptive immunity, important for lasting protection, also experiences delayed maturation with weakened Th1 and Th17 responses. These shortcomings result in a higher vulnerability to Candida infections during infancy. Murine models have been crucial in understanding the reasons behind this susceptibility. These models assist in examining how different immune elements, like neutrophils, macrophages, and T cells, and their interactions are involved in Candida infections. Moreover, they offer an understanding of how early-life exposure to Candida affects immune responses and may aid in developing possible therapeutic plans. In this article we review current results from research to provide a thorough summary and critical insights into neonatal immune response to Candida, highlighting the importance of using murine models in this field of study. Understanding these immune dynamics is essential for creating specific treatments and preventive strategies to prevent newborns from Candida infections, ultimately improving neonatal health outcomes.
Collapse
Affiliation(s)
| | - Ajay Kumar
- Isra University Faculty of Medicine and Allied Medical Sciences, Hyderabad, Sindh Pakistan
| | | | - Miguel A Becerril-Garcia
- Universidad Autónoma de Nuevo León School of Medicine, Department of Microbiology, Monterrey, Nuevo León México
| |
Collapse
|
6
|
Martínez-López R, Molero G, Parra-Giraldo CM, Cabeza MS, Castejón G, García-Durán C, Clemente LF, Hernáez ML, Gil C, Monteoliva L. From High Protection to Lethal Effect: Diverse Outcomes of Immunization Against Invasive Candidiasis with Different Candida albicans Extracellular Vesicles. Int J Mol Sci 2024; 26:244. [PMID: 39796100 PMCID: PMC11720215 DOI: 10.3390/ijms26010244] [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: 11/15/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Extracellular vesicles (EVs) from Candida albicans can elicit immune responses, positioning them as promising acellular vaccine candidates. We characterized EVs from an avirulent C. albicans cell wall mutant (ecm33Δ) and evaluated their protective potential against invasive candidiasis. EVs from the yeast (YEVs) and hyphal (HEVs) forms of the SC5314 wild-type strain were also tested, yielding high survival rates with SC5314 YEV (91%) and ecm33 YEV immunization (64%). Surprisingly, HEV immunization showed a dual effect, resulting in 36% protection but also causing premature death in some mice. Proteomic analyses revealed distinct profiles among the top 100 proteins in the different EVs, which may explain these effects: a shared core of 50 immunogenic proteins such as Pgk1, Cdc19, and Fba1; unique, relevant immunogenic proteins in SC5314 YEVs; and proteins linked to pathogenesis, like Ece1 in SC5314 HEVs. Sera from SC5314 YEV-immunized mice showed the highest IgG2a titers and moderate IL-17, IFN-γ, and TNF-α levels, indicating the importance of both humoral and cellular responses for protection. These findings highlight the distinct immunogenic properties of C. albicans EVs, suggesting their potential in acellular vaccine development while emphasizing the need to carefully evaluate pathogenic risks associated with certain EVs.
Collapse
Affiliation(s)
- Raquel Martínez-López
- Department of Microbiology and Parasitology, Pharmacy Faculty at Complutense University of Madrid, 28040 Madrid, Spain; (R.M.-L.); (G.M.); (C.M.P.-G.); (G.C.); (C.G.-D.); (C.G.)
| | - Gloria Molero
- Department of Microbiology and Parasitology, Pharmacy Faculty at Complutense University of Madrid, 28040 Madrid, Spain; (R.M.-L.); (G.M.); (C.M.P.-G.); (G.C.); (C.G.-D.); (C.G.)
| | - Claudia Marcela Parra-Giraldo
- Department of Microbiology and Parasitology, Pharmacy Faculty at Complutense University of Madrid, 28040 Madrid, Spain; (R.M.-L.); (G.M.); (C.M.P.-G.); (G.C.); (C.G.-D.); (C.G.)
| | - Matías Sebastián Cabeza
- Mycology and Molecular Diagnostics Laboratory, Biochemistry and Biological Science Faculty, Nacional del Litoral University, Santa Fe 3000, Argentina;
| | - Guillermo Castejón
- Department of Microbiology and Parasitology, Pharmacy Faculty at Complutense University of Madrid, 28040 Madrid, Spain; (R.M.-L.); (G.M.); (C.M.P.-G.); (G.C.); (C.G.-D.); (C.G.)
| | - Carmen García-Durán
- Department of Microbiology and Parasitology, Pharmacy Faculty at Complutense University of Madrid, 28040 Madrid, Spain; (R.M.-L.); (G.M.); (C.M.P.-G.); (G.C.); (C.G.-D.); (C.G.)
| | - Luis Felipe Clemente
- Proteomics Facility, Complutense University of Madrid, 28040 Madrid, Spain; (L.F.C.); (M.L.H.)
| | - María Luisa Hernáez
- Proteomics Facility, Complutense University of Madrid, 28040 Madrid, Spain; (L.F.C.); (M.L.H.)
| | - Concha Gil
- Department of Microbiology and Parasitology, Pharmacy Faculty at Complutense University of Madrid, 28040 Madrid, Spain; (R.M.-L.); (G.M.); (C.M.P.-G.); (G.C.); (C.G.-D.); (C.G.)
- Proteomics Facility, Complutense University of Madrid, 28040 Madrid, Spain; (L.F.C.); (M.L.H.)
| | - Lucía Monteoliva
- Department of Microbiology and Parasitology, Pharmacy Faculty at Complutense University of Madrid, 28040 Madrid, Spain; (R.M.-L.); (G.M.); (C.M.P.-G.); (G.C.); (C.G.-D.); (C.G.)
| |
Collapse
|
7
|
Penninger P, Brezovec H, Tsymala I, Teufl M, Phan-Canh T, Bitencourt T, Brinkmann M, Glaser W, Ellmeier W, Bonelli M, Kuchler K. HDAC1 fine-tunes Th17 polarization in vivo to restrain tissue damage in fungal infections. Cell Rep 2024; 43:114993. [PMID: 39580799 DOI: 10.1016/j.celrep.2024.114993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/13/2024] [Accepted: 11/04/2024] [Indexed: 11/26/2024] Open
Abstract
Histone deacetylases (HDACs) contribute to shaping many aspects of T cell lineage functions in anti-infective surveillance; however, their role in fungus-specific immune responses remains poorly understood. Using a T cell-specific deletion of HDAC1, we uncover its critical role in limiting polarization toward Th17 by restricting expression of the cytokine receptors gp130 and transforming growth factor β receptor 2 (TGF-βRII) in a fungus-specific manner, thus limiting Stat3 and Smad2/3 signaling. Controlled release of interleukin-17A (IL-17A) and granulocyte-macrophage colony-stimulating factor (GM-CSF) is vital to minimize apoptotic processes in renal tubular epithelial cells in vitro and in vivo. Consequently, animals harboring excess Th17-polarized HDCA1-deficient CD4+ T cells develop increased kidney pathology upon invasive Candida albicans infection. Importantly, pharmacological inhibition of class I HDACs similarly increased IL-17A release by both mouse and human CD4+ T cells. Collectively, this work shows that HDAC1 controls T cell polarization, thus playing a critical role in the antifungal immune defense and infection outcomes.
Collapse
Affiliation(s)
- Philipp Penninger
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030, Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Dr.-Bohr-Gasse 9, 1030, Vienna, Austria
| | - Helena Brezovec
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030, Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Dr.-Bohr-Gasse 9, 1030, Vienna, Austria
| | - Irina Tsymala
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030, Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Dr.-Bohr-Gasse 9, 1030, Vienna, Austria
| | - Magdalena Teufl
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030, Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Dr.-Bohr-Gasse 9, 1030, Vienna, Austria
| | - Trinh Phan-Canh
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030, Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Dr.-Bohr-Gasse 9, 1030, Vienna, Austria
| | - Tamires Bitencourt
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030, Vienna, Austria; CCRI - St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Marie Brinkmann
- Medical University of Vienna, Division of Rheumatology, Department of Internal Medicine III, 1090 Vienna, Austria
| | - Walter Glaser
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030, Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Dr.-Bohr-Gasse 9, 1030, Vienna, Austria
| | - Wilfried Ellmeier
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, 1090 Vienna, Austria
| | - Michael Bonelli
- Medical University of Vienna, Division of Rheumatology, Department of Internal Medicine III, 1090 Vienna, Austria
| | - Karl Kuchler
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030, Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Dr.-Bohr-Gasse 9, 1030, Vienna, Austria.
| |
Collapse
|
8
|
Sha S, Gao H, Zeng H, Chen F, Kang J, Jing Y, Liu X, Xu B. Adherent-invasive Escherichia coli LF82 disrupts the tight junctions of Caco-2 monolayers. Arab J Gastroenterol 2024; 25:383-389. [PMID: 39069423 DOI: 10.1016/j.ajg.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/14/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND AND STUDY AIMS Adherent invasive Escherichia coli (AIEC) are enriched in IBD (inflammatory bowel disease) patients, but the role and mechanism of AIEC in the intestinal epithelial barrier is poorly defined. We evaluated the role of the AIEC strain E. coli LF82 in vitro and investigated the role of Th17 in this process. MATERIAL AND METHODS After coincubation with AIEC, the epithelial barrier integrity was monitored by epithelial resistance measurements. The permeability of the barrier was evaluated by TEER (trans-epithelial electrical resistance) and mucosal-to-serosal flux rate. The presence of interepithelial tight junction proteins ZO-1 and Claudin-1 were determined by immunofluorescence and western blot analysis. Cytokines in the cell culture supernatant were assayed by enzyme-linked immunosorbent assay (ELISA). RESULTS AIEC infection decreased TEER and increased the mucosal-to-serosal flux rate of Lucifer yellow in the intestinal barrier model in a time- and dose-dependent manner. AIEC infection decreased the expression and changed the distribution of ZO-1 and claudin-1. It also induced the secretion of cytokines such as TNF-α and IL-17. CONCLUSION AIEC strain E. coli LF82 increased the permeability and disrupted the tight junctions of the intestinal epithelial barrier, revealing that AIEC plays an aggravative role in the inflammatory response.
Collapse
Affiliation(s)
- Sumei Sha
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Key Laboratory of Gastrointestinal Motility Disorders, Clinical Research Center of Gastrointestinal Diseases, Xi'an, Shaanxi Province 710004, PR China
| | - Huijun Gao
- Department of Gastroenterology, No. 988 Hospital of Joint Logistic Support Force, Jiaozuo, Henan Province 454000, PR China
| | - Hong Zeng
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Key Laboratory of Gastrointestinal Motility Disorders, Clinical Research Center of Gastrointestinal Diseases, Xi'an, Shaanxi Province 710004, PR China; Department of Gastroenterology, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, Shaanxi Province 710000, PR China
| | - Fenrong Chen
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Key Laboratory of Gastrointestinal Motility Disorders, Clinical Research Center of Gastrointestinal Diseases, Xi'an, Shaanxi Province 710004, PR China
| | - Junxiu Kang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Key Laboratory of Gastrointestinal Motility Disorders, Clinical Research Center of Gastrointestinal Diseases, Xi'an, Shaanxi Province 710004, PR China
| | - Yan Jing
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Key Laboratory of Gastrointestinal Motility Disorders, Clinical Research Center of Gastrointestinal Diseases, Xi'an, Shaanxi Province 710004, PR China
| | - Xin Liu
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Key Laboratory of Gastrointestinal Motility Disorders, Clinical Research Center of Gastrointestinal Diseases, Xi'an, Shaanxi Province 710004, PR China.
| | - Bin Xu
- Tangdu Hospital of the Air Force Medical University, Xi'an, Shaanxi, PR China; Department of General Surgery, the Chenggong Hospital Affiliated to Xiamen University (Central Hospital of the 73th Chinese People's Liberation Army), Xiamen Fujian Province 361003, PR China.
| |
Collapse
|
9
|
Schwartzman S, Puig L, Cohen AD, Khattri S, Jossart C, Diaz C, Garrelts A, Ngantcha M, Eberhart N, Eleftheriadi A, Tangsirisap N, Schuster C, Gottlieb AB. Treatment-emergent Candida infections in patients with psoriasis, psoriatic arthritis, and axial spondyloarthritis treated with ixekizumab: an integrated safety analysis of 25 clinical studies. Expert Opin Drug Saf 2024; 23:1347-1357. [PMID: 39234767 DOI: 10.1080/14740338.2024.2399092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND This safety analysis investigates treatment-emergent mucosal/cutaneous Candida infections in patients treated with ixekizumab (IXE), an anti-interleukin-17A monoclonal antibody, across the approved indications: psoriasis (PsO), psoriatic arthritis (PsA), and axial spondyloarthritis (axSpA). RESEARCH DESIGN AND METHODS Safety data were pooled from 25 clinical studies. Incidence rates (IRs) are expressed as per 100 patient-years (PY), using the entire duration of exposure. RESULTS Candida infections had an IR of 1.9 per 100 PY in patients with PsO (N = 6892; total PY = 18025.7), 2.0 per 100 PY in patients with PsA (N = 1401; total PY = 2247.7), and 1.2 per 100 PY in patients with axSpA (N = 932; total PY = 2097.7). The majority of treatment-emergent Candida infections were: (i) experienced only once by patients (IR = 1.3;IR = 1.6;IR = 1.0), (ii) mild/moderate in severity (IR = 0.8/0.9;IR = 1.5/0.4;IR = 0.8/0.5) as opposed to severe (IR = 0.0; IR = 0.0; IR = 0.0), (iii) oral Candida or genital Candida (IR = 0.9/0.6;IR = 1.0/0.7;IR = 0.4/0.6), (iv) marked as recovered/resolved during the studies (89.3%;93.8%;90.3%), (v) not leading to IXE discontinuation (0.0%;0.0%;0.1% discontinued), (vi) managed with topical (34.7%;22.2%;11.5%) or no anti-fungal medications (63.5%;77.8%;80.8%) as opposed to systemic therapies (1.5%;0.0%;7.7%), (vii) typically resolved before next visit. CONCLUSIONS This integrated safety analysis shows that the risk of developing Candida infections is low with IXE, and the severity is mild-to-moderate in most instances across the approved IXE indications. TRIAL REGISTRATION A comprehensive list of the clinical trials and their registration numbers is reported in Table S1 of the supplemental material.
Collapse
Affiliation(s)
| | - Luis Puig
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Arnon D Cohen
- Department of Quality Measurements and Research, Clalit Health Services, Tel Aviv, Israel
| | - Saakshi Khattri
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, USA
| | | | | | | | | | | | | | | | - Christopher Schuster
- Eli Lilly and Company, Indianapolis, USA
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Alice B Gottlieb
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
10
|
Jensen O, Trujillo E, Hanson L, Ost KS. Controlling Candida: immune regulation of commensal fungi in the gut. Infect Immun 2024; 92:e0051623. [PMID: 38647290 PMCID: PMC11385159 DOI: 10.1128/iai.00516-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
The intestinal microbiome harbors fungi that pose a significant risk to human health as opportunistic pathogens and drivers of inflammation. Inflammatory and autoimmune diseases are associated with dysbiotic fungal communities and the expansion of potentially pathogenic fungi. The gut is also the main reservoir for disseminated fungal infections. Immune interactions are critical for preventing commensal fungi from becoming pathogenic. Significant strides have been made in defining innate and adaptive immune pathways that regulate intestinal fungi, and these discoveries have coincided with advancements in our understanding of the fungal molecular pathways and effectors involved in both commensal colonization and pathogenesis within the gut. In this review, we will discuss immune interactions important for regulating commensal fungi, with a focus on how specific cell types and effectors interact with fungi to limit their colonization or pathogenic potential. This will include how innate and adaptive immune pathways target fungi and orchestrate antifungal immune responses, in addition to how secreted immune effectors, such as mucus and antimicrobial peptides, regulate fungal colonization and inhibit pathogenic potential. These immune interactions will be framed around our current understanding of the fungal effectors and pathways regulating colonization and pathogenesis within this niche. Finally, we highlight important unexplored mechanisms by which the immune system regulates commensal fungi in the gut.
Collapse
Affiliation(s)
- Owen Jensen
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Emma Trujillo
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Luke Hanson
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kyla S. Ost
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
11
|
Pastwińska J, Karwaciak I, Karaś K, Sałkowska A, Chałaśkiewicz K, Strapagiel D, Sobalska-Kwapis M, Dastych J, Ratajewski M. α-Hemolysin from Staphylococcus aureus Changes the Epigenetic Landscape of Th17 Cells. Immunohorizons 2024; 8:606-621. [PMID: 39240270 PMCID: PMC11447695 DOI: 10.4049/immunohorizons.2400061] [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: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/07/2024] Open
Abstract
The human body harbors a substantial population of bacteria, which may outnumber host cells. Thus, there are multiple interactions between both cell types. Given the common presence of Staphylococcus aureus in the human body and the role of Th17 cells in controlling this pathogen on mucous membranes, we sought to investigate the effect of α-hemolysin, which is produced by this bacterium, on differentiating Th17 cells. RNA sequencing analysis revealed that α-hemolysin influences the expression of signature genes for Th17 cells as well as genes involved in epigenetic regulation. We observed alterations in various histone marks and genome methylation levels via whole-genome bisulfite sequencing. Our findings underscore how bacterial proteins can significantly influence the transcriptome, epigenome, and phenotype of human Th17 cells, highlighting the intricate and complex nature of the interaction between immune cells and the microbiota.
Collapse
Affiliation(s)
- Joanna Pastwińska
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Iwona Karwaciak
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Kaja Karaś
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Anna Sałkowska
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Katarzyna Chałaśkiewicz
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Dominik Strapagiel
- Biobank Lab, Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Marta Sobalska-Kwapis
- Biobank Lab, Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Jarosław Dastych
- Laboratory of Cellular Immunology, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Marcin Ratajewski
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| |
Collapse
|
12
|
Rodríguez-Míguez Y, Lozano-Ordaz V, Ortiz-Cabrera AE, Barrios-Payan J, Mata-Espinosa D, Huerta-Yepez S, Baay-Guzman G, Hernández-Pando R. Effect of IL-17A on the immune response to pulmonary tuberculosis induced by high- and low-virulence strains of Mycobacterium bovis. PLoS One 2024; 19:e0307307. [PMID: 39024223 PMCID: PMC11257284 DOI: 10.1371/journal.pone.0307307] [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: 12/14/2023] [Accepted: 07/02/2024] [Indexed: 07/20/2024] Open
Abstract
Tuberculosis (TB) is an infectious, chronic, and progressive disease occurring globally. Human TB is caused mainly by Mycobacterium tuberculosis (M. tuberculosis), while the main causative agent of bovine TB is Mycobacterium bovis (M. bovis). The latter is one of the most important cattle pathogens and is considered the main cause of zoonotic TB worldwide. The mechanisms responsible for tissue damage (necrosis) during post-primary TB remain elusive. Recently, IL-17A was reported to be important for protection against M. tuberculosis infection, but it is also related to the production of an intense inflammatory response associated with necrosis. We used two M. bovis isolates with different levels of virulence and high IL-17A production to study this important cytokine's contrasting functions in a BALB/c mouse model of pulmonary TB. In the first part of the study, the gene expression kinetics and cellular sources of IL-17A were determined by real time PCR and immunohistochemistry respectively. Non-infected lungs showed low production of IL-17A, particularly by the bronchial epithelium, while lungs infected with the low-virulence 534 strain showed high IL-17A expression on Day 3 post-infection, followed by a decrease in expression in the early stage of the infection and another increase during late infection, on Day 60, when very low bacillary burdens were found. In contrast, infection with the highly virulent strain 04-303 induced a peak of IL-17A expression on Day 14 of infection, 1 week before extensive pulmonary necrosis was seen, being lymphocytes and macrophages the most important sources. In the second part of the study, the contribution of IL-17A to immune protection and pulmonary necrosis was evaluated by suppressing IL-17A via the administration of specific blocking antibodies. Infection with M. bovis strain 534 and treatment with IL-17A neutralizing antibodies did not affect mouse survival but produced a significant increase in bacillary load and a non-significant decrease in inflammatory infiltrate and granuloma area. In contrast, mice infected with the highly virulent 04-303 strain and treated with IL-17A blocking antibodies showed a significant decrease in survival, an increase in bacillary loads on Day 24 post-infection, and significantly more and earlier necrosis. Our results suggest that high expression of IL-17A is more related to protection than necrosis in a mouse model of pulmonary TB induced by M. bovis strains.
Collapse
Affiliation(s)
- Yadira Rodríguez-Míguez
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Departamento de Patología, Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
| | - Vasti Lozano-Ordaz
- Departamento de Patología, Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
| | - Angel E. Ortiz-Cabrera
- Departamento de Patología, Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
| | - Jorge Barrios-Payan
- Departamento de Patología, Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
| | - Dulce Mata-Espinosa
- Departamento de Patología, Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
| | - Sara Huerta-Yepez
- Unidad de Investigación en Enfermedades Oncológicas, Hospital Infantil de México “Federico Gómez”, Mexico City, Mexico
| | - Guillermina Baay-Guzman
- Unidad de Investigación en Enfermedades Oncológicas, Hospital Infantil de México “Federico Gómez”, Mexico City, Mexico
| | - Rogelio Hernández-Pando
- Departamento de Patología, Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
| |
Collapse
|
13
|
Huang C, Zhu W, Li Q, Lei Y, Chen X, Liu S, Chen D, Zhong L, Gao F, Fu S, He D, Li J, Xu H. Antibody Fc-receptor FcεR1γ stabilizes cell surface receptors in group 3 innate lymphoid cells and promotes anti-infection immunity. Nat Commun 2024; 15:5981. [PMID: 39013884 PMCID: PMC11252441 DOI: 10.1038/s41467-024-50266-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 07/03/2024] [Indexed: 07/18/2024] Open
Abstract
Group 3 innate lymphoid cells (ILC3) are crucial for maintaining mucosal homeostasis and regulating inflammatory diseases, but the molecular mechanisms governing their phenotype and function are not fully understood. Here, we show that ILC3s highly express Fcer1g gene, which encodes the antibody Fc-receptor common gamma chain, FcεR1γ. Genetic perturbation of FcεR1γ leads to the absence of critical cell membrane receptors NKp46 and CD16 in ILC3s. Alanine scanning mutagenesis identifies two residues in FcεR1γ that stabilize its binding partners. FcεR1γ expression in ILC3s is essential for effective protective immunity against bacterial and fungal infections. Mechanistically, FcεR1γ influences the transcriptional state and proinflammatory cytokine production of ILC3s, relying on the CD16-FcεR1γ signaling pathway. In summary, our findings highlight the significance of FcεR1γ as an adapter protein that stabilizes cell membrane partners in ILC3s and promotes anti-infection immunity.
Collapse
Affiliation(s)
- Chao Huang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
| | - Wenting Zhu
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Qing Li
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yuchen Lei
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Xi Chen
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Shaorui Liu
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Dianyu Chen
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Lijian Zhong
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Feng Gao
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Shujie Fu
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Danyang He
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Jinsong Li
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Heping Xu
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
14
|
Pathakumari B, Liu W, Wang Q, Kong X, Liang G, Chokkakula S, Pathakamuri V, Nunna V. Comparative Evaluation of Candida Species-Specific T-Cell Immune Response in Human Peripheral Blood Mononuclear Cells. Biomedicines 2024; 12:1487. [PMID: 39062060 PMCID: PMC11274682 DOI: 10.3390/biomedicines12071487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/19/2024] [Accepted: 05/28/2024] [Indexed: 07/28/2024] Open
Abstract
Non-albicans Candida (NAC) species are increasingly recognized as significant contributors to candidemia infections; however, relatively less is known about the immune responses induced by these species. In this study, we compared the cytokine production ability of human peripheral blood mononuclear cells (PBMCs) upon stimulation with different Candida species (Candida spp.). We measured secreted cytokines using ELISA and checked the functional profiles of T-cell responses using multicolor flow cytometry. Although there was a differential expression of cytokines against Candida spp., significant difference were observed in the levels of IFN-γ, TNF-α, IL-10, IL-12p40, and IL-23 (p < 0.05) between Candida spp. A significant difference was observed between C. albicans and C. glabrata (p = 0.026) in the levels of TNF-α. C. glabrata showed significant differences compared to C. albicans, C. parapsilosis, and C. krusei in the levels of IL-10 (p values of 0.02, 0.04, and 0.01, respectively). Despite the percentages of CD4+ and CD8+ expressing Th1, Th2, and Th17 cytokines being higher in stimulated PBMCs, none of the Candida spp. showed significant differences. The levels of secreted IL-17A and IL-23 were consistently lower in Candida spp. regardless of the stimulus used. Here, we showed the differential regulation of Th1, Th2, and Th17 during Candida spp. stimulation of the immune system ex vivo. Additionally, our findings suggest that C. albicans elicits an IFN-γ response, whereas C. glabrata promotes IL-10 cellular responses, but this warrants additional studies to conclude this association. This investigation holds the potential to advance our comprehension of the distinct immune responses induced by Candida spp., with probable implications in designing antifungal immunotherapeutics.
Collapse
Affiliation(s)
- Balaji Pathakumari
- Department of Medical Mycology, Institute of Dermatology, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China; (W.L.); (Q.W.); (X.K.); (G.L.)
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Weida Liu
- Department of Medical Mycology, Institute of Dermatology, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China; (W.L.); (Q.W.); (X.K.); (G.L.)
| | - Qiong Wang
- Department of Medical Mycology, Institute of Dermatology, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China; (W.L.); (Q.W.); (X.K.); (G.L.)
| | - Xue Kong
- Department of Medical Mycology, Institute of Dermatology, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China; (W.L.); (Q.W.); (X.K.); (G.L.)
| | - Guanzhao Liang
- Department of Medical Mycology, Institute of Dermatology, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China; (W.L.); (Q.W.); (X.K.); (G.L.)
| | - Santosh Chokkakula
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Republic of Korea;
| | - Vasundhara Pathakamuri
- Department of Radio-Diagnosis, Sri Venkateshwara Medical College, Tirupathi 517507, India;
| | - Venkatrao Nunna
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
| |
Collapse
|
15
|
Katsipoulaki M, Stappers MHT, Malavia-Jones D, Brunke S, Hube B, Gow NAR. Candida albicans and Candida glabrata: global priority pathogens. Microbiol Mol Biol Rev 2024; 88:e0002123. [PMID: 38832801 PMCID: PMC11332356 DOI: 10.1128/mmbr.00021-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024] Open
Abstract
SUMMARYA significant increase in the incidence of Candida-mediated infections has been observed in the last decade, mainly due to rising numbers of susceptible individuals. Recently, the World Health Organization published its first fungal pathogen priority list, with Candida species listed in medium, high, and critical priority categories. This review is a synthesis of information and recent advances in our understanding of two of these species-Candida albicans and Candida glabrata. Of these, C. albicans is the most common cause of candidemia around the world and is categorized as a critical priority pathogen. C. glabrata is considered a high-priority pathogen and has become an increasingly important cause of candidemia in recent years. It is now the second most common causative agent of candidemia in many geographical regions. Despite their differences and phylogenetic divergence, they are successful as pathogens and commensals of humans. Both species can cause a broad variety of infections, ranging from superficial to potentially lethal systemic infections. While they share similarities in certain infection strategies, including tissue adhesion and invasion, they differ significantly in key aspects of their biology, interaction with immune cells, host damage strategies, and metabolic adaptations. Here we provide insights on key aspects of their biology, epidemiology, commensal and pathogenic lifestyles, interactions with the immune system, and antifungal resistance.
Collapse
Affiliation(s)
- Myrto Katsipoulaki
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Mark H. T. Stappers
- MRC Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Dhara Malavia-Jones
- MRC Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| | - Neil A. R. Gow
- MRC Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
16
|
Banks LB, Sklarz T, Gohil M, O'Leary C, Behrens EM, Sun H, Chen YH, Koretzky GA, Jordan MS. Akt2 deficiency impairs Th17 differentiation, augments Th2 differentiation, and alters the peripheral response to immunization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.598023. [PMID: 38915532 PMCID: PMC11195075 DOI: 10.1101/2024.06.07.598023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Akt1 and Akt2, isoforms of the serine threonine kinase Akt, are essential for T cell development. However, their role in peripheral T cell differentiation remains undefined. Using mice with germline deletions of either Akt1 or Akt2, we found that both isoforms are important for Th17 differentiation, although Akt2 loss had a greater impact than loss of Akt1. In contrast to defective IL-17 production, Akt2 -/- T cells exhibited enhanced IL-4 production in vitro under Th2 polarizing conditions. In vivo , Akt2 -/- mice displayed significantly diminished IL-17A and GM-CSF production following immunization with myelin oligodendrocyte glycoprotein (MOG). This dampened response was associated with further alterations in Th cell differentiation including decreased IFNγ production but preserved IL-4 production, and preferential expansion of regulatory T cells compared to non-regulatory CD4 T cells. Taken together, we identify Akt2 as an important signaling molecule in regulating peripheral CD4 T cell responses.
Collapse
|
17
|
Peroumal D, Biswas PS. Kidney-Specific Interleukin-17 Responses During Infection and Injury. Annu Rev Immunol 2024; 42:35-55. [PMID: 37906942 DOI: 10.1146/annurev-immunol-052523-015141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
The kidneys are life-sustaining organs that are vital to removing waste from our bodies. Because of their anatomic position and high blood flow, the kidneys are vulnerable to damage due to infections and autoinflammatory conditions. Even now, our knowledge of immune responses in the kidney is surprisingly rudimentary. Studying kidney-specific immune events is challenging because of the poor regenerative capacity of the nephrons, accumulation of uremic toxins, and hypoxia- and arterial blood pressure-mediated changes, all of which have unexpected positive or negative impacts on the immune response in the kidney. Kidney-specific defense confers protection against pathogens. On the other hand, unresolved inflammation leads to kidney damage and fibrosis. Interleukin-17 is a proinflammatory cytokine that has been linked to immunity against pathogens and pathogenesis of autoinflammatory diseases. In this review, we discuss current knowledge of IL-17 activities in the kidney in the context of infections, autoinflammatory diseases, and renal fibrosis.
Collapse
Affiliation(s)
- Doureradjou Peroumal
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA;
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Partha S Biswas
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA;
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
18
|
Sahu SR, Dutta A, Peroumal D, Kumari P, Utakalaja BG, Patel SK, Acharya N. Immunogenicity and efficacy of CNA25 as a potential whole-cell vaccine against systemic candidiasis. EMBO Mol Med 2024; 16:1254-1283. [PMID: 38783167 PMCID: PMC11178797 DOI: 10.1038/s44321-024-00080-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Disseminated fungal infections account for ~1.5 million deaths per year worldwide, and mortality may increase further due to a rise in the number of immunocompromised individuals and drug-resistance fungal species. Since an approved antifungal vaccine is yet to be available, this study explored the immunogenicity and vaccine efficacy of a DNA polymerase mutant strain of Candida albicans. CNA25 is a pol32ΔΔ strain that exhibits growth defects and does not cause systemic candidiasis in mice. Immunized mice with live CNA25 were fully protected against C. albicans and C. parapsilosis but partially against C. tropicalis and C. glabrata infections. CNA25 induced steady expression of TLR2 and Dectin-1 receptors leading to a faster recognition and clearance by the immune system associated with the activation of protective immune responses mostly mediated by neutrophils, macrophages, NK cells, B cells, and CD4+ and CD8+ T cells. Molecular blockade of Dectin-1, IL-17, IFNγ, and TNFα abolished resistance to reinfection. Altogether, this study suggested that CNA25 collectively activates innate, adaptive, and trained immunity to be a promising live whole-cell vaccine against systemic candidiasis.
Collapse
Affiliation(s)
- Satya Ranjan Sahu
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
- Regional Center for Biotechnology, Faridabad, Haryana, 751021, India
| | - Abinash Dutta
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
| | - Doureradjou Peroumal
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
| | - Premlata Kumari
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
- Regional Center for Biotechnology, Faridabad, Haryana, 751021, India
| | - Bhabasha Gyanadeep Utakalaja
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
- Regional Center for Biotechnology, Faridabad, Haryana, 751021, India
| | - Shraddheya Kumar Patel
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
- Regional Center for Biotechnology, Faridabad, Haryana, 751021, India
| | - Narottam Acharya
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India.
| |
Collapse
|
19
|
Bittner-Eddy PD, Fischer LA, Parachuru PV, Costalonga M. MHC-II presentation by oral Langerhans cells impacts intraepithelial Tc17 abundance and Candida albicans oral infection via CD4 T cells. FRONTIERS IN ORAL HEALTH 2024; 5:1408255. [PMID: 38872986 PMCID: PMC11169704 DOI: 10.3389/froh.2024.1408255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/13/2024] [Indexed: 06/15/2024] Open
Abstract
In a murine model (LCΔMHC-II) designed to abolish MHC-II expression in Langerhans cells (LCs), ∼18% of oral LCs retain MHC-II, yet oral mucosal CD4 T cells numbers are unaffected. In LCΔMHC-II mice, we now show that oral intraepithelial conventional CD8αβ T cell numbers expand 30-fold. Antibody-mediated ablation of CD4 T cells in wild-type mice also resulted in CD8αβ T cell expansion in the oral mucosa. Therefore, we hypothesize that MHC class II molecules uniquely expressed on Langerhans cells mediate the suppression of intraepithelial resident-memory CD8 T cell numbers via a CD4 T cell-dependent mechanism. The expanded oral CD8 T cells co-expressed CD69 and CD103 and the majority produced IL-17A [CD8 T cytotoxic (Tc)17 cells] with a minority expressing IFN-γ (Tc1 cells). These oral CD8 T cells showed broad T cell receptor Vβ gene usage indicating responsiveness to diverse oral antigens. Generally supporting Tc17 cells, transforming growth factor-β1 (TGF-β1) increased 4-fold in the oral mucosa. Surprisingly, blocking TGF-β1 signaling with the TGF-R1 kinase inhibitor, LY364947, did not reduce Tc17 or Tc1 numbers. Nonetheless, LY364947 increased γδ T cell numbers and decreased CD49a expression on Tc1 cells. Although IL-17A-expressing γδ T cells were reduced by 30%, LCΔMHC-II mice displayed greater resistance to Candida albicans in early stages of oral infection. These findings suggest that modulating MHC-II expression in oral LC may be an effective strategy against fungal infections at mucosal surfaces counteracted by IL-17A-dependent mechanisms.
Collapse
Affiliation(s)
- Peter D. Bittner-Eddy
- Division of Basic Sciences, Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| | - Lori A. Fischer
- Division of Basic Sciences, Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| | - Praveen Venkata Parachuru
- Division of Periodontology, Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| | - Massimo Costalonga
- Division of Basic Sciences, Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
20
|
Bose S, Sahu SR, Dutta A, Acharya N. A chemically induced attenuated strain of Candida albicans generates robust protective immune responses and prevents systemic candidiasis development. eLife 2024; 13:RP93760. [PMID: 38787374 PMCID: PMC11126311 DOI: 10.7554/elife.93760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024] Open
Abstract
Despite current antifungal therapy, invasive candidiasis causes >40% mortality in immunocompromised individuals. Therefore, developing an antifungal vaccine is a priority. Here, we could for the first time successfully attenuate the virulence of Candida albicans by treating it with a fungistatic dosage of EDTA and demonstrate it to be a potential live whole cell vaccine by using murine models of systemic candidiasis. EDTA inhibited the growth and biofilm formation of C. albicans. RNA-seq analyses of EDTA-treated cells (CAET) revealed that genes mostly involved in metal homeostasis and ribosome biogenesis were up- and down-regulated, respectively. Consequently, a bulky cell wall with elevated levels of mannan and β-glucan, and reduced levels of total monosomes and polysomes were observed. CAET was eliminated faster than the untreated strain (Ca) as found by differential fungal burden in the vital organs of the mice. Higher monocytes, granulocytes, and platelet counts were detected in Ca- vs CAET-challenged mice. While hyper-inflammation and immunosuppression caused the killing of Ca-challenged mice, a critical balance of pro- and anti-inflammatory cytokines-mediated immune responses are the likely reasons for the protective immunity in CAET-infected mice.
Collapse
Affiliation(s)
- Swagata Bose
- Department of Infectious Disease Biology, Institute of Life SciencesBhubaneswarIndia
| | - Satya Ranjan Sahu
- Department of Infectious Disease Biology, Institute of Life SciencesBhubaneswarIndia
| | - Abinash Dutta
- Department of Infectious Disease Biology, Institute of Life SciencesBhubaneswarIndia
| | - Narottam Acharya
- Department of Infectious Disease Biology, Institute of Life SciencesBhubaneswarIndia
| |
Collapse
|
21
|
Zhou T, Solis NV, Marshall M, Yao Q, Garleb R, Yang M, Pearlman E, Filler SG, Liu H. Hyphal Als proteins act as CR3 ligands to promote immune responses against Candida albicans. Nat Commun 2024; 15:3926. [PMID: 38724513 PMCID: PMC11082240 DOI: 10.1038/s41467-024-48093-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 04/19/2024] [Indexed: 05/12/2024] Open
Abstract
Patients with decreased levels of CD18 (β2 integrins) suffer from life-threatening bacterial and fungal infections. CD11b, the α subunit of integrin CR3 (CD11b/CD18, αMβ2), is essential for mice to fight against systemic Candida albicans infections. Live elongating C. albicans activates CR3 in immune cells. However, the hyphal ligands that activate CR3 are not well defined. Here, we discovered that the C. albicans Als family proteins are recognized by the I domain of CD11b in macrophages. This recognition synergizes with the β-glucan-bound lectin-like domain to activate CR3, thereby promoting Syk signaling and inflammasome activation. Dectin-2 activation serves as the "outside-in signaling" for CR3 activation at the entry site of incompletely sealed phagosomes, where a thick cuff of F-actin forms to strengthen the local interaction. In vitro, CD18 partially contributes to IL-1β release from dendritic cells induced by purified hyphal Als3. In vivo, Als3 is vital for C. albicans clearance in mouse kidneys. These findings uncover a novel family of ligands for the CR3 I domain that promotes fungal clearance.
Collapse
Affiliation(s)
- Tingting Zhou
- Department of Biological Chemistry, University of California, Irvine, CA, USA
| | - Norma V Solis
- Division of Infectious Diseases, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Michaela Marshall
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
| | - Qing Yao
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Gilead Sciences Inc., Foster City, CA, USA
| | - Rachel Garleb
- Department of Biological Chemistry, University of California, Irvine, CA, USA
| | - Mengli Yang
- Department of Biological Chemistry, University of California, Irvine, CA, USA
- Zymo Research Corporation, Irvine, CA, USA
| | - Eric Pearlman
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
| | - Scott G Filler
- Division of Infectious Diseases, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Haoping Liu
- Department of Biological Chemistry, University of California, Irvine, CA, USA.
| |
Collapse
|
22
|
Zirmire RK, Saha D, Dey R, Tanimu H, Zaarour R, Bird D, Cherian P, Rana I, Roy N, Sanyal A, Misra N, Jamora C. Bacopa monnieri phytochemicals regulate fibroblast cell migration via modulation of focal adhesions. iScience 2024; 27:109489. [PMID: 38558933 PMCID: PMC10981128 DOI: 10.1016/j.isci.2024.109489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 01/03/2024] [Accepted: 03/08/2024] [Indexed: 04/04/2024] Open
Abstract
The Bacopa monnieri plant contains phytochemicals that have been used extensively in traditional medicine to treat various diseases. More recently it has been shown to accelerate wound healing, though its mechanism of action is largely unknown. Here we investigated the cellular pathways activated by a methanol extract of Bacopa monnieri in human dermal fibroblasts, which play many critical roles in the wound healing program. Gene expression analysis revealed that the Bacopa monnieri extract can modulate multiple processes involved in the wound healing program such as migration, proliferation, and angiogenesis. We discovered that the extract can increase migration of fibroblasts via modulating the size and number of focal adhesions. Bacopa monnieri-mediated changes in focal adhesions are dependent on α5β1 integrin activation and subsequent phosphorylation of focal adhesion kinase (FAK). Altogether our results suggest that Bacopa monnieri extract could enhance the wound healing rate via modulating fibroblast migration into the wound bed.
Collapse
Affiliation(s)
- Ravindra K. Zirmire
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka 560065, India
- Shanmugha Arts, Science, Technology and Research Academy (SASTRA) University, Thanjavur, Tamil Nadu 613401, India
| | - Dyuti Saha
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka 560065, India
- Department of Biology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Rakesh Dey
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka 560065, India
| | - Habibu Tanimu
- JAIN (Deemed-to-be University), #44/4, District Fund Road, Jayanagar 9th Block, Bangalore, Karnataka 560069, India
| | - Rania Zaarour
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka 560065, India
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, UAE
| | - Deborah Bird
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka 560065, India
| | - Prakash Cherian
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka 560065, India
| | - Isha Rana
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka 560065, India
- Shanmugha Arts, Science, Technology and Research Academy (SASTRA) University, Thanjavur, Tamil Nadu 613401, India
| | - Nita Roy
- L'Oréal, Research & Innovation, Bengaluru, India
| | | | - Namita Misra
- L’Oréal, Research and Innovation, Aulnay, France
| | - Colin Jamora
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka 560065, India
| |
Collapse
|
23
|
Shehab M, Hussein H, Fadlallah S, Rahal EA. An IL-17A-centric response to Epstein-Barr virus DNA mediated by dendritic Cell-T cell interactions. Front Mol Biosci 2024; 11:1243366. [PMID: 38638687 PMCID: PMC11024278 DOI: 10.3389/fmolb.2024.1243366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 03/20/2024] [Indexed: 04/20/2024] Open
Abstract
Introduction: The Epstein-Barr virus has been associated with a considerable number of autoimmune diseases. We have previously demonstrated that EBV DNA enhances the production of IL-17A, a pro-inflammatory cytokine, via endosomal Toll-like receptor signalling. Methods: We used RNA-seq to analyze the transcriptional profile of mouse immune cells treated with EBV DNA. Results: We observed that EBV DNA upregulates an IL-17A-centric network of mediators. Ensemble Gene Set Enrichment Analysis (EGSEA) showed enriched expression of sets involved in inflammatory responses including IFNγ and TNF-α-associated pathways as well as proinflammatory diseases. On the other hand, while macrophages and B cells were somewhat able to induce an IL-17A response from T cells to EBV DNA, they were less potent than dendritic cells. EBV virions were also capable of eliciting the production of inflammatory mediators from dendritic cell-T cell cultures largely mirroring responses to the viral DNA. Conclusions: Given the wide prevalence of EBV in the population, our analyses reveal a network of mediators and cell types that may serve as therapeutic targets in a large proportion of people affected by autoimmune diseases.
Collapse
Affiliation(s)
- Marwa Shehab
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut, Lebanon
| | - Hadi Hussein
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Sukayna Fadlallah
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Elias A. Rahal
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
24
|
Khan S, Bilal H, Khan MN, Fang W, Chang W, Yin B, Song NJ, Liu Z, Zhang D, Yao F, Wang X, Wang Q, Cai L, Hou B, Wang J, Mao C, Liu L, Zeng Y. Interleukin inhibitors and the associated risk of candidiasis. Front Immunol 2024; 15:1372693. [PMID: 38605952 PMCID: PMC11007146 DOI: 10.3389/fimmu.2024.1372693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
Interleukins (ILs) are vital in regulating the immune system, enabling to combat fungal diseases like candidiasis effectively. Their inhibition may cause enhanced susceptibility to infection. IL inhibitors have been employed to control autoimmune diseases and inhibitors of IL-17 and IL-23, for example, have been associated with an elevated risk of Candida infection. Thus, applying IL inhibitors might impact an individual's susceptibility to Candida infections. Variations in the severity of Candida infections have been observed between individuals with different IL inhibitors, necessitating careful consideration of their specific risk profiles. IL-1 inhibitors (anakinra, canakinumab, and rilonacept), IL-2 inhibitors (daclizumab, and basiliximab), and IL-4 inhibitors (dupilumab) have rarely been associated with Candida infection. In contrast, tocilizumab, an inhibitor of IL-6, has demonstrated an elevated risk in the context of coronavirus disease 2019 (COVID-19) treatment, as evidenced by a 6.9% prevalence of candidemia among patients using the drug. Furthermore, the incidence of Candida infections appeared to be higher in patients exposed to IL-17 inhibitors than in those exposed to IL-23 inhibitors. Therefore, healthcare practitioners must maintain awareness of the risk of candidiasis associated with using of IL inhibitors before prescribing them. Future prospective studies need to exhaustively investigate candidiasis and its associated risk factors in patients receiving IL inhibitors. Implementing enduring surveillance methods is crucial to ensure IL inhibitors safe and efficient utilization of in clinical settings.
Collapse
Affiliation(s)
- Sabir Khan
- Department of Dermatology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Hazrat Bilal
- Department of Dermatology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Muhammad Nadeem Khan
- Department of Microbiology, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan
| | - Wenjie Fang
- Department of Dermatology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Wenqiang Chang
- School of Pharmacy, Shandong University, Qingdao, Shandong, China
| | - Bin Yin
- Department of Dermatovenereology, Chengdu Second People’s Hospital, Chengdu, China
| | - Ning-jing Song
- Department of Dermatology, Tongren Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Zhongrong Liu
- Department of Dermatology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dongxing Zhang
- Department of Dermatology, Meizhou Dongshan Hospital, Meizhou, Guangdong, China
- Department of Dermatology, Meizhou People’s Hospital, Meizhou, Guangdong, China
| | - Fen Yao
- Department of Pharmacy, Shantou University School Medical College, Shantou, China
| | - Xun Wang
- Department of Dermatology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Qian Wang
- Department of Dermatology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Lin Cai
- Department of Dermatology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Bing Hou
- Department of Clinical Laboratory, Skin and Venereal Diseases Prevention and Control Hospital of Shantou City, Shantou, Guangdong, China
| | - Jiayue Wang
- Department of Dermatology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chunyan Mao
- Department of Dermatology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lingxi Liu
- Department of Dermatology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuebin Zeng
- Department of Dermatology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Department of Dermatology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
25
|
Zhang FY, Lian N, Li M. Macrophage pyroptosis induced by Candida albicans. Pathog Dis 2024; 82:ftae003. [PMID: 38499444 PMCID: PMC11162155 DOI: 10.1093/femspd/ftae003] [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: 07/02/2023] [Revised: 10/21/2023] [Accepted: 03/15/2024] [Indexed: 03/20/2024] Open
Abstract
Candida albicans (C. albicans) is a prevalent opportunistic pathogen that causes mucocutaneous and systemic infections, particularly in immunocompromised individuals. Macrophages play a crucial role in eliminating C. albicans in local and bloodstream contexts, while also regulating antifungal immune responses. However, C. albicans can induce macrophage lysis through pyroptosis, a type of regulated cell death. This process can enable C. albicans to escape from immune cells and trigger the release of IL-1β and IL-18, which can impact both the host and the pathogen. Nevertheless, the mechanisms by which C. albicans triggers pyroptosis in macrophages and the key factors involved in this process remain unclear. In this review, we will explore various factors that may influence or trigger pyroptosis in macrophages induced by C. albicans, such as hypha, ergosterol, cell wall remodeling, and other virulence factors. We will also examine the possible immune response following macrophage pyroptosis.
Collapse
Affiliation(s)
- Feng-yuan Zhang
- Hospital for Skin Diseases, Institute of Dermatology,Chinese Academy of Medical Sciences & Peking Union Medical College, 12th. JiangWangmiao street, Nanjing, 210042, China
| | - Ni Lian
- Hospital for Skin Diseases, Institute of Dermatology,Chinese Academy of Medical Sciences & Peking Union Medical College, 12th. JiangWangmiao street, Nanjing, 210042, China
| | - Min Li
- Hospital for Skin Diseases, Institute of Dermatology,Chinese Academy of Medical Sciences & Peking Union Medical College, 12th. JiangWangmiao street, Nanjing, 210042, China
- Center for Global Health, School of Public Health, Nanjing Medical University, 101st. LongMian Avenue, Nanjing, 211166, China
| |
Collapse
|
26
|
Marques PH, Tiwari S, Felice AG, Jaiswal AK, Aburjaile FF, Azevedo V, Silva-Vergara ML, Ferreira-Paim K, Soares SDC, Fonseca FM. Design of a Multi-Epitope Vaccine against Histoplasma capsulatum through Immunoinformatics Approaches. J Fungi (Basel) 2024; 10:43. [PMID: 38248954 PMCID: PMC10817582 DOI: 10.3390/jof10010043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
Histoplasmosis is a widespread systemic disease caused by Histoplasma capsulatum, prevalent in the Americas. Despite its significant morbidity and mortality rates, no vaccines are currently available. Previously, five vaccine targets and specific epitopes for H. capsulatum were identified. Immunoinformatics has emerged as a novel approach for determining the main immunogenic components of antigens through in silico methods. Therefore, we predicted the main helper and cytotoxic T lymphocytes and B-cell epitopes for these targets to create a potential multi-epitope vaccine known as HistoVAC-TSFM. A total of 38 epitopes were found: 23 common to CTL and B-cell responses, 11 linked to HTL and B cells, and 4 previously validated epitopes associated with the B subunit of cholera toxin, a potent adjuvant. In silico evaluations confirmed the stability, non-toxicity, non-allergenicity, and non-homology of these vaccines with the host. Notably, the vaccine exhibited the potential to trigger both innate and adaptive immune responses, likely involving the TLR4 pathway, as supported by 3D modeling and molecular docking. The designed HistoVAC-TSFM appears promising against Histoplasma, with the ability to induce important cytokines, such as IFN-γ, TNF-α, IL17, and IL6. Future studies could be carried out to test the vaccine's efficacy in in vivo models.
Collapse
Affiliation(s)
- Pedro Henrique Marques
- Postgraduate Interunits Program in Bioinformatics, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil; (P.H.M.); (A.K.J.)
- Department of Preventive Veterinary, Medicine, School of Veterinary Medicine, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Sandeep Tiwari
- Institute of Biology, Federal University of Bahia, Salvador 40170-115, Brazil;
- Institute of Health Sciences, Federal University of Bahia, Salvador 40170-115, Brazil
| | - Andrei Giacchetto Felice
- Department of Microbiology, Immunology and Parasitology, Federal University of Triangulo Mineiro, Uberaba 38015-050, Brazil; (A.G.F.); (S.d.C.S.)
| | - Arun Kumar Jaiswal
- Postgraduate Interunits Program in Bioinformatics, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil; (P.H.M.); (A.K.J.)
| | - Flávia Figueira Aburjaile
- Department of Preventive Veterinary, Medicine, School of Veterinary Medicine, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Vasco Azevedo
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Mario León Silva-Vergara
- Department of Infectious Diseases, Federal University of Triangulo Mineiro, Uberaba 38025-440, Brazil;
| | - Kennio Ferreira-Paim
- Department of Microbiology, Immunology and Parasitology, Federal University of Triangulo Mineiro, Uberaba 38015-050, Brazil; (A.G.F.); (S.d.C.S.)
| | - Siomar de Castro Soares
- Department of Microbiology, Immunology and Parasitology, Federal University of Triangulo Mineiro, Uberaba 38015-050, Brazil; (A.G.F.); (S.d.C.S.)
| | | |
Collapse
|
27
|
Khorshidvand Z, Shirian S, Amiri H, Zamani A, Maghsood AH. Immunomodulatory chitosan nanoparticles for Toxoplasma gondii infection: Novel application of chitosan in complex propranolol-hydrochloride as an adjuvant in vaccine delivery. Int J Biol Macromol 2023; 253:127228. [PMID: 37839605 DOI: 10.1016/j.ijbiomac.2023.127228] [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: 05/15/2023] [Revised: 09/28/2023] [Accepted: 10/01/2023] [Indexed: 10/17/2023]
Abstract
The study aimed to investigate the immunomodulatory effects of propranolol hydrochloride (PRO) in combination with chitosan nanoparticles (CS NPs) as an adjuvant to develop an effective vaccine against T. gondii. A total of 105 BALB/c mice were randomly divided into seven equal groups including PBS alone, CS NPs, SAG1 (Surface antigen 1), CS-SAG1 NPs, CS-PRO NPs, SAG1-PRO, and CS-SAG1-PRO NPs. The immunostimulatory effect of each adjuvant used for vaccine delivery was evaluated in a mice immunization model. The results showed that the mice immunized with CS-SAG1-PRO NPs exhibited the highest lymphocyte proliferation rate, along with increased secretion of IFN-γ, TNF-α, IL-6, IL-12, IL-17, and IL-23, as well as elevated levels of protective cytokines such as TGF-β, IL-27, and IL-10. Although, the CS-SAG1-PRO NPs immunized mice showed the highest level of T. gondii specific IgG compared to the other groups, a significant production of IgG2a and IgG1 was observed in the sera of mice immunized with the CS-SAG1-PRO NPs compared to the other group (p <0.001). The higher IgG2a/IgG1 ratio observed in the CS-SAG1-PRO NPs group indicates a bias towards Th1 cell polarization, suggesting the promotion of Th1 cell-mediated immune responses. Considering the combination of the highest lymphocyte proliferation and survival rates, IgG2a/IgG1 ratio, and cytokine levels in the mice immunized with CS-SAG1-PRO NPs, this approach holds promise for immunostimulation and vaccine delivery against T. gondii infection.
Collapse
Affiliation(s)
- Zohreh Khorshidvand
- Department of Parasitology and Mycology, School of Medicine Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sadegh Shirian
- Department of Pathology, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran; Shiraz Molecular Pathology Research Center, Dr Daneshbod Lab, Shiraz, Iran
| | - Hanieh Amiri
- Shiraz Molecular Pathology Research Center, Dr Daneshbod Lab, Shiraz, Iran; Department of Biology, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Alireza Zamani
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Amir Hossein Maghsood
- Department of Parasitology and Mycology, School of Medicine Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
28
|
Cossarini F, Aberg JA, Chen BK, Mehandru S. Viral Persistence in the Gut-Associated Lymphoid Tissue and Barriers to HIV Cure. AIDS Res Hum Retroviruses 2023; 40:54-65. [PMID: 37450338 PMCID: PMC10790554 DOI: 10.1089/aid.2022.0180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
More than 40 years after the first reported cases of what then became known as acquired immunodeficiency syndrome (AIDS), tremendous progress has been achieved in transforming the disease from almost universally fatal to a chronic manageable condition. Nonetheless, the efforts to find a preventative vaccine or a cure for the underlying infection with Human Immunodeficiency Virus (HIV) remain largely unsuccessful. Many challenges intrinsic to the virus characteristics and host response need to be overcome for either goal to be achieved. This article will review the obstacles to an effective HIV cure, specifically the steps involved in the generation of HIV latency, focusing on the role of the gut-associated lymphoid tissue, which has received less attention compared with the peripheral blood, despite being the largest repository of lymphoid tissue in the human body, and a large site for HIV persistence.
Collapse
Affiliation(s)
- Francesca Cossarini
- Division of Infectious Diseases, Department of Medicine, Icahn School at Mount Sinai, New York, New York, USA
- Precision Immunology Institute, Icahn School at Mount Sinai, New York, New York, USA
| | - Judith A. Aberg
- Division of Infectious Diseases, Department of Medicine, Icahn School at Mount Sinai, New York, New York, USA
| | - Benjamin K. Chen
- Division of Infectious Diseases, Department of Medicine, Icahn School at Mount Sinai, New York, New York, USA
- Precision Immunology Institute, Icahn School at Mount Sinai, New York, New York, USA
| | - Saurabh Mehandru
- Precision Immunology Institute, Icahn School at Mount Sinai, New York, New York, USA
- Division of Gastroenterology, Department of Medicine, Icahn School at Mount Sinai, New York, New York, USA
| |
Collapse
|
29
|
Pastwińska J, Karwaciak I, Karaś K, Bachorz RA, Ratajewski M. RORγT agonists as immune modulators in anticancer therapy. Biochim Biophys Acta Rev Cancer 2023; 1878:189021. [PMID: 37951483 DOI: 10.1016/j.bbcan.2023.189021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/26/2023] [Accepted: 11/04/2023] [Indexed: 11/14/2023]
Abstract
RORγT is a transcription factor that directs the development of Th17 lymphocytes and other IL-17-expressing cells (e.g., Tc17 and ILC3 cells). These cells are involved in the body's defense against pathogenic bacteria and fungi, but they also participate in maintaining the proinflammatory environment in some autoimmune diseases and play a role in the immune system's response to cancer. Similar to other members of the nuclear receptor superfamily, the activity of RORγT is regulated by low-molecular-weight ligands. Therefore, extensive efforts have been dedicated to identifying inverse agonists that diminish the activity of this receptor and subsequently inhibit the development of autoimmune diseases. Unfortunately, in the pursuit of an ideal inverse agonist, the development of agonists has been overlooked. It is important to remember that these types of compounds, by stimulating lymphocytes expressing RORγT (Th17 and Tc17), can enhance the immune system's response to tumors. In this review, we present recent advancements in the biology of RORγT agonists and their potential application in anticancer therapy.
Collapse
Affiliation(s)
- Joanna Pastwińska
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland
| | - Iwona Karwaciak
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland
| | - Kaja Karaś
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland
| | - Rafał A Bachorz
- Laboratory of Molecular Modeling, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland
| | - Marcin Ratajewski
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland.
| |
Collapse
|
30
|
Bachorz RA, Pastwińska J, Nowak D, Karaś K, Karwaciak I, Ratajewski M. The application of machine learning methods to the prediction of novel ligands for ROR γ/ROR γT receptors. Comput Struct Biotechnol J 2023; 21:5491-5505. [PMID: 38022699 PMCID: PMC10663739 DOI: 10.1016/j.csbj.2023.10.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
In this work, we developed and applied a computational procedure for creating and validating predictive models capable of estimating the biological activity of ligands. The combination of modern machine learning methods, experimental data, and the appropriate setup of molecular descriptors led to a set of well-performing models. We thoroughly inspected both the methodological space and various possibilities for creating a chemical feature space. The resulting models were applied to the virtual screening of the ZINC20 database to identify new, biologically active ligands of RORγ receptors, which are a subfamily of nuclear receptors. Based on the known ligands of RORγ, we selected candidates and calculate their predicted activities with the best-performing models. We chose two candidates that were experimentally verified. One of these candidates was confirmed to induce the biological activity of the RORγ receptors, which we consider proof of the efficacy of the proposed methodology.
Collapse
Affiliation(s)
- Rafał A. Bachorz
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, Łódź, 93-232, Poland
| | - Joanna Pastwińska
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, Łódź, 93-232, Poland
| | - Damian Nowak
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, Łódź, 93-232, Poland
| | - Kaja Karaś
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, Łódź, 93-232, Poland
| | - Iwona Karwaciak
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, Łódź, 93-232, Poland
| | - Marcin Ratajewski
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, Łódź, 93-232, Poland
| |
Collapse
|
31
|
Fan X, Shu P, Wang Y, Ji N, Zhang D. Interactions between neutrophils and T-helper 17 cells. Front Immunol 2023; 14:1279837. [PMID: 37920459 PMCID: PMC10619153 DOI: 10.3389/fimmu.2023.1279837] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/05/2023] [Indexed: 11/04/2023] Open
Abstract
Neutrophils comprise the majority of immune cells in human peripheral circulation, have potent antimicrobial activities, and are clinically significant in their abundance, heterogeneity, and subcellular localization. In the past few years, the role of neutrophils as components of the innate immune response has been studied in numerous ways, and these cells are crucial in fighting infections, autoimmune diseases, and cancer. T-helper 17 (Th17) cells that produce interleukin 17 (IL-17) are critical in fighting infections and maintaining mucosal immune homeostasis, whereas they mediate several autoimmune diseases. Neutrophils affect adaptive immune responses by interacting with adaptive immune cells. In this review, we describe the physiological roles of both Th17 cells and neutrophils and their interactions and briefly describe the pathological processes in which these two cell types participate. We provide a summary of relevant drugs targeting IL-17A and their clinical trials. Here, we highlight the interactions between Th17 cells and neutrophils in diverse pathophysiological situations.
Collapse
Affiliation(s)
- Xinzou Fan
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Panyin Shu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ying Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Ning Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Dunfang Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
32
|
Huangfu L, Li R, Huang Y, Wang S. The IL-17 family in diseases: from bench to bedside. Signal Transduct Target Ther 2023; 8:402. [PMID: 37816755 PMCID: PMC10564932 DOI: 10.1038/s41392-023-01620-3] [Citation(s) in RCA: 120] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/16/2023] [Accepted: 08/22/2023] [Indexed: 10/12/2023] Open
Abstract
The interleukin-17 (IL-17) family comprises six members (IL-17A-17F), and recently, all of its related receptors have been discovered. IL-17 was first discovered approximately 30 years ago. Members of this family have various biological functions, including driving an inflammatory cascade during infections and autoimmune diseases, as well as boosting protective immunity against various pathogens. IL-17 is a highly versatile proinflammatory cytokine necessary for vital processes including host immune defenses, tissue repair, inflammatory disease pathogenesis, and cancer progression. However, how IL-17 performs these functions remains controversial. The multifunctional properties of IL-17 have attracted research interest, and emerging data have gradually improved our understanding of the IL-17 signaling pathway. However, a comprehensive review is required to understand its role in both host defense functions and pathogenesis in the body. This review can aid researchers in better understanding the mechanisms underlying IL-17's roles in vivo and provide a theoretical basis for future studies aiming to regulate IL-17 expression and function. This review discusses recent progress in understanding the IL-17 signaling pathway and its physiological roles. In addition, we present the mechanism underlying IL-17's role in various pathologies, particularly, in IL-17-induced systemic lupus erythematosus and IL-17-related tumor cell transformation and metastasis. In addition, we have briefly discussed promising developments in the diagnosis and treatment of autoimmune diseases and tumors.
Collapse
Affiliation(s)
- Longjie Huangfu
- School of Stomatology, Harbin Medical University, Harbin, 150001, P. R. China
| | - Ruiying Li
- Department of Oral Pathology, School of Stomatology, Hainan Medical University, Haikou, 571199, P. R. China
| | - Yamei Huang
- Department of Oral Pathology, School of Stomatology, Hainan Medical University, Haikou, 571199, P. R. China
| | - Shan Wang
- Department of Oral Pathology, School of Stomatology, Hainan Medical University, Haikou, 571199, P. R. China.
- Department of Stomatology, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570216, P. R. China.
| |
Collapse
|
33
|
Song M, Liang J, Wang L, Li W, Jiang S, Xu S, Tang L, Du Q, Liu G, Meng H, Zhai D, Shi S, Yang Y, Zhang L, Zhang B. IL-17A functions and the therapeutic use of IL-17A and IL-17RA targeted antibodies for cancer treatment. Int Immunopharmacol 2023; 123:110757. [PMID: 37579542 DOI: 10.1016/j.intimp.2023.110757] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/16/2023]
Abstract
Interleukin 17A (IL-17A) is a major member of the IL-17 cytokine family and is produced mainly by T helper 17 (Th17) cells. Other cells such as CD8+ T cells, γδ T cells, natural killer T cells and innate lymphoid-like cells can also produce IL-17A. In healthy individuals, IL-17A has a host-protective capacity, but excessive elevation of IL-17A is associated with the development of autoimmune diseases and cancer. Monoclonal antibodies (mAbs) targeting IL-17A (e.g., ixekizumab and secukinumab) or IL-17A receptor (IL-17RA) (e.g., brodalumab) would be investigated as potential treatments for these diseases. Currently, the application of IL-17A-targeted drugs in autoimmune diseases will provide new ideas for the treatment of tumors, and its combined application with immune checkpoint inhibitors has become a research hotspot. This article reviews the mechanism of action of IL-17A and the application of anti-IL-17A antibodies, focusing on the research progress on the mechanism of action and therapeutic blockade of IL-17A in various tumors such as colorectal cancer (CRC), lung cancer, gastric cancer and breast cancer. Moreover, we also include the results of therapeutic blockade in the field of cancer as well as recent advances in the regulation of IL-17A signaling.
Collapse
Affiliation(s)
- Meiying Song
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, PR China
| | - Jie Liang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, PR China
| | - Luoyang Wang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, PR China
| | - Wei Li
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, PR China
| | - Suli Jiang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, PR China
| | - Shuo Xu
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, PR China
| | - Lei Tang
- Department of Special Medicine, School of Basic Medical College, Qingdao University, Qingdao, Shandong 266071, PR China
| | - Qiaochu Du
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, PR China
| | - Guixian Liu
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, PR China
| | - Haining Meng
- School of Emergency Medicine, Medical College of Qingdao University, Qingdao, Shandong 266071, PR China
| | - Dongchang Zhai
- Department of Special Medicine, School of Basic Medical College, Qingdao University, Qingdao, Shandong 266071, PR China
| | - Shangheng Shi
- Department of Liver Transplantation, School of Clinical Medicine, Qingdao University, Qingdao, Shandong 266071, PR China
| | - Yanyan Yang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, PR China
| | - Li Zhang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, PR China
| | - Bei Zhang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, PR China.
| |
Collapse
|
34
|
Wang J, Zhao X, Wan YY. Intricacies of TGF-β signaling in Treg and Th17 cell biology. Cell Mol Immunol 2023; 20:1002-1022. [PMID: 37217798 PMCID: PMC10468540 DOI: 10.1038/s41423-023-01036-7] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/27/2023] [Indexed: 05/24/2023] Open
Abstract
Balanced immunity is pivotal for health and homeostasis. CD4+ helper T (Th) cells are central to the balance between immune tolerance and immune rejection. Th cells adopt distinct functions to maintain tolerance and clear pathogens. Dysregulation of Th cell function often leads to maladies, including autoimmunity, inflammatory disease, cancer, and infection. Regulatory T (Treg) and Th17 cells are critical Th cell types involved in immune tolerance, homeostasis, pathogenicity, and pathogen clearance. It is therefore critical to understand how Treg and Th17 cells are regulated in health and disease. Cytokines are instrumental in directing Treg and Th17 cell function. The evolutionarily conserved TGF-β (transforming growth factor-β) cytokine superfamily is of particular interest because it is central to the biology of both Treg cells that are predominantly immunosuppressive and Th17 cells that can be proinflammatory, pathogenic, and immune regulatory. How TGF-β superfamily members and their intricate signaling pathways regulate Treg and Th17 cell function is a question that has been intensely investigated for two decades. Here, we introduce the fundamental biology of TGF-β superfamily signaling, Treg cells, and Th17 cells and discuss in detail how the TGF-β superfamily contributes to Treg and Th17 cell biology through complex yet ordered and cooperative signaling networks.
Collapse
Affiliation(s)
- Junying Wang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xingqi Zhao
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yisong Y Wan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
35
|
Sun L, Wang L, Moore BB, Zhang S, Xiao P, Decker AM, Wang HL. IL-17: Balancing Protective Immunity and Pathogenesis. J Immunol Res 2023; 2023:3360310. [PMID: 37600066 PMCID: PMC10439834 DOI: 10.1155/2023/3360310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/07/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
The biological role of interleukin 17 (IL-17) has been explored during recent decades and identified as a pivotal player in coordinating innate and adaptive immune responses. Notably, IL-17 functions as a double-edged sword with both destructive and protective immunological roles. While substantial progress has implicated unrestrained IL-17 in a variety of infectious diseases or autoimmune conditions, IL-17 plays an important role in protecting the host against pathogens and maintaining physiological homeostasis. In this review, we describe canonical IL-17 signaling mechanisms promoting neutrophils recruitment, antimicrobial peptide production, and maintaining the epithelium barrier integrity, as well as some noncanonical mechanisms involving IL-17 that elicit protective immunity.
Collapse
Affiliation(s)
- Lu Sun
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Lufei Wang
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill School of Dentistry, Chapel Hill, NC, USA
| | - Bethany B. Moore
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Shaoping Zhang
- Department of Periodontics, University of Iowa College of Dentistry, Iowa, IA, USA
| | - Peng Xiao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Immunological Disease Research Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ann M. Decker
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Hom-Lay Wang
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| |
Collapse
|
36
|
Lu H, Hong T, Jiang Y, Whiteway M, Zhang S. Candidiasis: From cutaneous to systemic, new perspectives of potential targets and therapeutic strategies. Adv Drug Deliv Rev 2023; 199:114960. [PMID: 37307922 DOI: 10.1016/j.addr.2023.114960] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/14/2023]
Abstract
Candidiasis is an infection caused by fungi from a Candida species, most commonly Candida albicans. C. albicans is an opportunistic fungal pathogen typically residing on human skin and mucous membranes of the mouth, intestines or vagina. It can cause a wide variety of mucocutaneous barrier and systemic infections; and becomes a severe health problem in HIV/AIDS patients and in individuals who are immunocompromised following chemotherapy, treatment with immunosuppressive agents or after antibiotic-induced dysbiosis. However, the immune mechanism of host resistance to C. albicans infection is not fully understood, there are a limited number of therapeutic antifungal drugs for candidiasis, and these have disadvantages that limit their clinical application. Therefore, it is urgent to uncover the immune mechanisms of the host protecting against candidiasis and to develop new antifungal strategies. This review synthesizes current knowledge of host immune defense mechanisms from cutaneous candidiasis to invasive C. albicans infection and documents promising insights for treating candidiasis through inhibitors of potential antifungal target proteins.
Collapse
Affiliation(s)
- Hui Lu
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Ting Hong
- Department of Anesthesiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yuanying Jiang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Malcolm Whiteway
- Department of Biology, Concordia University, Montreal, QC, Canada.
| | - Shiqun Zhang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China.
| |
Collapse
|
37
|
Riekert M, Almanzar G, Schmalzing M, Schütze N, Jakob F, Prelog M. Mesenchymal stem cells modulate IL-17 and IL-9 production induced by Th17-inducing cytokine conditions in autoimmune arthritis: an explorative analysis. Adv Rheumatol 2023; 63:37. [PMID: 37525265 DOI: 10.1186/s42358-023-00317-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 07/19/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND The importance of proinflammatory T-cells and their cytokine production in patients with autoimmune arthritis has been widely described. Due to their immunomodulatory properties, mesenchymal stem cells (MSCs) have come into focus as a potential therapeutic concept. The aim of this study was to investigate the influence of MSCs on the phenotype, cytokine profile, and functionality of naive and non-naive CD4+ T-cells from healthy donors (HD) and patients with autoimmune arthritis under Th17-cytokine polarizing conditions in an explorative way using a transwell system prohibiting any cell-cell-contact. METHODS Magnetically isolated naive and non-naive CD4+ T-cells were stimulated under Th17-polarizing proinflammatory cytokine conditions in presence and absence of bone marrow derived mesenchymal stromal cells (MSCs). After an incubation period of 6 days, the proportions of the T-cell subpopulations TEMRA (CD45RA+CD27-), memory (CD45RA-CD27+), effector (CD45RA-CD27-) and naive cells (CD45RA+CD27+) were determined. Quantitative immunofluorescence intensity was used as a measure for IL-9, IL-17 and IFN-γ production in each subpopulation. RESULTS In isolated naive CD4+ T-cells from HD and patients, MSCs suppressed the differentiation of naive towards an effector phenotype while memory and naive cells showed higher percentages in culture with MSCs. In patients, MSCs significantly decreased the proportion of IL-9 and IL-17 producing effector T-cells. MSCs also reduced IFN-γ production in the naive and memory phenotype from HD. CONCLUSION The results of the study indicate significant immunomodulatory properties of MSCs, as under Th17-polarizing conditions MSCs are still able to control T-cell differentiation and proinflammatory cytokine production in both HD and patients with autoimmune arthritis.
Collapse
Affiliation(s)
- Maximilian Riekert
- Department of Pediatrics, University Hospital Wuerzburg, Wuerzburg, Germany.
- Department of Oral and Craniomaxillofacial and Plastic Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Straße 62, 50924, Cologne, Germany.
| | - Giovanni Almanzar
- Department of Pediatrics, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Marc Schmalzing
- Department of Internal Medicine II, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Norbert Schütze
- Orthopedic Clinic, Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Franz Jakob
- Orthopedic Clinic, Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Martina Prelog
- Department of Pediatrics, University Hospital Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
38
|
Hipp AV, Bengsch B, Globig AM. Friend or Foe - Tc17 cell generation and current evidence for their importance in human disease. DISCOVERY IMMUNOLOGY 2023; 2:kyad010. [PMID: 38567057 PMCID: PMC10917240 DOI: 10.1093/discim/kyad010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/12/2023] [Accepted: 07/19/2023] [Indexed: 04/04/2024]
Abstract
The term Tc17 cells refers to interleukin 17 (IL-17)-producing CD8+ T cells. While IL-17 is an important mediator of mucosal defense, it is also centrally involved in driving the inflammatory response in immune-mediated diseases, such as psoriasis, multiple sclerosis, and inflammatory bowel disease. In this review, we aim to gather the current knowledge on the phenotypic and transcriptional profile, the in vitro and in vivo generation of Tc17 cells, and the evidence pointing towards a relevant role of Tc17 cells in human diseases such as infectious diseases, cancer, and immune-mediated diseases.
Collapse
Affiliation(s)
- Anna Veronika Hipp
- Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, University Medical Center Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Bertram Bengsch
- Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, University Medical Center Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Anna-Maria Globig
- Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, University Medical Center Freiburg, Faculty of Medicine, Freiburg, Germany
| |
Collapse
|
39
|
Sun L, Su Y, Jiao A, Wang X, Zhang B. T cells in health and disease. Signal Transduct Target Ther 2023; 8:235. [PMID: 37332039 PMCID: PMC10277291 DOI: 10.1038/s41392-023-01471-y] [Citation(s) in RCA: 311] [Impact Index Per Article: 155.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 06/20/2023] Open
Abstract
T cells are crucial for immune functions to maintain health and prevent disease. T cell development occurs in a stepwise process in the thymus and mainly generates CD4+ and CD8+ T cell subsets. Upon antigen stimulation, naïve T cells differentiate into CD4+ helper and CD8+ cytotoxic effector and memory cells, mediating direct killing, diverse immune regulatory function, and long-term protection. In response to acute and chronic infections and tumors, T cells adopt distinct differentiation trajectories and develop into a range of heterogeneous populations with various phenotype, differentiation potential, and functionality under precise and elaborate regulations of transcriptional and epigenetic programs. Abnormal T-cell immunity can initiate and promote the pathogenesis of autoimmune diseases. In this review, we summarize the current understanding of T cell development, CD4+ and CD8+ T cell classification, and differentiation in physiological settings. We further elaborate the heterogeneity, differentiation, functionality, and regulation network of CD4+ and CD8+ T cells in infectious disease, chronic infection and tumor, and autoimmune disease, highlighting the exhausted CD8+ T cell differentiation trajectory, CD4+ T cell helper function, T cell contributions to immunotherapy and autoimmune pathogenesis. We also discuss the development and function of γδ T cells in tissue surveillance, infection, and tumor immunity. Finally, we summarized current T-cell-based immunotherapies in both cancer and autoimmune diseases, with an emphasis on their clinical applications. A better understanding of T cell immunity provides insight into developing novel prophylactic and therapeutic strategies in human diseases.
Collapse
Affiliation(s)
- Lina Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Xin Wang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China.
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China.
| |
Collapse
|
40
|
Tangye SG, Puel A. The Th17/IL-17 Axis and Host Defense Against Fungal Infections. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:1624-1634. [PMID: 37116791 DOI: 10.1016/j.jaip.2023.04.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/30/2023]
Abstract
Chronic mucocutaneous candidiasis (CMC) was recognized as a primary immunodeficiency in the early 1970s. However, for almost 40 years, its genetic etiology remained unknown. The progressive molecular and cellular description of inborn errors of immunity (IEI) with syndromic CMC pointed toward a possible role of IL-17-mediated immunity in protecting against fungal infection and CMC. Since 2011, novel IEI affecting either the response to or production of IL-17A and/or IL-17F (IL-17A/F) in patients with isolated or syndromic CMC provided formal proof of the pivotal role of the IL-17 axis in mucocutaneous immunity to Candida spp, and, to a lesser extent, to Staphylococcus aureus in humans. In contrast, IL-17-mediated immunity seems largely redundant against other common microbes in humans. In this review, we outline the current knowledge of IEI associated with impaired IL-17A/F-mediated immunity, highlighting our current understanding of the role of IL-17A/F in human immunity.
Collapse
Affiliation(s)
- Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; School of Clinical Medicine, UNSW Faculty of Medicine & Health, Darlinghurst, NSW, Australia.
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France; Imagine Institute, University of Paris, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, the Rockefeller University, New York, NY, USA
| |
Collapse
|
41
|
Zhao J, Dong Z, Zhu L, Song W, Qi P. An Interleukin-17 Isoform from Thick Shell Mussel Mytilus coruscus Serves as a Mediator of Inflammatory Response. Molecules 2023; 28:molecules28041806. [PMID: 36838794 PMCID: PMC9965057 DOI: 10.3390/molecules28041806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/04/2023] [Accepted: 02/05/2023] [Indexed: 02/17/2023] Open
Abstract
The inflammatory cytokine interleukin-17 (IL17) plays an important role in innate immunity by binding to its receptors (IL17Rs) to activate immune defense signals. To date, information on members of the IL17 family is still very limited in molluscan species. Here, a novel member of the IL17 family was identified and characterized from thick shell mussel Mytilus coruscus, and this gene was designated as McIL17-1 by predicting structural domains and phylogenetic analysis. McIL17-1 transcripts existed in all examined tissues with high expression levels in gills, hemocytes and digestive glands. After the stimuli of different pathogen associated molecular patterns (PAMPs) for 72 h, transcriptional expression of McIL17-1 was significantly upregulated, except for poly I:C stimulation. Cytoplasm localization of McIL17-1 was shown in HEK293T cells by fluorescence microscopy. Further, in vivo and in vitro assays were performed to evaluate the potential function of McIL17-1 played in immune response. McIL17-1 was either knocked down or overexpressed in vivo through RNA inference (RNAi) and recombinant protein injection, respectively. With the infection of living Vibrio alginolyticus, a high mortality rate was exhibited in the McIL17-1 overexpressed group compared to the control group, while a lower mortality rate was observed in the McIL17-1 knocked down group than control group. In vitro, the flow cytometric analysis showed that the apoptosis rate of McIL17-1 inhibited hemocytes was significantly lower than that of the control group after lipopolysaccharide stimulation. These results collectively suggested that the newly identified IL17 isoform is involved in the inflammatory response to bacterial infection in M. coruscus.
Collapse
|
42
|
Akhter S, Tasnim FM, Islam MN, Rauf A, Mitra S, Emran TB, Alhumaydhi FA, Khalil AA, Aljohani ASM, Al Abdulmonem W, Thiruvengadam M. Role of Th17 and IL-17 Cytokines on Inflammatory and Auto-immune Diseases. Curr Pharm Des 2023; 29:2078-2090. [PMID: 37670700 DOI: 10.2174/1381612829666230904150808] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/21/2023] [Accepted: 07/24/2023] [Indexed: 09/07/2023]
Abstract
BACKGROUND The IL-17 (interleukin 17) family consists of six structurally related pro-inflammatory cytokines, namely IL-17A to IL-17F. These cytokines have garnered significant scientific interest due to their pivotal role in the pathogenesis of various diseases. Notably, a specific subset of T-cells expresses IL-17 family members, highlighting their importance in immune responses against microbial infections. INTRODUCTION IL-17 cytokines play a critical role in host defense mechanisms by inducing cytokines and chemokines, recruiting neutrophils, modifying T-cell differentiation, and stimulating the production of antimicrobial proteins. Maintaining an appropriate balance of IL-17 is vital for overall health. However, dysregulated production of IL-17A and other members can lead to the pathogenesis of numerous inflammatory and autoimmune diseases. METHOD This review provides a comprehensive overview of the IL-17 family and its involvement in several inflammatory and autoimmune diseases. Relevant literature and research studies were analyzed to compile the data presented in this review. RESULTS IL-17 cytokines, particularly IL-17A, have been implicated in the development of various inflammatory and autoimmune disorders, including multiple sclerosis, Hashimoto's thyroiditis, systemic lupus erythematosus, pyoderma gangrenosum, autoimmune hepatic disorders, rheumatoid arthritis, psoriasis, psoriatic arthritis, ankylosing spondylitis, osteoarthritis, and graft-versus-host disease. Understanding the role of IL-17 in these diseases is crucial for developing targeted therapeutic strategies. CONCLUSION The significant involvement of IL-17 cytokines in inflammatory and autoimmune diseases underscores their potential as therapeutic targets. Current treatments utilizing antibodies against IL-17 cytokines and IL-17RA receptors have shown promise in managing these conditions. This review consolidates the understanding of IL-17 family members and their roles, providing valuable insights for the development of novel immunomodulators to effectively treat inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Saima Akhter
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh
| | - Farhin Muntaha Tasnim
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh
| | - Mohammad Nazmul Islam
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Pakistan
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Science, Qassim University, Buraydah, Saudi Arabia
| | - Anees Ahmed Khalil
- University Institute of Diet and Nutritionals Sciences, Faculty of Allied Health Sciences, The University of Lahore, Lahore, Pakistan
| | - Abdullah S M Aljohani
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
43
|
Abstract
IL-17 cytokine family members have diverse biological functions, promoting protective immunity against many pathogens but also driving inflammatory pathology during infection and autoimmunity. IL-17A and IL-17F are produced by CD4+ and CD8+ T cells, γδ T cells, and various innate immune cell populations in response to IL-1β and IL-23, and they mediate protective immunity against fungi and bacteria by promoting neutrophil recruitment, antimicrobial peptide production and enhanced barrier function. IL-17-driven inflammation is normally controlled by regulatory T cells and the anti-inflammatory cytokines IL-10, TGFβ and IL-35. However, if dysregulated, IL-17 responses can promote immunopathology in the context of infection or autoimmunity. Moreover, IL-17 has been implicated in the pathogenesis of many other disorders with an inflammatory basis, including cardiovascular and neurological diseases. Consequently, the IL-17 pathway is now a key drug target in many autoimmune and chronic inflammatory disorders; therapeutic monoclonal antibodies targeting IL-17A, both IL-17A and IL-17F, the IL-17 receptor, or IL-23 are highly effective in some of these diseases. However, new approaches are needed to specifically regulate IL-17-mediated immunopathology in chronic inflammation and autoimmunity without compromising protective immunity to infection.
Collapse
Affiliation(s)
- Kingston H G Mills
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
44
|
Iraji D, Oftedal BE, Wolff ASB. Th17 Cells: Orchestrators of Mucosal Inflammation and Potential Therapeutic Targets. Crit Rev Immunol 2023; 43:25-52. [PMID: 37831521 DOI: 10.1615/critrevimmunol.2023050360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
T helper 17 (Th17) cells represent a specialized subgroup of effector CD4+ T cells known for their role in provoking neutrophil-driven tissue inflammation, particularly within mucosal tissues. Although they are pivotal for defending the host against extracellular bacteria and fungi, they have also been associated with development of various T cell-mediated inflammatory conditions, autoimmune diseases, and even cancer. Notably, Th17 cells exhibit a dual nature, with different Th17 cell subtypes showcasing distinct effector functions and varying capacities to incite autoimmune tissue inflammation. Furthermore, Th17 cells exhibit significant plasticity, which carries important functional implications, both in terms of their expression of cytokines typically associated with other effector T cell subsets and in their interactions with regulatory CD4+ T cells. The intricate balance of Th17 cytokines can also be a double-edged sword in inflammation, autoimmunity, and cancer. Within this article, we delve into the mechanisms that govern the differentiation, function, and adaptability of Th17 cells. We culminate with an exploration of therapeutic potentials in harnessing the power of Th17 cells and their cytokines. Targeted interventions to modulate Th17 responses are emerging as promising strategies for autoimmunity, inflammation, and cancer treatment. By precisely fine-tuning Th17-related pathways, we may unlock new avenues for personalized therapeutic approaches, aiming to restore immune balance, alleviate the challenges of these disorders, and ultimately enhance the quality of life for individuals affected by them.
Collapse
Affiliation(s)
- Dorsa Iraji
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bergithe E Oftedal
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anette S B Wolff
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
45
|
Hatabu T, Pham HHS, Aota W, Fujino S, Nishihara R, Kawamura G, Sakogawa Y, Taniguchi S, Matsubayashi M. Reduction of oocyte shedding and cecal inflammation by 5-aminolevulinic acid daily supplementation in laying hens infected with Eimeria tenella. Anim Sci J 2023; 94:e13806. [PMID: 36627207 DOI: 10.1111/asj.13806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/09/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023]
Abstract
The present study aimed to evaluate the effects of 5-aminolevulinic acid (5-ALA) on Eimeria tenella infection in laying hens. Oocyst shedding and histopathology were evaluated. A reduced oocyst shedding was observed 5 and 7 days post-infection (dpi) in the 5-ALA-administered group, but the total number of oocysts during the first infection period was not different between control and 5-ALA-treated groups. After E. tenella attack infection, the period of oocyst shedding in the 5-ALA-administered group lasted less long than that in controls. During the attack infection period, the total number of fecal oocysts in the 5-ALA-treated group was significantly lower than that in the control group. However, the parasite burden score in hens receiving 5-ALA was higher than that in controls after E. tenella attack infection. The lesion scores at 5 and 30 dpi in the control group were significantly lower than those in the 5-ALA-administered group. Therefore, 5-ALA administration might be beneficial against E. tenella infection in laying hens.
Collapse
Affiliation(s)
- Toshimitsu Hatabu
- Laboratory of Animal Physiology, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Hung Hoang Son Pham
- Department of Veterinary Medicine, Faculty of Animal Science and Veterinary Medicine, University of Agriculture and Forestry, Hue University, Hue, Vietnam
| | - Wataru Aota
- Laboratory of Animal Physiology, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Shota Fujino
- Laboratory of Animal Physiology, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Rio Nishihara
- Laboratory of Animal Physiology, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Go Kawamura
- Laboratory of Animal Physiology, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Yuudai Sakogawa
- Laboratory of Animal Physiology, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Shin Taniguchi
- Agricultural Promotion and Advisory Division, Agriculture, Forestry and Fisheries Department, Hokusatsu Regional Promotion Bureau, Satsumasendai, Japan
| | - Makoto Matsubayashi
- Department of Veterinary Science, Graduate School of Veterinary Sciences, Osaka Metropolitan University, Izumisano, Japan
| |
Collapse
|
46
|
Basso P, Dang EV, Urisman A, Cowen LE, Madhani HD, Noble SM. Deep tissue infection by an invasive human fungal pathogen requires lipid-based suppression of the IL-17 response. Cell Host Microbe 2022; 30:1589-1601.e5. [PMID: 36323314 PMCID: PMC9744107 DOI: 10.1016/j.chom.2022.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 08/17/2022] [Accepted: 10/05/2022] [Indexed: 11/07/2022]
Abstract
Candida albicans is the most common cause of fungal infection in humans. IL-17 is critical for defense against superficial fungal infections, but the role of this response in invasive disease is less understood. We show that C. albicans secretes a lipase, Lip2, that facilitates invasive disease via lipid-based suppression of the IL-17 response. Lip2 was identified as an essential virulence factor in a forward genetic screen in a mouse model of bloodstream infection. Murine infection with C. albicans strains lacking Lip2 display exaggerated IL-17 responses that lead to fungal clearance from solid organs and host survival. Both IL-17 signaling and lipase activity are required for Lip2-mediated suppression. Lip2 inhibits IL-17 production indirectly by suppressing IL-23 production by tissue-resident dendritic cells. The lipase hydrolysis product, palmitic acid, similarly suppresses dendritic cell activation in vitro. Thus, C. albicans suppresses antifungal IL-17 defense in solid organs by altering the tissue lipid milieu.
Collapse
Affiliation(s)
- Pauline Basso
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Eric V Dang
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Anatoly Urisman
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Hiten D Madhani
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Suzanne M Noble
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA; Division of Infectious Diseases, Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
47
|
Paroli M, Spadea L, Caccavale R, Spadea L, Paroli MP, Nante N. The Role of Interleukin-17 in Juvenile Idiopathic Arthritis: From Pathogenesis to Treatment. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:1552. [PMID: 36363508 PMCID: PMC9696590 DOI: 10.3390/medicina58111552] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/22/2022] [Accepted: 10/26/2022] [Indexed: 04/12/2024]
Abstract
Background and Objectives: Interleukin-17 (IL-17) is a cytokine family consisting of six members and five specific receptors. IL-17A was the first member to be identified in 1993. Since then, several studies have elucidated that IL-17 has predominantly pro-inflammatory activity and that its production is involved in both the defense against pathogens and the genesis of autoimmune processes. Materials and Methods: In this review, we provide an overview of the role of interleukin-17 in the pathogenesis of juvenile idiopathic arthritis (JIA) and its relationship with IL-23, the so-called IL-23-IL-17 axis, by reporting updated findings from the scientific literature. Results: Strong evidence supports the role of interleukin-17A in the pathogenesis of JIA after the deregulated production of this interleukin by both T helper 17 (Th17) cells and cells of innate immunity. The blocking of IL-17A was found to improve the course of JIA, leading to the approval of the use of the human anti-IL17A monoclonal antibody secukinumab in the treatment of the JIA subtypes juvenile psoriatic arthritis (JPsA) and enthesitis-related arthritis (ERA). Conclusions: IL-17A plays a central role in the pathogenesis of JIA. Blocking its production with specific biologic drugs enables the effective treatment of this disabling childhood rheumatic disease.
Collapse
Affiliation(s)
- Marino Paroli
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Faculty of Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Luca Spadea
- Post Graduate School of Public Health, University of Siena, 53100 Siena, Italy
| | - Rosalba Caccavale
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Faculty of Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Leopoldo Spadea
- Eye Clinic, Department of Sense Organs, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Pia Paroli
- Eye Clinic, Department of Sense Organs, Sapienza University of Rome, 00185 Rome, Italy
| | - Nicola Nante
- Post Graduate School of Public Health, University of Siena, 53100 Siena, Italy
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| |
Collapse
|
48
|
Taylor TC, Li Y, Li DD, Majumder S, McGeachy MJ, Biswas PS, Gingras S, Gaffen SL. Arid5a Mediates an IL-17-Dependent Pathway That Drives Autoimmunity but Not Antifungal Host Defense. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1138-1145. [PMID: 35940634 PMCID: PMC9492638 DOI: 10.4049/jimmunol.2200132] [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: 02/15/2022] [Accepted: 07/11/2022] [Indexed: 01/04/2023]
Abstract
IL-17 contributes to the pathogenesis of certain autoimmune diseases, but conversely is essential for host defense against fungi. Ab-based biologic drugs that neutralize IL-17 are effective in autoimmunity but can be accompanied by adverse side effects. Candida albicans is a commensal fungus that is the primary causative agent of oropharyngeal and disseminated candidiasis. Defects in IL-17 signaling cause susceptibility to candidiasis in mice and humans. A key facet of IL-17 receptor signaling involves RNA-binding proteins, which orchestrate the fate of target mRNA transcripts. In tissue culture models we showed that the RNA-binding protein AT-rich interaction domain 5A (Arid5a) promotes the stability and/or translation of multiple IL-17-dependent mRNAs. Moreover, during oropharyngeal candidiasis, Arid5a is elevated within the oral mucosa in an IL-17-dependent manner. However, the contribution of Arid5a to IL-17-driven events in vivo is poorly defined. In this study, we used CRISPR-Cas9 to generate mice lacking Arid5a. Arid5a -/- mice were fully resistant to experimental autoimmune encephalomyelitis, an autoimmune setting in which IL-17 signaling drives pathology. Surprisingly, Arid5a -/- mice were resistant to oropharyngeal candidiasis and systemic candidiasis, similar to immunocompetent wild-type mice and contrasting with mice defective in IL-17 signaling. Therefore, Arid5a-dependent signals mediate pathology in autoimmunity and yet are not required for immunity to candidiasis, indicating that selective targeting of IL-17 signaling pathway components may be a viable strategy for development of therapeutics that spare IL-17-driven host defense.
Collapse
Affiliation(s)
- Tiffany C Taylor
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA; and
| | - Yang Li
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA; and
| | - De-Dong Li
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA; and
| | - Saikat Majumder
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA; and
| | - Mandy J McGeachy
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA; and
| | - Partha S Biswas
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA; and
| | | | - Sarah L Gaffen
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA; and
| |
Collapse
|
49
|
Multicomponent Pseudomonas aeruginosa Vaccines Eliciting Th17 Cells and Functional Antibody Responses Confer Enhanced Protection against Experimental Acute Pneumonia in Mice. Infect Immun 2022; 90:e0020322. [PMID: 36069593 PMCID: PMC9584304 DOI: 10.1128/iai.00203-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Gram-negative pathogen Pseudomonas aeruginosa is a common cause of pneumonia in hospitalized patients. Its increasing antibiotic resistance and widespread occurrence present a pressing need for vaccines. We previously showed that a P. aeruginosa type III secretion system protein, PopB, elicits a strong Th17 response in mice after intranasal (IN) immunization and confers antibody-independent protection against pneumonia in mice. In the current study, we evaluated the immunogenicity and protective efficacy in mice of the combination of PopB (purified with its chaperone protein PcrH) and OprF/I, an outer membrane hybrid fusion protein, compared with immunization with the proteins individually either by the intranasal (IN) or subcutaneous (SC) routes. Our results show that after vaccination, a Th17 recall response from splenocytes was detected only in mice vaccinated with PopB/PcrH, either alone or in combination with OprF/I. Mice immunized with the combination of PopB/PcrH and OprF/I had enhanced protection in an acute lethal P. aeruginosa pneumonia model, regardless of vaccine route, compared with mice vaccinated with either alone or adjuvant control. Immunization generated IgG titers against the vaccine proteins and whole P. aeruginosa cells. Interestingly, none of these antisera had opsonophagocytic killing activity, but antisera from mice immunized with vaccines containing OprF/I, had the ability to block IFN-γ binding to OprF/I, a known virulence mechanism. Hence, vaccines combining PopB/PcrH with OprF/I that elicit functional antibodies lead to a broadly and potently protective vaccine against P. aeruginosa pulmonary infections.
Collapse
|
50
|
Falk-Mahapatra R, Gollnick SO. Photodynamic Therapy-Induced Cyclooxygenase 2 Expression in Tumor-Draining Lymph Nodes Regulates B-Cell Expression of Interleukin 17 and Neutrophil Infiltration. Photochem Photobiol 2022; 98:1207-1214. [PMID: 35103990 PMCID: PMC9484206 DOI: 10.1111/php.13601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/27/2022] [Indexed: 11/27/2022]
Abstract
Photodynamic therapy (PDT) is an effective anticancer modality approved by the U.S. Food and Drug Administration (FDA). Antitumor immunity can be augmented during PDT by inducing sterile inflammation in an acute manner, and this process is characterized by interleukin 17 (IL-17)-mediated neutrophil infiltration to tumor-draining lymph nodes (TDLNs). However, the inflammatory factors that influence IL-17 expression in TDLNs are poorly understood. Prior studies have linked the cyclooxygenase 2 (COX2)-driven prostaglandin E2 (PGE2) pathway to IL-17 expression. Here, we report that an immune-activating PDT regimen (imPDT) induces COX2/PGE2 expression in TDLNs, whereby IL-17 expression is facilitated without corresponding effects on the expression of RORγt, the transcriptional driver of the canonical IL-17 pathway. Pharmacologic inhibition with NS398, a COX2 inhibitor, was utilized to demonstrate that imPDT-induced COX2 regulates RORγt-independent expression of IL-17 by B cells and neutrophil entry into TDLNs. Depletion of B cells prior to imPDT significantly reduced neutrophil entry into TDLNs following treatment, and diminishes the efficacy of imPDT, which is dependent upon antitumor immunity. These findings are suggestive of a novel role for B cells in the augmentation of antitumor immunity by imPDT.
Collapse
Affiliation(s)
- Riddhi Falk-Mahapatra
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY 14263, USA
| | - Sandra O. Gollnick
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY 14263, USA,Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY 14263, USA,Corresponding author: (Sandra O. Gollnick)
| |
Collapse
|