1
|
Zhao Y, Zhou Z, Cai G, Zhang D, Yu X, Li D, Li S, Zhang Z, Zhang D, Luo J, Hu Y, Gao A, Zhang H. Systemic infection by Candida albicans requires FASN-α subunit induced cell wall remodeling to perturb immune response. PLoS Pathog 2025; 21:e1012865. [PMID: 40138332 PMCID: PMC11940687 DOI: 10.1371/journal.ppat.1012865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/29/2024] [Indexed: 03/29/2025] Open
Abstract
Invasive fungal infections are a leading cause of mortality and morbidity in patients with severely impaired host defenses, while treatment options remain limited. Fatty acid synthase (FASN), the key enzyme regulating de novo biosynthesis of fatty acids, is crucial for the lethal infection of fungi; however, its pathogenic mechanism is still far from clear. Here, we identified the α subunit of FASN as a potential immunotherapeutic target against systemic Candida albicans infection. The avirulence of the encoded gene (FAS2) -deleted mutant in a mouse model of systemic candidiasis is not due to its fitness defects, because sufficient exogenous fatty acids in serum can overcome FASN inhibition. However, the FAS2-deleted mutant displays increased circulating innate immune responses and enhances activated neutrophil fungicidal activity through the unmasking of immunogenic cell wall epitopes via the Rho-1 dependent Mkc1-MAPK signaling pathway, which facilitates fungal clearance, reduces renal tissue damage and inflammatory cell infiltration, ultimately lowers fungal pathogenicity. Priming with the FAS2-deleted mutant provided significant protection against subsequent lethal infection with wild-type C. albicans in mice as early as one week, and it was well-tolerated with limited toxicity. Our findings indicate that the FASN-α subunit plays key roles in the regulation of neutrophil-associated antifungal immunity and could be a potential target for immunotherapeutic intervention.
Collapse
Affiliation(s)
- Yajing Zhao
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Zhishan Zhou
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Guiyue Cai
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Dandan Zhang
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Xiaoting Yu
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Dongmei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington District of Columbia, United States of America
| | - Shuixiu Li
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Zhanpeng Zhang
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Dongli Zhang
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Jiyao Luo
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Yunfeng Hu
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Aili Gao
- Guangzhou Dermatology Hospital, Guangzhou, China
| | - Hong Zhang
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| |
Collapse
|
2
|
Lee KMC, Lupancu T, Chang L, Manthey CL, Zeeman M, Fourie AM, Hamilton JA. IL-23 regulation of myeloid cell biology during inflammation. Cytokine 2024; 179:156619. [PMID: 38669908 DOI: 10.1016/j.cyto.2024.156619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/15/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024]
Abstract
Interleukin (IL)-23 is implicated in the pathogenesis of several inflammatory diseases and is usually linked with helper T cell (Th17) biology. However, there is some data linking IL-23 with innate immune biology in such diseases. We therefore examined the effects of IL-23p19 genetic deletion and/or neutralization on in vitro macrophage activation and in an innate immune-driven peritonitis model. We report that endogenous IL-23 was required for maximal macrophage activation by zymosan as determined by pro-inflammatory cytokine production, including a dramatic upregulation of granulocyte-colony stimulating factor (G-CSF). Furthermore, both IL-23p19 genetic deletion and neutralization in zymosan-induced peritonitis (ZIP) led to a specific reduction in the neutrophil numbers, as well as a reduction in the G-CSF levels in exudate fluids. We conclude that endogenous IL-23 can contribute significantly to macrophage activation during an inflammatory response, mostly likely via an autocrine/paracrine mechanism; of note, endogenous IL-23 can directly up-regulate macrophage G-CSF expression, which in turn is likely to contribute to the regulation of IL-23-dependent neutrophil number and function during an inflammatory response, with potential significance for IL-23 targeting particularly in neutrophil-associated inflammatory diseases.
Collapse
Affiliation(s)
- Kevin M-C Lee
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria 3050, Australia.
| | - Tanya Lupancu
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria 3050, Australia
| | - Leon Chang
- Janssen Research & Development, LLC, La Jolla CA & Spring House PA, USA
| | - Carl L Manthey
- Janssen Research & Development, LLC, La Jolla CA & Spring House PA, USA
| | - Martha Zeeman
- Janssen Research & Development, LLC, La Jolla CA & Spring House PA, USA
| | - Anne M Fourie
- Janssen Research & Development, LLC, La Jolla CA & Spring House PA, USA
| | - John A Hamilton
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria 3050, Australia
| |
Collapse
|
3
|
Xia X, Zhu L, Xu M, Lei Z, Yu H, Li G, Wang X, Jia H, Yin Z, Huang F, Gao Y. ANKRD22 promotes resolution of psoriasiform skin inflammation by antagonizing NIK-mediated IL-23 production. Mol Ther 2024; 32:1561-1577. [PMID: 38454607 PMCID: PMC11081937 DOI: 10.1016/j.ymthe.2024.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 12/13/2023] [Accepted: 03/05/2024] [Indexed: 03/09/2024] Open
Abstract
Inflammation resolution is an essential process for preventing the development of chronic inflammatory diseases. However, the mechanisms that regulate inflammation resolution in psoriasis are not well understood. Here, we report that ANKRD22 is an endogenous negative orchestrator of psoriasiform inflammation because ANKRD22-deficient mice are more susceptible to IMQ-induced psoriasiform inflammation. Mechanistically, ANKRD22 deficiency leads to excessive activation of the TNFRII-NIK-mediated noncanonical NF-κB signaling pathway, resulting in the hyperproduction of IL-23 in DCs. This is due to ANKRD22 being a negative feedback regulator for NIK because it physically binds to and assists in the degradation of accumulated NIK. Clinically, ANKRD22 is negatively associated with IL-23A expression and psoriasis severity. Of greater significance, subcutaneous administration of an AAV carrying ANKRD22-overexpression vector effectively hastens the resolution of psoriasiform skin inflammation. Our findings suggest ANKRD22, an endogenous supervisor of NIK, is responsible for inflammation resolution in psoriasis, and may be explored in the context of psoriasis therapy.
Collapse
Affiliation(s)
- Xichun Xia
- Institute of Dermatology and Venereal Diseases, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China; The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China; Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Jinan University, Zhuhai 519050, China
| | - Leqing Zhu
- Guangzhou Laboratory, Bioland, Guangzhou 510005, China
| | - Miaomiao Xu
- The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China; Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Jinan University, Zhuhai 519050, China
| | - Zhiwei Lei
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China
| | - Hai Yu
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Guangqiang Li
- The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China
| | - Xiao Wang
- The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China
| | - Hongling Jia
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Zhinan Yin
- The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China; Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Jinan University, Zhuhai 519050, China.
| | - Fang Huang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Jinan University, Zhuhai 519050, China.
| | - Yunfei Gao
- Department of Oncology, Research Center of Cancer Diagnosis and Therapy, the First Affiliated Hospital, Jinan University, Guangzhou 510632, China; The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China.
| |
Collapse
|
4
|
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
|
5
|
Cheng A, Kashyap A, Salvator H, Rosen LB, Colby D, Ardeshir-Larijani F, Loehrer PJ, Ding L, Lugo Reyes SO, Riminton S, Ballman M, Rocco JM, Marciano BE, Freeman AF, Browne SK, Hsu AP, Zelazny A, Rajan A, Sereti I, Zerbe CS, Lionakis MS, Holland SM. Anti-Interleukin-23 Autoantibodies in Adult-Onset Immunodeficiency. N Engl J Med 2024; 390:1105-1117. [PMID: 38507753 DOI: 10.1056/nejmoa2210665] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
BACKGROUND Autoantibodies against interleukin-12 (anti-interleukin-12) are often identified in patients with thymoma, but opportunistic infections develop in only some of these patients. Interleukin-12 (with subunits p40 and p35) shares a common subunit with interleukin-23 (subunits p40 and p19). In a patient with disseminated Burkholderia gladioli infection, the identification of both anti-interleukin-23 and anti-interleukin-12 prompted further investigation. METHODS Among the patients (most of whom had thymoma) who were known to have anti-interleukin-12, we screened for autoantibodies against interleukin-23 (anti-interleukin-23). To validate the potential role of anti-interleukin-23 with respect to opportunistic infection, we tested a second cohort of patients with thymoma as well as patients without either thymoma or known anti-interleukin-12 who had unusual infections. RESULTS Among 30 patients with anti-interleukin-12 who had severe mycobacterial, bacterial, or fungal infections, 15 (50%) also had autoantibodies that neutralized interleukin-23. The potency of such neutralization was correlated with the severity of these infections. The neutralizing activity of anti-interleukin-12 alone was not associated with infection. In the validation cohort of 91 patients with thymoma, the presence of anti-interleukin-23 was associated with infection status in 74 patients (81%). Overall, neutralizing anti-interleukin-23 was detected in 30 of 116 patients (26%) with thymoma and in 30 of 36 patients (83%) with disseminated, cerebral, or pulmonary infections. Anti-interleukin-23 was present in 6 of 32 patients (19%) with severe intracellular infections and in 2 of 16 patients (12%) with unusual intracranial infections, including Cladophialophora bantiana and Mycobacterium avium complex. CONCLUSIONS Among patients with a variety of mycobacterial, bacterial, or fungal infections, the presence of neutralizing anti-interleukin-23 was associated with severe, persistent opportunistic infections. (Funded by the National Institute of Allergy and Infectious Diseases and others.).
Collapse
Affiliation(s)
- Aristine Cheng
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Anuj Kashyap
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Helene Salvator
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Lindsey B Rosen
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Devon Colby
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Fatemeh Ardeshir-Larijani
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Patrick J Loehrer
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Li Ding
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Saul O Lugo Reyes
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Sean Riminton
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Madison Ballman
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Joseph M Rocco
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Beatriz E Marciano
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Alexandra F Freeman
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Sarah K Browne
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Amy P Hsu
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Adrian Zelazny
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Arun Rajan
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Irini Sereti
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Christa S Zerbe
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Michail S Lionakis
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Steven M Holland
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| |
Collapse
|
6
|
Yazdi M, Behnaminia N, Nafari A, Sepahvand A. Genetic Susceptibility to Fungal Infections. Adv Biomed Res 2023; 12:248. [PMID: 38192892 PMCID: PMC10772798 DOI: 10.4103/abr.abr_259_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/15/2022] [Accepted: 08/20/2022] [Indexed: 01/10/2024] Open
Abstract
Reports of fungal infections have increased over the past decades, making them a major threat to human health. In this study, we review the effects of genetic defects on susceptibility to fungal diseases. To identify all relevant literature, we searched Google Scholar, PubMed, and Scopus and profiled studies published between 2008 and 2021. The results of several studies conducted on this subject have shown the significant effects of genetic variations such as hyper-IgE syndrome, Autoimmune polyendocrinopathy candidiasis ectodermal dystrophy syndrome, dectin-1 deficiency, CARD9 mutations, STAT1 mutations, and IL17 mutationson the host immune system's response, which has an important impact on susceptibility to fungal infections. The underlying immune system-related genetic profile affects the susceptibility of individuals to different fungal infections; therefore, this subject should be further studied for better treatment of fungal diseases.
Collapse
Affiliation(s)
- Mohammad Yazdi
- Department of Biochemistry, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Nima Behnaminia
- Student Research Committee, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Amirhossein Nafari
- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Asghar Sepahvand
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
7
|
Chen T, Feng Y, Sun W, Zhao G, Wu H, Cheng X, Zhao F, Zhang L, Zheng Y, Zhan P, Zhao W, Liu B, Gao C. The nucleotide receptor STING translocates to the phagosomes to negatively regulate anti-fungal immunity. Immunity 2023; 56:1727-1742.e6. [PMID: 37379835 DOI: 10.1016/j.immuni.2023.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 04/26/2023] [Accepted: 06/02/2023] [Indexed: 06/30/2023]
Abstract
STING (stimulator of interferon genes) exerts protective cellular responses to viral infection via induction of interferon production and autophagy. Here, we report the role of STING in modulating the immune responses toward fungal infection. Upon Candida albicans stimulation, STING transited alongside the endoplasmic reticulum (ER) to the phagosomes. In phagosomes, STING directly bound with Src via the N-terminal 18 amino acids of STING, and this binding prevented Src from recruiting and phosphorylating Syk. Consistently, Syk-associated signaling and production of pro-inflammatory cytokines and chemokines were increased in mouse BMDCs (bone-marrow-derived dendritic cells) lacking STING with fungal treatment. STING deficiency improved anti-fungal immunity in systemic C. albicans infection. Importantly, administration of the N-terminal 18-aa (amino acid) peptide of STING improved host outcomes in disseminated fungal infection. Overall, our study identifies a previously unrecognized function of STING in negatively regulating anti-fungal immune responses and offers a potential therapeutic strategy for controlling C. albicans infection.
Collapse
Affiliation(s)
- Tian Chen
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Pathogenic Biology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Yiting Feng
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Wanwei Sun
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Guimin Zhao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Han Wu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Xiaochen Cheng
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Fabao Zhao
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, P.R. China
| | - Lei Zhang
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Yi Zheng
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Peng Zhan
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, P.R. China
| | - Wei Zhao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Pathogenic Biology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Bingyu Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China.
| |
Collapse
|
8
|
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: 27] [Impact Index Per Article: 13.5] [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
|
9
|
Fink S, Fischer M, Spange S, Beier O, Horn K, Tittelbach J, Wiegand C. Cold Atmospheric Plasma Exerts Antimicrobial Effects in a 3D Skin Model of Cutaneous Candidiasis. Antibiotics (Basel) 2023; 12:antibiotics12050933. [PMID: 37237836 DOI: 10.3390/antibiotics12050933] [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: 03/28/2023] [Revised: 05/02/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Cutaneous candidiasis is characterized by an overgrowth of Candida leading to skin inflammation and infection. Similar to bacteria, Candida can develop tolerance to common antifungal drugs. Cold atmospheric plasma (CAP), with its proven antimicrobial properties, offers a promising alternative to the prevailing methods. Because of plasma heterogeneity each new device must be tested individually for its effectiveness. Antimicrobial activity is usually studied using planktonic microorganisms or animal models, making it difficult to extrapolate the results to the human system. Therefore, a 3D skin model of cutaneous candidiasis for the antimicrobial testing of CAP was established. First, the reaction of the 3D-skin model to Candida infection was examined using various histological and molecular-biological methods. Infection with C. albicans resulted in increased expression and secretion of pro-inflammatory cytokines and augmented expression of antimicrobial peptides. Within 48 h, hyphal growth spread throughout the model and caused tissue damage. Second, the CAP treatment was employed. It was shown that CAP significantly reduced the spread of the yeast in the infected skin models as well as decreased the expression and secretion of the infection markers. The plasma device exhibited a high antifungal activity by completely inhibiting hyphal growth and reducing inflammation at the highest treatment duration.
Collapse
Affiliation(s)
- Sarah Fink
- Department of Dermatology, Jena University Hospital, Friedrich Schiller University, 07747 Jena, Germany
| | - Michael Fischer
- Institute of Micro- and Nanotechnologies, Ilmenau University of Technology, 98639 Ilmenau, Germany
| | | | | | | | - Jörg Tittelbach
- Department of Dermatology, Jena University Hospital, Friedrich Schiller University, 07747 Jena, Germany
| | - Cornelia Wiegand
- Department of Dermatology, Jena University Hospital, Friedrich Schiller University, 07747 Jena, Germany
| |
Collapse
|
10
|
Lopes JP, Lionakis MS. Pathogenesis and virulence of Candida albicans. Virulence 2022; 13:89-121. [PMID: 34964702 PMCID: PMC9728475 DOI: 10.1080/21505594.2021.2019950] [Citation(s) in RCA: 191] [Impact Index Per Article: 63.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022] Open
Abstract
Candida albicans is a commensal yeast fungus of the human oral, gastrointestinal, and genital mucosal surfaces, and skin. Antibiotic-induced dysbiosis, iatrogenic immunosuppression, and/or medical interventions that impair the integrity of the mucocutaneous barrier and/or perturb protective host defense mechanisms enable C. albicans to become an opportunistic pathogen and cause debilitating mucocutaneous disease and/or life-threatening systemic infections. In this review, we synthesize our current knowledge of the tissue-specific determinants of C. albicans pathogenicity and host immune defense mechanisms.
Collapse
Affiliation(s)
- José Pedro Lopes
- From the Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA
| | - Michail S. Lionakis
- From the Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA
| |
Collapse
|
11
|
Lubkin A, Lionakis MS. Candida lipase packs a punch against IL-17. Cell Host Microbe 2022; 30:1503-1505. [DOI: 10.1016/j.chom.2022.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
12
|
Pathogenesis of Fungal Infections in the Central Nervous System: Host and Pathogen Factors in Neurotropism. CURRENT FUNGAL INFECTION REPORTS 2022. [DOI: 10.1007/s12281-022-00444-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
13
|
Millet N, Solis NV, Aguilar D, Lionakis MS, Wheeler RT, Jendzjowsky N, Swidergall M. IL-23 signaling prevents ferroptosis-driven renal immunopathology during candidiasis. Nat Commun 2022; 13:5545. [PMID: 36138043 PMCID: PMC9500047 DOI: 10.1038/s41467-022-33327-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 09/13/2022] [Indexed: 01/04/2023] Open
Abstract
During infection the host relies on pattern-recognition receptors to sense invading fungal pathogens to launch immune defense mechanisms. While fungal recognition and immune effector responses are organ and cell type specific, during disseminated candidiasis myeloid cells exacerbate collateral tissue damage. The β-glucan receptor ephrin type-A 2 receptor (EphA2) is required to initiate mucosal inflammatory responses during oral Candida infection. Here we report that EphA2 promotes renal immunopathology during disseminated candidiasis. EphA2 deficiency leads to reduced renal inflammation and injury. Comprehensive analyses reveal that EphA2 restrains IL-23 secretion from and migration of dendritic cells. IL-23 signaling prevents ferroptotic host cell death during infection to limit inflammation and immunopathology. Further, host cell ferroptosis limits antifungal effector functions via releasing the lipid peroxidation product 4-hydroxynonenal to induce various forms of cell death. Thus, we identify ferroptotic cell death as a critical pathway of Candida-mediated renal immunopathology that opens a new avenue to tackle Candida infection and inflammation.
Collapse
Affiliation(s)
- Nicolas Millet
- grid.239844.00000 0001 0157 6501Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA USA ,grid.513199.6The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA USA
| | - Norma V. Solis
- grid.239844.00000 0001 0157 6501Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA USA ,grid.513199.6The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA USA
| | - Diane Aguilar
- grid.513199.6The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA USA
| | - Michail S. Lionakis
- grid.419681.30000 0001 2164 9667Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD USA
| | - Robert T. Wheeler
- grid.21106.340000000121820794Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME USA
| | - Nicholas Jendzjowsky
- grid.513199.6The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA USA ,grid.19006.3e0000 0000 9632 6718David Geffen School of Medicine at UCLA, Los Angeles, CA USA
| | - Marc Swidergall
- grid.239844.00000 0001 0157 6501Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA USA ,grid.513199.6The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA USA ,grid.19006.3e0000 0000 9632 6718David Geffen School of Medicine at UCLA, Los Angeles, CA USA
| |
Collapse
|
14
|
Progranulin aggravates lethal Candida albicans sepsis by regulating inflammatory response and antifungal immunity. PLoS Pathog 2022; 18:e1010873. [PMID: 36121866 PMCID: PMC9521894 DOI: 10.1371/journal.ppat.1010873] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/29/2022] [Accepted: 09/09/2022] [Indexed: 11/19/2022] Open
Abstract
Candida albicans is the most frequent pathogen of fungal sepsis associated with substantial mortality in critically ill patients and those who are immunocompromised. Identification of novel immune-based therapeutic targets from a better understanding of its molecular pathogenesis is required. Here, we reported that the production of progranulin (PGRN) levels was significantly increased in mice after invasive C.albicans infection. Mice that lacked PGRN exhibited attenuated kidney injury and increased survival upon a lethal systemic infection with C. albicans. In mice, PGRN deficiency protected against systemic candidiasis by decreasing aberrant inflammatory reactions that led to renal immune cell apoptosis and kidney injury, and by enhancing antifungal capacity of macrophages and neutrophils that limited fungal burden in the kidneys. PGRN in hematopoietic cell compartment was important for this effect. Moreover, anti-PGRN antibody treatment limited renal inflammation and fungal burden and prolonged survival after invasive C. albicans infection. In vitro, PGRN loss increased phagocytosis, phagosome formation, reactive oxygen species production, neutrophil extracellular traps release, and killing activity in macrophages or neutrophils. Mechanistic studies demonstrated that PGRN loss up-regulated Dectin-2 expression, and enhanced spleen tyrosine kinase phosphorylation and extracellular signal-regulated kinase activation in macrophages and neutrophils. In summary, we identified PGRN as a critical factor that contributes to the immunopathology of invasive C.albicans infection, suggesting that targeting PGRN might serve as a novel treatment for fungal infection.
Collapse
|
15
|
A Fun-Guide to Innate Immune Responses to Fungal Infections. J Fungi (Basel) 2022; 8:jof8080805. [PMID: 36012793 PMCID: PMC9409918 DOI: 10.3390/jof8080805] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 11/22/2022] Open
Abstract
Immunocompromised individuals are at high risk of developing severe fungal infections with high mortality rates, while fungal pathogens pose little risk to most healthy people. Poor therapeutic outcomes and growing antifungal resistance pose further challenges for treatments. Identifying specific immunomodulatory mechanisms exploited by fungal pathogens is critical for our understanding of fungal diseases and development of new therapies. A gap currently exists between the large body of literature concerning the innate immune response to fungal infections and the potential manipulation of host immune responses to aid clearance of infection. This review considers the innate immune mechanisms the host deploys to prevent fungal infection and how these mechanisms fail in immunocompromised hosts. Three clinically relevant fungal pathogens (Candida albicans, Cryptococcus spp. and Aspergillus spp.) will be explored. This review will also examine potential mechanisms of targeting the host therapeutically to improve outcomes of fungal infection.
Collapse
|
16
|
Kischkel B, Lopes-Bezerra L, Taborda CP, A. B. Joosten L, Cristina dos Santos J, Netea MG. Differential recognition and cytokine induction by the peptidorhamnomannan from Sporothrix brasiliensis and S. schenckii. Cell Immunol 2022; 378:104555. [DOI: 10.1016/j.cellimm.2022.104555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/26/2022] [Accepted: 05/24/2022] [Indexed: 11/03/2022]
|
17
|
Hasan S, Hu L, Williams O, Eklund EA. Ruxolitinib ameliorates progressive anemia and improves survival during episodes of emergency granulopoiesis in Fanconi C−/− mice. Exp Hematol 2022; 109:55-67.e2. [PMID: 35278531 PMCID: PMC9064927 DOI: 10.1016/j.exphem.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 11/15/2022]
Abstract
Fanconi anemia (FA) is an inherited disorder of DNA repair with hematologic manifestations that range from anemia to bone marrow failure to acute myeloid leukemia. In a murine model of FA (Fancc-/- mice), we found bone marrow failure was accelerated by repeated attempts to induce emergency (stress) granulopoiesis, the process for granulocyte production during the innate immune response. Fancc-/- mice exhibited an impaired granulocytosis response and died with profound anemia during repeated challenge. In the current study, we found erythropoiesis and serum erythropoietin decreased in Fancc-/- and wild-type (Wt) mice as emergency granulopoiesis peaked. Serum erythropoietin returned to baseline during steady-state resumption, and compensatory proliferation of erythroid progenitors was associated with DNA damage and apoptosis in Fancc-/- mice, but not Wt mice. The erythropoietin receptor activates Janus kinase 2 (Jak2), and we found treatment of Fancc-/- mice with ruxolitinib (Jak1/2-inhibitor) decreased anemia, enhanced granulocytosis, delayed clonal progression and prolonged survival during repeated emergency granulopoiesis episodes. This was associated with a decrease in DNA damage and apoptosis in Fancc-/- erythroid progenitors during this process. Transcriptome analysis of these cells identified enhanced activity of pathways for metabolism of reactive oxygen species, and decreased apoptosis- and autophagy-related pathways, as major ruxolitinib-effects in Fancc-/- mice. In contrast, ruxolitinib influenced primarily pathways involved in proliferation and differentiation in Wt mice. Ruxolitinib is approved for treatment of myeloproliferative disorders and graft-versus-host disease, suggesting the possibility of translational use as a bone marrow protectant in FA.
Collapse
Affiliation(s)
- Shirin Hasan
- Department of Medicine, Northwestern University, Chicago, IL
| | - Liping Hu
- Department of Medicine, Northwestern University, Chicago, IL
| | | | - Elizabeth A Eklund
- Department of Medicine, Northwestern University, Chicago, IL; Jesse Brown VA Medical Center, Chicago, IL.
| |
Collapse
|
18
|
Nguyen NZN, Tran VG, Baek J, Kim Y, Youn EH, Na SW, Park SJ, Seo SK, Kwon B. IL-33 Coordinates Innate Defense to Systemic Candida albicans Infection by Regulating IL-23 and IL-10 in an Opposite Way. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:660-671. [PMID: 35022276 DOI: 10.4049/jimmunol.2100495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 10/06/2021] [Indexed: 11/19/2022]
Abstract
Invasive candidiasis has high mortality rates in immunocompromised patients, causing serious health problems. In mouse models, innate immunity protects the host by rapidly mobilizing a variety of resistance and tolerance mechanisms to systemic Candida albicans infection. We have previously demonstrated that exogenous IL-33 regulates multiple steps of innate immunity involving resistance and tolerance processes. In this study, we systematically analyzed the in vivo functions of endogenous IL-33 using Il33 -/- mice and in vitro immune cell culture. Tubular epithelial cells mainly secreted IL-33 in response to systemic C. albicans infection. Il33 -/- mice showed increased mortality and morbidity, which were due to impaired fungal clearance. IL-33 initiated an innate defense mechanism by costimulating dendritic cells to produce IL-23 after systemic C. albicans infection, which in turn promoted the phagocytosis of neutrophils through secretion of GM-CSF by NK cells. The susceptibility of Il33 -/- mice was also associated with increased levels of IL-10, and neutralization of IL-10 resulted in enhanced fungal clearance in Il33 -/- mice. However, depletion of IL-10 overrode the effect of IL-33 on fungal clearance. In Il10 -/- mouse kidneys, MHC class II+F4/80+ macrophages were massively differentiated after C. albicans infection, and these cells were superior to MHC class II-F4/80+ macrophages that were preferentially differentiated in wild-type mouse kidneys in killing of extracellular hyphal C. albicans Taken together, our results identify IL-33 as critical early regulator controlling a serial downstream signaling events of innate defense to C. albicans infection.
Collapse
Affiliation(s)
- Nu Z N Nguyen
- BK21 Integrated Immunomodulation Education and Research Team, School of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea
| | - Vuvi G Tran
- Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA
| | - Jiyeon Baek
- Biomedical Research Center, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan, Republic of Korea
| | - Younghee Kim
- Biomedical Research Center, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan, Republic of Korea
| | - Eun H Youn
- Department of Microbiology and Immunology, College of Medicine, Inje University, Busan, Republic of Korea
| | - Seung W Na
- Biomedical Research Center, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan, Republic of Korea.,Division of Pulmonology, Department of Internal Medicine, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan, Republic of Korea; and
| | - Sang J Park
- Biomedical Research Center, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan, Republic of Korea.,Department of Surgery, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan, Republic of Korea
| | - Su-Kil Seo
- Department of Microbiology and Immunology, College of Medicine, Inje University, Busan, Republic of Korea;
| | - Byungsuk Kwon
- BK21 Integrated Immunomodulation Education and Research Team, School of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea; .,Biomedical Research Center, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan, Republic of Korea
| |
Collapse
|
19
|
Šuler Baglama Š, Trčko K. Cutaneous adverse effects of biologic drugs in psoriasis: a literature review. ACTA DERMATOVENEROLOGICA ALPINA PANNONICA ET ADRIATICA 2021. [DOI: 10.15570/actaapa.2021.35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
20
|
Gordon KB, Lebwohl M, Papp KA, Bachelez H, Wu JJ, Langley RG, Blauvelt A, Kaplan B, Shah M, Zhao Y, Sinvhal R, Reich K. Long-term safety of risankizumab from 17 clinical trials in patients with moderate-to-severe plaque psoriasis. Br J Dermatol 2021; 186:466-475. [PMID: 34652810 PMCID: PMC9298814 DOI: 10.1111/bjd.20818] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2021] [Indexed: 01/14/2023]
Abstract
Background Risankizumab has demonstrated efficacy and safety in patients with moderate‐to‐severe plaque psoriasis in randomized clinical trials. Objectives To evaluate safety data from risankizumab psoriasis phase I–III clinical trials. Methods Short‐term safety (through week 16) was analysed using integrated data from five phase II and III clinical trials. Long‐term safety was evaluated using integrated data from 17 phase I–III completed and ongoing trials. Results Short‐term safety analyses included 1306 patients receiving risankizumab 150 mg and 300 patients receiving placebo [402·2 and 92·0 patient‐years (PY) of exposure, respectively]. Long‐term analyses included 3072 risankizumab‐treated patients (exposure: 7927 PY). The median (excluding four outliers) treatment duration was 2·9 years (range 2 days to 5·9 years). Exposure‐adjusted adverse event rates did not increase with long‐term treatment (318 vs. 171 events per 100 PY for short‐ and long‐term analyses). With long‐term risankizumab treatment, rates of serious adverse events were 7·8 per 100 PY, serious infections 1·2 per 100 PY, nonmelanoma skin cancer (NMSC) 0·7 per 100 PY, malignant tumours excluding NMSC 0·5 per 100 PY, and adjudicated major adverse cardiovascular events 0·3 per 100 PY, with no important identified risks. Limitations include that the study inclusion and exclusion criteria varied and that three studies enrolled ≤ 50 patients. Conclusions Risankizumab demonstrated a favourable safety profile over short‐ and long‐term treatment in patients with moderate‐to‐severe psoriasis. Whatis already known about this topic? In clinical trials of patients with moderate‐to‐severe plaque psoriasis, risankizumab, a selective interleukin‐23 inhibitor, was well tolerated and efficacious.
Whatdoes this study add? In this comprehensive evaluation of risankizumab safety in patients with moderate‐to‐severe psoriasis, adverse event rates were comparable between risankizumab (n = 1306, 402 patient‐years) and placebo (n = 300, 92 patient‐years) in the short‐term (16‐week) analysis set, and were consistent with those in the long‐term analysis (n = 3072, 7927 patient‐years of risankizumab exposure). These findings are consistent with the known safety profile of risankizumab and support its long‐term use in moderate‐to‐severe psoriasis.
Linked Comment: S. Mirali et al. Br J Dermatol 2022; 186:394–395. Plain language summary available online
Collapse
Affiliation(s)
- K B Gordon
- Department of Dermatology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - M Lebwohl
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - K A Papp
- K Papp Clinical Research and Probity Medical Research, Waterloo, ON, Canada
| | - H Bachelez
- Hôpital Saint-Louis, AP-HP, Université de Paris, Paris, France
| | - J J Wu
- Dermatology Research and Education Foundation, Irvine, CA, USA
| | - R G Langley
- Division of Dermatology, Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - A Blauvelt
- Oregon Medical Research Center, Portland, OR, USA
| | - B Kaplan
- AbbVie Inc., North Chicago, IL, USA
| | - M Shah
- AbbVie Inc., North Chicago, IL, USA
| | - Y Zhao
- AbbVie Inc., North Chicago, IL, USA
| | | | - K Reich
- Center for Translational Research in Inflammatory Skin Diseases, Institute for Health Services Research in Dermatology and Nursing, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
21
|
The Role of B-Cells and Antibodies against Candida Vaccine Antigens in Invasive Candidiasis. Vaccines (Basel) 2021; 9:vaccines9101159. [PMID: 34696267 PMCID: PMC8540628 DOI: 10.3390/vaccines9101159] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 01/08/2023] Open
Abstract
Systemic candidiasis is an invasive fungal infection caused by members of the genus Candida. The recent emergence of antifungal drug resistance and increased incidences of infections caused by non-albicans Candida species merit the need for developing immune therapies against Candida infections. Although the role of cellular immune responses in anti-Candida immunity is well established, less is known about the role of humoral immunity against systemic candidiasis. This review summarizes currently available information on humoral immune responses induced by several promising Candida vaccine candidates, which have been identified in the past few decades. The protective antibody and B-cell responses generated by polysaccharide antigens such as mannan, β-glucan, and laminarin, as well as protein antigens like agglutinin-like sequence gene (Als3), secreted aspartyl proteinase (Sap2), heat shock protein (Hsp90), hyphally-regulated protein (Hyr1), hyphal wall protein (Hwp1), enolase (Eno), phospholipase (PLB), pyruvate kinase (Pk), fructose bisphosphate aldolase (Fba1), superoxide dismutase gene (Sod5) and malate dehydrogenase (Mdh1), are outlined. As per studies reviewed, antibodies induced in response to leading Candida vaccine candidates contribute to protection against systemic candidiasis by utilizing a variety of mechanisms such as opsonization, complement fixation, neutralization, biofilm inhibition, direct candidacidal activity, etc. The contributions of B-cells in controlling fungal infections are also discussed. Promising results using anti-Candida monoclonal antibodies for passive antibody therapy reinforces the need for developing antibody-based therapeutics including anti-idiotypic antibodies, single-chain variable fragments, peptide mimotopes, and antibody-derived peptides. Future research involving combinatorial immunotherapies using humanized monoclonal antibodies along with antifungal drugs/cytokines may prove beneficial for treating invasive fungal infections.
Collapse
|
22
|
Lung eosinophils elicited during allergic and acute aspergillosis express RORγt and IL-23R but do not require IL-23 for IL-17 production. PLoS Pathog 2021; 17:e1009891. [PMID: 34464425 PMCID: PMC8437264 DOI: 10.1371/journal.ppat.1009891] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 09/13/2021] [Accepted: 08/12/2021] [Indexed: 12/19/2022] Open
Abstract
Exposure to the mold, Aspergillus, is ubiquitous and generally has no adverse consequences in immunocompetent persons. However, invasive and allergic aspergillosis can develop in immunocompromised and atopic individuals, respectively. Previously, we demonstrated that mouse lung eosinophils produce IL-17 in response to stimulation by live conidia and antigens of A. fumigatus. Here, we utilized murine models of allergic and acute pulmonary aspergillosis to determine the association of IL-23, IL-23R and RORγt with eosinophil IL-17 expression. Following A. fumigatus stimulation, a population of lung eosinophils expressed RORγt, the master transcription factor for IL-17 regulation. Eosinophil RORγt expression was demonstrated by flow cytometry, confocal microscopy, western blotting and an mCherry reporter mouse. Both nuclear and cytoplasmic localization of RORγt in eosinophils were observed, although the former predominated. A population of lung eosinophils also expressed IL-23R. While expression of IL-23R was positively correlated with expression of RORγt, expression of RORγt and IL-17 was similar when comparing lung eosinophils from A. fumigatus-challenged wild-type and IL-23p19-/- mice. Thus, in allergic and acute models of pulmonary aspergillosis, lung eosinophils express IL-17, RORγt and IL-23R. However, IL-23 is dispensable for production of IL-17 and RORγt. Humans regularly inhale spores of Aspergillus fumigatus, a common environmental fungus. While such exposure is of little consequence to most, in persons with impaired immune systems it can cause a spectrum of diseases ranging from invasive aspergillosis to allergic aspergillosis. A type of white blood cell called the eosinophil is a defining feature of allergic aspergillosis. Despite their importance, the contribution of eosinophils to this disease state is poorly understood. We previously demonstrated that eosinophils produce the cytokine IL-17 in murine models of aspergillosis models. Here, we defined the contributions of two molecules, the transcription factor RORγt and the cytokine IL-23, to eosinophil IL-17 production. These two molecules are important for optimal IL-17 production in other cell types. We discovered a population of lung eosinophils express RORγt. While expression of the receptor for IL-23 (IL-23R) was positively correlated with expression of RORγt, using mice deficient in IL-23, we showed IL-23 was not required for expression of RORγt and IL-17. Thus, challenge of lungs with live A. fumigatus or its antigens skews lung eosinophils towards IL-17 production by a pathway that is independent of IL-23. Our results advance our understanding of eosinophil plasticity and have implications for the development of therapeutic approaches for treating allergic lung diseases.
Collapse
|
23
|
Hu Y, Gu J, Wang Y, Lin J, Yu H, Yang F, Wu S, Yin J, Lv H, Ji X, Wang S. Promotion Effect of EGCG on the Raised Expression of IL-23 through the Signaling of STAT3-BATF2-c-JUN/ATF2. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:7898-7909. [PMID: 34227806 DOI: 10.1021/acs.jafc.1c02433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Tea polyphenol of epigallocatechin-3-gallate (EGCG) has been verified to possess multiple biological activities. Interleukin-23 (IL-23) is a heterodimeric cytokine consisting of two subunits of IL-23p19 and IL-12p40, with the functionality in regulating the production of cytokines under physiological or pathological conditions. By serendipity, the raised expression of IL-23 was observed after treating cells with EGCG, whereas the detailed mechanism remains poorly understood. This study was proposed to investigate the signaling related to EGCG-induced IL-23. The raised expression of IL-23 was confirmed primarily by intraperitoneally injecting with different concentrations of EGCG (0, 20, 50, 80 mg/kg) into BALB/c mice, and the raised expression was confirmed by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. Results from enzyme-linked immunosorbent assay (ELISA) revealed the increase of IL-23 in serum from 116.09 to 153.90 pg/mL after treating with EGCG. The same results were also observed in RAW264.7 and peritoneal macrophages after treating with EGCG (0, 1, 5, 10, 25 μM) with the increased tendency of IL-23 in cultural medium (7.98 to 25.38 pg/mL for RAW264.7; 3.64 to 260.93 pg/mL for peritoneal macrophages). After preliminary exploration of the signaling related to the increased IL-23, the classical signaling pathways and key transcription factors, such as nuclear factor kappa-B (NF-κB), mitogen-activated protein kinase (MAPK) signaling pathways, and interferon regulatory factor 5 (IRF5), were demonstrated with no relevant contribution. A further study revealed the involvement of the key transcription factor of BATF2, which could antagonistically modulate the transcription and translation of IL-23. The signaling of STAT3-BATF2-c-JUN/ATF2-IL-23 has been further verified in RAW264.7 macrophages using the STAT3 inhibitor of AG490 and the activator of Colivelin TFA. The results indicated that EGCG inhibits the phosphorylation of STAT3 to facilitate the decreased level of BATF2, which contributed to the increased level of IL-23 by the enhancing heterodimerization of c-JUN and ATF2.
Collapse
Affiliation(s)
- Yaozhong Hu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Jiaxin Gu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Yi Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Jing Lin
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Huaning Yu
- Guangdong Midea Kitchen Appliances Manufacturing Co., Ltd, Guangdong 528000, China
| | - Feier Yang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Sihao Wu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Jia Yin
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Huan Lv
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Xuemeng Ji
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
24
|
Jawale CV, Biswas PS. Local antifungal immunity in the kidney in disseminated candidiasis. Curr Opin Microbiol 2021; 62:1-7. [PMID: 33991758 DOI: 10.1016/j.mib.2021.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/12/2021] [Accepted: 04/15/2021] [Indexed: 12/30/2022]
Abstract
Disseminated candidiasis is a hospital-acquired infection that results in high degree of mortality despite antifungal treatment. Autopsy studies revealed that kidneys are the major target organs in disseminated candidiasis and death due to kidney damage is a frequent outcome in these patients. Thus, the need for effective therapeutic strategies to mitigate kidney damage in disseminated candidiasis is compelling. Recent studies have highlighted the essential contribution of kidney-specific immune response in host defense against systemic infection. Crosstalk between kidney-resident and infiltrating immune cells aid in the clearance of fungi and prevent tissue damage in disseminated candidiasis. In this review, we provide our recent understanding on antifungal immunity in the kidney with an emphasis on IL-17-mediated renal defense in disseminated candidiasis.
Collapse
Affiliation(s)
- Chetan V Jawale
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Partha S Biswas
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
25
|
Germination of a Field: Women in Candida albicans Research. CURRENT CLINICAL MICROBIOLOGY REPORTS 2021. [DOI: 10.1007/s40588-021-00169-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
26
|
Valand N, Girija UV. Candida Pathogenicity and Interplay with the Immune System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1313:241-272. [PMID: 34661898 DOI: 10.1007/978-3-030-67452-6_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Candida species are opportunistic fungal pathogens that are part of the normal skin and mucosal microflora. Overgrowth of Candida can cause infections such as thrush or life-threatening invasive candidiasis in immunocompromised patients. Though Candida albicans is highly prevalent, several non-albicans species are also isolated from nosocomial infections. Candida sp. are over presented in the gut of people with Crohn's disease and certain types of neurological disorders, with hyphal form and biofilms being the most virulent states. In addition, Candida uses several secreted and cell surface molecules such as pH related antigen 1, High affinity glucose transporter, Phosphoglycerate mutase 1 and lipases to establish pathogenicity. A strong innate immune response is elicited against Candida via dendritic cells, neutrophils and macrophages. All three complement pathways are also activated. Production of proinflammatory cytokines IL-10 and IL-12 signal differentiation of CD4+ cells into Th1 and Th2 cells, whereas IL-6, IL-17 and IL-23 induce Th17 cells. Importance of T-lymphocytes is reflected in depleted T-cell count patients being more prone to Candidiasis. Anti- Candida antibodies also play a role against candidiasis using various mechanisms such as targeting virulent enzymes and exhibiting direct candidacidal activity. However, the significance of antibody response during infection remains controversial. Furthermore, some of the Candida strains have evolved molecular strategies to evade the sophisticated host attack by proteolysis of components of immune system and interfering with immune signalling pathways. Emergence of several non-albicans species that are resistant to current antifungal agents makes treatment more difficult. Therefore, deeper insight into interactions between Candida and the host immune system is required for discovery of novel therapeutic options.
Collapse
Affiliation(s)
- Nisha Valand
- Leicester School of Allied Health and Life sciences, Faculty of Health and Life Sciences, De Montfort University, Leicester, UK
| | - Umakhanth Venkatraman Girija
- Leicester School of Allied Health and Life sciences, Faculty of Health and Life Sciences, De Montfort University, Leicester, UK.
| |
Collapse
|
27
|
Camilli G, Blagojevic M, Naglik JR, Richardson JP. Programmed Cell Death: Central Player in Fungal Infections. Trends Cell Biol 2020; 31:179-196. [PMID: 33293167 DOI: 10.1016/j.tcb.2020.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/08/2020] [Accepted: 11/11/2020] [Indexed: 12/26/2022]
Abstract
Fungal diseases contribute significantly to morbidity and mortality in humans. Although recent research has improved our understanding of the complex and dynamic interplay that occurs between pathogenic fungi and the human host, much remains to be elucidated concerning the molecular mechanisms that drive fungal pathogenicity and host responses to fungal infections. In recent times, there has been a significant increase in studies investigating the immunological functions of microbial-induced host cell death. In addition, pathogens use many strategies to manipulate host cell death pathways to facilitate their survival and dissemination. This review will focus on the mechanisms of host programmed cell death that occur during opportunistic fungal infections, and explore how cell death pathways may affect immunity towards pathogenic fungi.
Collapse
Affiliation(s)
- Giorgio Camilli
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 1UL, UK.
| | - Mariana Blagojevic
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 1UL, UK
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 1UL, UK
| | - Jonathan P Richardson
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 1UL, UK
| |
Collapse
|
28
|
Candida albicans Virulence Factors and Pathogenicity for Endodontic Infections. Microorganisms 2020; 8:microorganisms8091300. [PMID: 32858856 PMCID: PMC7563224 DOI: 10.3390/microorganisms8091300] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/16/2020] [Accepted: 08/19/2020] [Indexed: 02/07/2023] Open
Abstract
Candida albicans (C. albicans) is the fungus most frequently isolated from endodontic root canal infections. Although recognized by dental pulp and periradicular tissue cells that elicit immune responses, it eludes host defenses and elicits cell death. Then, C. albicans binds tooth dentin, forms biofilms, and invades dentinal tubules to resist intracanal disinfectants and endodontic treatments. Insensitive to most common medicaments, it survives sequestered within biofilms and intratubular dentin. Thus, C. albicans has been associated with cases of persistent or refractory root canal infections. Its treatment strategies may require alternative intracanal irrigants, intracanal medicaments such as chlorhexidine gel or human beta defensin-3 (HBD3), Ca-Si-based obturating materials, and microsurgical procedures.
Collapse
|
29
|
Wang H, Shah CA, Hu L, Huang W, Platanias LC, Eklund EA. An aberrantly sustained emergency granulopoiesis response accelerates postchemotherapy relapse in MLL1-rearranged acute myeloid leukemia in mice. J Biol Chem 2020; 295:9663-9675. [PMID: 32467231 PMCID: PMC7363149 DOI: 10.1074/jbc.ra120.013206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/21/2020] [Indexed: 11/06/2022] Open
Abstract
Acute myeloid leukemia (AML) with mixed lineage leukemia 1 (MLL1) gene rearrangement is characterized by increased expression of a set of homeodomain transcription factors, including homeobox A9 (HOXA9) and HOXA10. The target genes for these regulators include fibroblast growth factor 2 (FGF2) and Ariadne RBR E3 ubiquitin ligase 2 (ARIH2). FGF2 induces leukemia stem cell expansion in MLL1-rearranged AML. ARIH2 encodes TRIAD1, an E3 ubiquitin ligase required for termination of emergency granulopoiesis and leukemia suppressor function in MLL1-rearranged AML. Receptor tyrosine kinases (RTKs), including the FGF receptor, are TRIAD1 substrates that are possibly relevant to these activities. Using transcriptome analysis, we found increased activity of innate immune response pathways and RTK signaling in bone marrow progenitors from mice with MLL1-rearranged AML. We hypothesized that sustained RTK signaling, because of decreased TRIAD1 activity, impairs termination of emergency granulopoiesis during the innate immune response and contributes to leukemogenesis in this AML subtype. Consistent with this, we found aberrantly sustained emergency granulopoiesis in a murine model of MLL1-rearranged AML, associated with accelerated leukemogenesis. Treating these mice with an inhibitor of TRIAD1-substrate RTKs terminated emergency granulopoiesis, delayed leukemogenesis during emergency granulopoiesis, and normalized innate immune responses when combined with chemotherapy. Emergency granulopoiesis also hastened postchemotherapy relapse in mice with MLL1-rearranged AML, but remission was sustained by ongoing RTK inhibition. Our findings suggest that the physiological stress of infectious challenges may drive AML progression in molecularly defined subsets and identify RTK inhibition as a potential therapeutic approach to counteract this process.
Collapse
Affiliation(s)
- Hao Wang
- Department of Medicine, Northwestern University, Chicago, Illinois, USA.,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Chirag A Shah
- Department of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Liping Hu
- Department of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Weiqi Huang
- Department of Medicine, Northwestern University, Chicago, Illinois, USA.,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Leonidas C Platanias
- Department of Medicine, Northwestern University, Chicago, Illinois, USA.,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Elizabeth A Eklund
- Department of Medicine, Northwestern University, Chicago, Illinois, USA .,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| |
Collapse
|
30
|
Ramayo-Caldas Y, Prenafeta-Boldú F, Zingaretti LM, Gonzalez-Rodriguez O, Dalmau A, Quintanilla R, Ballester M. Gut eukaryotic communities in pigs: diversity, composition and host genetics contribution. Anim Microbiome 2020; 2:18. [PMID: 33499953 PMCID: PMC7807704 DOI: 10.1186/s42523-020-00038-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 04/29/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The pig gut microbiome harbors thousands of species of archaea, bacteria, viruses and eukaryotes such as protists and fungi. However, since the majority of published studies have been focused on prokaryotes, little is known about the diversity, host-genetic control, and contributions to host performance of the gut eukaryotic counterparts. Here we report the first study that aims at characterizing the diversity and composition of gut commensal eukaryotes in pigs, exploring their putative control by host genetics, and analyzing their association with piglets body weight. RESULTS Fungi and protists from the faeces of 514 healthy Duroc pigs of two sexes and two different ages were characterized by 18S and ITS ribosomal RNA gene sequencing. The pig gut mycobiota was dominated by yeasts, with a high prevalence and abundance of Kazachstania spp. Regarding protists, representatives of four genera (Blastocystis, Neobalantidium, Tetratrichomonas and Trichomitus) were predominant in more than the 80% of the pigs. Heritabilities for the diversity and abundance of gut eukaryotic communities were estimated with the subset of 60d aged piglets (N = 390). The heritabilities of α-diversity and of the abundance of fungal and protists genera were low, ranging from 0.15 to 0.28. A genome wide association study reported genetic variants related to the fungal α-diversity and to the abundance of Blastocystis spp. Annotated candidate genes were mainly associated with immunity, gut homeostasis and metabolic processes. Additionally, we explored the association of gut commensal eukaryotes with piglet body weight. Our results pointed to a positive contribution of fungi from the Kazachstania genus, while protists displayed both positive (Blastocystis and Entamoeba) and negative (Trichomitus) associations with piglet body weight. CONCLUSIONS Our results point towards a minor and taxa specific genetic control over the diversity and composition of the pig gut eukaryotic communities. Moreover, we provide evidences of the associations between piglets' body weight after weaning and members from the gut fungal and protist eukaryote community. Overall, this study highlights the relevance of considering, along with that of bacteria, the contribution of the gut eukaryote communities to better understand host-microbiome association and their role on pig performance, welfare and health.
Collapse
Affiliation(s)
- Yuliaxis Ramayo-Caldas
- Animal Breeding and Genetics Program, Institute of Agrifood Research and Technology (IRTA), Torre Marimon, 08140 Caldes de Montbui, Spain
| | | | - Laura M. Zingaretti
- Centre for Research in Agricultural Genomics, CSIC-IRTA-UAB-UB Consortium, Bellaterra, Spain
| | - Olga Gonzalez-Rodriguez
- Animal Breeding and Genetics Program, Institute of Agrifood Research and Technology (IRTA), Torre Marimon, 08140 Caldes de Montbui, Spain
| | - Antoni Dalmau
- Animal Welfare Subprogram, IRTA, 17121 Monells, Spain
| | - Raquel Quintanilla
- Animal Breeding and Genetics Program, Institute of Agrifood Research and Technology (IRTA), Torre Marimon, 08140 Caldes de Montbui, Spain
| | - Maria Ballester
- Animal Breeding and Genetics Program, Institute of Agrifood Research and Technology (IRTA), Torre Marimon, 08140 Caldes de Montbui, Spain
| |
Collapse
|