1
|
Zhang C, Yuan M, Rong W, Du H, Li X, Ji T, Li J, Dai B, Ma Z, Qi H, Zhang N, Yang J, Duan X, Bi Y. Synergistic effects of Lianhuaqingwen in combination with Oseltamivir and Baloxavir against seasonal influenza virus: In vitro and in vivo assessment. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119091. [PMID: 39528119 DOI: 10.1016/j.jep.2024.119091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lianhuaqingwen (LH), a traditional Chinese medicine, presents a broad-spectrum antiviral effect and has been widely used to treat influenza. Given the potential rise of drug-resistant influenza viruses, it is necessary to develop new antiviral drugs and explore combination therapies involving LH in tandem with existing antivirals such as Oseltamivir acid (Osel) or Baloxavir (Bal). These multidrug combinations could help effectively control the seasonal influenza epidemics and reduce the disease burden. AIM OF THE STUDY This study aimed to evaluate the antiviral effects of LH, alone and in combination with Osel or Bal, against human seasonal influenza viruses in vitro and in vivo models. MATERIALS AND METHODS The antiviral efficacy of LH alone and LH in combination with Osel/Bal against seasonal influenza A viruses (IAVs) (H1N1 and H3N2 subtypes) and influenza B viruses (IBVs) (BV- and BY-lineages) was assessed in vitro using MDCK cells. The median effective concentration (EC50) was determined, and the drug synergies were analyzed. Additionally, the antiviral activity of LH monotherapy and LH + Osel/Bal combination therapy were evaluated in vivo using an H1N1-infected BABL/c mouse model by monitoring changes in body weight, survival rate, lung viral titer, pathological damage, and inflammatory reaction. RESULTS In vitro, LH alone and in combination with Osel/Bal exhibited antiviral activity against both IAVs and IBVs. The addition of LH to Osel/Bal improved the therapeutic efficacy compared to Osel/Bal alone. In vivo, LH monotherapy reduced body weight loss and increased the survival rates of H1N1-infected mice. LH in combination with Osel/Bal resulted in lower virus titers, more effective relief of pathological damage, and comparable low expression of inflammatory factors in the lungs of H1N1-infected mice compared to the use of Osel/Bal alone. Transcriptomic analysis of the lungs revealed that LH + Osel/Bal significantly increased the expression of genes associated with antiviral and anti-inflammatory effects. CONCLUSIONS This study evaluated the antiviral effects of LH monotherapy and combination therapy with Osel/Bal against human seasonal influenza viruses in vitro and in vivo models. The results suggest that combining LH with Osel or Bal could enhance the antiviral efficiency for influenza viruses compared to the monotherapy using any of these three drugs.
Collapse
Affiliation(s)
- Cheng Zhang
- College of Life Science and Technology, Xinjiang University, Urumchi, 830046, China; CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing, 100101, China
| | - Manhua Yuan
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing, 100101, China
| | - Wenwan Rong
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Han Du
- College of Life Science and Technology, Xinjiang University, Urumchi, 830046, China
| | - Xuanxuan Li
- College of Life Science and Technology, Xinjiang University, Urumchi, 830046, China
| | - Tiannan Ji
- Department of Emergency, Department of Radiotherapy, The Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Jianxiong Li
- Department of Emergency, Department of Radiotherapy, The Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Bo Dai
- Department of Pharmacy, Air Force Medical Center, PLA, Beijing, 100142, China
| | - Zhenghai Ma
- College of Life Science and Technology, Xinjiang University, Urumchi, 830046, China
| | - Hui Qi
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Ning Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing, 100101, China
| | - Jing Yang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing, 100101, China
| | - Xuefeng Duan
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuhai Bi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
2
|
Zhao D, Duan X, Zhu L, Fang M, Qin T, Bi Y. IL-6-Caspase 3 Axis Plays an Important Role in Enteritis Caused by Legionella pneumophila Pulmonary Infection. Microorganisms 2025; 13:313. [PMID: 40005679 PMCID: PMC11858493 DOI: 10.3390/microorganisms13020313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Since Legionella pneumophila (Lp) is widely present in natural and artificial water environments, it has a high potential risk of outbreak. Diarrhea caused by Lp pulmonary infection is an important symptom of Legionnaires' disease (LD); however, the underlying mechanism of the diarrhea has not yet been revealed. This not only has a negative impact on clinical diagnosis and treatment, but may also cause misdiagnosis. METHODS In the present study, a mouse model of enteritis caused by pulmonary infection of Lp was established. By using this mouse model, we explored the underlying mechanisms of the enteritis caused by Lp pulmonary infection. RESULTS The results indicated that the systemic inflammatory response played a very important role in the enteritis phenotype caused by a strong-virulence strain of Lp. Furthermore, we found that the expression of Bcl-2 was downregulated by IL-6 through the p53 signaling pathway, thereby activating the caspase 3 of intestinal epithelial cells (IECs), causing the apoptosis of IECs, and ultimately leading to the enteritis phenotype. CONCLUSIONS The IL-6-caspase 3 axis plays an important role in enteritis caused by Lp pulmonary infection.
Collapse
Affiliation(s)
- Dahui Zhao
- University of Chinese Academy of Sciences, Beijing 100049, China; (D.Z.); (X.D.)
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-Warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China;
| | - Xuefeng Duan
- University of Chinese Academy of Sciences, Beijing 100049, China; (D.Z.); (X.D.)
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-Warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China;
| | - Li Zhu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-Warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China;
| | - Min Fang
- School of Life Sciences & Henan Key Laboratory of Synthetic Biology and Biomanufacturing, Henan University, Kaifeng 475004, China;
| | - Tian Qin
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Centre for Disease Control and Prevention, Beijing 102206, China
| | - Yuhai Bi
- University of Chinese Academy of Sciences, Beijing 100049, China; (D.Z.); (X.D.)
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-Warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China;
| |
Collapse
|
3
|
Ruiz Pérez M, Vandenabeele P, Tougaard P. The thymus road to a T cell: migration, selection, and atrophy. Front Immunol 2024; 15:1443910. [PMID: 39257583 PMCID: PMC11384998 DOI: 10.3389/fimmu.2024.1443910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/08/2024] [Indexed: 09/12/2024] Open
Abstract
The thymus plays a pivotal role in generating a highly-diverse repertoire of T lymphocytes while preventing autoimmunity. Thymus seeding progenitors (TSPs) are a heterogeneous group of multipotent progenitors that migrate to the thymus via CCR7 and CCR9 receptors. While NOTCH guides thymus progenitors toward T cell fate, the absence or disruption of NOTCH signaling renders the thymus microenvironment permissive to other cell fates. Following T cell commitment, developing T cells undergo multiple selection checkpoints by engaging with the extracellular matrix, and interacting with thymic epithelial cells (TECs) and other immune subsets across the different compartments of the thymus. The different selection checkpoints assess the T cell receptor (TCR) performance, with failure resulting in either repurposing (agonist selection), or cell death. Additionally, environmental cues such as inflammation and endocrine signaling induce acute thymus atrophy, contributing to the demise of most developing T cells during thymic selection. We discuss the occurrence of acute thymus atrophy in response to systemic inflammation. The thymus demonstrates high plasticity, shaping inflammation by abrogating T cell development and undergoing profound structural changes, and facilitating regeneration and restoration of T cell development once inflammation is resolved. Despite the challenges, thymic selection ensures a highly diverse T cell repertoire capable of discerning between self and non-self antigens, ultimately egressing to secondary lymphoid organs where they complete their maturation and exert their functions.
Collapse
Affiliation(s)
- Mario Ruiz Pérez
- Molecular Signaling and Cell Death Unit, VIB-UGent, Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Peter Vandenabeele
- Molecular Signaling and Cell Death Unit, VIB-UGent, Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Peter Tougaard
- Molecular Signaling and Cell Death Unit, VIB-UGent, Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| |
Collapse
|
4
|
Rizvi ZA, Sadhu S, Dandotiya J, Sharma P, Binayke A, Singh V, Das V, Khatri R, Kumar R, Samal S, Kalia M, Awasthi A. SARS-CoV-2 infection induces thymic atrophy mediated by IFN-γ in hACE2 transgenic mice. Eur J Immunol 2024; 54:e2350624. [PMID: 38655818 DOI: 10.1002/eji.202350624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/26/2024]
Abstract
Pathogenic infections cause thymic atrophy, perturb thymic T-cell development, and alter immunological response. Previous studies reported dysregulated T-cell function and lymphopenia in coronavirus disease-19 (COVID-19). However, immunopathological changes in the thymus associated with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection have not been elucidated. Here, we report that SARS-CoV-2 infects thymocytes, and induces CD4+CD8+ (double positive; DP) T-cell apoptosis leading to thymic atrophy and loss of peripheral TCR repertoire in K18-hACE2 transgenic mice. Infected thymus led to increased CD44+CD25- T-cells, indicating an early arrest in the T-cell maturation pathway. Thymic atrophy was notably higher in male hACE2-Tg mice than in females and involved an upregulated de-novo synthesis pathway of thymic glucocorticoid. Further, IFN-γ was crucial for thymic atrophy, as anti-IFN-γ -antibody neutralization blunted thymic involution. Therapeutic use of Remdesivir also rescued thymic atrophy. While the Omicron variant and its sub-lineage BA.5 variant caused marginal thymic atrophy, the delta variant of SARS-CoV-2 exhibited severe thymic atrophy characterized by severely depleted DP T-cells. Recently characterized broadly SARS-CoV-2 neutralizing monoclonal antibody P4A2 was able to rescue thymic atrophy and restore the thymic maturation pathway of T-cells. Together, we report SARS-CoV-2-associated thymic atrophy resulting from impaired T-cell maturation pathway which may contribute to dyregulated T cell response during COVID-19.
Collapse
Affiliation(s)
- Zaigham Abbas Rizvi
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Srikanth Sadhu
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Jyotsna Dandotiya
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Puja Sharma
- Regional Centre Biotechnology, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Akshay Binayke
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Virendra Singh
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Vinayaka Das
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Ritika Khatri
- Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Rajesh Kumar
- Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Sweety Samal
- Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Manjula Kalia
- Regional Centre Biotechnology, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Amit Awasthi
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| |
Collapse
|
5
|
Bjorgen JC, Dick JK, Cromarty R, Hart GT, Rhein J. NK cell subsets and dysfunction during viral infection: a new avenue for therapeutics? Front Immunol 2023; 14:1267774. [PMID: 37928543 PMCID: PMC10620977 DOI: 10.3389/fimmu.2023.1267774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/25/2023] [Indexed: 11/07/2023] Open
Abstract
In the setting of viral challenge, natural killer (NK) cells play an important role as an early immune responder against infection. During this response, significant changes in the NK cell population occur, particularly in terms of their frequency, location, and subtype prevalence. In this review, changes in the NK cell repertoire associated with several pathogenic viral infections are summarized, with a particular focus placed on changes that contribute to NK cell dysregulation in these settings. This dysregulation, in turn, can contribute to host pathology either by causing NK cells to be hyperresponsive or hyporesponsive. Hyperresponsive NK cells mediate significant host cell death and contribute to generating a hyperinflammatory environment. Hyporesponsive NK cell populations shift toward exhaustion and often fail to limit viral pathogenesis, possibly enabling viral persistence. Several emerging therapeutic approaches aimed at addressing NK cell dysregulation have arisen in the last three decades in the setting of cancer and may prove to hold promise in treating viral diseases. However, the application of such therapeutics to treat viral infections remains critically underexplored. This review briefly explores several therapeutic approaches, including the administration of TGF-β inhibitors, immune checkpoint inhibitors, adoptive NK cell therapies, CAR NK cells, and NK cell engagers among other therapeutics.
Collapse
Affiliation(s)
- Jacob C. Bjorgen
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Jenna K. Dick
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Ross Cromarty
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Geoffrey T. Hart
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Joshua Rhein
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
6
|
Dong Z, Ma J, Qiu J, Ren Q, Shan Q, Duan X, Li G, Zuo YY, Qi Y, Liu Y, Liu G, Lynch I, Fang M, Liu S. Airborne fine particles drive H1N1 viruses deep into the lower respiratory tract and distant organs. SCIENCE ADVANCES 2023; 9:eadf2165. [PMID: 37294770 PMCID: PMC10256160 DOI: 10.1126/sciadv.adf2165] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 05/05/2023] [Indexed: 06/11/2023]
Abstract
Mounting data suggest that environmental pollution due to airborne fine particles (AFPs) increases the occurrence and severity of respiratory virus infection in humans. However, it is unclear whether and how interactions with AFPs alter viral infection and distribution. We report synergetic effects between various AFPs and the H1N1 virus, regulated by physicochemical properties of the AFPs. Unlike infection caused by virus alone, AFPs facilitated the internalization of virus through a receptor-independent pathway. Moreover, AFPs promoted the budding and dispersal of progeny virions, likely mediated by lipid rafts in the host plasma membrane. Infected animal models demonstrated that AFPs favored penetration of the H1N1 virus into the distal lung, and its translocation into extrapulmonary organs including the liver, spleen, and kidney, thus causing severe local and systemic disorders. Our findings revealed a key role of AFPs in driving viral infection throughout the respiratory tract and beyond. These insights entail stronger air quality management and air pollution reduction policies.
Collapse
Affiliation(s)
- Zheng Dong
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Juan Ma
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiahuang Qiu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Quanzhong Ren
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Qing’e Shan
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Xuefeng Duan
- CAS Key Laboratory of Pathogenic Microbiology & Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Guangle Li
- Department of Mechanical Engineering, University of Hawaii at Mānoa, Honolulu, HI 96822, USA
| | - Yi Y. Zuo
- Department of Mechanical Engineering, University of Hawaii at Mānoa, Honolulu, HI 96822, USA
| | - Yu Qi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yajun Liu
- Beijing Jishuitan Hospital, Peking University Health Science Center, Beijing 100035, China
| | - Guoliang Liu
- Department of Pulmonary and Critical Care Medicine, Centre for Respiratory Diseases, China-Japan Friendship Hospital, Beijing 100029, China
- National Center for Respiratory Medicine, Beijing 100029, China
| | - Iseult Lynch
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Min Fang
- CAS Key Laboratory of Pathogenic Microbiology & Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| |
Collapse
|
7
|
Caruso B, Moran AE. Thymic expression of immune checkpoint molecules and their implication for response to immunotherapies. Trends Cancer 2023:S2405-8033(23)00063-8. [PMID: 37173189 DOI: 10.1016/j.trecan.2023.04.007] [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: 01/20/2023] [Revised: 04/07/2023] [Accepted: 04/14/2023] [Indexed: 05/15/2023]
Abstract
The thymus is responsible for generating a diverse T cell repertoire that is tolerant to self, but capable of responding to various immunologic insults, including cancer. Checkpoint blockade has changed the face of cancer treatment by targeting inhibitory molecules, which are known to regulate peripheral T cell responses. However, these inhibitory molecules and their ligands are expressed during T cell development in the thymus. In this review, we describe the underappreciated role of checkpoint molecule expression during the formation of the T cell repertoire and detail the importance of inhibitory molecules in regulating T cell lineage commitment. Understanding how these molecules function in the thymus may inform therapeutic strategies for better patient outcomes.
Collapse
Affiliation(s)
- Breanna Caruso
- Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Amy E Moran
- Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, USA; Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
8
|
Chiang KC, Kalantar-Zadeh K, Gupta A. Thymic Dysfunction and Atrophy in COVID-19 Disease Complicated by Inflammation, Malnutrition and Cachexia. Nutr Health 2022; 28:199-206. [PMID: 35234100 PMCID: PMC8891908 DOI: 10.1177/02601060221083160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Background: The current COVID-19 pandemic has put millions of people, especially children at risk of protein-energy malnutrition (PEM) by pushing them into poverty and disrupting the global food supply chain. The thymus is severely affected by nutritional deficiencies and is known as a barometer of malnutrition. Aim: The present commentary provides a novel perspective on the role of malnutrition-induced thymic dysfunction, involution and atrophy on the risk and severity of disease in children during the COVID-19 pandemic. Methods: A review of pertinent indexed literature including studies examining the effects of malnutrition on the thymus and immune dysfunction in COVID-19. Results: Protein-energy malnutrition and micronutrient deficiencies of zinc, iron and vitamin A are known to promote thymic dysfunction and thymocyte loss in children. Malnutrition- and infection-induced thymic atrophy and immune dysfunction may increase the risk of first, progression of COVID-19 disease to more severe forms including development of multisystem inflammatory syndrome in children (MIS-C); second, slow the recovery from COVID-19 disease; and third, increase the risk of other infections. Furthermore, malnourished children may be at increased risk of contracting SARS-CoV-2 infection due to socioeconomic conditions that promote viral transmission amongst contacts and create barriers to vaccination. Conclusion: National governments and international organizations including WHO, World Food Program, and UNICEF should institute measures to ensure provision of food and micronutrients for children at risk in order to limit the health impact of the ongoing COVID-19 pandemic.
Collapse
Affiliation(s)
| | - Kamyar Kalantar-Zadeh
- Division of Nephrology, Hypertension
and Kidney Transplantation and Department of Medicine,
University of
California Irvine (UCI) School of Medicine,
USA
| | - Ajay Gupta
- Charak
Foundation, Orange, CA, USA
- Division of Nephrology, Hypertension
and Kidney Transplantation and Department of Medicine,
University of
California Irvine (UCI) School of Medicine,
USA
| |
Collapse
|
9
|
Gulla S, Reddy MC, Reddy VC, Chitta S, Bhanoori M, Lomada D. Role of thymus in health and disease. Int Rev Immunol 2022; 42:347-363. [PMID: 35593192 DOI: 10.1080/08830185.2022.2064461] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/26/2022] [Accepted: 04/04/2022] [Indexed: 01/04/2023]
Abstract
The thymus is a primary lymphoid organ, essential for the development of T-cells that will protect from invading pathogens, immune disorders, and cancer. The thymus decreases in size and cellularity with age referred to as thymus involution or atrophy. This involution causes decreased T-cell development and decreased naive T-cell emigration to the periphery, increased proportion of memory T cells, and a restricted, altered T-cell receptor (TCR) repertoire. The changes in composition and function of the circulating T cell pool as a result of thymic involution led to increased susceptibility to infectious diseases including the recent COVID and a higher risk for autoimmune disorders and cancers. Thymic involution consisting of both structural and functional loss of the thymus has a deleterious effect on T cell development, T cell selection, and tolerance. The mechanisms which act on the structural (cortex and medulla) matrix of the thymus, the gradual accumulation of genetic mutations, and altered gene expressions may lead to immunosenescence as a result of thymus involution. Understanding the molecular mechanisms behind thymic involution is critical for identifying diagnostic biomarkers and targets for treatment help to develop strategies to mitigate thymic involution-associated complications. This review is focused on the consequences of thymic involution in infections, immune disorders, and diseases, identifying potential checkpoints and potential approaches to sustain or restore the function of the thymus particularly in elderly and immune-compromised individuals.
Collapse
Affiliation(s)
- Surendra Gulla
- Department of Biotechnology and Bioinformatics, Yogi Vemana University, Kadapa, Andhra Pradesh, India
| | - Madhava C Reddy
- Department of Biotechnology and Bioinformatics, Yogi Vemana University, Kadapa, Andhra Pradesh, India
| | - Vajra C Reddy
- Katuri Medical College and Hospital, Chinnakondrupadu, Guntur, India
| | | | - Manjula Bhanoori
- Department of Biochemistry, Osmania University, Hyderabad, Telangana State, India
| | - Dakshayani Lomada
- Department of Genetics and Genomics, Yogi Vemana University, Kadapa, Andhra Pradesh, India
| |
Collapse
|
10
|
Luo M, Xu L, Qian Z, Sun X. Infection-Associated Thymic Atrophy. Front Immunol 2021; 12:652538. [PMID: 34113341 PMCID: PMC8186317 DOI: 10.3389/fimmu.2021.652538] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022] Open
Abstract
The thymus is a vital organ of the immune system that plays an essential role in thymocyte development and maturation. Thymic atrophy occurs with age (physiological thymic atrophy) or as a result of viral, bacterial, parasitic or fungal infection (pathological thymic atrophy). Thymic atrophy directly results in loss of thymocytes and/or destruction of the thymic architecture, and indirectly leads to a decrease in naïve T cells and limited T cell receptor diversity. Thus, it is important to recognize the causes and mechanisms that induce thymic atrophy. In this review, we highlight current progress in infection-associated pathogenic thymic atrophy and discuss its possible mechanisms. In addition, we discuss whether extracellular vesicles/exosomes could be potential carriers of pathogenic substances to the thymus, and potential drugs for the treatment of thymic atrophy. Having acknowledged that most current research is limited to serological aspects, we look forward to the possibility of extending future work regarding the impact of neural modulation on thymic atrophy.
Collapse
Affiliation(s)
- Mingli Luo
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Lingxin Xu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Zhengyu Qian
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Xi Sun
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| |
Collapse
|
11
|
Elfaki Y, Robert PA, Binz C, Falk CS, Bruder D, Prinz I, Floess S, Meyer-Hermann M, Huehn J. Influenza A virus-induced thymus atrophy differentially affects dynamics of conventional and regulatory T-cell development in mice. Eur J Immunol 2021; 51:1166-1181. [PMID: 33638148 DOI: 10.1002/eji.202048981] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/23/2020] [Accepted: 02/18/2021] [Indexed: 01/26/2023]
Abstract
Foxp3+ Treg cells, which are crucial for maintenance of self-tolerance, mainly develop within the thymus, where they arise from CD25+ Foxp3- or CD25- Foxp3+ Treg cell precursors. Although it is known that infections can cause transient thymic involution, the impact of infection-induced thymus atrophy on thymic Treg (tTreg) cell development is unknown. Here, we infected mice with influenza A virus (IAV) and studied thymocyte population dynamics post infection. IAV infection caused a massive, but transient thymic involution, dominated by a loss of CD4+ CD8+ double-positive (DP) thymocytes, which was accompanied by a significant increase in the frequency of CD25+ Foxp3+ tTreg cells. Differential apoptosis susceptibility could be experimentally excluded as a reason for the relative tTreg cell increase, and mathematical modeling suggested that enhanced tTreg cell generation cannot explain the increased frequency of tTreg cells. Yet, an increased death of DP thymocytes and augmented exit of single-positive (SP) thymocytes was suggested to be causative. Interestingly, IAV-induced thymus atrophy resulted in a significantly reduced T-cell receptor (TCR) repertoire diversity of newly produced tTreg cells. Taken together, IAV-induced thymus atrophy is substantially altering the dynamics of major thymocyte populations, finally resulting in a relative increase of tTreg cells with an altered TCR repertoire.
Collapse
Affiliation(s)
- Yassin Elfaki
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Philippe A Robert
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Christoph Binz
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Christine S Falk
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
| | - Dunja Bruder
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Stefan Floess
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany.,Institute for Biochemistry, Biotechnology and Bioinformatics, Technical University Braunschweig, Braunschweig, Germany
| | - Jochen Huehn
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
12
|
Kinsella S, Dudakov JA. When the Damage Is Done: Injury and Repair in Thymus Function. Front Immunol 2020; 11:1745. [PMID: 32903477 PMCID: PMC7435010 DOI: 10.3389/fimmu.2020.01745] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/30/2020] [Indexed: 01/02/2023] Open
Abstract
Even though the thymus is exquisitely sensitive to acute insults like infection, shock, or common cancer therapies such as cytoreductive chemo- or radiation-therapy, it also has a remarkable capacity for repair. This phenomenon of endogenous thymic regeneration has been known for longer even than its primary function to generate T cells, however, the underlying mechanisms controlling the process have been largely unstudied. Although there is likely continual thymic involution and regeneration in response to stress and infection in otherwise healthy people, acute and profound thymic damage such as that caused by common cancer cytoreductive therapies or the conditioning regimes as part of hematopoietic cell transplantation (HCT), leads to prolonged T cell deficiency; precipitating high morbidity and mortality from opportunistic infections and may even facilitate cancer relapse. Furthermore, this capacity for regeneration declines with age as a function of thymic involution; which even at steady state leads to reduced capacity to respond to new pathogens, vaccines, and immunotherapy. Consequently, there is a real clinical need for strategies that can boost thymic function and enhance T cell immunity. One approach to the development of such therapies is to exploit the processes of endogenous thymic regeneration into novel pharmacologic strategies to boost T cell reconstitution in clinical settings of immune depletion such as HCT. In this review, we will highlight recent work that has revealed the mechanisms by which the thymus is capable of repairing itself and how this knowledge is being used to develop novel therapies to boost immune function.
Collapse
Affiliation(s)
- Sinéad Kinsella
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Jarrod A. Dudakov
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Department of Immunology, University of Washington, Seattle, WA, United States
| |
Collapse
|
13
|
Streptococcus suis Serotype 2 Infection Causes Host Immunomodulation through Induction of Thymic Atrophy. Infect Immun 2020; 88:IAI.00950-19. [PMID: 31932328 DOI: 10.1128/iai.00950-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 01/04/2020] [Indexed: 02/05/2023] Open
Abstract
Streptococcus suis serotype 2 is an important bacterial pathogen of swine and is also an emerging zoonotic agent that may be harmful to human health. Although the virulence genes of S. suis have been extensively studied, the mechanisms by which they damage the central immune organs have rarely been studied. In the current work, we wanted to uncover more details about the impact and mechanisms of S. suis on specific populations of thymic and immune cells in infected mice. Terminal deoxynucleotidyl transferase (TdT)-mediated dUTP-biotin nick end labeling (TUNEL) assays revealed that S. suis infection induced apoptosis in CD3+, CD14+, and epithelial cells from the thymus. S. suis infection resulted in a rapid depletion of mitochondrial permeability and release of cytochrome c (CytC) and apoptosis-inducing factor (AIF) through upregulation of Bax expression and downregulation of Bcl-xl and Bcl2 expression in thymocytes. Moreover, S. suis infection increased cleavage of caspase-3, caspase-8, and caspase-9. Thus, S. suis induced thymocyte apoptosis through a p53- and caspase-dependent pathway, which led to a decrease of CD3+ cells in the thymus, subsequently decreasing the numbers of CD4+ and CD8+ cells in the peripheral blood. Finally, expression dysregulation of proinflammatory cytokines in the serum, including interleukin 2 (IL-2), IL-6, IL-12 (p70), tumor necrosis factor (TNF), and IL-10, was observed in mice after S. suis type 2 infection. Taken together, these results suggest that S. suis infection can cause atrophy of the thymus and induce apoptosis of thymocytes in mice, thus likely suppressing host immunity.
Collapse
|
14
|
Insights into Thymus Development and Viral Thymic Infections. Viruses 2019; 11:v11090836. [PMID: 31505755 PMCID: PMC6784209 DOI: 10.3390/v11090836] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/03/2019] [Accepted: 09/06/2019] [Indexed: 12/16/2022] Open
Abstract
T-cell development in the thymus is a complex and highly regulated process, involving a wide variety of cells and molecules which orchestrate thymocyte maturation into either CD4+ or CD8+ single-positive (SP) T cells. Here, we briefly review the process regulating T-cell differentiation, which includes the latest advances in this field. In particular, we highlight how, starting from a pool of hematopoietic stem cells in the bone marrow, the sequential action of transcriptional factors and cytokines dictates the proliferation, restriction of lineage potential, T-cell antigen receptors (TCR) gene rearrangements, and selection events on the T-cell progenitors, ultimately leading to the generation of mature T cells. Moreover, this review discusses paradigmatic examples of viral infections affecting the thymus that, by inducing functional changes within this lymphoid gland, consequently influence the behavior of peripheral mature T-lymphocytes.
Collapse
|
15
|
Zhu L, Zhao W, Lu J, Li S, Zhou K, Jiang W, Duan X, Fu L, Yu B, Cai KQ, Gao GF, Liu W, Fang M. Influenza virus matrix protein M1 interacts with SLD5 to block host cell cycle. Cell Microbiol 2019; 21:e13038. [PMID: 31050118 DOI: 10.1111/cmi.13038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/16/2019] [Accepted: 04/29/2019] [Indexed: 01/09/2023]
Abstract
Influenza virus matrix 1 protein (M1) is highly conserved and plays essential roles at many stages of virus life cycle. Here, we used a yeast two-hybrid system to identify the host protein SLD5, a component of the GINS complex, which is essential for the initiation of DNA replication in eukaryotic cells, as a new M1 interacting protein. M1 from several different influenza virus strains all interacted with SLD5. Overexpression of SLD5 suppressed influenza virus replication. Transient, stable, or inducible expression of M1 induced host cell cycle blockade at G0/G1 phase. Moreover, SLD5 partially rescued M1 expression- or influenza virus infection-induced G0/G1 phase accumulation in cell lines and primary mouse embryonic fibroblasts. Importantly, SLD5 transgenic mice exhibited higher resistance and improved lung epithelial regeneration after virus infection compared with wild-type mice. Therefore, influenza virus M1 blocks host cell cycle process by interacting with SLD5. Our finding reveals the multifunctional nature of M1 and provides new insight for understanding influenza virus-host interaction.
Collapse
Affiliation(s)
- Li Zhu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wenming Zhao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jiao Lu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Shan Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Kai Zhou
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Wei Jiang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Xuefeng Duan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Lifeng Fu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Bolan Yu
- Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Kathy Q Cai
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - George Fu Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Wenjun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Min Fang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,International College, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
16
|
Majumdar S, Adiga V, Raghavan A, Rananaware SR, Nandi D. Comparative analysis of thymic subpopulations during different modes of atrophy identifies the reactive oxygen species scavenger, N-acetyl cysteine, to increase the survival of thymocytes during infection-induced and lipopolysaccharide-induced thymic atrophy. Immunology 2019; 157:21-36. [PMID: 30659606 DOI: 10.1111/imm.13043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/07/2018] [Accepted: 01/10/2019] [Indexed: 12/17/2022] Open
Abstract
The development of immunocompetent T cells entails a complex pathway of differentiation in the thymus. Thymic atrophy occurs with ageing and during conditions such as malnutrition, infections and cancer chemotherapy. The comparative changes in thymic subsets under different modes of thymic atrophy and the mechanisms involved are not well characterized. These aspects were investigated, using mice infected with Salmonella Typhimurium, injection with lipopolysaccharide (LPS), an inflammatory but non-infectious stimulus, etoposide (Eto), a drug used to treat some cancers, and dexamethasone (Dex), a steroid used in some inflammatory diseases. The effects on the major subpopulations of thymocytes based on multicolour flow cytometry studies were, first, the CD4- CD8- double-negative (DN) cells, mainly DN2-4, were reduced with infection, LPS and Eto treatment, but not with Dex. Second, the CD8+ CD3lo immature single-positive cells (ISPs) were highly sensitive to infection, LPS and Eto, but not Dex. Third, treatment with LPS, Eto and Dex reduced all three subpopulations of CD4+ CD8+ double-positive (DP) thymocytes, i.e. DP1, DP2 and DP3, but the DP3 subset was relatively more resistant during infection. Fourth, both CD4+ and CD8+ single-positive (SP) thymocytes were lowered by Eto and Dex, but not during infection. Notably, LPS lowered CD4+ SP subsets, whereas the CD8+ SP subsets were relatively more resistant. Interestingly, the reactive oxygen species quencher, N-acetyl cysteine, greatly improved the survival of thymocytes, especially DNs, ISPs and DPs, during infection and LPS treatment. The implications of these observations for the development of potential thymopoietic drugs are discussed.
Collapse
Affiliation(s)
- Shamik Majumdar
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Vasista Adiga
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Abinaya Raghavan
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | | | - Dipankar Nandi
- Department of Biochemistry, Indian Institute of Science, Bangalore, India.,Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| |
Collapse
|
17
|
Wang B, Zhang Y, Jiang W, Zhu L, Li K, Zhou K, Dai D, Chang S, Fang M. GALNT3 inhibits NF-κB signaling during influenza A virus infection. Biochem Biophys Res Commun 2018; 503:2872-2877. [PMID: 30100058 DOI: 10.1016/j.bbrc.2018.08.058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 08/06/2018] [Indexed: 01/13/2023]
Abstract
Protein glycosylation, attaching glycans covalently onto amino acid side chains of protein by various glycosyltransferase, is the most common post-translational modification. The UDP-GalNAc transferase 3 (GANLT3), encoded by Galnt3, transfers N-acetyl-d-galactosamine to hydroxyl groups of the side chains of Ser/Thr residues, initiating mucin type O-glycosylation of proteins. Most researches as yet focus on the involvement and abnormal expression of GALNT3 in various tumors. In this study, we found that GALNT3 was significantly decreased in the lungs after influenza A virus (IAV) infection in mice. Overexpression of GALNT3 in cell lines markedly inhibited IAV replication. Further experiments demonstrated that GALNT3 inhibited NF-κB signaling by preventing the translocation of phosphorylated P65 into nucleus. Therefore, our results reveal an important role of GALNT3 in regulating host responses during IAV infection, indicating the broad functions of the GALNT family, and the direct involvement of GALNTs during viral infections.
Collapse
Affiliation(s)
- Bin Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO.1 Beichen West Road, Chaoyang District, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yufan Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO.1 Beichen West Road, Chaoyang District, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Wei Jiang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO.1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Li Zhu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO.1 Beichen West Road, Chaoyang District, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Kaili Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO.1 Beichen West Road, Chaoyang District, Beijing, 100101, China; Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, China
| | - Kai Zhou
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO.1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Dongsheng Dai
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Shan Chang
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Min Fang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO.1 Beichen West Road, Chaoyang District, Beijing, 100101, China; International College, University of Chinese Academy of Sciences, Beijing, 100101, China; Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510150, China.
| |
Collapse
|
18
|
Lu J, Duan X, Zhao W, Wang J, Wang H, Zhou K, Fang M. Aged Mice are More Resistant to Influenza Virus Infection due to Reduced Inflammation and Lung Pathology. Aging Dis 2018; 9:358-373. [PMID: 29896425 PMCID: PMC5988592 DOI: 10.14336/ad.2017.0701] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/01/2017] [Indexed: 12/24/2022] Open
Abstract
Immune responses are a double-edged sword. Effective and appropriate immune responses capable of controlling viral infection while also largely preserving tissue integrity, are critical for host survival. Too strong immune responses might result in immune pathology, while too weak immune responses might cause viral persistence. Physiologic ageing is accompanied with a decline in the normal functioning of the immune system, which is termed as "immunosenescence". We show that aged mice (16-19 months old) are more resistant to influenza A virus (IAV) infection than the young mice. Strong immune responses in the young mice after IAV infection result in faster clearance of virus, but also cause severe lung injury and higher mortality rate. While in the aged mice, the delayed and milder immune responses contribute to reduced pulmonary damage, and are still capable to clear the infection even with a slower kinetics, displaying a more resistant phenotype during IAV infection. Hence, our work demonstrates that moderate immune responses as a decline with ageing in the aged mice balance the immune pathology and viral clearance, might be beneficial for the host during certain circumstances. Our results provide important insight to our basic knowledge of immunosenescence and immune defenses to invading pathogens. Further, our results indicate that age factors should be considered when investigating the vaccination and therapeutic strategies for severe IAV infection.
Collapse
Affiliation(s)
- Jiao Lu
- 1CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,2University of Chinese Academy of Sciences, Beijing, China
| | - Xuefeng Duan
- 1CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wenming Zhao
- 1CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jing Wang
- 1CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,2University of Chinese Academy of Sciences, Beijing, China
| | - Haoyu Wang
- 3Institute of Health Sciences, Anhui University, Hefei, China.,1CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Kai Zhou
- 1CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Min Fang
- 1CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,4International College, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
19
|
Abstract
Immunosuppression renders the host increased susceptible for secondary infections. It is becoming increasingly clear that not only bacterial sepsis, but also respiratory viruses with both severe and mild disease courses such as influenza, respiratory syncytial virus, and the human rhinovirus may induce immunosuppression. In this review, the current knowledge on (mechanisms of) bacterial- and virus-induced immunosuppression and the accompanying susceptibility toward various secondary infections is described. In addition, the frequently encountered secondary pathogens and their preferred localizations are presented. Finally, future perspectives in the context of the development of diagnostic markers and possibilities for personalized therapy to improve the diagnosis and treatment of immunocompromised patients are discussed.
Collapse
|
20
|
Duan X, Lu J, Wang H, Liu X, Wang J, Zhou K, Jiang W, Wang Y, Fang M. Bidirectional factors impact the migration of NK cells to draining lymph node in aged mice during influenza virus infection. Exp Gerontol 2017; 96:127-137. [PMID: 28669820 DOI: 10.1016/j.exger.2017.06.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/14/2017] [Accepted: 06/28/2017] [Indexed: 02/03/2023]
Abstract
Natural killer (NK) cells play an important role in controlling several viral diseases. Our previous studies demonstrated an age-dependent susceptibility to mousepox due to defective NK cell responses and trafficking. However, the mechanisms that underlie the age-related impairment in NK cell migration have yet to be identified. In the present study, we demonstrated that after influenza A virus (IAV) infection, NK cells from aged mice (17-19months old) failed to accumulate in draining lymph node (D-LN). We found that both environmental and intrinsic factors played roles for this defect. After infection, increase of chemokine transcripts, especially CXCL9, 10 and 11, which are important for NK cells homing to D-LN, was significantly lower in the D-LN of aged mice compared with those of young mice. Further, the expression levels of β2-integrins and β-actins, which play critical roles in NK cells homing to D-LN failed to be up-regulated in NK cells from aged mice. Finally, actin polymerization rates in NK cells from aged mice were also delayed compared to that of the young mice after IAV infection. Taken together, our data indicate that bi-directional factors play essential roles in the defective NK cell trafficking to the D-LN in the aged mice after IAV infection.
Collapse
Affiliation(s)
- Xuefeng Duan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO. 1 Beichen West Road, Chaoyang District, Beijing 100101, China.
| | - Jiao Lu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO. 1 Beichen West Road, Chaoyang District, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, China
| | - Haoyu Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO. 1 Beichen West Road, Chaoyang District, Beijing 100101, China; Institute of Health Sciences, Anhui University, Hefei, China
| | - Xiaofei Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, China.
| | - Jing Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO. 1 Beichen West Road, Chaoyang District, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, China
| | - Kai Zhou
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO. 1 Beichen West Road, Chaoyang District, Beijing 100101, China.
| | - Wei Jiang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO. 1 Beichen West Road, Chaoyang District, Beijing 100101, China.
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, China.
| | - Min Fang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO. 1 Beichen West Road, Chaoyang District, Beijing 100101, China; International College, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
21
|
Ansari AR, Liu H. Acute Thymic Involution and Mechanisms for Recovery. Arch Immunol Ther Exp (Warsz) 2017; 65:401-420. [PMID: 28331940 DOI: 10.1007/s00005-017-0462-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 03/12/2017] [Indexed: 12/14/2022]
Abstract
Acute thymic involution (ATI) is usually regarded as a virulence trait. It is caused by several infectious agents (bacteria, viruses, parasites, fungi) and other factors, including stress, pregnancy, malnutrition and chemotherapy. However, the complex mechanisms that operate during ATI differ substantially from each other depending on the causative agent. For instance, a transient reduction in the size and weight of the thymus and depletion of populations of T cell subsets are hallmarks of ATI in many cases, whereas severe disruption of the anatomical structure of the organ is also associated with some factors, including fungal, parasitic and viral infections. However, growing evidence shows that ATI may be therapeutically halted or reversed. In this review, we highlight the current progress in this field with respect to numerous pathological factors and discuss the possible mechanisms. Moreover, these new observations also show that ATI can be mechanistically reversed.
Collapse
Affiliation(s)
- Abdur Rahman Ansari
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, Hubei, China.,Section of Anatomy and Histology, Department of Basic Sciences, College of Veterinary and Animal Sciences (CVAS), Jhang, Pakistan.,University of Veterinary and Animal Sciences (UVAS), Lahore, Pakistan
| | - Huazhen Liu
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, Hubei, China.
| |
Collapse
|