1
|
Herrera-Uribe J, Convery O, ALmohammadi D, Weinberg FI, Stevenson NJ. The Neglected Suppressor of Cytokine Signalling (SOCS): SOCS4-7. Inflammation 2024:10.1007/s10753-024-02163-7. [PMID: 39460806 DOI: 10.1007/s10753-024-02163-7] [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: 07/22/2024] [Revised: 09/30/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024]
Abstract
SOCS proteins are essential for the regulation of oncogenic, anti-pathogenic, and proinflammatory signalling cascades, including the JAK/STAT and NF-kB pathways, where they act as negative feedback regulators. Given their powerful role in a broad spectrum of biological processes, it is surprising that the functions of many SOCS proteins have not been widely explored. While the mechanisms of action of CIS, SOCS1-3 are well-documented, information regarding SOCS4-7 remains limited. However, recent studies have begun to elucidate the regulatory functions of these proteins during infection and disease, such as influenza infection, cancer and diabetes. Therefore, this review aims to describe and discuss studies detailing our current understanding of SOCS4-7, painting a clearer picture of the biological processes these regulatory proteins maintain. Indeed, our review highlights important evidence proving that all SOCS play a role in biological processes that are essential for normal immunological homeostasis, clearance of infection and avoidance of disease. Understanding how SOCS proteins interact with other proteins or how they are dysregulated in disease is likely to provide valuable insights for advancing therapeutic approaches.
Collapse
Affiliation(s)
- Juber Herrera-Uribe
- Viral Immunology Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Orla Convery
- Viral Immunology Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Daniah ALmohammadi
- Viral Immunology Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Fabienne Ingrid Weinberg
- Viral Immunology Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Nigel J Stevenson
- Viral Immunology Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
2
|
Cirillo E, Tarallo A, Toriello E, Carissimo A, Giardino G, De Rosa A, Damiano C, Soresina A, Badolato R, Dellepiane RM, Baselli LA, Carrabba M, Fabio G, Bertolini P, Montin D, Conti F, Romano R, Pozzi E, Ferrero G, Roncarati R, Ferracin M, Brusco A, Parenti G, Pignata C. MicroRNA dysregulation in ataxia telangiectasia. Front Immunol 2024; 15:1444130. [PMID: 39224604 PMCID: PMC11366618 DOI: 10.3389/fimmu.2024.1444130] [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: 06/05/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Ataxia telangiectasia (AT) is a rare disorder characterized by neurodegeneration, combined immunodeficiency, a predisposition to malignancies, and high clinical variability. Profiling of microRNAs (miRNAs) may offer insights into the underlying mechanisms of complex rare human diseases, as miRNAs play a role in various biological functions including proliferation, differentiation, and DNA repair. In this study, we investigate the differential expression of miRNAs in samples from AT patients to identify miRNA patterns and analyze how these patterns are related to the disease. Methods We enrolled 20 AT patients (mean age 17.7 ± 9.6 years old) and collected clinical and genetic data. We performed short non-coding RNA-seq analysis on peripheral blood mononuclear cells (PBMCs) and fibroblasts to compare the miRNA expression profile between AT patients and controls. Results We observed 42 differentially expressed (DE)-miRNAs in blood samples and 26 in fibroblast samples. Among these, three DE-miRNAs, miR-342-3p, miR-30a-5p, and miR-195-5p, were further validated in additional AT samples, confirming their dysregulation. Discussion We identified an AT-related miRNA signature in blood cells and fibroblast samples collected from a group of AT patients. We also predicted several dysregulated pathways, primarily related to cancer, immune system control, or inflammatory processes. The findings suggest that miRNAs may provide insights into the pathophysiology and tumorigenesis of AT and have the potential to serve as useful biomarkers in cancer research.
Collapse
Affiliation(s)
- Emilia Cirillo
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Antonietta Tarallo
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Elisabetta Toriello
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | | | - Giuliana Giardino
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Antonio De Rosa
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Carla Damiano
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Annarosa Soresina
- Department of Clinical and Experimental Sciences, University of Brescia and Department of Pediatrics, ASST-Spedali Civili, Brescia, Italy
| | - Raffaele Badolato
- Department of Clinical and Experimental Sciences, University of Brescia and Department of Pediatrics, ASST-Spedali Civili, Brescia, Italy
| | - Rosa Maria Dellepiane
- Pediatric Area, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lucia A. Baselli
- Pediatric Area, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Carrabba
- Department of Internal Medicine, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giovanna Fabio
- Department of Internal Medicine, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Patrizia Bertolini
- Unità Operativa Complessa (U.O.C) di Pediatria e Oncoematologia, Azienda Ospedaliero Universitaria Parma, Parma, Italy
| | - Davide Montin
- Department of Pediatric and Public Health Sciences, University of Torino, Torino, Italy
| | - Francesca Conti
- Pediatric Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Roberta Romano
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Elisa Pozzi
- Centro Regionale di Biologia Molecolare – Arpa Piemonte, Torino, Italy
| | - Giulio Ferrero
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Roberta Roncarati
- Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche (CNR), Bologna, Italy
| | - Manuela Ferracin
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Alfredo Brusco
- Department of Neurosciences Rita Levi Montalcini, University of Torino, Torino, Italy
- Unit of Medical Genetics, Città della Salute e della Scienza University Hospital, Torino, Italy
| | - Giancarlo Parenti
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Claudio Pignata
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| |
Collapse
|
3
|
Ran F, Wang Y, Zhang G, Guo H, Li J, Zhang X, Wu Z, Bi L. Whole-transcriptome sequencing of phagocytes reveals a ceRNA network contributing to natural resistance to tuberculosis infection. Microb Pathog 2024; 192:106681. [PMID: 38754565 DOI: 10.1016/j.micpath.2024.106681] [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: 10/03/2023] [Revised: 03/14/2024] [Accepted: 05/06/2024] [Indexed: 05/18/2024]
Abstract
Tuberculosis (TB) is a major fatal infectious disease globally, exhibiting high morbidity rates and impacting public health and other socio-economic factors. However, some individuals are resistant to TB infection and are referred to as "Resisters". Resisters remain uninfected even after exposure to high load of Mycobacterium tuberculosis (Mtb). To delineate this further, this study aimed to investigate the factors and mechanisms influencing the Mtb resistance phenotype. We assayed the phagocytic capacity of peripheral blood mononuclear cells (PBMCs) collected from Resisters, patients with latent TB infection (LTBI), and patients with active TB (ATB), following infection with fluorescent Mycobacterium bovis Bacillus Calmette-Guérin (BCG). Phagocytosis was stronger in PBMCs from ATB patients, and comparable in LTBI patients and Resisters. Subsequently, phagocytes were isolated and subjected to whole transcriptome sequencing and small RNA sequencing to analyze transcriptional expression profiles and identify potential targets associated with the resistance phenotype. The results revealed that a total of 277 mRNAs, 589 long non-coding RNAs, 523 circular RNAs, and 35 microRNAs were differentially expressed in Resisters and LTBI patients. Further, the endogenous competitive RNA (ceRNA) network was constructed from differentially expressed genes after screening. Bioinformatics, statistical analysis, and quantitative real-time polymerase chain reaction were used for the identification and validation of potential crucial targets in the ceRNA network. As a result, we obtained a ceRNA network that contributes to the resistance phenotype. TCONS_00034796-F3, ENST00000629441-DDX43, hsa-ATAD3A_0003-CYP17A1, and XR_932996.2-CERS1 may be crucial association pairs for resistance to TB infection. Overall, this study demonstrated that the phagocytic capacity of PBMCs was not a determinant of the resistance phenotype and that some non-coding RNAs could be involved in the natural resistance to TB infection through a ceRNA mechanism.
Collapse
Affiliation(s)
- Fanlei Ran
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Yaguo Wang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; TB Healthcare Co., Ltd., Foshan, 528300, China
| | - Guoqin Zhang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haiyan Guo
- Department of Geriatrics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China; Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Jinlong Li
- TB Healthcare Co., Ltd., Foshan, 528300, China
| | - Xilin Zhang
- Foshan Fourth People's Hospital, Foshan, 528000, China.
| | - Zhilong Wu
- Foshan Fourth People's Hospital, Foshan, 528000, China.
| | - Lijun Bi
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Guangzhou National Laboratory, Guangzhou, 510005, China.
| |
Collapse
|
4
|
Zhao L, Fan K, Sun X, Li W, Qin F, Shi L, Gao F, Zheng C. Host-directed therapy against mycobacterium tuberculosis infections with diabetes mellitus. Front Immunol 2024; 14:1305325. [PMID: 38259491 PMCID: PMC10800548 DOI: 10.3389/fimmu.2023.1305325] [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: 10/01/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Tuberculosis (TB) is caused by the bacterial pathogen Mycobacterium tuberculosis (MTB) and is one of the principal reasons for mortality and morbidity worldwide. Currently, recommended anti-tuberculosis drugs include isoniazid, rifampicin, ethambutol, and pyrazinamide. TB treatment is lengthy and inflicted with severe side-effects, including reduced patient compliance with treatment and promotion of drug-resistant strains. TB is also prone to other concomitant diseases such as diabetes and HIV. These drug-resistant and complex co-morbid characteristics increase the complexity of treating MTB. Host-directed therapy (HDT), which effectively eliminates MTB and minimizes inflammatory tissue damage, primarily by targeting the immune system, is currently an attractive complementary approach. The drugs used for HDT are repositioned drugs in actual clinical practice with relative safety and efficacy assurance. HDT is a potentially effective therapeutic intervention for the treatment of MTB and diabetic MTB, and can compensate for the shortcomings of current TB therapies, including the reduction of drug resistance and modulation of immune response. Here, we summarize the state-of-the-art roles and mechanisms of HDT in immune modulation and treatment of MTB, with a special focus on the role of HDT in diabetic MTB, to emphasize the potential of HDT in controlling MTB infection.
Collapse
Affiliation(s)
- Li Zhao
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Ke Fan
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Xuezhi Sun
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Wei Li
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Fenfen Qin
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Liwen Shi
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Feng Gao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chunlan Zheng
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| |
Collapse
|
5
|
Wang J, Gan L, Li F, Li Q, Wu T, Wu Z, Chen P, Scicluna BP, Feng X, Gu J, Han W, Li N, Lei L. Tracheal epithelial cell-exosome-derived MiR-21-5p inhibits alveolar macrophage pyroptosis to resist pulmonary bacterial infection through PIK3CD-autophagy pathway. Life Sci 2024; 336:122340. [PMID: 38092143 DOI: 10.1016/j.lfs.2023.122340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/06/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023]
Abstract
AIMS Structural cells play an important role in regulating immune cells during infection. Our aim was to determine whether structural porcine tracheal epithelial cells (PTECs) can regulate alveolar macrophages (AMs) to prevent bacterial pneumonia, explore the underlying mechanism(s) and therapeutic target. MATERIALS AND METHODS Actinobacillus pleuropneumoniae (APP) was used as the model strain for infection studies. Small RNA sequencing was used to identify differentially abundant exosome-derived miRNAs. The role of PTECs exosome-derived miR-21-5p in regulating AMs autophagy, pyroptosis, reactive oxygen species (ROS) was determined using RT-qPCR, western-blotting, flow cytometry, immunohistochemistry. Luciferase reporter assays were conducted to identify potential binding targets of miR-21-5p. The universality of miR-21-5p action on resistance to bacterial pulmonary infection was demonstrated using Klebsiella pneumoniae or Staphylococcus aureus in vitro and in vivo infection models. KEY FINDINGS MiR-21-5p was enriched in PETCs-derived exosomes, which protected AMs against pulmonary bacterial infection. Mechanistically, miR-21-5p targeted PIK3CD, to promote autophagy of AMs, which reduced the pyroptosis induced by APP infection via inhibiting the over-production of ROS, which in turn suppressed the over-expression of pro-inflammatory cytokines, and increased bacterial clearance. Importantly, the protective effect and mechanism of miR-21-5p were universal as they also occurred upon challenge with Klebsiella pneumoniae and Staphylococcus aureus. SIGNIFICANCE Our data reveals miR-21-5p can promote pulmonary resistance to bacterial infection by inhibiting pyroptosis of alveolar macrophages through the PIK3CD-autophagy-ROS pathway, suggesting PIK3CD may be a potential therapeutic target for bacterial pneumonia.
Collapse
Affiliation(s)
- Jun Wang
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Lin Gan
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Fengyang Li
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Qin Li
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Tong Wu
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zengshuai Wu
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Peiru Chen
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Brendon P Scicluna
- Department of Applied Biomedical Science, Faculty of Health Sciences, Mater Dei Hospital, University of Malta, Msida, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Xin Feng
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jingmin Gu
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Wenyu Han
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Na Li
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Liancheng Lei
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China.
| |
Collapse
|
6
|
Wei W, Bai H, Zhang T, Cai S, Zhou Y, Liu M, Zhang Y, Chen Y, Hua J, He J, Ding N, Miao G, Wang J. Regulation of Circulating miR-342-3p Alleviates the Radiation-Induced Immune System Injury. Radiat Res 2023; 200:556-568. [PMID: 37874034 DOI: 10.1667/rade-23-00125.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/12/2023] [Indexed: 10/25/2023]
Abstract
Ionizing radiation in space, radiation devices or nuclear disasters are major threats to human health and public security. Expanding countermeasures for dealing with accidental or occupational radiation exposure is crucial for the protection of radiation injuries. Circulating microRNAs (miRNAs) have emerged as promising radiation biomarkers in recent years. However, the origin, distribution and functions of radiosensitive circulating miRNAs remain unclear, which obstructs their clinical applications in the future. In this study, we found that mmu-miR-342-3p (miR-342) in mouse serum presents a stable and significant decrease after X-ray total-body irradiation (TBI). Focusing on this miRNA, we investigated the influences of circulating miR-342 on the radiation-induced injury. Through tail vein injection of Cy5-labeled synthetic miR-342, we found the exogenous miR-342-Cy5 was mainly enriched in metabolic and immune organs. Besides, the bioinformatic analysis predicted that miR-342 might involve in immune-related processes or pathways. Further, mice were tail vein injected with synthetic miR-342 mimetics (Ago-miR-342) after irradiation to upregulate the level of miR-342 in circulating blood. The results showed that the upregulation of circulating miR-342 alleviated the radiation-induced depletion of CD3+CD4+ T lymphocytes and influenced the levels of IL-2 and IL-6 in irradiated mice. Moreover, the injection of Ago-miR-342 improved the survival rates of mice with acute radiation injury. Our findings demonstrate that upregulation of circulating miR-342 alleviates the radiation-induced immune system injury, which provides us new insights into the functions of circulating miRNAs and the prospect as the targets for mitigation of radiation injuries.
Collapse
Affiliation(s)
- Wenjun Wei
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Hao Bai
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Tianyi Zhang
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Shufan Cai
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
- School of Nuclear Science and Technology, Lanzhou University, Lanzhou 730000, China
| | - Yumeng Zhou
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China
| | - Min Liu
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Yanan Zhang
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Yaxiong Chen
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Junrui Hua
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Jinpeng He
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Nan Ding
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - GuoYing Miao
- Department of Radiation Oncology, Gansu Provincial Central Hospital, Lanzhou 730000, China
| | - Jufang Wang
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| |
Collapse
|
7
|
Fu B, Xiong Y, Sha Z, Xue W, Xu B, Tan S, Guo D, Lin F, Wang L, Ji J, Luo Y, Lin X, Wu H. SEPTIN2 suppresses an IFN-γ-independent, proinflammatory macrophage activation pathway. Nat Commun 2023; 14:7441. [PMID: 37978190 PMCID: PMC10656488 DOI: 10.1038/s41467-023-43283-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023] Open
Abstract
Interferon-gamma (IFN-γ) signaling is necessary for the proinflammatory activation of macrophages but IFN-γ-independent pathways, for which the initiating stimuli and downstream mechanisms are lesser known, also contribute. Here we identify, by high-content screening, SEPTIN2 (SEPT2) as a negative regulation of IFN-γ-independent macrophage autoactivation. Mechanistically, endoplasmic reticulum (ER) stress induces the expression of SEPT2, which balances the competition between acetylation and ubiquitination of heat shock protein 5 at position Lysine 327, thereby alleviating ER stress and constraining M1-like polarization and proinflammatory cytokine release. Disruption of this negative feedback regulation leads to the accumulation of unfolded proteins, resulting in accelerated M1-like polarization, excessive inflammation and tissue damage. Our study thus uncovers an IFN-γ-independent macrophage proinflammatory autoactivation pathway and suggests that SEPT2 may play a role in the prevention or resolution of inflammation during infection.
Collapse
Affiliation(s)
- Beibei Fu
- School of Life Sciences, Chongqing University, 401331, Chongqing, China
| | - Yan Xiong
- School of Life Sciences, Chongqing University, 401331, Chongqing, China
| | - Zhou Sha
- School of Life Sciences, Chongqing University, 401331, Chongqing, China
| | - Weiwei Xue
- School of Pharmaceutical Sciences, Chongqing University, 401331, Chongqing, China
| | - Binbin Xu
- School of Pharmaceutical Sciences, Chongqing University, 401331, Chongqing, China
| | - Shun Tan
- Chongqing Public Health Medical Center, 400036, Chongqing, China
| | - Dong Guo
- School of Life Sciences, Chongqing University, 401331, Chongqing, China
| | - Feng Lin
- School of Life Sciences, Chongqing University, 401331, Chongqing, China
| | - Lulu Wang
- School of Life Sciences, Chongqing University, 401331, Chongqing, China
| | - Jianjian Ji
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, 210023, Nanjing, China
| | - Yang Luo
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing University, 400044, Chongqing, China.
| | - Xiaoyuan Lin
- Institut für Virologie, Freie Universität Berlin, Robert-von-Ostertag-Str. 7-13, 14163, Berlin, Germany.
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), 400038, Chongqing, China.
| | - Haibo Wu
- School of Life Sciences, Chongqing University, 401331, Chongqing, China.
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing University, 400044, Chongqing, China.
| |
Collapse
|
8
|
Liu W, Hu C, Zhang B, Li M, Deng F, Zhao S. Exosomal microRNA-342-5p secreted from adipose-derived mesenchymal stem cells mitigates acute kidney injury in sepsis mice by inhibiting TLR9. Biol Proced Online 2023; 25:10. [PMID: 37085762 PMCID: PMC10120132 DOI: 10.1186/s12575-023-00198-y] [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: 10/13/2022] [Accepted: 04/11/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND Sepsis-related acute kidney injury (AKI) is an inflammatory disease associated with extremely high mortality and health burden. This study explored the possibility of exosomes secreted by adipose-derived mesenchymal stem cells (AMSCs) serving as a carrier for microRNA (miR)-342-5p to alleviate sepsis-related AKI and investigated the possible mechanism. METHODS Serum was obtained from 30 patients with sepsis-associated AKI and 30 healthy volunteers for the measurement of miR-342-5p, blood urea nitrogen (BUN), and serum creatinine (SCr) levels. For in vitro experiments, AMSCs were transfected with LV-miR-342-5p or LV-miR-67 to acquire miR-342-5p-modified AMSCs and miR-67-modified AMSCs, from which the exosomes (AMSC-Exo-342 and AMSC-Exo-67) were isolated. The human renal proximal tubular epithelial cell line HK-2 was induced by lipopolysaccharide (LPS) to construct a cellular model of sepsis. The expression of Toll-like receptor 9 (TLR9) was also detected in AKI cells and mouse models. The interaction between miR-342-5p and TLR9 was predicted by dual luciferase reporter gene assay. RESULTS Detection on clinical serum samples showed that BUN, SCr, and TLR9 were elevated and miR-342-5p level was suppressed in the serum of patients with sepsis-associated AKI. Transfection with LV-miR-342-5p reinforced miR-342-5p expression in AMSCs and AMSC-secreted exosomes. miR-342-5p negatively targeted TLR9. LPS treatment enhanced TLR9 expression, reduced miR-342-5p levels, suppressed autophagy, and increased inflammation in HK-2 cells, while the opposite trends were observed in LPS-induced HK-2 cells exposed to AMSC-Exo-342, Rapa, miR-342-5p mimic, or si-TLR9. Additionally, the effects of AMSC-Exo-342 on autophagy and inflammation in LPS-induced cells could be weakened by 3-MA or pcDNA3.1-TLR9 treatment. Injection of AMSC-Exo-342 enhanced autophagy, mitigated kidney injury, suppressed inflammation, and reduced BUN and SCr levels in sepsis-related AKI mouse models. CONCLUSION miR-342-5p transferred by exosomes from miR-342-5p-modified AMSCs ameliorated AKI by inhibiting TLR9 to accelerate autophagy.
Collapse
Affiliation(s)
- Wei Liu
- Department of Critical Care Medicine, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Chenghuan Hu
- Department of Critical Care Medicine, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Buyao Zhang
- Department of Critical Care Medicine, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Mingxia Li
- Department of Critical Care Medicine, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Fuxing Deng
- Department of Critical Care Medicine, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Shuangping Zhao
- Department of Critical Care Medicine, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China.
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
9
|
Peng ZM, Zhang YY, Wei D, Zhang XJ, Liu B, Peng J, Luo XJ. MALT1 promotes necroptosis in stroke rat brain via targeting the A20/RIPK3 pathway. Arch Biochem Biophys 2023; 735:109502. [PMID: 36603698 DOI: 10.1016/j.abb.2023.109502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/30/2022] [Accepted: 01/01/2023] [Indexed: 01/04/2023]
Abstract
Necroptosis has been demonstrated to contribute to brain injury in ischemic stroke, whereas A20 can exert anti-necroptosis effect via deubiquitinating receptor-interacting protein kinase (RIPK3) at k63 and it can be cleaved by MALT1. This study aims to explore whether MALT1 is upregulated in the brain during ischemic stroke and promotes brain cell necroptosis through enhancing the degradation of A20. Ischemic stroke model was established in Sprague Dawley rats by occlusion of the middle cerebral artery (MCA) for 2 h, followed by 24 h reperfusion, which showed brain injury (increase in neurological deficit score and infarct volume) concomitant with an upregulation of MALT1, a decrease in A20 level, and increases in necroptosis-associated protein levels [RIPK3, mixed lineage kinase domain-like protein (MLKL) and p-MLKL] and k63-ubiquitination of RIPK3 in brain tissues. Administration of MALT1 inhibitor (Ml-2) at 8 or 15 mg/kg (i.p.) at 1 h after ischemia significantly improved neurological function and reduced infarct volume together with a downregulation of MALT1, an increase in A20 level and decreases in necroptosis-associated protein levels and k63-ubiquitination of RIPK3. Similarly, knockdown of MALT1 could also reduce oxygen-glucose deprivation/reoxygenation (OGD/R)-induced injury in the cultured HT22 cells coincident with an increase in A20 level and decreases in necroptosis-associated protein levels and k63-ubiquitination of RIPK3. Based on these observations, we conclude that MALT1 promotes necroptosis in stroke rat brain via enhancing the degradation of A20, which leads to a decrease in the capability of A20 to deubiquitinate RIPK3 at k63 and a subsequent compromise in counteraction against the brain cell necroptosis.
Collapse
Affiliation(s)
- Zi-Mei Peng
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China; Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Yi-Yue Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Dan Wei
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Xiao-Jie Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Department of Pharmacy, Xiangya Hospital, Central South University, Hunan, China
| | - Bin Liu
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Department of Pharmacy, Xiangya Hospital, Central South University, Hunan, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| |
Collapse
|
10
|
Zhang J, Han L, Ma Q, Wang X, Yu J, Xu Y, Zhang X, Wu X, Deng G. RIP3 impedes Mycobacterium tuberculosis survival and promotes p62-mediated autophagy. Int Immunopharmacol 2023; 115:109696. [PMID: 36638666 DOI: 10.1016/j.intimp.2023.109696] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 12/18/2022] [Accepted: 01/03/2023] [Indexed: 01/13/2023]
Abstract
Macrophage is believed to play a vital role in the fight against Mycobacterium tuberculosis (M.tb) infection by activating autophagy. Recently, receptor-interacting protein kinase-3 (RIP3), an essential kinase for necroptotic cell death signaling, has been demonstrated to be involved in autophagy. However, RIP3's role in fighting against M.tb infection remains elusive. Here we show that a substantial increase in inflammatory cell infiltration and higher bacterial burden are observed in the lungs of RIP3-/- mice with Mycobacterium bovis Bacillus Calmette-Guerin (BCG) infection. Meanwhile, RIP3 ameliorates lung injury and promote autophagy via induce autophagosome and autophagolysosome formation which indicate that RIP3 is indispensable for host clearance of BCG via autophagy. Mechanically, RIP3 enhances p62 binding to ubiquitylated proteins and LC3 by interacting with p62, and RHIM domain is required for RIP3-p62 interaction. Hence, our results conclusively show that RIP3 impedes M.tb survival and promotes p62-mediated autophagy. The findings provide further insight into understanding the mechanism of M.tb immune escape and pathogenesis of tuberculosis.
Collapse
Affiliation(s)
- Jiamei Zhang
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, NingXia 750021, China; School of Life Science, NingXia University, Yinchuan, NingXia 750021, China
| | - Lu Han
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, NingXia 750021, China; School of Life Science, NingXia University, Yinchuan, NingXia 750021, China
| | - Qinmei Ma
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, NingXia 750021, China; School of Life Science, NingXia University, Yinchuan, NingXia 750021, China
| | - Xiaoping Wang
- Tuberculosis Reference Laboratory, Ningxia Institute for Tuberculosis Control, The Fourth People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia 750021, China
| | - Jialin Yu
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, NingXia 750021, China; School of Life Science, NingXia University, Yinchuan, NingXia 750021, China
| | - Yanan Xu
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, NingXia 750021, China; School of Life Science, NingXia University, Yinchuan, NingXia 750021, China
| | - Xu Zhang
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Xiaoling Wu
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, NingXia 750021, China; School of Life Science, NingXia University, Yinchuan, NingXia 750021, China.
| | - Guangcun Deng
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, NingXia 750021, China; School of Life Science, NingXia University, Yinchuan, NingXia 750021, China.
| |
Collapse
|
11
|
Xiong Y, Wang L, Xu S, Fu B, Che Y, Zaky MY, Tian R, Yao R, Guo D, Sha Z, Lin F, Lin X, Wu H. Small molecule Z363 co-regulates TAF10 and MYC via the E3 ligase TRIP12 to suppress tumour growth. Clin Transl Med 2023; 13:e1153. [PMID: 36639831 PMCID: PMC9839843 DOI: 10.1002/ctm2.1153] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/17/2022] [Accepted: 08/12/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The MYC oncoprotein, also known as the master regulator of genes, is a transcription factor that regulates numerous physiological processes, including cell cycle control, apoptosis, protein synthesis and cell adhesion, among others. MYC is overexpressed in approximately 70% of human cancers. Given its pervasive role in cancer biology, MYC down-regulation has become an attractive cancer treatment strategy. METHODS The CRISPR/Cas9 method was used to produce KO cell models. Western blot was used to analyzed the expressions of MYC and TATA-binding proteinassociated factors 10 (TAF10) in cancer cells (MCF7, A549, HepG2 cells) Cell culture studies were performed to determine the mechanisms by which small molecules (Z363119456, Z363) affects MYC and TAF10 expressions and functions. Mouse studies were carried out to investigate the impact of Z363 regulation on tumor growth. RESULTS Z363 activate Thyroid hormone Receptor-interacting Protein 12 (TRIP12), which phosphorylates MYC at Thr58, resulting in MYC ubiquitination and degradation and thereby regulating MYC target genes. Importantly, TRIP12 also induces TAF10 degradation, which reduces MYC protein levels. TRIP12, an E3 ligase, controls MYC levels both directly and indirectly by inhibiting MYC or TAF10 activity. CONCLUSIONS In summary,these results demonstrate the anti-cancer properties of Z363, a small molecule that is co-regulated by TAF10 and MYC.
Collapse
Affiliation(s)
- Yan Xiong
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Lulu Wang
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Shiyao Xu
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Beibei Fu
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Yuchen Che
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Mohamed Y. Zaky
- Molecular Physiology DivisionZoology DepartmentFaculty of ScienceBeni‐Suef UniversityBeni‐SuefEgypt,Department of OncologyFaculty of MedicineLinköping UniversitySweden,Department of Biomedical and Clinical SciencesFaculty of MedicineLinköping UniversitySweden
| | - Rong Tian
- Department of Biomedical and Clinical SciencesFaculty of MedicineLinköping UniversitySweden
| | - Rui Yao
- Department of PathologyChongqing Hygeia HospitalChongqingChina
| | - Dong Guo
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Zhou Sha
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Feng Lin
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Xiaoyuan Lin
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Haibo Wu
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| |
Collapse
|
12
|
Dong W, Wang G, Bai Y, Li Y, Huo X, Zhao J, Lu W, Lu H, Wang C, Wang X, Chen H, Tan C. Analysis of the noncoding RNA regulatory networks of H37Rv- and H37Rv△1759c-infected macrophages. Front Microbiol 2023; 14:1106643. [PMID: 36992931 PMCID: PMC10042141 DOI: 10.3389/fmicb.2023.1106643] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/13/2023] [Indexed: 03/31/2023] Open
Abstract
Noncoding RNAs regulate the process of Mycobacterium tuberculosis (M. tb) infecting the host, but there is no simultaneous transcriptional information of long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) and the global regulatory networks of non-coding RNA. Rv1759c, a virulence factor, is a member of protein family containing the proline-glutamic acid (PE) in M. tb, which can increase M. tb survival. To reveal the noncoding RNA regulatory networks and the effect of Rv1759c on non-coding RNA expression during M. tb infection, we collected samples of H37Rv- and H37Rv△1759c-infected macrophages and explored the full transcriptome expression profile. We found 356 mRNAs, 433 lncRNAs, 168 circRNAs, and 12 miRNAs differentially expressed during H37Rv infection, 356 mRNAs, 433 lncRNAs, 168 circRNAs, and 12 miRNAs differentially expressed during H37Rv△1759c infection. We constructed lncRNA/circRNA-miRNA-mRNA regulatory networks during H37Rv and H37Rv△1759c infection. We demonstrated the role of one of the hubs of the networks, hsa-miR-181b-3p, for H37Rv survival in macrophages. We discovered that the expression changes of 68 mRNAs, 92 lncRNAs, 26 circRNAs, and 3 miRNAs were only related to the deletion of Rv1759c by comparing the transcription profiles of H37Rv and H37Rv△1759c. Here, our study comprehensively characterizes the transcriptional profiles in THP1-derived-macrophages infected with H37Rv and H37Rv△1759c, which provides support and new directions for in-depth exploration of noncoding RNA and PE/PPE family functions during the infection process.
Collapse
Affiliation(s)
- Wenqi Dong
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
| | - Gaoyan Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yajuan Bai
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yuxin Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xinyu Huo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Jing Zhao
- WuHan Animal Disease Control Center, Wuhan, Hubei, China
| | - Wenjia Lu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Hao Lu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Chenchen Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xiangru Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
| | - Chen Tan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- *Correspondence: Chen Tan,
| |
Collapse
|
13
|
周 琴, 刘 健, 孙 艳, 陈 晓, 张 先, 丁 香. [Expression of miR-342-3p in rheumatoid arthritis patients and its effect on synovial fibroblast inflammation and migration]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:1712-1719. [PMID: 36504065 PMCID: PMC9742770 DOI: 10.12122/j.issn.1673-4254.2022.11.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To investigate the expression level of miR-342-3p in peripheral blood mononuclear cells (PBMCs) of patients with rheumatoid arthritis (RA) and its effect on inflammatory response and migration of synovial fibroblasts in RA (RA-FLS). METHODS PBMCs were collected from 30 healthy individuals and 50 RA patients for detecting the expression of miR-342-3p, and its correlation with the clinical indicators RF, ESR, anti-CCP, hs-CRP, C3, DAS-28, SAS, and SDS was analyzed. In RA-FLS cultures, the effect of transfection of miR-342-3p mimics and inhibitor on TNF-α-induced inflammatory response of the cells was evaluated by detecting the expressions of IL-1β, IL-6, IL-10, and TNF-α using ELISA. CCK8 assay and Transwell assay were used for detecting the changes in cell viability and migration ability of the synovial cells. RESULTS In RA patients, the expression level of miR-342-3p was significantly lowered in the PBMCs (P < 0.05) with an area under the ROC curve of 97.53% and showed inverse correlations with RF (r=-0.321), ESR(r=-0.284), anti-CCP (r=-0.355), hs-CRP (r=-0.320), C3 (r=-0.294), DAS-28 (r=-0.395), SAS (r=-0.366), and SDS (r=-0.397) (all P < 0.05); a low expression of miR-342-3p was strongly associated with elevated levels of anti-CCP, DAS-28, SDS, and SAS (all with a rule support greater than 85%, confidence greater than 88%, and lift greater than 1). In cultured RA-FLS, TNF-α stimulation significantly increased the cell viability (P < 0.05), upregulated the expressions of IL-1β, IL-6, and TNF-α, and lowered the expression of IL-10 (P < 0.05). These changes were significantly suppressed by transfection of the cells with miR-342-3p mimics (P < 0.05) but enhanced by transfection with miR-342-3p inhibitor (P < 0.05). CONCLUSION The expression of miR-342-3p is decreased in the PBMCs of RA patients. A lowered expression of miR-342-3p contributes to the pathogenesis of RA by promoting inflammatory responses and migration of RA-FLS.
Collapse
Affiliation(s)
- 琴 周
- />安徽中医药大学第一附属医院,安徽 合肥 230012First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei 230012, China
| | - 健 刘
- />安徽中医药大学第一附属医院,安徽 合肥 230012First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei 230012, China
| | - 艳秋 孙
- />安徽中医药大学第一附属医院,安徽 合肥 230012First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei 230012, China
| | - 晓露 陈
- />安徽中医药大学第一附属医院,安徽 合肥 230012First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei 230012, China
| | - 先恒 张
- />安徽中医药大学第一附属医院,安徽 合肥 230012First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei 230012, China
| | - 香 丁
- />安徽中医药大学第一附属医院,安徽 合肥 230012First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei 230012, China
| |
Collapse
|
14
|
Fang J, Dong C, Xiong S. Mycobacterium tuberculosis Rv0790c inhibits the cellular autophagy at its early stage and facilitates mycobacterial survival. Front Cell Infect Microbiol 2022; 12:1014897. [DOI: 10.3389/fcimb.2022.1014897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/25/2022] [Indexed: 11/11/2022] Open
Abstract
Rv0790c is predicted to be a conserved hypothetical protein encoded by Mycobacterium tuberculosis (Mtb). However, its function in Mtb infection remains largely unknown. In this study, we found that Rv0790c promoted bacillary survival of M. smegmatis (Ms), both in vitro and in vivo. The bacillary burden of Ms exogenously expressing Rv0790c increased, whereas in Rv0790c-knockouts the bacillary burden decreased in infected macrophages. Multiple cellular processes were analyzed to explore the underlying mechanisms. We found that neither inflammatory regulation nor apoptotic induction were responsible for the promotion of bacillary survival mediated by Rv0790c. Interestingly, we found that Rv0790c facilitates mycobacterial survival through cellular autophagy at its early stage. Immunoprecipitation assay of autophagy initiation-related proteins indicated that Rv0790c interacted with mTOR and enhanced its activity, as evidenced by the increased phosphorylation level of mTOR downstream substrates, ULK-1, at Ser757 and P70S6K, at Thr389. Our study uncovers a novel autophagy suppressor encoded by mycobacterial Rv0790c, which inhibits the early stage of cellular autophagy induction upon Mtb infection and takes an important role in maintaining intracellular mycobacterial survival. It may aid in understanding the mechanism of Mtb evasion of host cellular degradation, as well as hold the potential to develop new targets for the prevention and treatment of tuberculosis.
Collapse
|
15
|
Liang S, Ma J, Gong H, Shao J, Li J, Zhan Y, Wang Z, Wang C, Li W. Immune regulation and emerging roles of noncoding RNAs in Mycobacterium tuberculosis infection. Front Immunol 2022; 13:987018. [PMID: 36311754 PMCID: PMC9608867 DOI: 10.3389/fimmu.2022.987018] [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: 07/05/2022] [Accepted: 08/29/2022] [Indexed: 05/10/2024] Open
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis, engenders an onerous burden on public hygiene. Congenital and adaptive immunity in the human body act as robust defenses against the pathogens. However, in coevolution with humans, this microbe has gained multiple lines of mechanisms to circumvent the immune response to sustain its intracellular persistence and long-term survival inside a host. Moreover, emerging evidence has revealed that this stealthy bacterium can alter the expression of demic noncoding RNAs (ncRNAs), leading to dysregulated biological processes subsequently, which may be the rationale behind the pathogenesis of tuberculosis. Meanwhile, the differential accumulation in clinical samples endows them with the capacity to be indicators in the time of tuberculosis suffering. In this article, we reviewed the nearest insights into the impact of ncRNAs during Mycobacterium tuberculosis infection as realized via immune response modulation and their potential as biomarkers for the diagnosis, drug resistance identification, treatment evaluation, and adverse drug reaction prediction of tuberculosis, aiming to inspire novel and precise therapy development to combat this pathogen in the future.
Collapse
Affiliation(s)
- Shufan Liang
- Department of Respiratory and Critical Care Medicine, Med-X Center for Manufacturing, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Jiechao Ma
- Artificial Intelligence (AI) Lab, Deepwise Healthcare, Beijing, China
| | - Hanlin Gong
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jun Shao
- Department of Respiratory and Critical Care Medicine, Med-X Center for Manufacturing, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Jingwei Li
- Department of Respiratory and Critical Care Medicine, Med-X Center for Manufacturing, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Yuejuan Zhan
- Department of Respiratory and Critical Care Medicine, Med-X Center for Manufacturing, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Zhoufeng Wang
- Department of Respiratory and Critical Care Medicine, Med-X Center for Manufacturing, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Chengdi Wang
- Department of Respiratory and Critical Care Medicine, Med-X Center for Manufacturing, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, Med-X Center for Manufacturing, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Simper JD, Perez E, Schlesinger LS, Azad AK. Resistance and Susceptibility Immune Factors at Play during Mycobacterium tuberculosis Infection of Macrophages. Pathogens 2022; 11:pathogens11101153. [PMID: 36297211 PMCID: PMC9611686 DOI: 10.3390/pathogens11101153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/27/2022] [Accepted: 10/01/2022] [Indexed: 11/28/2022] Open
Abstract
Tuberculosis (TB), caused by infection with Mycobacterium tuberculosis (M.tb), is responsible for >1.5 million deaths worldwide annually. Innate immune cells, especially macrophages, are the first to encounter M.tb, and their response dictates the course of infection. During infection, macrophages exert a variety of immune factors involved in either controlling or promoting the growth of M.tb. Research on this topic has been performed in both in vitro and in vivo animal models with discrepant results in some cases based on the model of study. Herein, we review macrophage resistance and susceptibility immune factors, focusing primarily on recent advances in the field. We include macrophage cellular pathways, bioeffector proteins and molecules, cytokines and chemokines, associated microbiological factors and bacterial strains, and host genetic factors in innate immune genes. Recent advances in mechanisms underlying macrophage resistance and susceptibility factors will aid in the successful development of host-directed therapeutics, a topic emphasized throughout this review.
Collapse
Affiliation(s)
- Jan D. Simper
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
- Department of Microbiology, Immunology and Molecular Genetics, UT Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Esteban Perez
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
- Translational Sciences Program, UT Health San Antonio Graduate School of Biomedical Sciences, San Antonio, TX 78229, USA
| | - Larry S. Schlesinger
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
- Correspondence: (L.S.S.); (A.K.A.); Tel.: +1-210-258-9578 (L.S.S.); +1-210-258-9467 (A.K.A.)
| | - Abul K. Azad
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
- Correspondence: (L.S.S.); (A.K.A.); Tel.: +1-210-258-9578 (L.S.S.); +1-210-258-9467 (A.K.A.)
| |
Collapse
|
17
|
Tao YC, Wang YH, Wang ML, Jiang W, Wu DB, Chen EQ, Tang H. Upregulation of microRNA-125b-5p alleviates acute liver failure by regulating the Keap1/Nrf2/HO-1 pathway. Front Immunol 2022; 13:988668. [PMID: 36268033 PMCID: PMC9578503 DOI: 10.3389/fimmu.2022.988668] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/15/2022] [Indexed: 12/14/2022] Open
Abstract
Background Acute liver failure (ALF) and acute-on-chronic liver failure (ACLF) are the two most common subtypes of liver failure. They are both life-threatening clinical problems with high short-term mortality. Although liver transplantation is an effective therapeutic, its application is limited due to the shortage of donor organs. Given that both ACLF and ALF are driven by excessive inflammation in the initial stage, molecules targeting inflammation may benefit the two conditions. MicroRNAs (miRNAs) are a group of small endogenous noncoding interfering RNA molecules. Regulation of miRNAs related to inflammation may serve as promising interventions for the treatment of liver failure. Aims To explore the role and mechanism of miR-125b-5p in the development of liver failure. Methods Six human liver tissues were categorized into HBV-non-ACLF and HBV-ACLF groups. Differentially expressed miRNAs (DE-miRNAs) were screened and identified through high-throughput sequencing analysis. Among these DE-miRNAs, miR-125b-5p was selected for further study of its role and mechanism in lipopolysaccharide (LPS)/D-galactosamine (D-GalN) -challenged Huh7 cells and mice in vitro and in vivo. Results A total of 75 DE-miRNAs were obtained. Of these DE-miRNAs, miR-125b-5p was the focus of further investigation based on our previous findings and preliminary results. We preliminarily observed that the levels of miR-125b-5p were lower in the HBV-ACLF group than in the HBV-non-ACLF group. Meanwhile, LPS/D-GalN-challenged mice and Huh7 cells both showed decreased miR-125b-5p levels when compared to their untreated control group, suggesting that miR-125b-5p may have a protective role against liver injury, regardless of ACLF or ALF. Subsequent results revealed that miR-125b-5p not only inhibited Huh7 cell apoptosis in vitro but also relieved mouse ALF in vivo with evidence of improved liver histology, decreased alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, and reduced tumor necrosis factor-α (TNF-α) and IL-1β levels. Based on the results of a biological prediction website, microRNA.org, Kelch-like ECH-associated protein 1 (Keap1) was predicted to be one of the target genes of miR-125b-5p, which was verified by a dual-luciferase reporter gene assay. Western blot results in vitro and in vivo showed that miR-125b-5p could decrease the expression of Keap1 and cleaved caspase-3 while upregulating the expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and heme oxygenase-1(HO-1). Conclusion Upregulation of miR-125b-5p can alleviate acute liver failure by regulating the Keap1/Nrf2/HO-1 pathway, and regulation of miR-125b-5p may serve as an alternative intervention for liver failure.
Collapse
Affiliation(s)
- Ya-Chao Tao
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China,Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Yong-Hong Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China,Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Meng-Lan Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China,Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Wei Jiang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China,Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Dong-Bo Wu
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China,Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - En-Qiang Chen
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China,Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China,*Correspondence: Hong Tang, ; En-Qiang Chen,
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China,Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China,*Correspondence: Hong Tang, ; En-Qiang Chen,
| |
Collapse
|
18
|
Han M, Chen Z, He P, Li Z, Chen Q, Tong Z, Wang M, Du H, Zhang H. YgiM may act as a trigger in the sepsis caused by Klebsiella pneumoniae through the membrane-associated ceRNA network. Front Genet 2022; 13:973145. [PMID: 36212144 PMCID: PMC9537587 DOI: 10.3389/fgene.2022.973145] [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: 06/19/2022] [Accepted: 09/07/2022] [Indexed: 11/27/2022] Open
Abstract
Sepsis is one of the diseases that can cause serious mortality. In E. coli, an inner membrane protein YgiM encoded by gene ygiM can target the eukaryotic peroxisome. Peroxisome is a membrane-enclosed organelle associated with the ROS metabolism and was reported to play the key role in immune responses and inflammation during the development of sepsis. Klebsiella pneumoniae (K. pneumoniae) is one of the important pathogens causing sepsis. However, the function of gene vk055_4013 which is highly homologous to ygiM of E. coli has not been demonstrated in K. pneumoniae. In this study, we prepared ΔygiM of K. pneumoniae ATCC43816, and found that the deletion of ygiM did not affect bacterial growth and mouse mortality in the mouse infection model. Interestingly, ΔygiM not only resulted in reduced bacterial resistance to macrophages, but also attenuated pathological manifestations in mouse organs. Furthermore, based on the data of Gene Expression Omnibus, the expression profiles of micro RNAs (miRNAs) and messenger RNAs (mRNAs) in the serum of 44 sepsis patients caused by K. pneumoniae infection were analyzed, and 11 differently expressed miRNAs and 8 DEmRNAs associated with the membrane function were found. Finally, the membrane-associated competing endogenous RNAs (ceRNAs) network was constructed. In this ceRNAs network, DEmiRNAs (hsa-miR-7108-5p, hsa-miR-6780a-5p, hsa-miR-6756-5p, hsa-miR-4433b-3p, hsa-miR-3652, hsa-miR-342-3p, hsa-miR-32-5p) and their potential downstream target DEmRNAs (VNN1, CEACAM8, PGLYRP1) were verified in the cell model infected by wild type and ΔygiM of K. pneumoniae, respectively. Taken together, YgiM may trigger the sepsis caused by K. pneumoniae via membrane-associated ceRNAs. This study provided new insights into the role of YgiM in the process of K. pneumoniae induced sepsis.
Collapse
Affiliation(s)
- Mingxiao Han
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhihao Chen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Ping He
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Clinical Laboratory, Sichuan Province Science City Hospital, Chengdu, China
| | - Ziyuan Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zelei Tong
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hong Du
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
19
|
Activating transcription factor 3 protects alveolar epithelial type II cells from Mycobacterium tuberculosis infection-induced inflammation. Tuberculosis (Edinb) 2022; 135:102227. [DOI: 10.1016/j.tube.2022.102227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/07/2022] [Accepted: 06/19/2022] [Indexed: 11/22/2022]
|
20
|
Bai W, Huo S, Li J, Shao J. Advances in the Study of the Ubiquitin-Editing Enzyme A20. Front Pharmacol 2022; 13:845262. [PMID: 35592427 PMCID: PMC9110840 DOI: 10.3389/fphar.2022.845262] [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: 12/29/2021] [Accepted: 02/28/2022] [Indexed: 11/30/2022] Open
Abstract
Ubiquitin modification is a common post-translational protein modification and an important mechanism whereby the body regulates protein levels and functions. As a common enzyme associated with ubiquitin modification, the ubiquitin-editing enzyme A20 may be closely associated with the development of numerous pathological processes through its different structural domains. The aim of this paper is to provide an overview of the following: advances in ubiquitination research, the structure and function of A20, and the relationships between A20 and immune inflammatory response, apoptosis, necroptosis, pyroptosis, and autophagy.
Collapse
Affiliation(s)
- Wenya Bai
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Siying Huo
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Junjie Li
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jianlin Shao
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
21
|
Identification of Unique Key miRNAs, TFs, and mRNAs in Virulent MTB Infection Macrophages by Network Analysis. Int J Mol Sci 2021; 23:ijms23010382. [PMID: 35008808 PMCID: PMC8745702 DOI: 10.3390/ijms23010382] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/25/2021] [Accepted: 12/27/2021] [Indexed: 12/13/2022] Open
Abstract
Although Mycobacterium tuberculosis (MTB) has existed for thousands of years, its immune escape mechanism remains obscure. Increasing evidence signifies that microRNAs (miRNAs) play pivotal roles in the progression of tuberculosis (TB). RNA sequencing was used to sequence miRNAs in human acute monocytic leukemia cells (THP-1) infected by the virulent MTB-1458 strain and the avirulent vaccine strain Mycobacterium bovis Bacillus Calmette-Guérin (BCG). Sets of differentially expressed miRNAs (DE-miRNAs) between MTB-1458/BCG-infected groups and uninfected groups were identified, among which 18 were differentially expressed only in the MTB-1458-infected THP-1 group. Then, 13 transcription factors (TFs) and 81 target genes of these 18 DE-miRNAs were matched. Gene Ontology classification as well as Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis showed that the candidate targets were predominantly involved in apoptotic-associated and interferon-γ-mediated signaling pathways. A TF-miRNA-mRNA interaction network was constructed to analyze the relationships among these 18 DE-miRNAs and their targets and TFs, as well as display the hub miRNAs, TFs, and target genes. Considering the degrees from network analysis and the reported functions, this study focused on the BHLHE40-miR-378d-BHLHE40 regulation axis and confirmed that BHLHE40 was a target of miR-378d. This cross-talk among DE-miRNAs, mRNAs, and TFs might be an important feature in TB, and the findings merited further study and provided new insights into immune defense and evasion underlying host-pathogen interactions.
Collapse
|