1
|
Munke K, Wulff L, Lienard J, Carlsson F, Agace WW. In vivo regulation of the monocyte phenotype by Mycobacterium marinum and the ESX-1 type VII secretion system. Sci Rep 2025; 15:4545. [PMID: 39915532 PMCID: PMC11802795 DOI: 10.1038/s41598-025-88212-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/25/2025] [Indexed: 02/09/2025] Open
Abstract
Pathogenic mycobacteria require the conserved ESX-1 type VII secretion system to cause disease. In a murine Mycobacterium marinum infection model we previously demonstrated that infiltrating monocytes and neutrophils represent the major bacteria-harbouring cell populations in infected tissue. In the current study we use this model, in combination with scRNA sequencing, to assess the impact of M. marinum infection on the transcriptional profile of infiltrating Ly6C⁺MHCII⁺ monocytes in vivo. Our findings demonstrate that infection of infiltrating monocytes with M. marinum alters their cytokine expression profile, induces glycolytic metabolism, hypoxia-mediated signaling, nitric oxide synthesis, tissue remodeling, and suppresses responsiveness to IFNγ. We further show that the transcriptional response of bystander monocytes is influenced by ESX-1-dependent mechanisms, including a reduced responsiveness to IFNγ. These findings suggest that mycobacterial infection has pleiotropic effects on monocyte phenotype, with potential implications in bacterial growth restriction and granuloma formation.
Collapse
Affiliation(s)
- Kristina Munke
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Line Wulff
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Julia Lienard
- Department of Biology, Lund University, Lund, Sweden
| | | | - William W Agace
- Department of Experimental Medical Science, Lund University, Lund, Sweden.
- Department of Immunology and Microbiology, LEO Foundation Skin Immunology Research Centre, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
2
|
Wang MS, Li-Hunnam J, Chen YL, Gilmour B, Alene KA, Zhang YA, Nicol MP. Conversion or Reversion of Interferon γ Release Assays for Mycobacterium tuberculosis Infection: A Systematic Review and Meta-analysis. Clin Infect Dis 2025; 80:168-179. [PMID: 38954503 DOI: 10.1093/cid/ciae357] [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: 02/29/2024] [Revised: 06/05/2024] [Accepted: 06/28/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Interferon γ release assays (IGRAs) are widely used for diagnosis of latent tuberculosis infection. However, with repeated testing, IGRA transformation (conversion or reversion) may be detected and is challenging to interpret. We reviewed the frequency of and risk factors for IGRA transformation. METHODS We screened public databases for studies of human participants that reported the frequency of IGRA transformation. We extracted study and participant characteristics, details of IGRA testing and results. We calculated the pooled frequency of IGRA transformation (and transient transformation) and examined associated risk factors. RESULTS The pooled frequency of IGRA conversion or reversion from 244 studies was estimated at 7.3% (95% confidence interval [CI], 6.1%-8.5%) or 22.8% (20.1%-25.7%), respectively. Transient conversion or reversion were estimated at 46.0% (95% CI, 35.7%-56.4%) or 19.6% (9.2%-31.7%) of conversion or reversion events respectively. Indeterminate results seldom reverted to positive (1.2% [95% CI, .1%-3.5%]). IGRA results in the borderline-positive or borderline-negative range were associated with increased risk of conversion or reversion (pooled odds ratio [OR] for conversion, 4.15 [95% CI, 3.00-5.30]; pooled OR for reversion, 4.06 [3.07-5.06]). BCG vaccination was associated with decreased risk of conversion (OR, 0.70 [95% CI, .56-.84]), cigarette smoking with decreased risk of reversion (0.44 [.06-.82]), and female sex with decreased risk of either conversion or reversion (OR for conversion, 0.66 [.58-.75]; OR for reversion, 0.46 [.31-.61]). CONCLUSIONS IGRA conversion is less common than reversion, and frequently transient. Research is needed to determine whether individuals with reversion would benefit from tuberculosis-preventive treatment. Retesting of people with indeterminate results is probably not indicated, because indeterminate results seldom revert to positive.
Collapse
Affiliation(s)
- Mao-Shui Wang
- Department of Lab Medicine, Shandong Public Health Clinical Center, Shandong University, Jinan, China
- Shandong Key Laboratory of Infectious Respiratory Disease, Jinan, China
| | - Jarrod Li-Hunnam
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Ya-Li Chen
- Department of Lab Medicine, Shandong Public Health Clinical Center, Shandong University, Jinan, China
- Shandong Key Laboratory of Infectious Respiratory Disease, Jinan, China
| | - Beth Gilmour
- Faculty of Health Sciences, School of Population Health, Curtin University, Perth, WA, Australia
- Geospatial and Tuberculosis Research Team, Telethon Kids Institute, Perth, WA, Australia
| | - Kefyalew Addis Alene
- Faculty of Health Sciences, School of Population Health, Curtin University, Perth, WA, Australia
- Geospatial and Tuberculosis Research Team, Telethon Kids Institute, Perth, WA, Australia
| | - Yan-An Zhang
- Shandong Key Laboratory of Infectious Respiratory Disease, Jinan, China
- Department of Cardiovascular Surgery, Shandong Public Health Clinical Center, Shandong University, Jinan, China
| | - Mark P Nicol
- Marshall Centre, Division of Infection and Immunity, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
3
|
Vance RE. Tuberculosis as an unconventional interferonopathy. Curr Opin Immunol 2025; 92:102508. [PMID: 39637776 DOI: 10.1016/j.coi.2024.102508] [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: 05/12/2024] [Revised: 10/31/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024]
Abstract
Tuberculosis is caused by Mycobacterium tuberculosis, a bacterium that accounts for more human mortality than any other. Evidence is accumulating for the view that tuberculosis is an interferonopathy - a disease driven by type I interferons. However, how type I interferons exacerbate tuberculosis remains poorly understood. As an infection, tuberculosis is distinct from conventional interferonopathies, which are autoinflammatory diseases. Here I consider the hypothesis that type I interferons promote bacterial replication by impairing key antibacterial immune responses, including those orchestrated by interleukin-1 and interferon γ. Paradoxically, during tuberculosis, the underlying state of impaired antibacterial immunity co-exists with overt (but ineffective) inflammation. Conceiving of tuberculosis as an unconventional interferonopathy may suggest fruitful avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Russell E Vance
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, CA USA.
| |
Collapse
|
4
|
Silva RCMC. The dichotomic role of cytokines in aging. Biogerontology 2024; 26:17. [PMID: 39621124 DOI: 10.1007/s10522-024-10152-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/30/2024] [Indexed: 12/11/2024]
Abstract
The chronic inflammation present in aged individuals is generally depicted as a detrimental player for longevity. Here, it is discussed several beneficial effects associated with the cytokines that are chronically elevated in inflammaging. These cytokines, such as IL-1β, type I interferons, IL-6 and TNF positively regulate macroautophagy, mitochondrial function, anti-tumor immune responses and skeletal muscle biogenesis, possibly contributing to longevity. On the other side, the detrimental and antagonistic role of these cytokines including the induction of sarcopenia, tissue damage and promotion of tumorigenesis are also discussed, underscoring the dichotomy associated with inflammaging and its players. In addition, it is discussed the role of the anti-inflammatory cytokine IL-10 and other cytokines that affect aging in a more linear way, such as IL-11, which promotes senescence, and IL-4 and IL-15, which promotes longevity. It is also discussed more specific regulators of aging that are downstream cytokines-mediated signaling.
Collapse
|
5
|
Han X, Wang X, Han F, Yan H, Sun J, Zhang X, Moog C, Zhang C, Su B. The cGAS-STING pathway in HIV-1 and Mycobacterium tuberculosis coinfection. Infection 2024:10.1007/s15010-024-02429-0. [PMID: 39509013 DOI: 10.1007/s15010-024-02429-0] [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: 04/23/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
Mycobacterium tuberculosis (M. tuberculosis) infection is the most common opportunistic infection in human immunodeficiency virus-1 (HIV-1)-infected individuals, and the mutual reinforcement of these two pathogens may accelerate disease progression and lead to rapid mortality. Therefore, HIV-1/M. tuberculosis coinfection is one of the major global public health concerns. HIV-1 infection is the greatest risk factor for M. tuberculosis infection and increases the likelihood of endogenous relapse and exogenous reinfection with M. tuberculosis. Moreover, M. tuberculosis further increases HIV-1 replication and the occurrence of chronic immune activation, accelerating the progression of HIV-1 disease. Exploring the pathogenesis of HIV-1/M. tuberculosis coinfections is essential for the development of novel treatments to reduce the global burden of tuberculosis. Innate immunity, which is the first line of host immune defense, plays a critical role in resisting HIV-1 and M. tuberculosis infections. The role of the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway, which is a major DNA-sensing innate immune signaling pathway, in HIV-1 infection and M. tuberculosis infection has been intensively studied. This paper reviews the role of the cGAS-STING signaling pathway in HIV-1 infection and M. tuberculosis infection and discusses the possible role of this pathway in HIV-1/M. tuberculosis coinfection to provide new insight into the pathogenesis of HIV-1/M. tuberculosis coinfection and the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoxu Han
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Xiuwen Wang
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Fangping Han
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Hongxia Yan
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Jin Sun
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Xin Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Christiane Moog
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Laboratoire d'ImmunoRhumatologie Moléculaire, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1109, Institut Thématique Interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Fédération Hospitalo-Universitaire OMICARE, Université de Strasbourg, Transplantex, Strasbourg, NG, 67000, France
- Vaccine Research Institute (VRI), Créteil, 94000, France
| | - Conggang Zhang
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China.
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
6
|
Darboe F, Reijneveld JF, Maison DP, Martinez L, Suliman S. Unmasking the hidden impact of viruses on tuberculosis risk. Trends Immunol 2024; 45:649-661. [PMID: 39181733 PMCID: PMC11769684 DOI: 10.1016/j.it.2024.07.008] [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: 06/28/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024]
Abstract
Tuberculosis (TB) is a leading cause of mortality from an infectious disease. In this opinion article, we focus on accumulating scientific evidence indicating that viral infections may contribute to TB progression, possibly allowing novel preventive interventions. Viruses can remodel the mammalian immune system, potentially modulating the risk of reactivating latent microbes such as Mycobacterium tuberculosis (Mtb). Evidence is mixed regarding the impact of emergent viruses such as SARS-CoV-2 on the risk of TB. Therefore, we posit that important knowledge gaps include elucidating which viral families increase TB risk and whether these provide unique or shared immune mechanisms. We also propose potential future research to define the contribution of viruses to TB pathogenesis.
Collapse
Affiliation(s)
- Fatoumatta Darboe
- Zuckerberg San Francisco General Hospital, Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Josephine F Reijneveld
- Zuckerberg San Francisco General Hospital, Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - David P Maison
- Zuckerberg San Francisco General Hospital, Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Leonardo Martinez
- Boston University School of Public Health, Department of Epidemiology, Boston, MA, USA.
| | - Sara Suliman
- Zuckerberg San Francisco General Hospital, Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
7
|
Yadav S, El Hamra R, Alturki NA, Ariana A, Bhan A, Hurley K, Gaestel M, Blackshear PJ, Blais A, Sad S. Regulation of Zfp36 by ISGF3 and MK2 restricts the expression of inflammatory cytokines during necroptosis stimulation. Cell Death Dis 2024; 15:574. [PMID: 39117638 PMCID: PMC11310327 DOI: 10.1038/s41419-024-06964-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024]
Abstract
Necrosome activation following TLR- or cytokine receptor-signaling results in cell death by necroptosis which is characterized by the rupture of cell membranes and the consequent release of intracellular contents to the extracellular milieu. While necroptosis exacerbates various inflammatory diseases, the mechanisms through which the inflammatory responses are regulated are not clear. We show that the necrosome activation of macrophages results in an upregulation of various pathways, including the mitogen-activated protein kinase (MAPK) cascade, which results in an elevation of the inflammatory response and consequent expression of several cytokines and chemokines. Programming for this upregulation of inflammatory response occurs during the early phase of necrosome activation and proceeds independently of cell death but depends on the activation of the receptor-interacting protein kinase-1 (RipK1). Interestingly, necrosome activation also results in an upregulation of IFNβ, which in turn exerts an inhibitory effect on the maintenance of inflammatory response through the repression of MAPK-signaling and an upregulation of Zfp36. Activation of the interferon-induced gene factor-3 (ISGF3) results in the expression of ZFP36 (TTP), which induces the post-transcriptional degradation of mRNAs of various inflammatory cytokines and chemokines through the recognition of AU-rich elements in their 3'UTR. Furthermore, ZFP-36 inhibits IFNβ-, but not TNFα- induced necroptosis. Overall, these results reveal the molecular mechanism through which IFNβ, a pro-inflammatory cytokine, induces the expression of ZFP-36, which in turn inhibits necroptosis and halts the maintenance of the inflammatory response.
Collapse
Affiliation(s)
- Sahil Yadav
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Rayan El Hamra
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Norah A Alturki
- Clinical Laboratory Science Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ardeshir Ariana
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Avni Bhan
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Kate Hurley
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Matthias Gaestel
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Perry J Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina, United States of America
| | - Alexandre Blais
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Ottawa Institute of Systems Biology, Ottawa, ON, Canada
- University of Ottawa, Centre for Infection Immunity and Inflammation, Ottawa, ON, Canada
- University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Subash Sad
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
- University of Ottawa, Centre for Infection Immunity and Inflammation, Ottawa, ON, Canada.
| |
Collapse
|
8
|
Anes E, Azevedo-Pereira JM, Pires D. Role of Type I Interferons during Mycobacterium tuberculosis and HIV Infections. Biomolecules 2024; 14:848. [PMID: 39062562 PMCID: PMC11275242 DOI: 10.3390/biom14070848] [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: 06/18/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Tuberculosis and AIDS remain two of the most relevant human infectious diseases. The pathogens that cause them, Mycobacterium tuberculosis (Mtb) and HIV, individually elicit an immune response that treads the line between beneficial and detrimental to the host. Co-infection further complexifies this response since the different cytokines acting on one infection might facilitate the dissemination of the other. In these responses, the role of type I interferons is often associated with antiviral mechanisms, while for bacteria such as Mtb, their importance and clinical relevance as a suitable target for manipulation are more controversial. In this article, we review the recent knowledge on how these interferons play distinct roles and sometimes have opposite consequences depending on the stage of the pathogenesis. We highlight the dichotomy between the acute and chronic infections displayed by both infections and how type I interferons contribute to an initial control of each infection individually, while their chronic induction, particularly during HIV infection, might facilitate Mtb primo-infection and progression to disease. We expect that further findings and their systematization will allow the definition of windows of opportunity for interferon manipulation according to the stage of infection, contributing to pathogen clearance and control of immunopathology.
Collapse
Affiliation(s)
- Elsa Anes
- Host-Pathogen Interactions Unit, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.M.A.-P.); (D.P.)
| | - José Miguel Azevedo-Pereira
- Host-Pathogen Interactions Unit, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.M.A.-P.); (D.P.)
| | - David Pires
- Host-Pathogen Interactions Unit, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.M.A.-P.); (D.P.)
- Center for Interdisciplinary Research in Health, Católica Medical School, Universidade Católica Portuguesa, Estrada Octávio Pato, 2635-631 Rio de Mouro, Portugal
| |
Collapse
|
9
|
Lee J, Seo H, Kim D, Hwang J, Kwon JW. Influenza and the risk of active tuberculosis occurrence among individuals with latent tuberculosis infection: A national cohort study in South Korea (2015-2020). JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2024; 57:437-445. [PMID: 38627123 DOI: 10.1016/j.jmii.2024.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/15/2024] [Accepted: 04/02/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Influenza's potential impact on active tuberculosis (TB) development has been debated, with limited clinical evidence. To address this, we explored the association between influenza episodes and TB incidence in a national cohort of individuals with latent TB infection (LTBI). METHODS We examined adults (≥20 years) diagnosed with LTBI between 2015 and 2020, using the Health Insurance Review and Assessment Service's national database in South Korea. We collected demographic data, comorbidities, and influenza episodes within 6 months before and after the initial LTBI diagnosis (prior vs. subsequent episode). We stratified the analysis into groups with and without TB preventive therapy (TPT). RESULTS Among 220,483 LTBI subjects, 49% received TPT, while 51% did not. The average age was 48.4 years, with 52% having comorbidities. A prior and subsequent influenza episode was identified in 3221 and 4580 individuals, respectively. Of these, 1159 (0.53%) developed incident TB over an average follow-up of 1.86 years. The incidence rates of TB were comparable between individuals with and without prior and/or subsequent influenza episodes in the TPT group, but 1.4 times higher in the non-TPT group for those with such episodes. Cox proportional-hazards regression analysis indicated that influenza was not a risk factor for incident TB in the TPT group. However, a subsequent influenza episode significantly increased TB risk in the non-TPT group (hazard ratio: 1.648 [95% CI, 1.053-2.580]). CONCLUSIONS In individuals with LTBI not receiving TPT, experiencing an influenza episode may elevate the risk of developing active TB.
Collapse
Affiliation(s)
- Jaehee Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
| | - Hyewon Seo
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
| | - Dohyang Kim
- Department of Statistics, Daegu University, Gyeongbuk, Republic of Korea.
| | - Jinseub Hwang
- Department of Statistics, Daegu University, Gyeongbuk, Republic of Korea.
| | - Jin-Won Kwon
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
10
|
Silva RCMC, Travassos LH, Dutra FF. The dichotomic role of single cytokines: Fine-tuning immune responses. Cytokine 2024; 173:156408. [PMID: 37925788 DOI: 10.1016/j.cyto.2023.156408] [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: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/07/2023]
Abstract
Cytokines are known for their pleiotropic effects. They can be classified by their function as pro-inflammatory, such as tumor necrosis factor (TNF), interleukin (IL) 1 and IL-12, or anti-inflammatory, like IL-10, IL-35 and transforming growth factor β (TGF-β). Though this type of classification is an important simplification for the understanding of the general cytokine's role, it can be misleading. Here, we discuss recent studies that show a dichotomic role of the so-called pro and anti-inflammatory cytokines, highlighting that their function can be dependent on the microenvironment and their concentrations. Furthermore, we discuss how the back-and-forth interplay between cytokines and immunometabolism can influence the dichotomic role of inflammatory responses as an important target to complement cytokine-based therapies.
Collapse
Affiliation(s)
| | - Leonardo Holanda Travassos
- Laboratório de Receptores e Sinalização intracelular, Instituto de Biofísica Carlos Chagas Filho, UFRJ, Rio de Janeiro, Brazil
| | - Fabianno Ferreira Dutra
- Laboratório de Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, UFRJ, Rio de Janeiro, Brazil
| |
Collapse
|
11
|
Wang R, Fan X, Xu D, Li M, Zhao X, Cao B, Qian C, Yu J, Fang D, Gu Y, Wan K, Liu H. Comparison of the Immunogenicity and Efficacy of rBCG-EPCP009, BCG Prime-EPCP009 Booster, and EPCP009 Protein Regimens as Tuberculosis Vaccine Candidates. Vaccines (Basel) 2023; 11:1738. [PMID: 38140143 PMCID: PMC10747267 DOI: 10.3390/vaccines11121738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/04/2023] [Accepted: 11/15/2023] [Indexed: 12/24/2023] Open
Abstract
Bacillus Calmette-Guérin (BCG) is the only widely used prophylactic tuberculosis (TB) vaccine that can prevent severe TB in infants. However, it provides poor protection in adults, and therefore, there is ongoing research into new TB vaccines and immunization strategies with more durable immune effects. The recombinant BCG and BCG prime-protein booster are two important vaccine strategies that have recently been developed based on BCG and could improve immune responses. In this study, three immune strategies based on four protective antigens, namely, ESAT-6, CFP-10, nPPE18, and nPstS1, were applied to construct recombinant rBCG-EPCP009, EPCP009 subunit protein, and BCG prime-EPCP009 booster vaccine candidates. The short- and long-term immune effects after vaccination in Balb/c mice were evaluated based on humoral immunity, cellular immunity, and the ability of spleen cells to inhibit in vitro mycobacterial growth. At 8 and 12 weeks after the initial immunization, splenocytes from mice inoculated with the BCG prime-EPCP009 protein booster secreted higher levels of PPD- and EPCP009-specific IFN-γ, IL-2, TNF-α, IL-17, GM-CSF, and IL-12 and had a higher IFN-γ+CD4+ TEM:IL-2+CD8+ TCM cell ratio than splenocytes from mice inoculated with the rBCG-EPCP009 and EPCP009 proteins. In addition, the EPCPE009-specific IgG2a/IgG1 ratio was slightly higher in the BCG prime-EPCP009 protein booster group than in the other two groups. The in vitro mycobacterial inhibition assay showed that the splenocytes of mice from the BCG prime-EPCP009 protein booster group exhibited stronger inhibition of Mycobacterium tuberculosis (M. tuberculosis) growth than the splenocytes of mice from the other two groups. These results indicate that the BCG prime-EPCP009 protein booster exhibited superior immunogenicity and M. tuberculosis growth inhibition to the parental BCG, rBCG-EPCP009, and EPCP009 proteins under in vitro conditions. Thus, the BCG prime-EPCP009 protein booster may be important for the development of a more effective adult TB vaccine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Kanglin Wan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (R.W.); (X.F.); (D.X.); (M.L.); (X.Z.); (B.C.); (C.Q.); (J.Y.); (D.F.); (Y.G.)
| | - Haican Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (R.W.); (X.F.); (D.X.); (M.L.); (X.Z.); (B.C.); (C.Q.); (J.Y.); (D.F.); (Y.G.)
| |
Collapse
|
12
|
Wang Y, De Labastida Rivera F, Edwards CL, Frame TC, Engel JA, Bukali L, Na J, Ng SS, Corvino D, Montes de Oca M, Bunn PT, Soon MS, Andrew D, Loughland JR, Zhang J, Amante FH, Barber BE, McCarthy JS, Lopez JA, Boyle MJ, Engwerda CR. STING activation promotes autologous type I interferon-dependent development of type 1 regulatory T cells during malaria. J Clin Invest 2023; 133:e169417. [PMID: 37781920 PMCID: PMC10541195 DOI: 10.1172/jci169417] [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: 02/03/2023] [Accepted: 08/08/2023] [Indexed: 10/03/2023] Open
Abstract
The development of highly effective malaria vaccines and improvement of drug-treatment protocols to boost antiparasitic immunity are critical for malaria elimination. However, the rapid establishment of parasite-specific immune regulatory networks following exposure to malaria parasites hampers these efforts. Here, we identified stimulator of interferon genes (STING) as a critical mediator of type I interferon production by CD4+ T cells during blood-stage Plasmodium falciparum infection. The activation of STING in CD4+ T cells by cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) stimulated IFNB gene transcription, which promoted development of IL-10- and IFN-γ-coproducing CD4+ T (type I regulatory [Tr1]) cells. The critical role for type I IFN signaling for Tr1 cell development was confirmed in vivo using a preclinical malaria model. CD4+ T cell sensitivity to STING phosphorylation was increased in healthy volunteers following P. falciparum infection, particularly in Tr1 cells. These findings identified STING expressed by CD4+ T cells as an important mediator of type I IFN production and Tr1 cell development and activation during malaria.
Collapse
Affiliation(s)
- Yulin Wang
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Griffith University, School of Environment and Science, Nathan, Australia
| | | | - Chelsea L. Edwards
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- University of Queensland, School of Medicine, Brisbane, Australia
| | - Teija C.M. Frame
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- University of Queensland, School of Medicine, Brisbane, Australia
| | | | - Luzia Bukali
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- University of Queensland, School of Medicine, Brisbane, Australia
| | - Jinrui Na
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- University of Queensland, School of Medicine, Brisbane, Australia
| | - Susanna S. Ng
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Dillon Corvino
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Marcela Montes de Oca
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Patrick T. Bunn
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Megan S.F. Soon
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Dean Andrew
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - Jia Zhang
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Fiona H. Amante
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - James S. McCarthy
- Victorian Infectious Diseases Services, Doherty Institute, University of Melbourne, Melbourne, Australia
| | - J. Alejandro Lopez
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Griffith University, School of Environment and Science, Nathan, Australia
| | - Michelle J. Boyle
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Life Sciences Division, Burnet Institute, Melbourne, Australia
| | | |
Collapse
|
13
|
Shimoda M, Inagaki T, Davis RR, Merleev A, Tepper CG, Maverakis E, Izumiya Y. Virally encoded interleukin-6 facilitates KSHV replication in monocytes and induction of dysfunctional macrophages. PLoS Pathog 2023; 19:e1011703. [PMID: 37883374 PMCID: PMC10602306 DOI: 10.1371/journal.ppat.1011703] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic double-stranded DNA virus and the etiologic agent of Kaposi's sarcoma and hyperinflammatory lymphoproliferative disorders. Understanding the mechanism by which KSHV increases the infected cell population is crucial for curing KSHV-associated diseases. Using scRNA-seq, we demonstrate that KSHV preferentially infects CD14+ monocytes, sustains viral lytic replication through the viral interleukin-6 (vIL-6), which activates STAT1 and 3, and induces an inflammatory gene expression program. To study the role of vIL-6 in monocytes upon KSHV infection, we generated recombinant KSHV with premature stop codon (vIL-6(-)) and its revertant viruses (vIL-6(+)). Infection of the recombinant viruses shows that both vIL-6(+) and vIL-6(-) KSHV infection induced indistinguishable host anti-viral response with STAT1 and 3 activations in monocytes; however, vIL-6(+), but not vIL-6(-), KSHV infection promoted the proliferation and differentiation of KSHV-infected monocytes into macrophages. The macrophages derived from vIL-6(+) KSHV infection showed a distinct transcriptional profile of elevated IFN-pathway activation with immune suppression and were compromised in T-cell stimulation function compared to those from vIL-6(-) KSHV infection or uninfected control. Notably, a viral nuclear long noncoding RNA (PAN RNA), which is required for sustaining KSHV gene expression, was substantially reduced in infected primary monocytes upon vIL-6(-) KSHV infection. These results highlight the critical role of vIL-6 in sustaining KSHV transcription in primary monocytes. Our findings also imply a clever strategy in which KSHV utilizes vIL-6 to secure its viral pool by expanding infected monocytes via differentiating into longer-lived dysfunctional macrophages. This mechanism may facilitate KSHV to escape from host immune surveillance and to support a lifelong infection.
Collapse
Affiliation(s)
- Michiko Shimoda
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, California, United States of America
- UC Davis Comprehensive Cancer Center, Sacramento, California, United States of America
| | - Tomoki Inagaki
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, California, United States of America
| | - Ryan R. Davis
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, California, United States of America
| | - Alexander Merleev
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, California, United States of America
| | - Clifford G. Tepper
- UC Davis Comprehensive Cancer Center, Sacramento, California, United States of America
- Department of Biochemistry and Molecular Medicine, School of Medicine, UC Davis, Sacramento, California, United States of America
| | - Emanual Maverakis
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, California, United States of America
- UC Davis Comprehensive Cancer Center, Sacramento, California, United States of America
| | - Yoshihiro Izumiya
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, California, United States of America
- UC Davis Comprehensive Cancer Center, Sacramento, California, United States of America
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, California, United States of America
| |
Collapse
|
14
|
Bloom BR. A half-century of research on tuberculosis: Successes and challenges. J Exp Med 2023; 220:e20230859. [PMID: 37552470 PMCID: PMC10407785 DOI: 10.1084/jem.20230859] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/09/2023] Open
Abstract
Great progress has been made over the past half-century, but TB remains a formidable global health problem, particularly in low- and middle-income countries. Understanding the mechanisms of pathogenesis and necessary and sufficient conditions for protection are critical. The need for inexpensive and sensitive point-of-care diagnostic tests for earlier detection of infection and disease, shorter and less-toxic drug regimens for drug-sensitive and -resistant TB, and a more effective vaccine than BCG is immense. New and better tools, greater support for international research, collaborations, and training will be required to dramatically reduce the burden of this devastating disease which still kills 1.6 million people annually.
Collapse
Affiliation(s)
- Barry R. Bloom
- Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
15
|
Trono P, Tocci A, Palermo B, Di Carlo A, D'Ambrosio L, D'Andrea D, Di Modugno F, De Nicola F, Goeman F, Corleone G, Warren S, Paolini F, Panetta M, Sperduti I, Baldari S, Visca P, Carpano S, Cappuzzo F, Russo V, Tripodo C, Zucali P, Gregorc V, Marchesi F, Nistico P. hMENA isoforms regulate cancer intrinsic type I IFN signaling and extrinsic mechanisms of resistance to immune checkpoint blockade in NSCLC. J Immunother Cancer 2023; 11:e006913. [PMID: 37612043 PMCID: PMC10450042 DOI: 10.1136/jitc-2023-006913] [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] [Accepted: 08/08/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Understanding how cancer signaling pathways promote an immunosuppressive program which sustains acquired or primary resistance to immune checkpoint blockade (ICB) is a crucial step in improving immunotherapy efficacy. Among the pathways that can affect ICB response is the interferon (IFN) pathway that may be both detrimental and beneficial. The immune sensor retinoic acid-inducible gene I (RIG-I) induces IFN activation and secretion and is activated by actin cytoskeleton disturbance. The actin cytoskeleton regulatory protein hMENA, along with its isoforms, is a key signaling hub in different solid tumors, and recently its role as a regulator of transcription of genes encoding immunomodulatory secretory proteins has been proposed. When hMENA is expressed in tumor cells with low levels of the epithelial specific hMENA11a isoform, identifies non-small cell lung cancer (NSCLC) patients with poor prognosis. Aim was to identify cancer intrinsic and extrinsic pathways regulated by hMENA11a downregulation as determinants of ICB response in NSCLC. Here, we present a potential novel mechanism of ICB resistance driven by hMENA11a downregulation. METHODS Effects of hMENA11a downregulation were tested by RNA-Seq, ATAC-Seq, flow cytometry and biochemical assays. ICB-treated patient tumor tissues were profiled by Nanostring IO 360 Panel enriched with hMENA custom probes. OAK and POPLAR datasets were used to validate our discovery cohort. RESULTS Transcriptomic and biochemical analyses demonstrated that the depletion of hMENA11a induces IFN pathway activation, the production of different inflammatory mediators including IFNβ via RIG-I, sustains the increase of tumor PD-L1 levels and activates a paracrine loop between tumor cells and a unique macrophage subset favoring an epithelial-mesenchymal transition (EMT). Notably, when we translated our results in a clinical setting of NSCLC ICB-treated patients, transcriptomic analysis revealed that low expression of hMENA11a, high expression of IFN target genes and high macrophage score identify patients resistant to ICB therapy. CONCLUSIONS Collectively, these data establish a new function for the actin cytoskeleton regulator hMENA11a in modulating cancer cell intrinsic type I IFN signaling and extrinsic mechanisms that promote protumoral macrophages and favor EMT. These data highlight the role of actin cytoskeleton disturbance in activating immune suppressive pathways that may be involved in resistance to ICB in NSCLC.
Collapse
Affiliation(s)
- Paola Trono
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Institute of Biochemistry and Cell Biology, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Annalisa Tocci
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Belinda Palermo
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Anna Di Carlo
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Lorenzo D'Ambrosio
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Daniel D'Andrea
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Francesca Di Modugno
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Frauke Goeman
- SAFU Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giacomo Corleone
- SAFU Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Sarah Warren
- NanoString Technologies Inc, Seattle, Washington, USA
| | - Francesca Paolini
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Mariangela Panetta
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Isabella Sperduti
- Biostatistics Unit, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Baldari
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Paolo Visca
- Pathology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Carpano
- Second Division of Medical Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Federico Cappuzzo
- Second Division of Medical Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Vincenzo Russo
- Department of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Claudio Tripodo
- Department of Health Sciences, Human Pathology Section, Tumor Immunology Unit, University of Palermo, Palermo, Italy
| | - Paolo Zucali
- Department of Oncology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Vanesa Gregorc
- Department of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Federica Marchesi
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Paola Nistico
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
16
|
Thirunavukkarasu S, Ahmed M, Rosa BA, Boothby M, Cho SH, Rangel-Moreno J, Mbandi SK, Schreiber V, Gupta A, Zuniga J, Mitreva M, Kaushal D, Scriba TJ, Khader SA. Poly(ADP-ribose) polymerase 9 mediates early protection against Mycobacterium tuberculosis infection by regulating type I IFN production. J Clin Invest 2023; 133:e158630. [PMID: 37200107 PMCID: PMC10266794 DOI: 10.1172/jci158630] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/19/2023] [Indexed: 05/20/2023] Open
Abstract
The ADP ribosyltransferases (PARPs 1-17) regulate diverse cellular processes, including DNA damage repair. PARPs are classified on the basis of their ability to catalyze poly-ADP-ribosylation (PARylation) or mono-ADP-ribosylation (MARylation). Although PARP9 mRNA expression is significantly increased in progressive tuberculosis (TB) in humans, its participation in host immunity to TB is unknown. Here, we show that PARP9 mRNA encoding the MARylating PARP9 enzyme was upregulated during TB in humans and mice and provide evidence of a critical modulatory role for PARP9 in DNA damage, cyclic GMP-AMP synthase (cGAS) expression, and type I IFN production during TB. Thus, Parp9-deficient mice were susceptible to Mycobacterium tuberculosis infection and exhibited increased TB disease, cGAS and 2'3'-cyclic GMP-AMP (cGAMP) expression, and type I IFN production, along with upregulation of complement and coagulation pathways. Enhanced M. tuberculosis susceptibility is type I IFN dependent, as blockade of IFN α receptor (IFNAR) signaling reversed the enhanced susceptibility of Parp9-/- mice. Thus, in sharp contrast to PARP9 enhancement of type I IFN production in viral infections, this member of the MAR family plays a protective role by limiting type I IFN responses during TB.
Collapse
Affiliation(s)
| | | | - Bruce A. Rosa
- McDonnell Genome Institute, and
- Division of Infectious Diseases, Department of Internal Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Mark Boothby
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sung Hoon Cho
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Javier Rangel-Moreno
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Stanley K. Mbandi
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Valérie Schreiber
- Biotechnology and Cell Signaling, CNRS UMR7242, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, INSERM U1258, Université de Strasbourg, Illkirch, France
| | | | - Joaquin Zuniga
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Makedonka Mitreva
- McDonnell Genome Institute, and
- Division of Infectious Diseases, Department of Internal Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | | |
Collapse
|
17
|
Shimoda M, Inagaki T, Davis RR, Merleev A, Tepper CG, Maverakis E, Izumiya Y. KSHV uses viral IL6 to expand infected immunosuppressive macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.05.531224. [PMID: 36945595 PMCID: PMC10028810 DOI: 10.1101/2023.03.05.531224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic double-stranded DNA virus and the etiologic agent of Kaposi's sarcoma and hyperinflammatory lymphoproliferative disorders. Understanding the mechanism by which KSHV increases the infected cell population is crucial for curing KSHV-associated diseases. Here we demonstrate that KSHV preferentially infects CD14 + monocytes and sustains viral replication through the viral interleukin-6 (vIL6)-mediated activation of STAT1 and 3. Using vIL6-sufficient and vIL6-deficient recombinant KSHV, we demonstrated that vIL6 plays a critical role in promoting the proliferation and differentiation of KSHV-infected monocytes into macrophages. The macrophages derived from vIL6-sufficient KSHV infection showed a distinct transcriptional profile of elevated IFN-pathway activation with immune suppression and were compromised in T-cell stimulation function compared to those from vIL6-deficient KSHV infection or uninfected control. These results highlight a clever strategy, in which KSHV utilizes vIL6 to secure its viral pool by expanding infected dysfunctional macrophages. This mechanism also facilitates KSHV to escape from host immune surveillance and to establish a lifelong infection. 160. Summary KSHV causes multiple inflammatory diseases, however, the underlying mechanism is not clear. Shimoda et al. demonstrate that KSHV preferentially infects monocytes and utilizes virally encoded interleukin-6 to expand and deregulate infected monocytes. This helps the virus escape from host immune surveillance.
Collapse
|
18
|
Mundra A, Yegiazaryan A, Karsian H, Alsaigh D, Bonavida V, Frame M, May N, Gargaloyan A, Abnousian A, Venketaraman V. Pathogenicity of Type I Interferons in Mycobacterium tuberculosis. Int J Mol Sci 2023; 24:3919. [PMID: 36835324 PMCID: PMC9965986 DOI: 10.3390/ijms24043919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Tuberculosis (TB) is a leading cause of mortality due to infectious disease and rates have increased during the emergence of COVID-19, but many of the factors determining disease severity and progression remain unclear. Type I Interferons (IFNs) have diverse effector functions that regulate innate and adaptive immunity during infection with microorganisms. There is well-documented literature on type I IFNs providing host defense against viruses; however, in this review, we explore the growing body of work that indicates high levels of type I IFNs can have detrimental effects to a host fighting TB infection. We report findings that increased type I IFNs can affect alveolar macrophage and myeloid function, promote pathological neutrophil extracellular trap responses, inhibit production of protective prostaglandin 2, and promote cytosolic cyclic GMP synthase inflammation pathways, and discuss many other relevant findings.
Collapse
Affiliation(s)
- Akaash Mundra
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Aram Yegiazaryan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Haig Karsian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Dijla Alsaigh
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Victor Bonavida
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Mitchell Frame
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Nicole May
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Areg Gargaloyan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Arbi Abnousian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91768, USA
| |
Collapse
|
19
|
Qu M, Zhu H, Zhang X. Extracellular vesicle-mediated regulation of macrophage polarization in bacterial infections. Front Microbiol 2022; 13:1039040. [PMID: 36619996 PMCID: PMC9815515 DOI: 10.3389/fmicb.2022.1039040] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022] Open
Abstract
Extracellular vesicles (EVs) are nanoscale membrane-enveloped vesicles secreted by prokaryotic and eukaryotic cells, which are commonly defined as membrane vesicles (MVs) and exosomes, respectively. They play critical roles in the bacteria-bacteria and bacteria-host interactions. In infectious diseases caused by bacteria, as the first line of defense against pathogens, the macrophage polarization mode commonly determines the success or failure of the host's response to pathogen aggression. M1-type macrophages secrete pro-inflammatory factors that support microbicidal activity, while alternative M2-type macrophages secrete anti-inflammatory factors that perform an antimicrobial immune response but partially allow pathogens to replicate and survive intracellularly. Membrane vesicles (MVs) released from bacteria as a distinctive secretion system can carry various components, including bacterial effectors, nucleic acids, or lipids to modulate macrophage polarization in host-pathogen interaction. Similar to MVs, bacteria-infected macrophages can secrete exosomes containing a variety of components to manipulate the phenotypic polarization of "bystander" macrophages nearby or long distance to differentiate into type M1 or M2 to regulate the course of inflammation. Exosomes can also repair tissue damage associated with the infection by upregulating the levels of anti-inflammatory factors, downregulating the pro-inflammatory factors, and regulating cellular biological behaviors. The study of the mechanisms by which EVs modulate macrophage polarization has opened new frontiers in delineating the molecular machinery involved in bacterial pathogenesis and challenges in providing new strategies for diagnosis and therapy.
Collapse
Affiliation(s)
- Mingjuan Qu
- School of Life Sciences, Ludong University, Yantai, China,Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai, China
| | - Hongwei Zhu
- School of Life Sciences, Ludong University, Yantai, China,Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai, China,Shandong Provincial Key Laboratory of Quality Safety Monitoring and Risk Assessment for Animal Products, Jinan, China
| | - Xingxiao Zhang
- School of Life Sciences, Ludong University, Yantai, China,Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai, China,Shandong Breeding Environmental Control Engineering Laboratory, Yantai, China,*Correspondence: Xingxiao Zhang, ✉
| |
Collapse
|
20
|
Llibre A, Smith N, Rouilly V, Musvosvi M, Nemes E, Posseme C, Mabwe S, Charbit B, Mbandi SK, Filander E, Africa H, Saint-André V, Bondet V, Bost P, Mulenga H, Bilek N, Albert ML, Scriba TJ, Duffy D. Tuberculosis alters immune-metabolic pathways resulting in perturbed IL-1 responses. Front Immunol 2022; 13:897193. [PMID: 36591308 PMCID: PMC9795069 DOI: 10.3389/fimmu.2022.897193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 09/12/2022] [Indexed: 12/15/2022] Open
Abstract
Tuberculosis (TB) remains a major public health problem and we lack a comprehensive understanding of how Mycobacterium tuberculosis (M. tb) infection impacts host immune responses. We compared the induced immune response to TB antigen, BCG and IL-1β stimulation between latently M. tb infected individuals (LTBI) and active TB patients. This revealed distinct responses between TB/LTBI at transcriptomic, proteomic and metabolomic levels. At baseline, we identified a novel immune-metabolic association between pregnane steroids, the PPARγ pathway and elevated plasma IL-1ra in TB. We observed dysregulated IL-1 responses after BCG stimulation in TB patients, with elevated IL-1ra responses being explained by upstream TNF differences. Additionally, distinct secretion of IL-1α/IL-1β in LTBI/TB after BCG stimulation was associated with downstream differences in granzyme mediated cleavage. Finally, IL-1β driven signalling was dramatically perturbed in TB disease but was completely restored after successful treatment. This study improves our knowledge of how immune responses are altered during TB disease, and may support the design of improved preventive and therapeutic tools, including host-directed strategies.
Collapse
Affiliation(s)
- Alba Llibre
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Nikaïa Smith
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | | | - Munyaradzi Musvosvi
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Elisa Nemes
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Céline Posseme
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Simbarashe Mabwe
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Bruno Charbit
- Cytometry and Biomarkers UTechS, CRT, Institut Pasteur, Université Paris Cité, Paris, France
| | - Stanley Kimbung Mbandi
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Elizabeth Filander
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Hadn Africa
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Violaine Saint-André
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France,Bioinformatics and Biostatistics HUB, Computational Biology Department, Institut Pasteur, Université Paris Cité, Paris, France
| | - Vincent Bondet
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Pierre Bost
- Sorbonne Université, Complexité du vivant, Paris, France,Systems Biology Group, Computational Biology Department, Institut Pasteur, Université Paris Cité, Paris, France
| | - Humphrey Mulenga
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Nicole Bilek
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | | | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France,Cytometry and Biomarkers UTechS, CRT, Institut Pasteur, Université Paris Cité, Paris, France,*Correspondence: Darragh Duffy,
| |
Collapse
|
21
|
Shankaran D, Arumugam P, Vasanthakumar RP, Singh A, Bothra A, Gandotra S, Rao V. Modern Clinical Mycobacterium tuberculosisStrains Leverage Type I IFN Pathway for a Proinflammatory Response in the Host. THE JOURNAL OF IMMUNOLOGY 2022; 209:1736-1745. [DOI: 10.4049/jimmunol.2101029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 08/16/2022] [Indexed: 11/15/2022]
|
22
|
Feriotti C, Sá-Pessoa J, Calderón-González R, Gu L, Morris B, Sugisawa R, Insua JL, Carty M, Dumigan A, Ingram RJ, Kissenpfening A, Bowie AG, Bengoechea JA. Klebsiella pneumoniae hijacks the Toll-IL-1R protein SARM1 in a type I IFN-dependent manner to antagonize host immunity. Cell Rep 2022; 40:111167. [PMID: 35947948 PMCID: PMC9638020 DOI: 10.1016/j.celrep.2022.111167] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 05/18/2022] [Accepted: 07/14/2022] [Indexed: 02/06/2023] Open
Abstract
Many bacterial pathogens antagonize host defense responses by translocating effector proteins into cells. It remains an open question how those pathogens not encoding effectors counteract anti-bacterial immunity. Here, we show that Klebsiella pneumoniae exploits the evolutionary conserved innate protein SARM1 to regulate negatively MyD88- and TRIF-governed inflammation, and the activation of the MAP kinases ERK and JNK. SARM1 is required for Klebsiella induction of interleukin-10 (IL-10) by fine-tuning the p38-type I interferon (IFN) axis. SARM1 inhibits the activation of Klebsiella-induced absent in melanoma 2 inflammasome to limit IL-1β production, suppressing further inflammation. Klebsiella exploits type I IFNs to induce SARM1 in a capsule and lipopolysaccharide O-polysaccharide-dependent manner via the TLR4-TRAM-TRIF-IRF3-IFNAR1 pathway. Absence of SARM1 reduces the intracellular survival of K. pneumoniae in macrophages, whereas sarm1-deficient mice control the infection. Altogether, our results illustrate an anti-immunology strategy deployed by a human pathogen. SARM1 inhibition will show a beneficial effect to treat Klebsiella infections.
Collapse
Affiliation(s)
- Claudia Feriotti
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, UK
| | - Joana Sá-Pessoa
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, UK
| | - Ricardo Calderón-González
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, UK
| | - Lili Gu
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Brenda Morris
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, UK
| | - Ryoichi Sugisawa
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Jose L Insua
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, UK
| | - Michael Carty
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Amy Dumigan
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, UK
| | - Rebecca J Ingram
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, UK
| | - Adrien Kissenpfening
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, UK
| | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - José A Bengoechea
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, UK.
| |
Collapse
|
23
|
Maciag K, Raychowdhury R, Smith K, Schneider AM, Coers J, Mumbach MR, Schwartz S, Hacohen N. IRF3 inhibits IFN-γ-mediated restriction of intracellular pathogens in macrophages independently of IFNAR. J Leukoc Biol 2022; 112:257-271. [PMID: 34826345 PMCID: PMC9550582 DOI: 10.1002/jlb.3a0218-069rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 10/25/2021] [Accepted: 10/30/2021] [Indexed: 01/14/2023] Open
Abstract
Macrophages use an array of innate immune sensors to detect intracellular pathogens and to tailor effective antimicrobial responses. In addition, extrinsic activation with the cytokine IFN-γ is often required as well to tip the scales of the host-pathogen balance toward pathogen restriction. However, little is known about how host-pathogen sensing impacts the antimicrobial IFN-γ-activated state. It was observed that in the absence of IRF3, a key downstream component of pathogen sensing pathways, IFN-γ-primed macrophages more efficiently restricted the intracellular bacterium Legionella pneumophila and the intracellular protozoan parasite Trypanosoma cruzi. This effect did not require IFNAR, the receptor for Type I IFNs known to be induced by IRF3, nor the sensing adaptors MyD88/TRIF, MAVS, or STING. This effect also did not involve differential activation of STAT1, the major signaling protein downstream of both Type 1 and Type 2 IFN receptors. IRF3-deficient macrophages displayed a significantly altered IFN-γ-induced gene expression program, with up-regulation of microbial restriction factors such as Nos2. Finally, we found that IFN-γ-primed but not unprimed macrophages largely excluded the activated form of IRF3 from the nucleus following bacterial infection. These data are consistent with a relationship of mutual inhibition between IRF3 and IFN-γ-activated programs, possibly as a component of a partially reversible mechanism for modulating the activity of potent innate immune effectors (such as Nos2) in the context of intracellular infection.
Collapse
Affiliation(s)
- Karolina Maciag
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Program in Immunology, Harvard Medical School, Boston, MA 02115, USA,Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | | | - Karen Smith
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alexis M. Schneider
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jörn Coers
- Departments of Molecular Genetics and Microbiology and Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Program in Immunology, Harvard Medical School, Boston, MA 02115, USA,Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Charlestown, MA 02129, USA
| |
Collapse
|
24
|
Study on the Correlation between Interleukin-27 and CXCL10 in Pulmonary Tuberculosis. J Immunol Res 2022; 2022:2932837. [PMID: 35785034 PMCID: PMC9242752 DOI: 10.1155/2022/2932837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 03/21/2022] [Accepted: 05/11/2022] [Indexed: 11/18/2022] Open
Abstract
Objective. To investigate the correlation between interleukin-27 and CXCL10 and other cytokines in pulmonary tuberculosis and to further explore the related miRNAs through bioinformatics. Methods. Collect the lesion tissue and peripheral blood of pulmonary tuberculosis patients and the peripheral blood of healthy controls. Immunohistochemical staining and qRT-PCR were used to observe the expression of interleukin-27, CXCL9, CXCL10, and CXCL11. Then, predict the key miRNA, qRT-PCR was used to verify the expression of miRNA in the peripheral blood and evaluated the correlation between them. Results. Both immunohistochemical staining and qRT-PCR indicated that the expressions of IL-27, CXCL9, CXCL10, and CXCL11 were significantly increased in tuberculosis patients, and IL-27 was significantly correlated with CXCL10 (
). Key molecules such as has-let-7b-5p, has-miR-30a-3p, and has-miR-320b were screened out. Among them, has-let-7b-5p was significantly downregulated, and has-miR-30a-3p was significantly upregulated; they were related to interleukin-27 and CXCL10. Conclusion. Our data shows that interleukin-27 and CXCL10 are significantly related in pulmonary tuberculosis, and has-let-7b-5p and has-miR-30a-3p are also related to interleukin-27 and CXCL10. It laid the foundation for subsequently exploiting the potential biomarkers in tuberculosis disease.
Collapse
|
25
|
Pellegrini JM, Gorvel JP, Mémet S. Immunosuppressive Mechanisms in Brucellosis in Light of Chronic Bacterial Diseases. Microorganisms 2022; 10:1260. [PMID: 35888979 PMCID: PMC9324529 DOI: 10.3390/microorganisms10071260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 01/27/2023] Open
Abstract
Brucellosis is considered one of the major zoonoses worldwide, constituting a critical livestock and human health concern with a huge socio-economic burden. Brucella genus, its etiologic agent, is composed of intracellular bacteria that have evolved a prodigious ability to elude and shape host immunity to establish chronic infection. Brucella's intracellular lifestyle and pathogen-associated molecular patterns, such as its specific lipopolysaccharide (LPS), are key factors for hiding and hampering recognition by the immune system. Here, we will review the current knowledge of evading and immunosuppressive mechanisms elicited by Brucella species to persist stealthily in their hosts, such as those triggered by their LPS and cyclic β-1,2-d-glucan or involved in neutrophil and monocyte avoidance, antigen presentation impairment, the modulation of T cell responses and immunometabolism. Attractive strategies exploited by other successful chronic pathogenic bacteria, including Mycobacteria, Salmonella, and Chlamydia, will be also discussed, with a special emphasis on the mechanisms operating in brucellosis, such as granuloma formation, pyroptosis, and manipulation of type I and III IFNs, B cells, innate lymphoid cells, and host lipids. A better understanding of these stratagems is essential to fighting bacterial chronic infections and designing innovative treatments and vaccines.
Collapse
|
26
|
Kang TG, Kwon KW, Kim K, Lee I, Kim MJ, Ha SJ, Shin SJ. Viral coinfection promotes tuberculosis immunopathogenesis by type I IFN signaling-dependent impediment of Th1 cell pulmonary influx. Nat Commun 2022; 13:3155. [PMID: 35672321 PMCID: PMC9174268 DOI: 10.1038/s41467-022-30914-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 05/06/2022] [Indexed: 01/09/2023] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is often exacerbated upon coinfection, but the underlying immunological mechanisms remain unclear. Here, to elucidate these mechanisms, we use an Mtb and lymphocytic choriomeningitis virus coinfection model. Viral coinfection significantly suppresses Mtb-specific IFN-γ production, with elevated bacterial loads and hyperinflammation in the lungs. Type I IFN signaling blockade rescues the Mtb-specific IFN-γ response and ameliorates lung immunopathology. Single-cell sequencing, tissue immunofluorescence staining, and adoptive transfer experiments indicate that viral infection-induced type I IFN signaling could inhibit CXCL9/10 production in myeloid cells, ultimately impairing pulmonary migration of Mtb-specific CD4+ T cells. Thus, our study suggests that augmented and sustained type I IFNs by virus coinfection prior to the pulmonary localization of Mtb-specific Th1 cells exacerbates TB immunopathogenesis by impeding the Mtb-specific Th1 cell influx. Our study highlights a negative function of viral coinfection-induced type I IFN responses in delaying Mtb-specific Th1 responses in the lung. Viral coinfection alongside mycobacterium tuberculosis (Mtb) infection may lead to immune complications or interference with immune responses. Here the authors show that in mice infected with Mtb and LCMV virus the specific TH1 response to MTb is reduced through a type I IFN response to the infecting virus.
Collapse
Affiliation(s)
- Tae Gun Kang
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.,Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul, 03722, Republic of Korea
| | - Kee Woong Kwon
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Kyungsoo Kim
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.,Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Insuk Lee
- Department of Biotechnology, College of Life Science & Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Myeong Joon Kim
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.,Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea. .,Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul, 03722, Republic of Korea.
| | - Sung Jae Shin
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea. .,Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| |
Collapse
|
27
|
Synthesis, Characterization, and In Vivo Cytokinome Profile of IL-12-Loaded PLGA Nanospheres. J Immunol Res 2022; 2022:6993187. [PMID: 35465347 PMCID: PMC9023212 DOI: 10.1155/2022/6993187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/23/2022] [Indexed: 01/19/2023] Open
Abstract
We report the successful encapsulation and elution of recombinant murine IL-12 (rmIL-12) from poly(lactide-co-glycolic) acid (PLGA) nanospheres (IL-12-NS) synthesized using the double emulsion/solvent evaporation (DESE) technique with microsphere depletion through ultracentrifugation. Images obtained with scanning electron microscopy (SEM) showcased a characteristic spherical shape with a mean particle diameter of 138.1 ± 10.8 nm and zeta potential of −15.1 ± 1.249 mV. These values suggest minimal flocculation when in solution, which was reflected in an in vivo biodistribution study that reported no observed morbidity/mortality. Encapsulation efficiency (EE) was determined to be 0.101 ± 0.009% with average particle concentration obtained per batch of 1.66 × 109 ± 4.45 × 108 particles/mL. Disparate zeta (ζ) potentials obtained from both protein-loaded and protein-unloaded batches suggested surface adsorption of protein, and confocal microscopy of BSA-FITC-loaded nanospheres confirmed the presence of protein within the polymeric shell. Furthermore, elution of rmIL-12 from IL-12-NS at a concentration of 500 million particles/mL was characterized using enzyme-linked immunosorbent assay (ELISA). When IL-12-NS was administered in vivo to female BALB/c mice through retroorbital injection, IL-12-NS produced a favorable systemic cytokine profile for tumoricidal activity within the peripheral blood. Whereas IFN-γ nadir occurred at 72 hours, levels recovered quickly and displayed positive correlations postburst out to 25 days postinjection. IL-12-NS administration induced proinflammatory changes while prompting minimal counterregulatory increases in anti-inflammatory IL-10 and IL-4 cytokine levels. Further, while IL-6 levels increased to 30 folds of the baseline during the burst phase, they normalized by 72 hours and trended negatively throughout the sill phase. Similar trends were observed with IL-1β and CXCL-1, suggesting a decreased likelihood of progression to a systemic inflammatory response syndrome-like state. As IL-12-NS delivers logarithmically lower amounts of IL-12 than previously administered during human clinical trials, our data reflect the importance of IL-12-NS which safely create a systemic immunostimulatory environment.
Collapse
|
28
|
Rinaldi S, Pallikkuth S, De Armas L, Richardson B, Pan L, Pahwa R, Williams S, Cameron M, Pahwa S. Distinct Molecular Signatures of Aging in Healthy and HIV-Infected Individuals. J Acquir Immune Defic Syndr 2022; 89:S47-S55. [PMID: 35015745 PMCID: PMC8751284 DOI: 10.1097/qai.0000000000002864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Virally suppressed chronic HIV-infected individuals on antiretroviral therapy experience similar immune impairments as HIV-uninfected elderly. However, they manifest symptoms of premature immune aging such as suboptimal responses to vaccination at a younger age. Mechanisms underlying premature immune aging are unclear. SETTING The study site was University of Miami Miller School of Medicine. METHODS In this study, we aimed to identify molecular signatures of aging in HIV-infected (HIV) individuals compared with age-matched healthy control (HC) participants. Transcriptomic profiles of peripheral blood mononuclear cells collected cross-sectionally from study participants were evaluated using RNA sequencing, and genes and pathways associated with age and HIV status were identified and compared between study groups. Generalized linear modeling was used to identify transcriptional signatures associated with age. RESULTS Despite that fewer differentially expressed genes between young (<40 yrs) and old (>59 yrs) were observed in the HIV group, metabolic and innate immune activation pathways were associated with increasing age in both HIV and HC. Age was also associated with pathways involved with T-cell immune activation in HC and with interferon signaling pathways in HIV. We observed signs of precocious immune aging at the transcriptional level in HIV and defined a transcriptional perturbation associated with innate immunity and glucose metabolism induced by aging in both HC and HIV. CONCLUSION In this study, we identified distinct molecular signatures predictive of age in HIV versus HC, which suggest precocious immune aging in HIV. Overall, our results highlight the molecular pathways of immune aging in both HC and HIV that may be targeted for additional mechanistic insights or in a therapeutic setting.
Collapse
Affiliation(s)
- Stefano Rinaldi
- Department of Microbiology & Immunology, University of Miami Miller School of Medicine, Miami, FL
| | - Suresh Pallikkuth
- Department of Microbiology & Immunology, University of Miami Miller School of Medicine, Miami, FL
- Miami Center for AIDS Research, University of Miami Miller School of Medicine, Miami, FL
| | - Lesley De Armas
- Department of Microbiology & Immunology, University of Miami Miller School of Medicine, Miami, FL
| | - Brian Richardson
- Department of Population & Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH.
| | - Li Pan
- Department of Microbiology & Immunology, University of Miami Miller School of Medicine, Miami, FL
| | - Rajendra Pahwa
- Department of Microbiology & Immunology, University of Miami Miller School of Medicine, Miami, FL
| | - Sion Williams
- Department of Microbiology & Immunology, University of Miami Miller School of Medicine, Miami, FL
- Miami Center for AIDS Research, University of Miami Miller School of Medicine, Miami, FL
| | - Mark Cameron
- Miami Center for AIDS Research, University of Miami Miller School of Medicine, Miami, FL
- Department of Population & Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH.
| | - Savita Pahwa
- Department of Microbiology & Immunology, University of Miami Miller School of Medicine, Miami, FL
- Miami Center for AIDS Research, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
29
|
Unterman A, Sumida TS, Nouri N, Yan X, Zhao AY, Gasque V, Schupp JC, Asashima H, Liu Y, Cosme C, Deng W, Chen M, Raredon MSB, Hoehn KB, Wang G, Wang Z, DeIuliis G, Ravindra NG, Li N, Castaldi C, Wong P, Fournier J, Bermejo S, Sharma L, Casanovas-Massana A, Vogels CBF, Wyllie AL, Grubaugh ND, Melillo A, Meng H, Stein Y, Minasyan M, Mohanty S, Ruff WE, Cohen I, Raddassi K, Niklason LE, Ko AI, Montgomery RR, Farhadian SF, Iwasaki A, Shaw AC, van Dijk D, Zhao H, Kleinstein SH, Hafler DA, Kaminski N, Dela Cruz CS. Single-cell multi-omics reveals dyssynchrony of the innate and adaptive immune system in progressive COVID-19. Nat Commun 2022; 13:440. [PMID: 35064122 PMCID: PMC8782894 DOI: 10.1038/s41467-021-27716-4] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/03/2021] [Indexed: 02/06/2023] Open
Abstract
Dysregulated immune responses against the SARS-CoV-2 virus are instrumental in severe COVID-19. However, the immune signatures associated with immunopathology are poorly understood. Here we use multi-omics single-cell analysis to probe the dynamic immune responses in hospitalized patients with stable or progressive course of COVID-19, explore V(D)J repertoires, and assess the cellular effects of tocilizumab. Coordinated profiling of gene expression and cell lineage protein markers shows that S100Ahi/HLA-DRlo classical monocytes and activated LAG-3hi T cells are hallmarks of progressive disease and highlights the abnormal MHC-II/LAG-3 interaction on myeloid and T cells, respectively. We also find skewed T cell receptor repertories in expanded effector CD8+ clones, unmutated IGHG+ B cell clones, and mutated B cell clones with stable somatic hypermutation frequency over time. In conclusion, our in-depth immune profiling reveals dyssynchrony of the innate and adaptive immune interaction in progressive COVID-19.
Collapse
MESH Headings
- Adaptive Immunity/drug effects
- Adaptive Immunity/genetics
- Adaptive Immunity/immunology
- Aged
- Antibodies, Monoclonal, Humanized/therapeutic use
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- COVID-19/genetics
- COVID-19/immunology
- Cells, Cultured
- Female
- Gene Expression Profiling/methods
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/immunology
- Humans
- Immunity, Innate/drug effects
- Immunity, Innate/genetics
- Immunity, Innate/immunology
- Male
- RNA-Seq/methods
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- SARS-CoV-2/drug effects
- SARS-CoV-2/immunology
- SARS-CoV-2/physiology
- Single-Cell Analysis/methods
- COVID-19 Drug Treatment
Collapse
Affiliation(s)
- Avraham Unterman
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA.
- Pulmonary Institute, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel.
| | - Tomokazu S Sumida
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA.
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA.
| | - Nima Nouri
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Center for Medical Informatics, Yale School of Medicine, New Haven, CT, USA
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Xiting Yan
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Amy Y Zhao
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Victor Gasque
- Department of Computer Science, Yale University, New Haven, CT, USA
- Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Jonas C Schupp
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
- Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease Hannover, German Lung Research Center (DZL), Hannover, Germany
| | - Hiromitsu Asashima
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Yunqing Liu
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Carlos Cosme
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Wenxuan Deng
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Ming Chen
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Micha Sam Brickman Raredon
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA
| | - Kenneth B Hoehn
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Guilin Wang
- Yale Center for Genome Analysis/Keck Biotechnology Resource Laboratory, Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, CT, USA
| | - Zuoheng Wang
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Giuseppe DeIuliis
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Neal G Ravindra
- Department of Computer Science, Yale University, New Haven, CT, USA
- Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Ningshan Li
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, USA
- SJTU-Yale Joint Center for Biostatistics and Data Science, Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | | | - Patrick Wong
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - John Fournier
- School of Medicine, Yale University, New Haven, CT, USA
| | - Santos Bermejo
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Lokesh Sharma
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Arnau Casanovas-Massana
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Chantal B F Vogels
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Anne L Wyllie
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Nathan D Grubaugh
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Anthony Melillo
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Hailong Meng
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Yan Stein
- Pulmonary Institute, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Maksym Minasyan
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Subhasis Mohanty
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - William E Ruff
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Inessa Cohen
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Khadir Raddassi
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Laura E Niklason
- Departments of Anesthesiology & Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Albert I Ko
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Ruth R Montgomery
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Shelli F Farhadian
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Albert C Shaw
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - David van Dijk
- Department of Computer Science, Yale University, New Haven, CT, USA
- Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- SJTU-Yale Joint Center for Biostatistics and Data Science, Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Inter-Departmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Steven H Kleinstein
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Inter-Departmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - David A Hafler
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Charles S Dela Cruz
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
- West Haven Veterans Affair Medical Center, West Haven, CT, USA
| |
Collapse
|
30
|
Swartzwelter BJ, Michelini S, Frauenlob T, Barbero F, Verde A, De Luca AC, Puntes V, Duschl A, Horejs-Hoeck J, Italiani P, Boraschi D. Innate Memory Reprogramming by Gold Nanoparticles Depends on the Microbial Agents That Induce Memory. Front Immunol 2021; 12:751683. [PMID: 34804037 PMCID: PMC8600232 DOI: 10.3389/fimmu.2021.751683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/14/2021] [Indexed: 01/14/2023] Open
Abstract
Innate immune memory, the ability of innate cells to react in a more protective way to secondary challenges, is induced by exposure to infectious and other exogeous and endogenous agents. Engineered nanoparticles are particulate exogenous agents that, as such, could trigger an inflammatory reaction in monocytes and macrophages and could therefore be also able to induce innate memory. Here, we have evaluated the capacity of engineered gold nanoparticles (AuNPs) to induce a memory response or to modulate the memory responses induced by microbial agents. Microbial agents used were in soluble vs. particulate form (MDP and the gram-positive bacteria Staphylococcus aureus; β-glucan and the β-glucan-producing fungi C. albicans), and as whole microrganisms that were either killed (S. aureus, C. albicans) or viable (the gram-negative bacteria Helicobacter pylori). The memory response was assessed in vitro, by exposing human primary monocytes from 2-7 individual donors to microbial agents with or without AuNPs (primary response), then resting them for 6 days to allow return to baseline, and eventually challenging them with LPS (secondary memory response). Primary and memory responses were tested as production of the innate/inflammatory cytokine TNFα and other inflammatory and anti-inflammatory factors. While inactive on the response induced by soluble microbial stimuli (muramyl dipeptide -MDP-, β-glucan), AuNPs partially reduced the primary response induced by whole microorganisms. AuNPs were also unable to directly induce a memory response but could modulate stimulus-induced memory in a circumscribed fashion, limited to some agents and some cytokines. Thus, the MDP-induced tolerance in terms of TNFα production was further exacerbated by co-priming with AuNPs, resulting in a less inflammatory memory response. Conversely, the H. pylori-induced tolerance was downregulated by AuNPs only relative to the anti-inflammatory cytokine IL-10, which would lead to an overall more inflammatory memory response. These effects of AuNPs may depend on a differential interaction/association between the reactive particle surfaces and the microbial components and agents, which may lead to a change in the exposure profiles. As a general observation, however, the donor-to-donor variability in memory response profiles and reactivity to AuNPs was substantial, suggesting that innate memory depends on the individual history of exposures.
Collapse
Affiliation(s)
- Benjamin J. Swartzwelter
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
- Department Biosciences, Paris Lodron University of Salzburg (PLUS), Salzburg, Austria
| | - Sara Michelini
- Department Biosciences, Paris Lodron University of Salzburg (PLUS), Salzburg, Austria
| | - Tobias Frauenlob
- Department Biosciences, Paris Lodron University of Salzburg (PLUS), Salzburg, Austria
| | - Francesco Barbero
- Institut Català de Nanociència i Nanotecnologia (ICN2), Consejo Superior de Investigaciones Científicas (CSIC) and The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Alessandro Verde
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Anna Chiara De Luca
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Victor Puntes
- Institut Català de Nanociència i Nanotecnologia (ICN2), Consejo Superior de Investigaciones Científicas (CSIC) and The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Vall d’Hebron Research Institute (VHIR), Barcelona, Spain
- Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Spain
| | - Albert Duschl
- Department Biosciences, Paris Lodron University of Salzburg (PLUS), Salzburg, Austria
| | - Jutta Horejs-Hoeck
- Department Biosciences, Paris Lodron University of Salzburg (PLUS), Salzburg, Austria
| | - Paola Italiani
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Diana Boraschi
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
- Stazione Zoologica Anton Dohrn, Napoli, Italy
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| |
Collapse
|
31
|
Tang P, Virtue S, Goie JYG, Png CW, Guo J, Li Y, Jiao H, Chua YL, Campbell M, Moreno-Navarrete JM, Shabbir A, Fernández-Real JM, Gasser S, Kemeny DM, Yang H, Vidal-Puig A, Zhang Y. Regulation of adipogenic differentiation and adipose tissue inflammation by interferon regulatory factor 3. Cell Death Differ 2021; 28:3022-3035. [PMID: 34091599 PMCID: PMC8563729 DOI: 10.1038/s41418-021-00798-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 02/04/2023] Open
Abstract
Dysfunction of adipocytes and adipose tissue is a primary defect in obesity and obesity-associated metabolic diseases. Interferon regulatory factor 3 (IRF3) has been implicated in adipogenesis. However, the role of IRF3 in obesity and obesity-associated disorders remains unclear. Here, we show that IRF3 expression in human adipose tissues is positively associated with insulin sensitivity and negatively associated with type 2 diabetes. In mouse pre-adipocytes, deficiency of IRF3 results in increased expression of PPARγ and PPARγ-mediated adipogenic genes, leading to increased adipogenesis and altered adipocyte functionality. The IRF3 knockout (KO) mice develop obesity, insulin resistance, glucose intolerance, and eventually type 2 diabetes with aging, which is associated with the development of white adipose tissue (WAT) inflammation. Increased macrophage accumulation with M1 phenotype which is due to the loss of IFNβ-mediated IL-10 expression is observed in WAT of the KO mice compared to that in wild-type mice. Bone-marrow reconstitution experiments demonstrate that the nonhematopoietic cells are the primary contributors to the development of obesity and both hematopoietic and nonhematopoietic cells contribute to the development of obesity-related complications in IRF3 KO mice. This study demonstrates that IRF3 regulates the biology of multiple cell types including adipocytes and macrophages to prevent the development of obesity and obesity-related complications and hence, could be a potential target for therapeutic interventions for the prevention and treatment of obesity-associated metabolic disorders.
Collapse
Affiliation(s)
- Peng Tang
- Department of Microbiology & Immunology, and NUSMED Immunology Translational Research Programme,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Sam Virtue
- Institute of Metabolic Science, Wellcome Trust-MRC MDU Metabolic Disease Unit, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Jian Yi Gerald Goie
- Department of Microbiology & Immunology, and NUSMED Immunology Translational Research Programme,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Chin Wen Png
- Department of Microbiology & Immunology, and NUSMED Immunology Translational Research Programme,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Jing Guo
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Ying Li
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Huipeng Jiao
- Department of Microbiology & Immunology, and NUSMED Immunology Translational Research Programme,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Yen Leong Chua
- Department of Microbiology & Immunology, and NUSMED Immunology Translational Research Programme,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Mark Campbell
- Institute of Metabolic Science, Wellcome Trust-MRC MDU Metabolic Disease Unit, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - José Maria Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigacio Biomedica de Girona (IDIBGI), CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn, CB06/03/010), Instituto de Salud Carlos III, and Department of Medical Sciences, Faculty of Medicine, Girona, Spain
| | - Asim Shabbir
- Department of Surgery, National University Hospital, Singapore, Singapore
| | - José-Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigacio Biomedica de Girona (IDIBGI), CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn, CB06/03/010), Instituto de Salud Carlos III, and Department of Medical Sciences, Faculty of Medicine, Girona, Spain
| | - Stephan Gasser
- Department of Microbiology & Immunology, and NUSMED Immunology Translational Research Programme,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - David Michael Kemeny
- Department of Microbiology & Immunology, and NUSMED Immunology Translational Research Programme,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Antonio Vidal-Puig
- Institute of Metabolic Science, Wellcome Trust-MRC MDU Metabolic Disease Unit, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.
| | - Yongliang Zhang
- Department of Microbiology & Immunology, and NUSMED Immunology Translational Research Programme,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
32
|
Ahn JH, Park JY, Kim DY, Lee TS, Jung DH, Kim YJ, Lee YJ, Lee YJ, Seo IS, Song EJ, Jang AR, Yang SJ, Shin SJ, Park JH. Type I Interferons Are Involved in the Intracellular Growth Control of Mycobacterium abscessus by Mediating NOD2-Induced Production of Nitric Oxide in Macrophages. Front Immunol 2021; 12:738070. [PMID: 34777348 PMCID: PMC8581665 DOI: 10.3389/fimmu.2021.738070] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/30/2021] [Indexed: 12/20/2022] Open
Abstract
Mycobacterium abscessus (MAB) is one of the rapidly growing, multidrug-resistant non-tuberculous mycobacteria (NTM) causing various diseases including pulmonary disorder. Although it has been known that type I interferons (IFNs) contribute to host defense against bacterial infections, the role of type I IFNs against MAB infection is still unclear. In the present study, we show that rIFN-β treatment reduced the intracellular growth of MAB in macrophages. Deficiency of IFN-α/β receptor (IFNAR) led to the reduction of nitric oxide (NO) production in MAB-infected macrophages. Consistently, rIFN-β treatment enhanced the expression of iNOS gene and protein, and NO production in response to MAB. We also found that NO is essential for the intracellular growth control of MAB within macrophages in an inhibitor assay using iNOS-deficient cells. In addition, pretreatment of rIFN-β before MAB infection in mice increased production of NO in the lungs at day 1 after infection and promoted the bacterial clearance at day 5. However, when alveolar macrophages were depleted by treatment of clodronate liposome, rIFN-β did not promote the bacterial clearance in the lungs. Moreover, we found that a cytosolic receptor nucleotide-binding oligomerization domain 2 (NOD2) is required for MAB-induced TANK binding kinase 1 (TBK1) phosphorylation and IFN-β gene expression in macrophages. Finally, increase in the bacterial loads caused by reduction of NO levels was reversed by rIFN-β treatment in the lungs of NOD2-deficient mice. Collectively, our findings suggest that type I IFNs act as an intermediator of NOD2-induced NO production in macrophages and thus contribute to host defense against MAB infection.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Disease Models, Animal
- Female
- Host-Pathogen Interactions
- Interferon Type I/metabolism
- Lung/immunology
- Lung/metabolism
- Lung/microbiology
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/metabolism
- Macrophages, Alveolar/microbiology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mycobacterium Infections, Nontuberculous/immunology
- Mycobacterium Infections, Nontuberculous/metabolism
- Mycobacterium Infections, Nontuberculous/microbiology
- Mycobacterium abscessus/growth & development
- Mycobacterium abscessus/immunology
- Mycobacterium abscessus/metabolism
- Nitric Oxide/metabolism
- Nitric Oxide Synthase Type II/genetics
- Nitric Oxide Synthase Type II/metabolism
- Nod2 Signaling Adaptor Protein/genetics
- Nod2 Signaling Adaptor Protein/metabolism
- Receptor, Interferon alpha-beta/genetics
- Receptor, Interferon alpha-beta/metabolism
- Signal Transduction
- Mice
Collapse
Affiliation(s)
- Jae-Hun Ahn
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, South Korea
| | - Ji-Yeon Park
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, South Korea
| | - Dong-Yeon Kim
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, South Korea
| | - Tae-Sung Lee
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, South Korea
| | - Do-Hyeon Jung
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, South Korea
| | - Yeong-Jun Kim
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, South Korea
| | - Yeon-Ji Lee
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, South Korea
| | - Yun-Ji Lee
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, South Korea
| | - In-Su Seo
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, South Korea
| | - Eun-Jung Song
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, South Korea
| | - Ah-Ra Jang
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, South Korea
| | - Soo-Jin Yang
- Department of Veterinary Microbiology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
33
|
Rajan DK, Mohan K, Zhang S, Ganesan AR. Dieckol: a brown algal phlorotannin with biological potential. Biomed Pharmacother 2021; 142:111988. [PMID: 34371307 DOI: 10.1016/j.biopha.2021.111988] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/24/2021] [Accepted: 07/30/2021] [Indexed: 12/21/2022] Open
Abstract
Dieckol [C36H22O18], is a naturally occurring phlorotannin found in some brown algal species. Dieckol is gaining more attention in the scientific community for its potential biological activities. It has been exhibited a broad spectrum of therapeutic functions including anti-bacterial, anti-cancer, anti-oxidant, anti-aging, anti-diabetic, neuroprotective, and other medicinal applications. Distinct emphasis has been given to extraction, purification, and biomedical applications of dieckol. This critical review comprises of in vitro, in vivo, and in silico biological properties of dieckol. An attempt has been made to evaluate the effectiveness, therapeutical application, and mechanism of dieckol against various diseases. The pharmacological significance, current status and the dosage of multifunctional dieckol and its mechanisms have been discussed in this review. Dieckol plays an important role in apoptosis induction via inhibiting the PI3K, AKT, mTOR and FAK signaling molecules. Dieckol remarkably inhibited the lipid accumulation in high fat diet induced animal models. Dieckol, a multifaceted compound will be beneficial in attenuating the action of various diseases and it could be a potential pharmaceutical and nutraceutical compound. Therefore, the combined effects of dieckol with existing drugs and natural compounds will be studied in future to optimize its benefits. Besides limited information on the toxicological action and dosage administration of dieckol on the human was reported to date. Overall, dieckol is a prospective health-promoting compound for the development of a novel drug against numerous diseases.
Collapse
Affiliation(s)
- Durairaj Karthick Rajan
- Centre of Advanced Study in Marine Biology, Faculty of Marine Sciences, Annamalai University, Parangipettai 608502, Tamil Nadu, India.
| | - Kannan Mohan
- PG and Research Department of Zoology, Sri Vasavi College, Erode 638316, Tamil Nadu, India.
| | - Shubing Zhang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Abirami Ramu Ganesan
- Group of Fermentation and Distillation, Laimburg Research Center, Laimburg 6, I-39040 Post Auer, BZ, Italy
| |
Collapse
|
34
|
Alexander AF, Kelsey I, Forbes H, Miller-Jensen K. Single-cell secretion analysis reveals a dual role for IL-10 in restraining and resolving the TLR4-induced inflammatory response. Cell Rep 2021; 36:109728. [PMID: 34551303 DOI: 10.1016/j.celrep.2021.109728] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 06/18/2021] [Accepted: 08/26/2021] [Indexed: 01/22/2023] Open
Abstract
Following Toll-like receptor 4 (TLR4) stimulation of macrophages, negative feedback mediated by the anti-inflammatory cytokine interleukin-10 (IL-10) limits the inflammatory response. However, extensive cell-to-cell variability in TLR4-stimulated cytokine secretion raises questions about how negative feedback is robustly implemented. To explore this, we characterize the TLR4-stimulated secretion program in primary murine macrophages using a single-cell microwell assay that enables evaluation of functional autocrine IL-10 signaling. High-dimensional analysis of single-cell data reveals three tiers of TLR4-induced proinflammatory activation based on levels of cytokine secretion. Surprisingly, while IL-10 inhibits TLR4-induced activation in the highest tier, it also contributes to the TLR4-induced activation threshold by regulating which cells transition from non-secreting to secreting states. This role for IL-10 in restraining TLR4 inflammatory activation is largely mediated by intermediate interferon (IFN)-β signaling, while TNF likely mediates response resolution by IL-10. Thus, cell-to-cell variability in cytokine regulatory motifs provides a means to tailor the TLR4-induced inflammatory response.
Collapse
Affiliation(s)
- Amanda F Alexander
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Ilana Kelsey
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Hannah Forbes
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Kathryn Miller-Jensen
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA; Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06511, USA; Systems Biology Institute, Yale University, New Haven, CT 06511, USA.
| |
Collapse
|
35
|
Taghipour A, Malih N, Köksal F, Jokelainen P, Ghaffarifar F. Toxoplasma gondii seroprevalence among tuberculosis patients: A systematic review and meta-analysis. Microb Pathog 2021; 159:105083. [PMID: 34246749 DOI: 10.1016/j.micpath.2021.105083] [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: 06/06/2021] [Revised: 06/27/2021] [Accepted: 07/02/2021] [Indexed: 11/26/2022]
Abstract
Toxoplasma gondii and Mycobacterium tuberculosis are intracellular pathogens, both infecting a substantial proportion of human population. We conducted a systematic review and meta-analysis to estimate the pooled T. gondii seroprevalence in tuberculosis patients. Three international databases were systematically searched for literature on prevalence of T. gondii in tuberculosis patients. A total of 1389 documents were identified, and eight papers were eligible to be included in the systematic review and meta-analysis. Geographical data gaps were evident, as no studies were identified from many countries where both infections are important. The pooled seroprevalence of IgG, IgM, and both IgG and IgM antibodies against T. gondii in tuberculosis patients were estimated to be 35.9% (95% confidence interval [CI], 19.3-56.7%), 35.0% (95% CI, 3.0-90.3%), and 13.4% (95% CI, 2.4-49.0%), respectively. In the included case-control studies, the pooled T. gondii seroprevalence (proportion anti- T. gondii IgG antibody positive) was higher in tuberculosis patients than in their controls, with an odds ratio by random effects model of 1.63 (95% CI, 1.28-2.08). The results of our work suggest an association between T. gondii seropositivity and being a tuberculosis patient, which should however be interpreted with caution because the timeline of the infections and the disease process are not accounted for. Our work showed that T. gondii seropositivity, indicating chronic infection with the zoonotic parasite, was relatively common among tuberculosis patients.
Collapse
Affiliation(s)
- Ali Taghipour
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Narges Malih
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatih Köksal
- Department of Clinical Bacteriology, Faculty of Medicine, Çukurova University, Adana, Turkey; TR Ministry of Health Regional Tuberculosis Laboratories and the Director of the Tropical Diseases Research and Application Center, Turkey
| | - Pikka Jokelainen
- Department of Bacteria, Parasites & Fungi, Infectious Disease Preparedness, Statens Serum Institut, Copenhagen, Denmark.
| | - Fatemeh Ghaffarifar
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
36
|
Parihar SP, Ozturk M, Höft MA, Chia JE, Guler R, Keeton R, van Rensburg IC, Loxton AG, Brombacher F. IL-4-Responsive B Cells Are Detrimental During Chronic Tuberculosis Infection in Mice. Front Immunol 2021; 12:611673. [PMID: 34220793 PMCID: PMC8243286 DOI: 10.3389/fimmu.2021.611673] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 05/25/2021] [Indexed: 12/02/2022] Open
Abstract
In tuberculosis, T cell-mediated immunity is extensively studied whilst B cells received limited attention in human and mice. Of interest, Mycobacterium tuberculosis (Mtb) does increase IL-4 Receptor-alpha (IL4Rα) expression in murine B cells. To better understand the role of IL4Rα signalling in B cells, we compared wild type mice with B cell-specific IL4Rα deficient mice (mb1creIL-4Rα-/lox mice). Chronic Mtb aerosol infection in mb1creIL-4Rα-/lox mice reduced lung and spleen bacterial burdens, compared to littermate (IL-4Rα-/lox) control animals. Consequently, lung pathology, inflammation and inducible nitric oxide synthase (iNOS) expression were reduced in the lungs of mb1creIL-4Rα-/lox mice, which was also accompanied by increased lung IgA and decreased IgG1 levels. Furthermore, intratracheal adoptive transfer of wild-type B cells into B cell-specific IL4Rα deficient mice reversed the protective phenotype. Moreover, constitutively mCherry expressing Mtb showed decreased association with B cells from mb1creIL-4Rα-/lox mice ex vivo. In addition, supernatants from Mtb-exposed B cells of mb1creIL-4Rα-/lox mice also increased the ability of macrophages to produce nitric oxide, IL-1β, IL-6 and TNF. Together, this demonstrates that IL-4-responsive B cells are detrimental during the chronic phase of tuberculosis in mice with perturbed antibody profiles, inflammatory cytokines and tnf and stat1 levels in the lungs.
Collapse
Affiliation(s)
- Suraj P. Parihar
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Division of Medical Microbiology, Department of Pathology, Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
| | - Mumin Ozturk
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Maxine A. Höft
- AFGrica Medical Mycology Research Unit, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
| | - Julius E. Chia
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Reto Guler
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Department of Pathology, Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
| | - Roanne Keeton
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
| | - Ilana C. van Rensburg
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Andre G. Loxton
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Department of Pathology, Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
| |
Collapse
|
37
|
Rasouli J, Casella G, Ishikawa LLW, Thome R, Boehm A, Ertel A, Melo-Silva CR, Mari ER, Porazzi P, Zhang W, Xiao D, Sigal LJ, Fortina P, Zhang GX, Rostami A, Ciric B. IFN-β Acts on Monocytes to Ameliorate CNS Autoimmunity by Inhibiting Proinflammatory Cross-Talk Between Monocytes and Th Cells. Front Immunol 2021; 12:679498. [PMID: 34149716 PMCID: PMC8213026 DOI: 10.3389/fimmu.2021.679498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/12/2021] [Indexed: 01/18/2023] Open
Abstract
IFN-β has been the treatment for multiple sclerosis (MS) for almost three decades, but understanding the mechanisms underlying its beneficial effects remains incomplete. We have shown that MS patients have increased numbers of GM-CSF+ Th cells in circulation, and that IFN-β therapy reduces their numbers. GM-CSF expression by myelin-specific Th cells is essential for the development of experimental autoimmune encephalomyelitis (EAE), an animal model of MS. These findings suggested that IFN-β therapy may function via suppression of GM-CSF production by Th cells. In the current study, we elucidated a feedback loop between monocytes and Th cells that amplifies autoimmune neuroinflammation, and found that IFN-β therapy ameliorates central nervous system (CNS) autoimmunity by inhibiting this proinflammatory loop. IFN-β suppressed GM-CSF production in Th cells indirectly by acting on monocytes, and IFN-β signaling in monocytes was required for EAE suppression. IFN-β increased IL-10 expression by monocytes, and IL-10 was required for the suppressive effects of IFN-β. IFN-β treatment suppressed IL-1β expression by monocytes in the CNS of mice with EAE. GM-CSF from Th cells induced IL-1β production by monocytes, and, in a positive feedback loop, IL-1β augmented GM-CSF production by Th cells. In addition to GM-CSF, TNF and FASL expression by Th cells was also necessary for IL-1β production by monocyte. IFN-β inhibited GM-CSF, TNF, and FASL expression by Th cells to suppress IL-1β secretion by monocytes. Overall, our study describes a positive feedback loop involving several Th cell- and monocyte-derived molecules, and IFN-β actions on monocytes disrupting this proinflammatory loop.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Autoimmunity/drug effects
- Cell Communication/genetics
- Cell Communication/immunology
- Cytokines/metabolism
- Disease Models, Animal
- Disease Susceptibility/immunology
- Encephalomyelitis, Autoimmune, Experimental/etiology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis
- Interferon-beta/metabolism
- Interferon-beta/pharmacology
- Mice
- Mice, Knockout
- Monocytes/drug effects
- Monocytes/immunology
- Monocytes/metabolism
- T-Lymphocytes, Helper-Inducer/drug effects
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
Collapse
Affiliation(s)
- Javad Rasouli
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Giacomo Casella
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | | | - Rodolfo Thome
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Alexandra Boehm
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Adam Ertel
- Sidney Kimmel Cancer Center, Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Carolina R. Melo-Silva
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Elisabeth R. Mari
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Patrizia Porazzi
- Sidney Kimmel Cancer Center, Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Weifeng Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Dan Xiao
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Luis J. Sigal
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Paolo Fortina
- Sidney Kimmel Cancer Center, Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
- Department of Translation and Precision Medicine, Sapienza University, Rome, Italy
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
38
|
Min B, Kim D, Feige MJ. IL-30 † (IL-27A): a familiar stranger in immunity, inflammation, and cancer. Exp Mol Med 2021; 53:823-834. [PMID: 34045653 PMCID: PMC8178335 DOI: 10.1038/s12276-021-00630-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/12/2021] [Accepted: 03/29/2021] [Indexed: 12/16/2022] Open
Abstract
Over the years, interleukin (IL)-27 has received much attention because of its highly divergent, sometimes even opposing, functions in immunity. IL-30, the p28 subunit that forms IL-27 together with Ebi3 and is also known as IL-27p28 or IL-27A, has been considered a surrogate to represent IL-27. However, it was later discovered that IL-30 can form complexes with other protein subunits, potentially leading to overlapping or discrete functions. Furthermore, there is emerging evidence that IL-30 itself may perform immunomodulatory functions independent of Ebi3 or other binding partners and that IL-30 production is strongly associated with certain cancers in humans. In this review, we will discuss the biology of IL-30 and other IL-30-associated cytokines and their functions in inflammation and cancer. Studying the ways that interleukin IL-30 regulates immune responses may provide novel insights into tumor development and inflammatory conditions. Interleukins are a diverse family of proteins involved in intercellular communications and immunity, where they can exert divergent and even opposing functions. Booki Min at Northwestern University in Chicago, USA, and co-workers reviewed the current understanding of IL-30 and its links to inflammation and cancer. IL-30 forms the IL-27 complex with the Ebi3 protein and was thought to be a surrogate for IL-27 in terms of activity. However, recent insights suggest that IL-30 may perform discrete immune modulation functions. Elevated IL-30 secretion is linked to prostate and breast cancer development. Extensive research is needed into the formation of IL-30, its associated protein interactions, and the development of a suitable animal model.
Collapse
Affiliation(s)
- Booki Min
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA. .,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| | - Dongkyun Kim
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Matthias J Feige
- Department of Chemistry and Institute for Advanced Study, Technical University of Munich, 85748, Garching, Germany
| |
Collapse
|
39
|
Mandala JP, Thada S, Sivangala R, Ponnana M, Myakala R, Gaddam S. Influence of NOD-like receptor 2 gene polymorphisms on muramyl dipeptide induced pro-inflammatory response in patients with active pulmonary tuberculosis and household contacts. Immunobiology 2021; 226:152096. [PMID: 34058448 DOI: 10.1016/j.imbio.2021.152096] [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: 11/02/2020] [Revised: 03/20/2021] [Accepted: 04/16/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE The immune response induced by nucleotide-binding oligomerization domain-2(NOD2) is associated with the production of cytokines affected by the host's genetic background. The present study aimed to examine the effects of NOD2; 802C > T, 2105G > A polymorphisms associated with altered cytokine levels in patients with active pulmonary tuberculosis disease, Latent TB subjects (household contacts(HHC) and healthy controls(HC). METHODS Genetic polymorphisms were analyzed by Restriction Fragment Length Polymorphism(RFLP) in 102-PTB patients, 102-HHC, and 132-HC. QuantiFERON-TB Gold In-Tube test was performed to identify latent TB infection in 60-HHC. Estimated their cytokine levels by ELISA in MDP (muramyl dipeptide) stimulated culture supernatants of all the groups. Further, we studied pre-mRNA structures by insilico analysis and relative gene expression by RT-PCR. RESULTS Recessive genetic models of NOD2 802C > T SNP with TT genotype and AA genotype of NOD2 2105G > A SNP were significantly associated with increased TB risk in PTB patients and HHC compared with HC. In vitro stimulations were performed with NOD2 ligand MDP in PTB patients and latent TB subjects: QuantiFERON positive household contacts (QFT + ve HHC)and QuantiFERON negative household contacts(QFT-ve HHC). The results showed that reduced TNF-α and enhanced IL-12, IL-1β indicate that these cytokines may play an essential role in the initial maintenance of cell-mediated immunity. Our study demonstrated the correlation between NOD2 polymorphism with IL-1β, TNF-α, IL-12 levels. Insilico analysis represents the pre-mRNA secondary structures affected by NOD2 SNPs. We also observed the difference in m RNA levels in variant and wild genotypes. CONCLUSION This finding may lead to the forthcoming development of immunotherapy and may be used as predictive markers to identify high-risk individuals for TB disease.
Collapse
Affiliation(s)
- Jyothi Priya Mandala
- Bhagwan Mahavir Medical Research Centre, Hyderabad, India; Department of Genetics, Osmania University, Hyderabad, India
| | - Shruthi Thada
- Bhagwan Mahavir Medical Research Centre, Hyderabad, India; Institute of Microbiology and Hygiene, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | - Meenakshi Ponnana
- Bhagwan Mahavir Medical Research Centre, Hyderabad, India; Department of Genetics, Osmania University, Hyderabad, India
| | | | - SumanLatha Gaddam
- Bhagwan Mahavir Medical Research Centre, Hyderabad, India; Department of Genetics, Osmania University, Hyderabad, India.
| |
Collapse
|
40
|
Type I Interferons Suppress Anti-parasitic Immunity and Can Be Targeted to Improve Treatment of Visceral Leishmaniasis. Cell Rep 2021; 30:2512-2525.e9. [PMID: 32101732 PMCID: PMC7981274 DOI: 10.1016/j.celrep.2020.01.099] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 11/28/2019] [Accepted: 01/28/2020] [Indexed: 12/13/2022] Open
Abstract
Type I interferons (IFNs) play critical roles in anti-viral and anti-tumor immunity. However, they also suppress protective immune responses in some infectious diseases. Here, we identify type I IFNs as major upstream regulators of CD4+ T cells from visceral leishmaniasis (VL) patients. Furthermore, we report that mice deficient in type I IFN signaling have significantly improved control of Leishmania donovani, a causative agent of human VL, associated with enhanced IFNγ but reduced IL-10 production by parasite-specific CD4+ T cells. Importantly, we identify a small-molecule inhibitor that can be used to block type I IFN signaling during established infection and acts synergistically with conventional anti-parasitic drugs to improve parasite clearance and enhance anti-parasitic CD4+ T cell responses in mice and humans. Thus, manipulation of type I IFN signaling is a promising strategy for improving disease outcome in VL patients.
Collapse
|
41
|
Ravesloot-Chávez MM, Van Dis E, Stanley SA. The Innate Immune Response to Mycobacterium tuberculosis Infection. Annu Rev Immunol 2021; 39:611-637. [PMID: 33637017 DOI: 10.1146/annurev-immunol-093019-010426] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Infection with Mycobacterium tuberculosis causes >1.5 million deaths worldwide annually. Innate immune cells are the first to encounter M. tuberculosis, and their response dictates the course of infection. Dendritic cells (DCs) activate the adaptive response and determine its characteristics. Macrophages are responsible both for exerting cell-intrinsic antimicrobial control and for initiating and maintaining inflammation. The inflammatory response to M. tuberculosis infection is a double-edged sword. While cytokines such as TNF-α and IL-1 are important for protection, either excessive or insufficient cytokine production results in progressive disease. Furthermore, neutrophils-cells normally associated with control of bacterial infection-are emerging as key drivers of a hyperinflammatory response that results in host mortality. The roles of other innate cells, including natural killer cells and innate-like T cells, remain enigmatic. Understanding the nuances of both cell-intrinsic control of infection and regulation of inflammation will be crucial for the successful development of host-targeted therapeutics and vaccines.
Collapse
Affiliation(s)
| | - Erik Van Dis
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, USA; ,
| | - Sarah A Stanley
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, USA; , .,Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, California 94720, USA
| |
Collapse
|
42
|
Mikhalkevich N, O’Carroll IP, Tkavc R, Lund K, Sukumar G, Dalgard CL, Johnson KR, Li W, Wang T, Nath A, Iordanskiy S. Response of human macrophages to gamma radiation is mediated via expression of endogenous retroviruses. PLoS Pathog 2021; 17:e1009305. [PMID: 33556144 PMCID: PMC7895352 DOI: 10.1371/journal.ppat.1009305] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/19/2021] [Accepted: 01/11/2021] [Indexed: 01/11/2023] Open
Abstract
Ionizing radiation-induced tissue damage recruits monocytes into the exposed area where they are differentiated to macrophages. These implement phagocytic removal of dying cells and elicit an acute inflammatory response, but can also facilitate tumorigenesis due to production of anti-inflammatory cytokines. Using primary human monocyte-derived macrophages (MDMs) and the THP1 monocytic cell line, we demonstrate that gamma radiation triggers monocyte differentiation toward the macrophage phenotype with increased expression of type I interferons (IFN-I) and both pro- and anti-inflammatory macrophage activation markers. We found that these changes correlate with significantly upregulated expression of 622 retroelements from various groups, particularly of several clades of human endogenous retroviruses (HERVs). Elevated transcription was detected in both sense and antisense directions in the HERV subgroups tested, including the most genetically homogeneous clade HML-2. The level of antisense transcription was three- to five-fold higher than of the sense strand levels. Using a proximity ligation assay and immunoprecipitation followed by RNA quantification, we identified an increased amount of the dsRNA receptors MDA-5 and TLR3 bound to an equivalent number of copies of sense and antisense chains of HERVK HML-2 RNA. This binding triggered MAVS-associated signaling pathways resulting in increased expression of IFN-I and inflammation related genes that enhanced the cumulative inflammatory effect of radiation-induced senescence. HML-2 knockdown was accompanied with reduced expression and secretion of IFNα, pro-inflammatory (IL-1β, IL-6, CCL2, CCL3, CCL8, and CCL20) and anti-inflammatory (IL10) modulators in irradiated monocytes and MDMs. Taken together, our data indicate that radiation stress-induced HERV expression enhances the IFN-I and cytokine response and results in increased levels of pro-inflammatory modulators along with expression of anti-inflammatory factors associated with the macrophage tumorigenic phenotype. Ionizing radiation is a powerful stressogenic factor that induces massive cell damage. The signals released from radiation-damaged tissues recruit the monocytes, which are differentiated into macrophages that remove dying cells via phagocytosis and facilitate inflammation but can also contribute to tumorigenesis through anti-inflammatory and regenerative activities. The mechanism of this dual response of macrophages to irradiation is not fully understood. Using primary human macrophages and a monocytic cell line, we demonstrated that gamma radiation doses activate expression of various human endogenous retroviruses (HERVs). At the molecular level, we have shown that increased numbers of sense and antisense transcripts of tested HERV subgroups bind to double-stranded RNA receptors inducing the expression of type I interferons, multiple pro-inflammatory and some anti-inflammatory factors. At the phenotypic level, polarized macrophages exhibit a potent inflammatory response along with potentially tumorigenic characteristics. Our data suggest that endogenous retroviruses represent an important contributor of the macrophage-mediated inflammation in response to radiation-induced stress but may also indirectly influence tumorigenesis via biased macrophage polarization.
Collapse
Affiliation(s)
- Natallia Mikhalkevich
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Ina P. O’Carroll
- Department of Chemistry, United States Naval Academy, Annapolis, Maryland, United States of America
| | - Rok Tkavc
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Kateryna Lund
- Biomedical Instrumentation Center, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Gauthaman Sukumar
- The American Genome Center (TAGC), Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Clifton L. Dalgard
- The American Genome Center (TAGC), Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Kory R. Johnson
- Bioinformatics Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Wenxue Li
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Tongguang Wang
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Avindra Nath
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (AN); (SI)
| | - Sergey Iordanskiy
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- * E-mail: (AN); (SI)
| |
Collapse
|
43
|
Kinsella RL, Zhu DX, Harrison GA, Mayer Bridwell AE, Prusa J, Chavez SM, Stallings CL. Perspectives and Advances in the Understanding of Tuberculosis. ANNUAL REVIEW OF PATHOLOGY 2021; 16:377-408. [PMID: 33497258 DOI: 10.1146/annurev-pathol-042120-032916] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), remains a leading cause of death due to infection in humans. To more effectively combat this pandemic, many aspects of TB control must be developed, including better point of care diagnostics, shorter and safer drug regimens, and a protective vaccine. To address all these areas of need, better understanding of the pathogen, host responses, and clinical manifestations of the disease is required. Recently, the application of cutting-edge technologies to the study of Mtb pathogenesis has resulted in significant advances in basic biology, vaccine development, and antibiotic discovery. This leaves us in an exciting era of Mtb research in which our understanding of this deadly infection is improving at a faster rate than ever, and renews hope in our fight to end TB. In this review, we reflect on what is known regarding Mtb pathogenesis, highlighting recent breakthroughs that will provide leverage for the next leaps forward in the field.
Collapse
Affiliation(s)
- Rachel L Kinsella
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA;
| | - Dennis X Zhu
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA;
| | - Gregory A Harrison
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA;
| | - Anne E Mayer Bridwell
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA;
| | - Jerome Prusa
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA;
| | - Sthefany M Chavez
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA;
| | - Christina L Stallings
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA;
| |
Collapse
|
44
|
Fox LE, Locke MC, Lenschow DJ. Context Is Key: Delineating the Unique Functions of IFNα and IFNβ in Disease. Front Immunol 2020; 11:606874. [PMID: 33408718 PMCID: PMC7779635 DOI: 10.3389/fimmu.2020.606874] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022] Open
Abstract
Type I interferons (IFNs) are critical effector cytokines of the immune system and were originally known for their important role in protecting against viral infections; however, they have more recently been shown to play protective or detrimental roles in many disease states. Type I IFNs consist of IFNα, IFNβ, IFNϵ, IFNκ, IFNω, and a few others, and they all signal through a shared receptor to exert a wide range of biological activities, including antiviral, antiproliferative, proapoptotic, and immunomodulatory effects. Though the individual type I IFN subtypes possess overlapping functions, there is growing appreciation that they also have unique properties. In this review, we summarize some of the mechanisms underlying differential expression of and signaling by type I IFNs, and we discuss examples of differential functions of IFNα and IFNβ in models of infectious disease, cancer, and autoimmunity.
Collapse
Affiliation(s)
- Lindsey E. Fox
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States
| | - Marissa C. Locke
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States
| | - Deborah J. Lenschow
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| |
Collapse
|
45
|
Mary A, Hénaut L, Macq PY, Badoux L, Cappe A, Porée T, Eckes M, Dupont H, Brazier M. Rationale for COVID-19 Treatment by Nebulized Interferon-β-1b-Literature Review and Personal Preliminary Experience. Front Pharmacol 2020; 11:592543. [PMID: 33329000 PMCID: PMC7734101 DOI: 10.3389/fphar.2020.592543] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022] Open
Abstract
The inflammatory response to COVID-19 is specifically associated with an impaired type I interferon (IFN) response and complete blockade of IFN-β secretion. Clinically, nebulization of IFN-α-2b has been historically used in China to treat viral pneumonia associated with SARS-CoV. Very recent data show that the use of inhaled type I IFN is associated with decreased mortality in Chinese COVID-19 patients. However, IFN nebulization is currently not standard in Europe and the United States. Therefore, our group has set up a project aimed to evaluate the possibility to nebulize IFN-β-1b (a drug currently used in Europe to treat multiple sclerosis via subcutaneous injections) and to assess the safety of this new mode of administration in SARS-CoV-2 infected patients. We present here literature data that allowed us to build our hypothesis and to develop collaboration between clinical pharmacists, intensivists and nebulization engineers in order to gain first pre-clinical and clinical experience of IFN-β-1b nebulization. After validation of the nebulization method and verification of droplet size compatible with nebulization, the method has been applied to four intensive care patients treated at our university hospital, for whom none of the COVID-19 therapies initially used in France led to significant clinical improvement. All patients exhibited negative viral carriage and experienced clinical improvement 7-16 days after having initiated nebulized IFN-β-1b inhalation therapy. No side effects were observed. All patients were alive within a 90-days follow-up. Although it is not possible to draw firm conclusions on treatment efficacy based on this case report, our study shows that pulmonary IFN-β-1b administration is feasible, with a good safety profile. This procedure, which presents the advantage of directly targeting the lungs and reducing the risks of systemic side effects, may represent a promising therapeutic strategy for the care of patients with severe COVID-19. However, our preliminary observation requires confirmation by randomized controlled trials.
Collapse
Affiliation(s)
- Aurélien Mary
- Clinical Critical Care Pharmacy Department, Amiens-Picardie University Hospital, Amiens, France
- UR UPJV 7517, MP3CV, CURS, University of Picardie Jules Verne, Amiens, France
| | - Lucie Hénaut
- UR UPJV 7517, MP3CV, CURS, University of Picardie Jules Verne, Amiens, France
| | - Pierre Yves Macq
- Surgical Critical Care Department, Amiens-Picardie University Hospital, Amiens, France
| | - Louise Badoux
- Surgical Critical Care Department, Amiens-Picardie University Hospital, Amiens, France
| | - Arnaud Cappe
- Clinical Critical Care Pharmacy Department, Amiens-Picardie University Hospital, Amiens, France
| | | | | | - Hervé Dupont
- UR UPJV 7517, MP3CV, CURS, University of Picardie Jules Verne, Amiens, France
- Surgical Critical Care Department, Amiens-Picardie University Hospital, Amiens, France
| | - Michel Brazier
- UR UPJV 7517, MP3CV, CURS, University of Picardie Jules Verne, Amiens, France
- Department of Biochemistry, Amiens-Picardie University Hospital, Amiens, France
| |
Collapse
|
46
|
Akhade AS, Atif SM, Lakshmi BS, Dikshit N, Hughes KT, Qadri A, Subramanian N. Type 1 interferon-dependent repression of NLRC4 and iPLA2 licenses down-regulation of Salmonella flagellin inside macrophages. Proc Natl Acad Sci U S A 2020; 117:29811-29822. [PMID: 33177235 PMCID: PMC7703570 DOI: 10.1073/pnas.2002747117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Inflammasomes have been implicated in the detection and clearance of a variety of bacterial pathogens, but little is known about whether this innate sensing mechanism has any regulatory effect on the expression of stimulatory ligands by the pathogen. During infection with Salmonella and many other pathogens, flagellin is a major activator of NLRC4 inflammasome-mediated macrophage pyroptosis and pathogen eradication. Salmonella switches to a flagellin-low phenotype as infection progresses to avoid this mechanism of clearance by the host. However, the host cues that Salmonella perceives to undergo this switch remain unclear. Here, we report an unexpected role of the NLRC4 inflammasome in promoting expression of its microbial ligand, flagellin, and identify a role for type 1 IFN signaling in switching of Salmonella to a flagellin-low phenotype. Early in infection, activation of NLRC4 by flagellin initiates pyroptosis and concomitant release of lysophospholipids which in turn enhance expression of flagellin by Salmonella thereby amplifying its ability to elicit cell death. TRIF-dependent production of type 1 IFN, however, later represses NLRC4 and the lysophospholipid biosynthetic enzyme iPLA2, causing a decline in intracellular lysophospholipids that results in down-regulation of flagellin expression by Salmonella These findings reveal a previously unrecognized immune-modulating regulatory cross-talk between endosomal TLR signaling and cytosolic NLR activation with significant implications for the establishment of infection with Salmonella.
Collapse
Affiliation(s)
| | - Shaikh M Atif
- Hybridoma Laboratory, National Institute of Immunology, 110067 New Delhi, India
| | | | - Neha Dikshit
- Hybridoma Laboratory, National Institute of Immunology, 110067 New Delhi, India
| | - Kelly T Hughes
- Department of Biology, University of Utah, Salt Lake City, UT 84112
| | - Ayub Qadri
- Hybridoma Laboratory, National Institute of Immunology, 110067 New Delhi, India;
| | - Naeha Subramanian
- Institute for Systems Biology, Seattle, WA 98109;
- Department of Immunology, University of Washington, Seattle, WA 98109
- Department of Global Health, University of Washington, Seattle, WA 98109
| |
Collapse
|
47
|
Type I IFN exacerbates disease in tuberculosis-susceptible mice by inducing neutrophil-mediated lung inflammation and NETosis. Nat Commun 2020; 11:5566. [PMID: 33149141 PMCID: PMC7643080 DOI: 10.1038/s41467-020-19412-6] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Tuberculosis (TB) is a leading cause of mortality due to infectious disease, but the factors determining disease progression are unclear. Transcriptional signatures associated with type I IFN signalling and neutrophilic inflammation were shown to correlate with disease severity in mouse models of TB. Here we show that similar transcriptional signatures correlate with increased bacterial loads and exacerbate pathology during Mycobacterium tuberculosis infection upon GM-CSF blockade. Loss of GM-CSF signalling or genetic susceptibility to TB (C3HeB/FeJ mice) result in type I IFN-induced neutrophil extracellular trap (NET) formation that promotes bacterial growth and promotes disease severity. Consistently, NETs are present in necrotic lung lesions of TB patients responding poorly to antibiotic therapy, supporting the role of NETs in a late stage of TB pathogenesis. Our findings reveal an important cytokine-based innate immune effector network with a central role in determining the outcome of M. tuberculosis infection. GM-CSF is involved in control over M. tuberculosis infection. Here the authors show that GM-CSF reduces type 1 interferon driven neutrophil recruitment, NETosis and bacterial growth in the lungs of infected mice, and provide evidence that this NETosis occurs in infected humans who are not responsive to antibiotic therapy.
Collapse
|
48
|
Nguyen H, Gazy N, Venketaraman V. A Role of Intracellular Toll-Like Receptors (3, 7, and 9) in Response to Mycobacterium tuberculosis and Co-Infection with HIV. Int J Mol Sci 2020; 21:E6148. [PMID: 32858917 PMCID: PMC7503332 DOI: 10.3390/ijms21176148] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) is a highly infectious acid-fast bacillus and is known to cause tuberculosis (TB) in humans. It is a leading cause of death from a sole infectious agent, with an estimated 1.5 million deaths yearly worldwide, and up to one third of the world's population has been infected with TB. The virulence and susceptibility of Mtb are further amplified in the presence of Human Immunodeficiency Virus (HIV). Coinfection with Mtb and HIV forms a lethal combination. Previous studies had demonstrated the synergistic effects of Mtb and HIV, with one disease accelerating the disease progression of the other through multiple mechanisms, including the modulation of the immune response to these two pathogens. The response of the endosomal pattern recognition receptors to these two pathogens, specifically toll-like receptors (TLR)-3, -7, and -9, has not been elucidated, with some studies producing mixed results. This article seeks to review the roles of TLR-3, -7, and -9 in response to Mtb infection, as well as Mtb-HIV-coinfection via Toll-interleukin 1 receptor (TIR) domain-containing adaptor inducing INF-β (TRIF)-dependent and myeloid differentiation factor 88 (MyD88)-dependent pathways.
Collapse
Affiliation(s)
- Huy Nguyen
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| | - Nicky Gazy
- Beaumont Health System, 5450 Fort St, Trenton, MI 48183, USA
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| |
Collapse
|
49
|
LPS restores protective immunity in macrophages against Mycobacterium tuberculosis via autophagy. Mol Immunol 2020; 124:18-24. [DOI: 10.1016/j.molimm.2020.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 04/24/2020] [Accepted: 05/03/2020] [Indexed: 12/12/2022]
|
50
|
Taneja V, Kalra P, Goel M, Khilnani GC, Saini V, Prasad GBKS, Gupta UD, Krishna Prasad H. Impact and prognosis of the expression of IFN-α among tuberculosis patients. PLoS One 2020; 15:e0235488. [PMID: 32667932 PMCID: PMC7363073 DOI: 10.1371/journal.pone.0235488] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 06/16/2020] [Indexed: 12/03/2022] Open
Abstract
Mycobacterium tuberculosis (M.tb) infection stimulates the release of cytokines, including interferons (IFNs). IFNs are initiators, regulators, and effectors of innate and adaptive immunity. Accordingly, the expression levels of Type I (α, β) and II (γ) IFNs, among untreated tuberculosis (TB) patients and household contacts (HHC) clinically free of TB was assessed. A total of 264 individuals (TB patients-123; HHC-86; laboratory volunteers-55; Treated TB patients-36) were enrolled for this study. IFN-α mRNA expression levels predominated compared to IFN-γ and IFN-β among untreated TB patients. IFN-α transcripts were ~3.5 folds higher in TB patients compared to HHC, (p<0.0001). High expression of IFN-α was seen among 46% (56/ 123) of the TB patients and 26%, (22/86) of HHCs. The expression levels of IFN-α correlated with that of IFN transcriptional release factor 7 (IRF) (p<0.0001). In contrast, an inverse relationship exists between PGE2 and IFN-α expression levels; high IFN-α expressers were associated with low levels of PGE2 and vice-versa (Spearman’s rho = -0.563; p<0.0001). In-vitro, IFN-α failed to restrict the replication of intracellular M.tb. The anti-mycobacterial activity of IFN-γ was compromised in the presence of IFN-α, but not by IFN-β. The expression of IFN-α and β diminished or is absent, among successfully treated TB patients. These observations suggest the utility of assessment of Type I IFNs expression levels as a prognostic marker to monitor tuberculosis patient response to chemotherapy because changes in Type I IFNs expression are expected to precede the clearance and /reduction in bacterial load.
Collapse
Affiliation(s)
- Vibha Taneja
- National JALMA Institute of Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, India
- Department of Biochemistry, Jiwaji University, Gwalior, Madhya Pradesh, India
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Priya Kalra
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Manish Goel
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Gopi Chand Khilnani
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Vikram Saini
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - G. B. K. S. Prasad
- Department of Biochemistry, Jiwaji University, Gwalior, Madhya Pradesh, India
| | - Umesh Datta Gupta
- National JALMA Institute of Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, India
| | | |
Collapse
|