1
|
Al-Janabi H, Moyes K, Allen R, Fisher M, Crespo M, Gurel B, Rescigno P, de Bono J, Nunns H, Bailey C, Junker-Jensen A, Muthana M, Phillips WA, Pearson HB, Taplin ME, Brown JE, Lewis CE. Targeting a STING agonist to perivascular macrophages in prostate tumors delays resistance to androgen deprivation therapy. J Immunother Cancer 2024; 12:e009368. [PMID: 39060021 DOI: 10.1136/jitc-2024-009368] [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] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Androgen deprivation therapy (ADT) is a front-line treatment for prostate cancer. In some men, their tumors can become refractory leading to the development of castration-resistant prostate cancer (CRPC). This causes tumors to regrow and metastasize, despite ongoing treatment, and impacts negatively on patient survival. ADT is known to stimulate the accumulation of immunosuppressive cells like protumoral tumor-associated macrophages (TAMs), myeloid-derived suppressor cells and regulatory T cells in prostate tumors, as well as hypofunctional T cells. Protumoral TAMs have been shown to accumulate around tumor blood vessels during chemotherapy and radiotherapy in other forms of cancer, where they drive tumor relapse. Our aim was to see whether such perivascular (PV) TAMs also accumulate in ADT-treated prostate tumors prior to CRPC, and, if so, whether selectively inducing them to express a potent immunostimulant, interferon beta (IFNβ), would stimulate antitumor immunity and delay CRPC. METHODS We used multiplex immunofluorescence to assess the effects of ADT on the distribution and activation status of TAMs, CD8+T cells, CD4+T cells and NK cells in mouse and/or human prostate tumors. We then used antibody-coated, lipid nanoparticles (LNPs) to selectively target a STING agonist, 2'3'-cGAMP (cGAMP), to PV TAMs in mouse prostate tumors during ADT. RESULTS TAMs accumulated at high density around blood vessels in response to ADT and expressed markers of a protumoral phenotype including folate receptor-beta (FR-β), MRC1 (CD206), CD169 and VISTA. Additionally, higher numbers of inactive (PD-1-) CD8+T cells and reduced numbers of active (CD69+) NK cells were present in these PV tumor areas. LNPs coated with an antibody to FR-β selectively delivered cGAMP to PV TAMs in ADT-treated tumors, where they activated STING and upregulated the expression of IFNβ. This resulted in a marked increase in the density of active CD8+T cells (along with CD4+T cells and NK cells) in PV tumor areas, and significantly delayed the onset of CRPC. Antibody depletion of CD8+T cells during LNP administration demonstrated the essential role of these cells in delay in CRPC induced by LNPs. CONCLUSION Together, our data indicate that targeting a STING agonist to PV TAMs could be used to extend the treatment window for ADT in prostate cancer.
Collapse
Affiliation(s)
- Haider Al-Janabi
- Divsion of Clinical Medicine, The University of Sheffield, Sheffield, UK
| | - Katy Moyes
- Divsion of Clinical Medicine, The University of Sheffield, Sheffield, UK
| | - Richard Allen
- Divsion of Clinical Medicine, The University of Sheffield, Sheffield, UK
| | - Matthew Fisher
- Divsion of Clinical Medicine, The University of Sheffield, Sheffield, UK
| | | | - Bora Gurel
- The Institute of Cancer Research, London, UK
| | - Pasquale Rescigno
- The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, The Institute of Cancer Research, London, UK
| | | | - Harry Nunns
- NeoGenomics Laboratories Inc Aliso Viejo, Aliso Viejo, California, USA
| | | | | | - Munitta Muthana
- Division of Clinical Medicine, The University of Sheffield, Sheffield, UK
| | | | | | | | - Janet E Brown
- Division of Clinical Medicine, The University of Sheffield, Sheffield, UK
| | - Claire E Lewis
- Divsion of Clinical Medicine, The University of Sheffield, Sheffield, UK
| |
Collapse
|
2
|
Hou Y, Yang K, Wang L, Wang J, Huang X, Piffko A, Luo SZ, Yu X, Rao E, Martinez C, Bugno J, Mack M, Vokes EE, Pitroda SP, Chmura SJ, Weichselbaum RR, Liang HL. Radiotherapy Enhances Metastasis Through Immune Suppression by Inducing PD-L1 and MDSC in Distal Sites. Clin Cancer Res 2024; 30:1945-1958. [PMID: 38427437 PMCID: PMC11062826 DOI: 10.1158/1078-0432.ccr-23-3206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/22/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024]
Abstract
PURPOSE Radiotherapy (RT) is a widely employed anticancer treatment. Emerging evidence suggests that RT can elicit both tumor-inhibiting and tumor-promoting immune effects. The purpose of this study is to investigate immune suppressive factors of radiotherapy. EXPERIMENTAL DESIGN We used a heterologous two-tumor model in which adaptive concomitant immunity was eliminated. RESULTS Through analysis of PD-L1 expression and myeloid-derived suppressor cells (MDSC) frequencies using patient peripheral blood mononuclear cells and murine two-tumor and metastasis models, we report that local irradiation can induce a systemic increase in MDSC, as well as PD-L1 expression on dendritic cells and myeloid cells, and thereby increase the potential for metastatic dissemination in distal, nonirradiated tissue. In a mouse model using two distinct tumors, we found that PD-L1 induction by ionizing radiation was dependent on elevated chemokine CXCL10 signaling. Inhibiting PD-L1 or MDSC can potentially abrogate RT-induced metastasis and improve clinical outcomes for patients receiving RT. CONCLUSIONS Blockade of PD-L1/CXCL10 axis or MDSC infiltration during irradiation can enhance abscopal tumor control and reduce metastasis.
Collapse
Affiliation(s)
- Yuzhu Hou
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University; Xi’an, ShaanXi 710061, China
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Kaiting Yang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Liangliang Wang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Jiaai Wang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Xiaona Huang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Andras Piffko
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Sean Z. Luo
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
- Current address: Biomedical Engineering program, Northwestern University; Evanston, IL 60201, USA
| | - Xinshuang Yu
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
- Current address: Department of Oncology, First Affiliated Hospital of Shandong, First Medical University and Shandong Provincial Qianfoshan Hospital; Jinan, Shandong 250014, China
| | - Enyu Rao
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
- Current address: Cancer Institute, Xuzhou Medical University; Xuzhou, Jiangsu 221004, China
| | - Carlos Martinez
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
- Current address: University of Illinois at Chicago, Chicago, IL, 60607 USA
| | - Jason Bugno
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
- The Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, IL 600637, USA
| | - Matthias Mack
- Department of Nephrology, University Hospital Regensburg, 93042 Regensburg, Germany
| | - Everett E. Vokes
- Department of Medicine, University of Chicago, Chicago, IL, 60637 USA
| | - Sean P. Pitroda
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Steven J. Chmura
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
| | - Ralph R. Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Hua Laura Liang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| |
Collapse
|
3
|
Chen X, Qi D, Fan S, He Y, Jing H, Wang D. Interferon regulatory factor 1 (IRF1) inhibits lung endothelial regeneration following inflammation-induced acute lung injury. Clin Sci (Lond) 2023; 137:367-383. [PMID: 36857175 PMCID: PMC10011169 DOI: 10.1042/cs20220876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/02/2023]
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a respiratory condition caused by severe endothelial barrier dysfunction within the lung. In ARDS, excessive inflammation, tissue edema, and immune cell influx prevents endothelial cell regeneration that is crucial in repairing the endothelial barrier. However, little is known about the molecular mechanism that underpin endothelial cell regeneration in ARDS. METHODS R-based bioinformatics tools were used to analyze microarray-derived transcription profiles in human lung microvascular endothelial cells (HLMVECs) subjected to non-treatment or lipopolysaccharide (LPS) exposure. We generated endothelial cell-specific interferon regulatory factor 1 (Irf1) knockout (Irf1EC-/-) and Irf1fl/fl control mice for use in an endotoxemic murine model of acute lung injury (ALI). In vitro studies (qPCR, immunoblotting, and ChIP-qPCR) were conducted in mouse lung endothelial cells (MLECs) and HLMVECs. Dual-luciferase promoter reporter assays were performed in HLMVECs. RESULTS Bioinformatics analyses identified IRF1 as a key up-regulated gene in HLMVECs post-LPS exposure. Endothelial-specific knockout of Irf1 in ALI mice resulted in enhanced regeneration of lung endothelium, while liposomal delivery of endothelial-specific Irf1 to wild-type ALI mice inhibited lung endothelial regeneration in a leukemia inhibitory factor (Lif)-dependent manner. Mechanistically, we demonstrated that LPS-induced Stat1Ser727 phosphorylation promotes Irf1 transactivation, resulting in downstream up-regulation of Lif that inhibits endothelial cell proliferation. CONCLUSIONS These results demonstrate the existence of a p-Stat1Ser727-Irf1-Lif axis that inhibits lung endothelial cell regeneration post-LPS injury. Thus, direct inhibition of IRF1 or LIF may be a promising strategy for enhancing endothelial cell regeneration and improving clinical outcomes in ARDS patients.
Collapse
Affiliation(s)
- Xiaorui Chen
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Correspondence: Xiaorui Chen () or Daoxin Wang ()
| | - Di Qi
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Shulei Fan
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yirui He
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hekun Jing
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Daoxin Wang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Correspondence: Xiaorui Chen () or Daoxin Wang ()
| |
Collapse
|
4
|
Kula A, Makuch E, Lisowska M, Reniewicz P, Lipiński T, Siednienko J. Pellino3 ligase negatively regulates influenza B dependent RIG-I signalling through downregulation of TRAF3-mediated induction of the transcription factor IRF3 and IFNβ production. Immunology 2023. [PMID: 36861386 DOI: 10.1111/imm.13637] [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: 10/06/2022] [Accepted: 02/19/2023] [Indexed: 03/03/2023] Open
Abstract
Viral infection activates the innate immune system, which recognizes viral components by a variety of pattern recognition receptors and initiates signalling cascades leading to the production of pro-inflammatory cytokines. To date, signalling cascades triggered after virus recognition are not fully characterized and are investigated by many research groups. The critical role of the E3 ubiquitin ligase Pellino3 in antibacterial and antiviral response is now widely accepted, but the precise mechanism remains elusive. In this study, we sought to explore Pellino3 role in the retinoic acid-inducible gene I (RIG-I)-dependent signalling pathway. In this work, the molecular mechanisms of the innate immune response, regulated by Pellino3, were investigated in lung epithelial cells during influenza B virus infection. We used wild-type and Pellino3-deficient A549 cells as model cell lines to examine the role of Pellino3 ligase in the type I interferon (IFN) signalling pathway. Our results indicate that Pellino3 is involved in direct ubiquitination and degradation of the TRAF3, suppressing interferon regulatory factor 3 (IRF3) activation and interferon beta (IFNβ) production.
Collapse
Affiliation(s)
- Anna Kula
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland.,Laboratory of Medical Microbiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Edyta Makuch
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Marta Lisowska
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Patryk Reniewicz
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Tomasz Lipiński
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Jakub Siednienko
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| |
Collapse
|
5
|
IFNβ-Induced CXCL10 Chemokine Expression Is Regulated by Pellino3 Ligase in Monocytes and Macrophages. Int J Mol Sci 2022; 23:ijms232314915. [PMID: 36499241 PMCID: PMC9741470 DOI: 10.3390/ijms232314915] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/17/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
IFN-I is the key regulatory component activating and modulating the response of innate and adaptive immune system to bacterial as well as viral pathogens. IFN-I promotes the expression of IFN-induced genes (ISG) and, consequently, the production of chemokines, e.g., CXCL10. Those chemokines control migration and localization of immune cells in tissues, and, thus, are critical to the function of the innate immune system during infection. Consequently, the regulation of IFN-I signaling is essential for the proper induction of an immune response. Our previous study has shown that E3 ubiquitin ligase Pellino3 positively regulates IFNβ expression and secretion. Herein, we examined the role of Pellino3 ligase in regulating CXCL10 expression in response to IFNβ stimulation. Our experiments were carried out on murine macrophage cell line (BMDM) and human monocytes cell line (THP-1) using IFNβ as a IFNAR ligand. We demonstrate that Pellino3 is important for IFNβ-induced phosphorylation and nuclear translocation of STAT1/STAT2/IRF9 complex which interacts with CXCL10 promoter and enhances its expression. In this study, we characterize a novel molecular mechanism allowing Pellino3-dependent modulation of the IFNβ-induced response in BMDM and THP-1 cell lines.
Collapse
|
6
|
Alum-anchored intratumoral retention improves the tolerability and antitumor efficacy of type I interferon therapies. Proc Natl Acad Sci U S A 2022; 119:e2205983119. [PMID: 36037341 PMCID: PMC9457244 DOI: 10.1073/pnas.2205983119] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Effective antitumor immunity in mice requires activation of the type I interferon (IFN) response pathway. IFNα and IFNβ therapies have proven promising in humans, but suffer from limited efficacy and high toxicity. Intratumoral IFN retention ameliorates systemic toxicity, but given the complexity of IFN signaling, it was unclear whether long-term intratumoral retention of type I IFNs would promote or inhibit antitumor responses. To this end, we compared the efficacy of IFNα and IFNβ that exhibit either brief or sustained retention after intratumoral injection in syngeneic mouse tumor models. Significant enhancement in tumor retention, mediated by anchoring these IFNs to coinjected aluminum-hydroxide (alum) particles, greatly improved both their tolerability and efficacy. The improved efficacy of alum-anchored IFNs could be attributed to sustained pleiotropic effects on tumor cells, immune cells, and nonhematopoietic cells. Alum-anchored IFNs achieved high cure rates of B16F10 tumors upon combination with either anti-PD-1 antibody or interleukin-2. Interestingly however, these alternative combination immunotherapies yielded disparate T cell phenotypes and differential resistance to tumor rechallenge, highlighting important distinctions in adaptive memory formation for combinations of type I IFNs with other immunotherapies.
Collapse
|
7
|
Semple SL, Au SKW, Jacob RA, Mossman KL, DeWitte-Orr SJ. Discovery and Use of Long dsRNA Mediated RNA Interference to Stimulate Antiviral Protection in Interferon Competent Mammalian Cells. Front Immunol 2022; 13:859749. [PMID: 35603190 PMCID: PMC9120774 DOI: 10.3389/fimmu.2022.859749] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/04/2022] [Indexed: 12/20/2022] Open
Abstract
In invertebrate cells, RNA interference (RNAi) acts as a powerful immune defense that stimulates viral gene knockdown thereby preventing infection. With this pathway, virally produced long dsRNA (dsRNA) is cleaved into short interfering RNA (siRNA) by Dicer and loaded into the RNA-induced silencing complex (RISC) which can then destroy/disrupt complementary viral mRNA sequences. Comparatively, in mammalian cells it is believed that the type I interferon (IFN) pathway is the cornerstone of the innate antiviral response. In these cells, dsRNA acts as a potent inducer of the IFN system, which is dependent on dsRNA length, but not sequence, to stimulate an antiviral state. Although the cellular machinery for RNAi is intact and functioning in mammalian cells, its role to trigger an antiviral response using long dsRNA (dsRNAi) remains controversial. Here we show that dsRNAi is not only functional but has a significant antiviral effect in IFN competent mammalian cells. We found that pre-soaking mammalian cells with concentrations of sequence specific dsRNA too low to induce IFN production could significantly inhibit vesicular stomatitis virus expressing green fluorescent protein (VSV-GFP), and the human coronaviruses (CoV) HCoV-229E and SARS-CoV-2 replication. This phenomenon was shown to be dependent on dsRNA length, was comparable in effect to transfected siRNAs, and could knockdown multiple sequences at once. Additionally, knockout cell lines revealed that functional Dicer was required for viral inhibition, revealing that the RNAi pathway was indeed responsible. These results provide the first evidence that soaking with gene-specific long dsRNA can generate viral knockdown in mammalian cells. We believe that this novel discovery provides an explanation as to why the mammalian lineage retained its RNAi machinery and why vertebrate viruses have evolved methods to suppress RNAi. Furthermore, demonstrating RNAi below the threshold of IFN induction has uses as a novel therapeutic platform, both antiviral and gene targeting in nature.
Collapse
Affiliation(s)
- Shawna L. Semple
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada
| | - Sarah K. W. Au
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada
| | - Rajesh A. Jacob
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Karen L. Mossman
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Stephanie J. DeWitte-Orr
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada
- *Correspondence: Stephanie J. DeWitte-Orr,
| |
Collapse
|
8
|
Steiner A, Hrovat-Schaale K, Prigione I, Yu CH, Laohamonthonkul P, Harapas CR, Low RRJ, De Nardo D, Dagley LF, Mlodzianoski MJ, Rogers KL, Zillinger T, Hartmann G, Gantier MP, Gattorno M, Geyer M, Volpi S, Davidson S, Masters SL. Deficiency in coatomer complex I causes aberrant activation of STING signalling. Nat Commun 2022; 13:2321. [PMID: 35484149 PMCID: PMC9051092 DOI: 10.1038/s41467-022-29946-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 04/05/2022] [Indexed: 12/15/2022] Open
Abstract
Coatomer complex I (COPI) mediates retrograde vesicular trafficking from Golgi to the endoplasmic reticulum (ER) and within Golgi compartments. Deficiency in subunit alpha causes COPA syndrome and is associated with type I IFN signalling, although the upstream innate immune sensor involved was unknown. Using in vitro models we find aberrant activation of the STING pathway due to deficient retrograde but probably not intra-Golgi transport. Further we find the upstream cytosolic DNA sensor cGAS as essentially required to drive type I IFN signalling. Genetic deletion of COPI subunits COPG1 or COPD similarly induces type I IFN activation in vitro, which suggests that inflammatory diseases associated with mutations in other COPI subunit genes may exist. Finally, we demonstrate that inflammation in COPA syndrome patient peripheral blood mononuclear cells and COPI-deficient cell lines is ameliorated by treatment with the small molecule STING inhibitor H-151, suggesting targeted inhibition of the cGAS/STING pathway as a promising therapeutic approach.
Collapse
Affiliation(s)
- Annemarie Steiner
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
- Institute of Structural Biology, University Hospital Bonn, 53127, Bonn, Germany
| | - Katja Hrovat-Schaale
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Ignazia Prigione
- Centre for Autoinflammatory Diseases and Primary Immunodeficiencies, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
| | - Chien-Hsiung Yu
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Pawat Laohamonthonkul
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Cassandra R Harapas
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Ronnie Ren Jie Low
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Dominic De Nardo
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3168, Australia
| | - Laura F Dagley
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
- Advanced Technology and Biology, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Michael J Mlodzianoski
- Center for Dynamic Imaging, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Kelly L Rogers
- Center for Dynamic Imaging, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Thomas Zillinger
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
- Institute of Immunology, Philipps-University Marburg, BMFZ, 35043, Marburg, Germany
| | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, 53127, Bonn, Germany
| | - Michael P Gantier
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, 3168, Australia
| | - Marco Gattorno
- Centre for Autoinflammatory Diseases and Primary Immunodeficiencies, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
| | - Matthias Geyer
- Institute of Structural Biology, University Hospital Bonn, 53127, Bonn, Germany
| | - Stefano Volpi
- Centre for Autoinflammatory Diseases and Primary Immunodeficiencies, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
- University of Genoa, 16126, Genoa, Italy
| | - Sophia Davidson
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Seth L Masters
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
9
|
High Expression of Interferon Pathway Genes CXCL10 and STAT2 Is Associated with Activated T-Cell Signature and Better Outcome of Oral Cancer Patients. J Pers Med 2022; 12:jpm12020140. [PMID: 35207629 PMCID: PMC8877377 DOI: 10.3390/jpm12020140] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 02/05/2023] Open
Abstract
To improve the survival rate of cancer patients, biomarkers for both early diagnosis and patient stratification for appropriate therapeutics play crucial roles in precision oncology. Investigation of altered gene expression and the relevant molecular pathways in cancer cells are helpful for discovering such biomarkers. In this study, we explore the potential prognostic biomarkers for oral cancer patients through systematically analyzing five oral cancer transcriptomic data sets (TCGA, GSE23558, GSE30784, GSE37991, and GSE138206). Gene Set Enrichment Analysis (GSEA) was individually applied to each data set and the upregulated Hallmark molecular pathways of each data set were intersected to generate 13 common pathways including interferon-α/γ pathways. Among the 5 oral cancer data sets, 43 interferon pathway genes were commonly upregulated and 17 genes exhibited prognostic values in TCGA cohort. After validating in another oral cancer cohort (GSE65858), high expressions of C-X-C motif chemokine ligand 10 (CXCL10) and Signal transducer and activator of transcription 2 (STAT2) were confirmed to be good prognostic biomarkers. GSEA of oral cancers stratified by CXCL10/STAT2 expression showed that activation of T-cell pathways and increased tumor infiltration scores of Type 1 T helper (Th1) and CD8+ T cells were associated with high CXCL10/STAT2 expression. These results suggest that high CXCL10/STAT2 expression can predict a favorable outcome in oral cancer patients.
Collapse
|
10
|
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
|
11
|
Chen J, Ye C, Yang Z, Wang T, Xu B, Li P, Zhang S, Xue X. Study on the Effect of Macrophages on Vascular Endothelium in Mice With Different TCM Syndromes of Dyslipidemia and its Biological Basis Based on RNA-Seq Technology. Front Pharmacol 2021; 12:665635. [PMID: 34512320 PMCID: PMC8427158 DOI: 10.3389/fphar.2021.665635] [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: 02/08/2021] [Accepted: 08/12/2021] [Indexed: 12/12/2022] Open
Abstract
Background: “Treating the same disease with different methods” is a Traditional Chinese medicine (TCM) therapeutic concept suggesting that, while patients may be diagnosed with the same disease, they may also have different syndromes that require distinct drug administrations. Objective: This study aimed to identify the differentially expressed genes and related biological processes in dyslipidemia in relation to phlegm–dampness retention (PDR) syndrome and spleen and kidney Yang deficiency (SKYD) syndrome using transcriptomic analysis. Methods: Ten ApoE−/− mice were used for the establishment of dyslipidemic disease–syndrome models via multifactor-hybrid modeling, with five in the PDR group and five in the SKYD group. Additionally, five C57BL/6J mice were employed as a normal control group. Test model-quality aortic endothelial macrophages in mice were screened using flow cytometry. Transcriptomic analysis was performed for macrophages using RNA-Seq. Results: A quality assessment of the disease–syndrome model showed that levels of lipids significantly increased in the PDR and SKYD groups, compared to the normal control group, p < 0.05. Applying, in addition, hematoxylin and eosin staining of aorta, the disease model was also successfully established. A quality assessment of the syndrome models showed that mice in the PDR group presented with typical manifestations of PDR syndrome, and mice in the SKYD group had related manifestations of SKYD syndrome, indicating that the syndrome models were successfully constructed as well. After comparing the differentially expressed gene expressions in macrophages of the dyslipidemic mice with different syndromes, 4,142 genes were identified with statistical significance, p < 0.05. Gene ontology analysis for the differentially expressed genes showed that the biological process of difference between the PDR group and the SKYD group included both adverse and protective processes. Conclusion: The differentially expressed genes between PDR syndrome and SKYD syndrome indicate different biological mechanisms between the onsets of the two syndromes. They have distinctive biological processes, including adverse and protective processes that correspond to the invasion of pathogenic factors into the body and the fight of healthy Qi against pathogenic factors, respectively, according to TCM theory. Our results provide biological evidence for the TCM principle of “treating the same disease with different treatments.”
Collapse
Affiliation(s)
- Jing Chen
- Preventive Treatment of Disease Department, The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Chao Ye
- Orthopedics Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zheng Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tieshan Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Bing Xu
- Traditional Chinese Medicine Department, Tibetology Research Center of Beijing Tibetan Medicine Hospital, Beijing, China
| | - Pengyang Li
- Orthopedics Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shan Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaolin Xue
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
12
|
Wu J, Zhu K, Luo X, Han Y, Zhang B, Wang Z, Dong S, Zou X, Chen X, Liu H, Wu T, Zheng Z, Xie Y, Zhao J, Liu Y, Wen Z, Liu D, Wang Y, Zheng S, Huang X, Jing C, Yang G. PM2.5 promotes replication of VSV by ubiquitination degradation of phospho-IRF3 in A549 cells. Toxicol In Vitro 2020; 62:104698. [DOI: 10.1016/j.tiv.2019.104698] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 09/25/2019] [Accepted: 10/22/2019] [Indexed: 01/10/2023]
|
13
|
Albakri MM, Veliz FA, Fiering SN, Steinmetz NF, Sieg SF. Endosomal toll-like receptors play a key role in activation of primary human monocytes by cowpea mosaic virus. Immunology 2020; 159:183-192. [PMID: 31630392 PMCID: PMC6954739 DOI: 10.1111/imm.13135] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/20/2019] [Accepted: 10/14/2019] [Indexed: 12/30/2022] Open
Abstract
The plant virus, cowpea mosaic virus (CPMV), has demonstrated a remarkable capacity to induce anti-tumour immune responses following direct administration into solid tumours. The molecular pathways that account for these effects and the capacity of CPMV to activate human cells are not well defined. Here, we examine the ability of CPMV particles to activate human monocytes, dendritic cells (DCs) and macrophages. Monocytes in peripheral blood mononuclear cell cultures and purified CD14+ monocytes were readily activated by CPMV in vitro, leading to induction of HLA-DR, CD86, PD-L1, IL-15R and CXCL10 expression. Monocytes released chemokines, CXCL10, MIP-1α and MIP-1β into cell culture supernatants after incubation with CPMV. DC subsets (pDC and mDC) and monocyte-derived macrophages also demonstrated evidence of activation after incubation with CPMV. Inhibitors of spleen tyrosine kinase (SYK), endocytosis or endocytic acidification impaired the capacity of CPMV to activate monocytes. Furthermore, CPMV activation of monocytes was partially blocked by a TLR7/8 antagonist. These data demonstrate that CPMV activates human monocytes in a manner dependent on SYK signalling, endosomal acidification and with an important contribution from TLR7/8 recognition.
Collapse
Affiliation(s)
- Marwah M. Albakri
- Department of PathologySchool of MedicineCase Western Reserve UniversityClevelandOHUSA
- Department of Medical Laboratory TechnologyCollege of Applied Medical SciencesTaibah UniversityMedinaSaudi Arabia
| | - Frank A. Veliz
- Department of Biomedical EngineeringSchool of MedicineCase Western Reserve UniversityClevelandOHUSA
| | - Steven N. Fiering
- Department of Microbiology and ImmunologyGeisel School of Medicine at DartmouthNorris Cotton Cancer CenterLebanonNHUSA
| | - Nicole F. Steinmetz
- Department of NanoEngineeringUniversity of California San DiegoLa JollaCAUSA
- Department of RadiologyUniversity of California San DiegoLa JollaCAUSA
- Department of BioengineeringUniversity of California San DiegoLa JollaCAUSA
- Moores Cancer CenterUniversity of California San DiegoLa JollaCAUSA
| | - Scott F. Sieg
- Division of Infectious Diseases and HIV MedicineSchool of MedicineCase Western Reserve UniversityClevelandOHUSA
| |
Collapse
|
14
|
African Swine Fever Virus Armenia/07 Virulent Strain Controls Interferon Beta Production through the cGAS-STING Pathway. J Virol 2019; 93:JVI.02298-18. [PMID: 30918080 PMCID: PMC6613762 DOI: 10.1128/jvi.02298-18] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/21/2019] [Indexed: 12/12/2022] Open
Abstract
African swine fever, a devastating disease for domestic pigs and wild boar, is currently spreading in Europe, Russia, and China, becoming a global threat with huge economic and ecological consequences. One interesting aspect of ASFV biology is the molecular mechanism leading to high virulence of some strains compared to more attenuated strains, which produce subclinical infections. In this work, we show that the presently circulating virulent Armenia/07 virus blocks the synthesis of IFN-β, a key mediator between the innate and adaptive immune response. Armenia/07 inhibits the cGAS-STING pathway by impairing STING activation during infection. In contrast, the cGAS-STING pathway is efficiently activated during NH/P68 attenuated strain infection, leading to the production of large amounts of IFN-β. Our results show for the first time the relationship between the cGAS-STING pathway and ASFV virulence, contributing to uncover the molecular mechanisms of ASFV virulence and to the rational development of ASFV vaccines. African swine fever virus (ASFV) is a complex, cytoplasmic double-stranded DNA (dsDNA) virus that is currently expanding throughout the world. Currently, circulating virulent genotype II Armenia/07-like viruses cause fatal disease in pigs and wild boar, whereas attenuated strains induce infections with various levels of chronic illness. Sensing cytosolic dsDNA, mainly by the key DNA sensor cyclic GMP-AMP synthase (cGAS), leads to the synthesis of type I interferon and involves signaling through STING, TBK1, and IRF3. After phosphorylation, STING translocates from the endoplasmic reticulum to the Golgi compartment and to the perinuclear region, acting as an indispensable adaptor connecting the cytosolic detection of DNA to the TBK1-IRF3 signaling pathway. We demonstrate here that attenuated NH/P68, but not virulent Armenia/07, activates the cGAS-STING-IRF3 cascade very early during infection, inducing STING phosphorylation and trafficking through a mechanism involving cGAMP. Both TBK1 and IRF3 are subsequently activated and, in response to this, a high level of beta interferon (IFN-β) was produced during NH/P68 infection; in contrast, Armenia/07 infection generated IFN-β levels below those of uninfected cells. Our results show that virulent Armenia/07 ASFV controls the cGAS-STING pathway, but these mechanisms are not at play when porcine macrophages are infected with attenuated NH/P68 ASFV. These findings show for the first time the involvement of the cGAS-STING-IRF3 route in ASFV infection, where IFN-β production or inhibition was found after infection by attenuated or virulent ASFV strains, respectively, thus reinforcing the idea that ASFV virulence versus attenuation may be a phenomenon grounded in ASFV-mediated innate immune modulation where the cGAS-STING pathway might play an important role. IMPORTANCE African swine fever, a devastating disease for domestic pigs and wild boar, is currently spreading in Europe, Russia, and China, becoming a global threat with huge economic and ecological consequences. One interesting aspect of ASFV biology is the molecular mechanism leading to high virulence of some strains compared to more attenuated strains, which produce subclinical infections. In this work, we show that the presently circulating virulent Armenia/07 virus blocks the synthesis of IFN-β, a key mediator between the innate and adaptive immune response. Armenia/07 inhibits the cGAS-STING pathway by impairing STING activation during infection. In contrast, the cGAS-STING pathway is efficiently activated during NH/P68 attenuated strain infection, leading to the production of large amounts of IFN-β. Our results show for the first time the relationship between the cGAS-STING pathway and ASFV virulence, contributing to uncover the molecular mechanisms of ASFV virulence and to the rational development of ASFV vaccines.
Collapse
|
15
|
Becker J, Kinast V, Döring M, Lipps C, Duran V, Spanier J, Tegtmeyer PK, Wirth D, Cicin-Sain L, Alcamí A, Kalinke U. Human monocyte-derived macrophages inhibit HCMV spread independent of classical antiviral cytokines. Virulence 2019; 9:1669-1684. [PMID: 30403913 PMCID: PMC7000197 DOI: 10.1080/21505594.2018.1535785] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Infection of healthy individuals with human cytomegalovirus (HCMV) is usually unnoticed and results in life-long latency, whereas HCMV reactivation as well as infection of newborns or immunocompromised patients can cause life-threatening disease. To better understand HCMV pathogenesis we studied mechanisms that restrict HCMV spread. We discovered that HCMV-infected cells can directly trigger plasmacytoid dendritic cells (pDC) to mount antiviral type I interferon (IFN-I) responses, even in the absence of cell-free virus. In contrast, monocyte-derived cells only expressed IFN-I when stimulated by cell-free HCMV, or upon encounter of HCMV-infected cells that already produced cell-free virus. Nevertheless, also in the absence of cell-free virus, i.e., upon co-culture of infected epithelial/endothelial cells and monocyte-derived macrophages (moMΦ) or dendritic cells (moDC), antiviral responses were induced that limited HCMV spread. The induction of this antiviral effect was dependent on cell-cell contact, whereas cell-free supernatants from co-culture experiments also inhibited virus spread, implying that soluble factors were critically needed. Interestingly, the antiviral effect was independent of IFN-γ, TNF-α, and IFN-I as indicated by cytokine inhibition experiments using neutralizing antibodies or the vaccinia virus-derived soluble IFN-I binding protein B18R, which traps human IFN-α and IFN-β. In conclusion, our results indicate that human macrophages and dendritic cells can limit HCMV spread by IFN-I dependent as well as independent mechanisms, whereas the latter ones might be particularly relevant for the restriction of HCMV transmission via cell-to-cell spread.
Collapse
Affiliation(s)
- Jennifer Becker
- a Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School , Hannover , Germany
| | - Volker Kinast
- a Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School , Hannover , Germany
| | - Marius Döring
- a Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School , Hannover , Germany
| | - Christoph Lipps
- b Model Systems for Infection and Immunity , Helmholtz Centre for Infection Research , Braunschweig , Germany
| | - Veronica Duran
- a Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School , Hannover , Germany
| | - Julia Spanier
- a Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School , Hannover , Germany
| | - Pia-Katharina Tegtmeyer
- a Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School , Hannover , Germany
| | - Dagmar Wirth
- b Model Systems for Infection and Immunity , Helmholtz Centre for Infection Research , Braunschweig , Germany
| | - Luka Cicin-Sain
- c Department of Vaccinology , Helmholtz Centre for Infection Research , Braunschweig , Germany.,d German Center for Infection Research (DZIF) , Hannover-Braunschweig site , Germany.,e Institute for Virology , Hannover Medical School , Hannover , Germany
| | - Antonio Alcamí
- f Centro de Biología Molecular Severo Ochoa , Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid , Madrid , Spain
| | - Ulrich Kalinke
- a Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School , Hannover , Germany
| |
Collapse
|
16
|
Annibali V, Umeton R, Palermo A, Severa M, Etna MP, Giglio S, Romano S, Ferraldeschi M, Buscarinu MC, Vecchione A, Annese A, Policano C, Mechelli R, Pizzolato Umeton R, Fornasiero A, Angelini DF, Guerrera G, Battistini L, Coccia EM, Salvetti M, Ristori G. Analysis of coding and non-coding transcriptome of peripheral B cells reveals an altered interferon response factor (IRF)-1 pathway in multiple sclerosis patients. J Neuroimmunol 2018; 324:165-171. [PMID: 30270021 DOI: 10.1016/j.jneuroim.2018.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/13/2018] [Accepted: 09/10/2018] [Indexed: 01/15/2023]
Abstract
Several evidences emphasize B-cell pathogenic roles in multiple sclerosis (MS). We performed transcriptome analyses on peripheral B cells from therapy-free patients and age/sex-matched controls. Down-regulation of two transcripts (interferon response factor 1-IRF1, and C-X-C motif chemokine 10-CXCL10), belonging to the same pathway, was validated by RT-PCR in 26 patients and 21 controls. IRF1 and CXCL10 transcripts share potential seeding sequences for hsa-miR-424, that resulted up-regulated in MS patients. We confirmed this interaction and its functional effect by transfection experiments. Consistent findings indicate down-regulation of IRF1/CXCL10 axis, that may plausibly contribute to a pro-survival status of B cells in MS.
Collapse
Affiliation(s)
- Viviana Annibali
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Renato Umeton
- Department of Informatics, Dana-Farber Cancer Institute, Boston, MA, United States; Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Antonia Palermo
- Department of Mathematics and Computer Science, University of Calabria
| | - Martina Severa
- Department of Infectious, Parasitic and Immune-mediated Disease, Istituto Superiore di Sanità, Rome, Italy
| | - Marilena Paola Etna
- Department of Infectious, Parasitic and Immune-mediated Disease, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Giglio
- Division of Pathology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Silvia Romano
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Michela Ferraldeschi
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Maria Chiara Buscarinu
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Andrea Vecchione
- Division of Pathology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Anita Annese
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Claudia Policano
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Rosella Mechelli
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | | | - Arianna Fornasiero
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | | | | | | | - Eliana Marina Coccia
- Department of Infectious, Parasitic and Immune-mediated Disease, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Salvetti
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy; IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed (M.S.), Pozzilli, IS, Italy.
| | - Giovanni Ristori
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
17
|
Longitudinal association of type 1 interferon-induced chemokines with disease activity in systemic lupus erythematosus. Sci Rep 2018; 8:3268. [PMID: 29459655 PMCID: PMC5818532 DOI: 10.1038/s41598-018-20203-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 01/15/2018] [Indexed: 12/19/2022] Open
Abstract
Type I interferon (IFN) pathways are significant in SLE pathogenesis. Less is known about the utility of measuring markers of IFN activity in patients, or whether patient subsets with different profiles exist. We explored the longitudinal associations of IFN-induced chemokines with disease activity in a cohort of SLE patients. We calculated a validated composite score (IFN-CK) of three type I IFN-inducible chemokines (CCL2/CXCL10/CCL19) measured in 109 SLE patients (median 7 occasions over 3.2 years). Longitudinal associations of IFN-CK score with disease activity (SLEDAI-2K) and other variables were assessed using general estimating equation (GEE) methods. IFN-CK was detectable in all patients. SLEDAI-2K was significantly associated with IFN-CK, damage score and prednisolone dose. SLEDAI-2K remained significantly associated with IFN-CK over time after adjustment of covariates. Patients with high time-adjusted mean IFN-CK had lower complement and higher time-adjusted disease activity. Concordance between IFN-CK and SLEDAI-2K varied widely among patients, with some individuals having none, others weak, and a subset very high concordance. In summary in our cohort of SLE patients, serum IFN-CK varied over time with disease activity, but with wide variation in concordance. Differing relationships between IFN pathway activation and disease activity may be valuable in assigning patients to emerging IFN-pathway targeting treatments.
Collapse
|
18
|
Cruz SA, Hari A, Qin Z, Couture P, Huang H, Lagace DC, Stewart AFR, Chen HH. Loss of IRF2BP2 in Microglia Increases Inflammation and Functional Deficits after Focal Ischemic Brain Injury. Front Cell Neurosci 2017; 11:201. [PMID: 28769762 PMCID: PMC5515910 DOI: 10.3389/fncel.2017.00201] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 06/27/2017] [Indexed: 12/17/2022] Open
Abstract
Ischemic stroke causes neuronal cell death and triggers a cascade of inflammatory signals that contribute to secondary brain damage. Microglia, the brain-resident macrophages that remove dead neurons, play a critical role in the brain’s response to ischemic injury. Our previous studies showed that IRF2 binding protein 2 (IRF2BP2) regulates peripheral macrophage polarization, limits their inflammatory response and reduces susceptibility to atherosclerosis. Here, we show that loss of IRF2BP2 in microglia leads to increased inflammatory cytokine expression in response to lipopolysaccharide challenge and impaired activation of anti-inflammatory markers in response to interleukin-4 (IL4) stimulation. Focal ischemic brain injury of the sensorimotor cortex induced by photothrombosis caused more severe functional deficits in mice with IRF2BP2 ablated in macrophages/microglia, associated with elevated expression of inflammatory cytokines in the brain. These mutant mice had larger infarctions 4 days after stroke associated with fewer anti-inflammatory M2 microglia/macrophages recruited to the peri-infarct area, suggesting an impaired clearance of injured tissues. Since IRF2BP2 modulates interferon signaling, and interferon beta (IFNβ) has been reported to be anti-inflammatory and reduce ischemic brain injury, we asked whether loss of IRF2BP2 in macrophages/microglia would affect the response to IFNβ in our stroke model. IFNβ suppressed inflammatory cytokine production of macrophages and reduced infarct volumes at 4 days after photothrombosis in wild type mice. The anti-inflammatory effect of IFNβ was lost in IRF2BP2-deficient macrophages and IFNβ failed to protect mice lacking IRF2BP2 in macrophages/microglia from ischemic injury. In summary, IRF2BP2 expression in macrophages/microglia is important to limit inflammation and stroke injury, in part by mediating the beneficial effect of IFNβ.
Collapse
Affiliation(s)
- Shelly A Cruz
- Ottawa Hospital Research InstituteOttawa, ON, Canada.,Brain and Mind Institute, University of OttawaOttawa, ON, Canada.,Canadian Partnership for Stroke RecoveryOttawa, ON, Canada
| | - Aswin Hari
- Ottawa Hospital Research InstituteOttawa, ON, Canada.,Brain and Mind Institute, University of OttawaOttawa, ON, Canada.,Canadian Partnership for Stroke RecoveryOttawa, ON, Canada
| | - Zhaohong Qin
- Ottawa Hospital Research InstituteOttawa, ON, Canada.,Brain and Mind Institute, University of OttawaOttawa, ON, Canada
| | - Pascal Couture
- Ottawa Hospital Research InstituteOttawa, ON, Canada.,Brain and Mind Institute, University of OttawaOttawa, ON, Canada.,Cellular and Molecular Medicine, University of OttawaOttawa, ON, Canada
| | - Hua Huang
- Ottawa Hospital Research InstituteOttawa, ON, Canada.,Brain and Mind Institute, University of OttawaOttawa, ON, Canada.,University of Ottawa Heart InstituteOttawa, ON, Canada
| | - Diane C Lagace
- Brain and Mind Institute, University of OttawaOttawa, ON, Canada.,Canadian Partnership for Stroke RecoveryOttawa, ON, Canada.,Cellular and Molecular Medicine, University of OttawaOttawa, ON, Canada
| | - Alexandre F R Stewart
- University of Ottawa Heart InstituteOttawa, ON, Canada.,Biochemistry, Microbiology and Immunology, University of OttawaOttawa, ON, Canada
| | - Hsiao-Huei Chen
- Ottawa Hospital Research InstituteOttawa, ON, Canada.,Brain and Mind Institute, University of OttawaOttawa, ON, Canada.,Canadian Partnership for Stroke RecoveryOttawa, ON, Canada.,Cellular and Molecular Medicine, University of OttawaOttawa, ON, Canada.,Medicine, University of OttawaOttawa, ON, Canada
| |
Collapse
|
19
|
Zhang Y, Li H. Reprogramming Interferon Regulatory Factor Signaling in Cardiometabolic Diseases. Physiology (Bethesda) 2017; 32:210-223. [PMID: 28404737 DOI: 10.1152/physiol.00038.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/06/2017] [Accepted: 02/09/2017] [Indexed: 01/12/2023] Open
Abstract
Interferon regulatory factors (IRFs) are evolutionarily conserved proteins expressed not only in immune cells but also in other tissues and organs outside the immune system. In this review, we discuss mechanisms responsible for IRF-mediated innate immune responses and the function and mechanism of IRFs in cardiometabolic diseases. We focus on the role of IRFs in innate immunity and cardiometabolic homeostasis, and highlight reprogrammed IRF signaling.
Collapse
Affiliation(s)
- Yaxing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Model Animal, Wuhan University, Wuhan, People's Republic of China; and
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, People's Republic of China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Model Animal, Wuhan University, Wuhan, People's Republic of China; and
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
20
|
Walker EC, Johnson RW, Hu Y, Brennan HJ, Poulton IJ, Zhang JG, Jenkins BJ, Smyth GK, Nicola NA, Sims NA. Murine Oncostatin M Acts via Leukemia Inhibitory Factor Receptor to Phosphorylate Signal Transducer and Activator of Transcription 3 (STAT3) but Not STAT1, an Effect That Protects Bone Mass. J Biol Chem 2016; 291:21703-21716. [PMID: 27539849 DOI: 10.1074/jbc.m116.748483] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/15/2016] [Indexed: 12/31/2022] Open
Abstract
Oncostatin M (OSM) and leukemia inhibitory factor (LIF) are IL-6 family members with a wide range of biological functions. Human OSM (hOSM) and murine LIF (mLIF) act in mouse cells via a LIF receptor (LIFR)-glycoprotein 130 (gp130) heterodimer. In contrast, murine OSM (mOSM) signals mainly via an OSM receptor (OSMR)-gp130 heterodimer and binds with only very low affinity to mLIFR. hOSM and mLIF stimulate bone remodeling by both reducing osteocytic sclerostin and up-regulating the pro-osteoclastic factor receptor activator of NF-κB ligand (RANKL) in osteoblasts. In the absence of OSMR, mOSM still strongly suppressed sclerostin and stimulated bone formation but did not induce RANKL, suggesting that intracellular signaling activated by the low affinity interaction of mOSM with mLIFR is different from the downstream effects when mLIF or hOSM interacts with the same receptor. Both STAT1 and STAT3 were activated by mOSM in wild type cells or by mLIF/hOSM in wild type and Osmr-/- cells. In contrast, in Osmr-/- primary osteocyte-like cells stimulated with mOSM (therefore acting through mLIFR), microarray expression profiling and Western blotting analysis identified preferential phosphorylation of STAT3 and induction of its target genes but not of STAT1 and its target genes; this correlated with reduced phosphorylation of both gp130 and LIFR. In a mouse model of spontaneous osteopenia caused by hyperactivation of STAT1/3 signaling downstream of gp130 (gp130Y757F/Y757F), STAT1 deletion rescued the osteopenic phenotype, indicating a beneficial effect of promoting STAT3 signaling over STAT1 downstream of gp130 in this low bone mass condition, and this may have therapeutic value.
Collapse
Affiliation(s)
- Emma C Walker
- From the St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Rachelle W Johnson
- From the St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Yifang Hu
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Holly J Brennan
- From the St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Ingrid J Poulton
- From the St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Jian-Guo Zhang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Medical Biology, and
| | - Brendan J Jenkins
- Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.,Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton 3168, Victoria, Australia, and
| | - Gordon K Smyth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Departments of Mathematics and Statistics
| | - Nicos A Nicola
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Medical Biology, and
| | - Natalie A Sims
- From the St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia, .,Medicine at St. Vincent's Hospital, The University of Melbourne, Victoria 3010, Australia
| |
Collapse
|
21
|
Haarmann A, Nehen M, Deiß A, Buttmann M. Fumaric Acid Esters Do Not Reduce Inflammatory NF-κB/p65 Nuclear Translocation, ICAM-1 Expression and T-Cell Adhesiveness of Human Brain Microvascular Endothelial Cells. Int J Mol Sci 2015; 16:19086-95. [PMID: 26287168 PMCID: PMC4581287 DOI: 10.3390/ijms160819086] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/31/2015] [Accepted: 08/10/2015] [Indexed: 11/30/2022] Open
Abstract
Dimethyl fumarate (DMF) is approved for disease-modifying treatment of patients with relapsing-remitting multiple sclerosis. Animal experiments suggested that part of its therapeutic effect is due to a reduction of T-cell infiltration of the central nervous system (CNS) by uncertain mechanisms. Here we evaluated whether DMF and its primary metabolite monomethyl fumarate (MMF) modulate pro-inflammatory intracellular signaling and T-cell adhesiveness of nonimmortalized single donor human brain microvascular endothelial cells at low passages. Neither DMF nor MMF at concentrations of 10 or 50 µM blocked the IL-1β-induced nuclear translocation of NF-κB/p65, whereas the higher concentration of DMF inhibited the nuclear entry of p65 in human umbilical vein endothelium cultured in parallel. DMF and MMF also did not alter the IL-1β-stimulated activation of p38 MAPK in brain endothelium. Furthermore, neither DMF nor MMF reduced the basal or IL-1β-inducible expression of ICAM-1. In accordance, both fumaric acid esters did not reduce the adhesion of activated Jurkat T cells to brain endothelium under basal or inflammatory conditions. Therefore, brain endothelial cells probably do not directly mediate a potential blocking effect of fumaric acid esters on the inflammatory infiltration of the CNS by T cells.
Collapse
Affiliation(s)
- Axel Haarmann
- Department of Neurology, University of Würzburg, Josef-Schneider-Str. 11, Würzburg 97080, Germany.
| | - Mathias Nehen
- Department of Neurology, University of Würzburg, Josef-Schneider-Str. 11, Würzburg 97080, Germany.
| | - Annika Deiß
- Department of Neurology, University of Würzburg, Josef-Schneider-Str. 11, Würzburg 97080, Germany.
| | - Mathias Buttmann
- Department of Neurology, University of Würzburg, Josef-Schneider-Str. 11, Würzburg 97080, Germany.
| |
Collapse
|
22
|
Wright FL, Gamboni F, Moore EE, Nydam TL, Mitra S, Silliman CC, Banerjee A. Hyperosmolarity invokes distinct anti-inflammatory mechanisms in pulmonary epithelial cells: evidence from signaling and transcription layers. PLoS One 2014; 9:e114129. [PMID: 25479425 PMCID: PMC4257597 DOI: 10.1371/journal.pone.0114129] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 11/04/2014] [Indexed: 12/29/2022] Open
Abstract
Hypertonic saline (HTS) has been used intravenously to reduce organ dysfunction following injury and as an inhaled therapy for cystic fibrosis lung disease. The role and mechanism of HTS inhibition was explored in the TNFα and IL-1β stimulation of pulmonary epithelial cells. Hyperosmolar (HOsm) media (400 mOsm) inhibited the production of select cytokines stimulated by TNFα and IL-1β at the level of mRNA translation, synthesis and release. In TNFα stimulated A549 cells, HOsm media inhibited I-κBα phosphorylation, NF-κB translocation into the nucleus and NF-κB nuclear binding. In IL-1β stimulated cells HOsm inhibited I-κBα phosphorylation without affecting NF-κB translocation or nuclear binding. Incubation in HOsm conditions inhibited both TNFα and IL-1β stimulated nuclear localization of interferon response factor 1 (IRF-1). Additional transcription factors such as AP-1, Erk-1/2, JNK and STAT-1 were unaffected by HOsm. HTS and sorbitol supplemented media produced comparable outcomes in all experiments, indicating that the effects of HTS were mediated by osmolarity, not by sodium. While not affecting MAPK modules discernibly in A549 cells, both HOsm conditions inhibit IRF-1 against TNFα or IL-1β, but inhibit p65 NF-kB translocation only against TNFα but not IL-1β. Thus, anti-inflammatory mechanisms of HTS/HOsm appear to disrupt cytokine signals at distinct intracellular steps.
Collapse
Affiliation(s)
- Franklin L. Wright
- Department of Surgery/Trauma Research Center, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Fabia Gamboni
- Department of Surgery/Trauma Research Center, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Ernest E. Moore
- Department of Surgery, Denver Health Medical Center, Denver, Colorado, United States of America
| | - Trevor L. Nydam
- Department of Surgery/Trauma Research Center, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Sanchayita Mitra
- Department of Surgery/Trauma Research Center, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Christopher C. Silliman
- Department of Pediatrics, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Anirban Banerjee
- Department of Surgery/Trauma Research Center, University of Colorado Denver, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
23
|
Huang H, Langenkamp E, Georganaki M, Loskog A, Fuchs PF, Dieterich LC, Kreuger J, Dimberg A. VEGF suppresses T-lymphocyte infiltration in the tumor microenvironment through inhibition of NF-κB-induced endothelial activation. FASEB J 2014; 29:227-38. [PMID: 25361735 DOI: 10.1096/fj.14-250985] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Antiangiogenic treatment targeting the vascular endothelial growth factor (VEGF) signaling pathway is in clinical use, but its effect on vascular function and the tumor microenvironment is poorly understood. Here, we investigate cross-talk between VEGF and proinflammatory TNF-α signaling in endothelial cells and its impact on leukocyte recruitment. We found that cotreatment with VEGF decreased TNF-α-induced Jurkat cell adhesion to human microvascular endothelial cells by 40%. This was associated with inhibition of TNF-α-mediated regulation of 86 genes, including 2 T-lymphocyte-attracting chemokines, CXCL10 and CXCL11 [TNF-α concentration 1 ng/ml; 50% inhibition/inhibitory concentration (IC50) VEGF, 3 ng/ml]. Notably, VEGF directly suppressed TNF-α-induced gene expression through negative cross-talk with the NF-κB-signaling pathway, leading to an early decrease in IFN regulatory factor 1 (IRF-1) expression and reduced phosphorylation of signal transducer and activator of transcription 1 (p-Stat1) at later times. Inhibition of VEGF signaling in B16 melanoma tumor-bearing mice by sunitinib treatment resulted in up-regulation of CXCL10 and CXCL11 in tumor vessels, accompanied by up to 18-fold increased infiltration of CD3(+) T-lymphocytes in B16 tumors. Our results demonstrate a novel role of VEGF in negative regulation of NF-κB signaling and endothelial activation in the tumor microenvironment and provide evidence that pharmacological inhibition of VEGF signaling enhances T-lymphocyte recruitment through up-regulation of chemokines CXCL10 and CXCL11.
Collapse
Affiliation(s)
- Hua Huang
- *Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, and Department of Medical Biochemistry and Microbiology, Biomedical Center, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Elise Langenkamp
- *Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, and Department of Medical Biochemistry and Microbiology, Biomedical Center, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Maria Georganaki
- *Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, and Department of Medical Biochemistry and Microbiology, Biomedical Center, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Angelica Loskog
- *Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, and Department of Medical Biochemistry and Microbiology, Biomedical Center, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Peder Fredlund Fuchs
- *Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, and Department of Medical Biochemistry and Microbiology, Biomedical Center, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lothar C Dieterich
- *Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, and Department of Medical Biochemistry and Microbiology, Biomedical Center, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Johan Kreuger
- *Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, and Department of Medical Biochemistry and Microbiology, Biomedical Center, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Anna Dimberg
- *Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, and Department of Medical Biochemistry and Microbiology, Biomedical Center, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
24
|
Kalinowski A, Ueki I, Min-Oo G, Ballon-Landa E, Knoff D, Galen B, Lanier LL, Nadel JA, Koff JL. EGFR activation suppresses respiratory virus-induced IRF1-dependent CXCL10 production. Am J Physiol Lung Cell Mol Physiol 2014; 307:L186-96. [PMID: 24838750 DOI: 10.1152/ajplung.00368.2013] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Airway epithelial cells are the primary cell type involved in respiratory viral infection. Upon infection, airway epithelium plays a critical role in host defense against viral infection by contributing to innate and adaptive immune responses. Influenza A virus, rhinovirus, and respiratory syncytial virus (RSV) represent a broad range of human viral pathogens that cause viral pneumonia and induce exacerbations of asthma and chronic obstructive pulmonary disease. These respiratory viruses induce airway epithelial production of IL-8, which involves epidermal growth factor receptor (EGFR) activation. EGFR activation involves an integrated signaling pathway that includes NADPH oxidase activation of metalloproteinase, and EGFR proligand release that activates EGFR. Because respiratory viruses have been shown to activate EGFR via this signaling pathway in airway epithelium, we investigated the effect of virus-induced EGFR activation on airway epithelial antiviral responses. CXCL10, a chemokine produced by airway epithelial cells in response to respiratory viral infection, contributes to the recruitment of lymphocytes to target and kill virus-infected cells. While respiratory viruses activate EGFR, the interaction between CXCL10 and EGFR signaling pathways is unclear, and the potential for EGFR signaling to suppress CXCL10 has not been explored. Here, we report that respiratory virus-induced EGFR activation suppresses CXCL10 production. We found that influenza virus-, rhinovirus-, and RSV-induced EGFR activation suppressed IFN regulatory factor (IRF) 1-dependent CXCL10 production. In addition, inhibition of EGFR during viral infection augmented IRF1 and CXCL10. These findings describe a novel mechanism that viruses use to suppress endogenous antiviral defenses, and provide potential targets for future therapies.
Collapse
Affiliation(s)
| | - Iris Ueki
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, California
| | - Gundula Min-Oo
- Department of Microbiology and Immunology, and Cancer Research Institute, University of California, San Francisco, California; and
| | | | - David Knoff
- Department of Medicine, Yale University, New Haven, Connecticut
| | - Benjamin Galen
- Department of Medicine, Yale University, New Haven, Connecticut
| | - Lewis L Lanier
- Department of Microbiology and Immunology, and Cancer Research Institute, University of California, San Francisco, California; and
| | - Jay A Nadel
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, California
| | - Jonathan L Koff
- Department of Medicine, Yale University, New Haven, Connecticut;
| |
Collapse
|
25
|
Bhattacharya S, Katlinski KV, Reichert M, Takano S, Brice A, Zhao B, Yu Q, Zheng H, Carbone CJ, Katlinskaya YV, Leu NA, McCorkell KA, Srinivasan S, Girondo M, Rui H, May MJ, Avadhani NG, Rustgi AK, Fuchs SY. Triggering ubiquitination of IFNAR1 protects tissues from inflammatory injury. EMBO Mol Med 2014; 6:384-97. [PMID: 24480543 PMCID: PMC3958312 DOI: 10.1002/emmm.201303236] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Type 1 interferons (IFN) protect the host against viruses by engaging a cognate receptor (consisting of IFNAR1/IFNAR2 chains) and inducing downstream signaling and gene expression. However, inflammatory stimuli can trigger IFNAR1 ubiquitination and downregulation thereby attenuating IFN effects in vitro. The significance of this paradoxical regulation is unknown. Presented here results demonstrate that inability to stimulate IFNAR1 ubiquitination in the Ifnar1(SA) knock-in mice renders them highly susceptible to numerous inflammatory syndromes including acute and chronic pancreatitis, and autoimmune and toxic hepatitis. Ifnar1(SA) mice (or their bone marrow-receiving wild type animals) display persistent immune infiltration of inflamed tissues, extensive damage and gravely inadequate tissue regeneration. Pharmacologic stimulation of IFNAR1 ubiquitination is protective against from toxic hepatitis and fulminant generalized inflammation in wild type but not Ifnar1(SA) mice. These results suggest that endogenous mechanisms that trigger IFNAR1 ubiquitination for limiting the inflammation-induced tissue damage can be purposely mimicked for therapeutic benefits.
Collapse
Affiliation(s)
- Sabyasachi Bhattacharya
- Department of Animal Biology, School of Veterinary Medicine University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Voigt E, Inankur B, Baltes A, Yin J. A quantitative infection assay for human type I, II, and III interferon antiviral activities. Virol J 2013; 10:224. [PMID: 23829314 PMCID: PMC3716869 DOI: 10.1186/1743-422x-10-224] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 06/24/2013] [Indexed: 12/11/2022] Open
Abstract
Background Upon virus infection, cells secrete a diverse group of antiviral molecules that signal proximal cells to enter into an antiviral state, slowing or preventing viral spread. These paracrine signaling molecules can work synergistically, so measurement of any one antiviral molecule does not reflect the total antiviral activity of the system. Results We have developed an antiviral assay based on replication inhibition of an engineered fluorescent vesicular stomatitis virus reporter strain on A549 human lung epithelial cells. Our assay provides a quantitative functional readout of human type I, II, and III interferon activities, and it provides better sensitivity, intra-, and inter-assay reproducibility than the traditional crystal violet based assay. Further, it eliminates cell fixation, rinsing, and staining steps, and is inexpensive to implement. Conclusions A dsRed2-strain of vesicular stomatitis virus that is sensitive to type I, II, and III interferons was used to develop a convenient and sensitive assay for interferon antiviral activity. We demonstrate use of the assay to quantify the kinetics of paracrine antiviral signaling from human prostate cancer (PC3) cells in response to viral infection. The assay is applicable to high-throughput screening for anti-viral compounds as well as basic studies of cellular antiviral signaling.
Collapse
Affiliation(s)
- Emily Voigt
- Department of Chemical and Biological Engineering, University of Wisconsin, Madison, USA
| | | | | | | |
Collapse
|
27
|
Lobular panniculitis and lipoatrophy of the thighs with interferon-ß1a for intramuscular injection in a patient with multiple sclerosis. J Clin Neurosci 2012; 19:1312-3. [PMID: 22721889 DOI: 10.1016/j.jocn.2011.11.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2011] [Accepted: 11/14/2011] [Indexed: 11/23/2022]
Abstract
Multiple sclerosis (MS) patients may experience severe local inflammatory skin reactions during disease-modifying therapy with subcutaneously injected interferon-β (IFN-β). It is common clinical practice to switch those patients to an intramuscularly administered formulation, where severe local skin reactions have not been described. Here we report a 42-year-old woman with stable relapsing-remitting MS, who was switched from subcutaneously to intramuscularly injected IFN-β1a due to abdominal skin necroses and slight multifocal lipoatrophy. After two years of complication-free therapy with intramuscular IFN-β1a, the patient slowly developed painful lobular panniculitis and severe lipoatrophy of both lateral thighs. A careful diagnostic workup identified misguided subcutaneous injections due to a wrong injection angle as the most plausible cause. Upon correction of her injection technique, pain and skin reddening resolved, while her disfiguring lipoatrophy was irreversible. This report should enhance awareness that severe skin adverse effects may also occur, although rarely, with IFN-β for intramuscular injection. Early recognition and correction of the injection technique may help to prevent severe complications.
Collapse
|
28
|
Brock LG, Karron RA, Krempl CD, Collins PL, Buchholz UJ. Evaluation of pneumonia virus of mice as a possible human pathogen. J Virol 2012; 86:5829-43. [PMID: 22438539 PMCID: PMC3347304 DOI: 10.1128/jvi.00163-12] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 03/08/2012] [Indexed: 01/22/2023] Open
Abstract
Pneumonia virus of mice (PVM), a relative of human respiratory syncytial virus (RSV), causes respiratory disease in mice. There is serologic evidence suggesting widespread exposure of humans to PVM. To investigate replication in primates, African green monkeys (AGM) and rhesus macaques (n = 4) were inoculated with PVM by the respiratory route. Virus was shed intermittently at low levels by a subset of animals, suggesting poor permissiveness. PVM efficiently replicated in cultured human cells and inhibited the type I interferon (IFN) response in these cells. This suggests that poor replication in nonhuman primates was not due to a general nonpermissiveness of primate cells or poor control of the IFN response. Seroprevalence in humans was examined by screening sera from 30 adults and 17 young children for PVM-neutralizing activity. Sera from a single child (6%) and 40% of adults had low neutralizing activity against PVM, which could be consistent with increasing incidence of exposure following early childhood. There was no cross-reaction of human or AGM sera between RSV and PVM and no cross-protection in the mouse model. In native Western blots, human sera reacted with RSV but not PVM proteins under conditions in which AGM immune sera reacted strongly. Serum reactivity was further evaluated by flow cytometry using unfixed Vero cells infected with PVM or RSV expressing green fluorescent protein (GFP) as a measure of viral gene expression. The reactivity of human sera against RSV-infected cells correlated with GFP expression, whereas reactivity against PVM-infected cells was low and uncorrelated with GFP expression. Thus, PVM specificity was not evident. Our results indicate that the PVM-neutralizing activity of human sera is not due to RSV- or PVM-specific antibodies but may be due to low-affinity, polyreactive natural antibodies of the IgG subclass. The absence of PVM-specific antibodies and restriction in nonhuman primates makes PVM unlikely to be a human pathogen.
Collapse
Affiliation(s)
- Linda G. Brock
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Ruth A. Karron
- Center for Immunization Research, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Christine D. Krempl
- Institute of Virology and Immunobiology, Julius-Maximilian University, Würzburg, Germany
| | - Peter L. Collins
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Ursula J. Buchholz
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
29
|
Comini-Frota ER, Teixeira AL, Angelo JPA, Andrade MV, Brum DG, Kaimen-Maciel DR, Foss NT, Donadi EA. Evaluation of serum levels of chemokines during interferon-β treatment in multiple sclerosis patients: a 1-year, observational cohort study. CNS Drugs 2011; 25:971-81. [PMID: 22054120 DOI: 10.2165/11595060-000000000-00000] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND The molecules that provide access to activated T cells in the CNS, including chemokines, have been considered to be a crucial step in the pathogenesis of multiple sclerosis (MS). AIMS In this study, we investigated serial serum chemokine levels in patients with relapsing-remitting MS over 1 year and the association of these chemokine levels with treatment regimens, lesions on MRI and patients' characteristics. METHODS Serum CXCL9, CXCL10, CCL2, CCL4 and CCL5 levels were evaluated using ELISA every 2 months for a year in 28 healthy controls and 28 MS patients during their treatment with interferon (IFN)-β. Patients underwent MRI and were evaluated using the Expanded Disability Status Scale (EDSS) at the first and final evaluations. RESULTS CXCL10 serum levels were higher in MS patients compared with controls, were positively correlated with T2 lesions on MRI and were slightly increased during relapses. Treatment with IFNβ-1a or IFNβ-1b was associated with increased CXCL10 levels when evaluated more than 36 hours after subcutaneous injection. The CXCL9 levels were higher after MS relapse. There was significant variability in CCL4 and CCL5 levels in the serial evaluations, associated with gender and treatment. CCL2 levels were higher in treated MS patients than healthy controls, particularly among those patients with a stable form of the disease. CONCLUSION Serum is a feasible resource for searching for an immunological marker in MS. Peripheral chemokine levels correlated in different ways with IFNβ therapy and with disease and patient characteristics. CLINICAL TRIAL REGISTRATION NUMBER ISRCTN45526724.
Collapse
Affiliation(s)
- Elizabeth R Comini-Frota
- School of Medicine, Federal University of Minas Gerais, and Department of Neurology, Hospital das Clínicas, Belo Horizonte, Minas Gerais, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Bulcao CF, D'Souza KM, Malhotra R, Staron M, Duffy JY, Pandalai PK, Jeevanandam V, Akhter SA. Activation of JAK-STAT and nitric oxide signaling as a mechanism for donor heart dysfunction. J Heart Lung Transplant 2010; 29:346-51. [PMID: 20022263 DOI: 10.1016/j.healun.2009.09.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 08/31/2009] [Accepted: 09/01/2009] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Donor heart dysfunction (DHD) precluding procurement for transplantation occurs in up to 25% of brain-dead (BD) donors. The molecular mechanisms of DHD remain unclear. We investigated the potential role of myocardial interleukin (IL)-6 signaling through the JAK2-STAT3 pathway, which can lead to the generation of nitric oxide (NO) and decreased cardiac myocyte contractility. METHODS Hearts were procured using standard technique with University of Wisconsin (UW) solution from 14 donors with a left ventricular (LV) ejection fraction of <35% (DHD). Ten hearts with normal function (NF) after BD served as controls. LV IL-6 was quantitated by enzyme-linked immunoassay (ELISA) and JAK2-STAT3 signaling was assessed by expression of phosphorylated STAT3. Inducible NO synthase (iNOS) and caspase-3 were measured by activity assays. RESULTS Myocardial IL-6 expression was 8-fold greater in the DHD group vs NF controls. Phosphorylated STAT3 expression was 5-fold higher in DHD than in NF, indicating increased JAK2-STAT3 signaling. LV activity of iNOS was 2.5-fold greater in DHD than in NF. LV expression of the pro-apoptotic gene Bnip3 and caspase-3 activity were 3-fold greater in the DHD group than in the NF group. CONCLUSIONS Myocardial IL-6 expression is significantly higher in the setting of DHD compared with hearts procured with normal function. This may lead to increased JAK2-STAT3 signaling and upregulation of iNOS, which has been shown to decrease cardiac myocyte contractility. Increased NO production may also lead to increased apoptosis through upregulation of Bnip3 gene expression. Increased iNOS signaling may be an important mechanism of DHD and represents a novel therapeutic target to improve cardiac function after BD.
Collapse
Affiliation(s)
- Christian F Bulcao
- Department of Surgery, Section of Cardiothoracic Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Lang PA, Recher M, Honke N, Scheu S, Borkens S, Gailus N, Krings C, Meryk A, Kulawik A, Cervantes-Barragan L, Van Rooijen N, Kalinke U, Ludewig B, Hengartner H, Harris N, Häussinger D, Ohashi PS, Zinkernagel RM, Lang KS. Tissue macrophages suppress viral replication and prevent severe immunopathology in an interferon-I-dependent manner in mice. Hepatology 2010; 52:25-32. [PMID: 20578253 DOI: 10.1002/hep.23640] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
UNLABELLED The innate immune response plays an essential role in the prevention of early viral dissemination. We used the lymphocytic choriomeningitis virus model system to analyze the role of tissue macrophages/Kupffer cells in this process. Our findings demonstrated that Kupffer cells are essential for the efficient capture of infectious virus and for preventing viral replication. The latter process involved activation of Kupffer cells by interferon (IFN)-I and prevented viral spread to neighboring hepatocytes. In the absence of Kupffer cells, hepatocytes were not able to suppress virus replication, even in the presence of IFN-I, leading to prolonged viral replication and severe T cell-dependent immunopathology. CONCLUSION Tissue-resident macrophages play a crucial role in early viral capture and represent the major liver cell type exhibiting responsiveness to IFN-I and providing control of viral replication.
Collapse
Affiliation(s)
- Philipp A Lang
- Institute of Experimental Immunology, University Hospital of Zurich, Zurich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Repression of IP-10 by interactions between histone deacetylation and hypermethylation in idiopathic pulmonary fibrosis. Mol Cell Biol 2010; 30:2874-86. [PMID: 20404089 DOI: 10.1128/mcb.01527-09] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Targeted repression of a subset of key genes involved in tissue remodeling is a cardinal feature of idiopathic pulmonary fibrosis (IPF). The mechanism is unclear but is potentially important in disease pathogenesis and therapeutic targeting. We have previously reported that defective histone acetylation is responsible for the repression of the antifibrotic cyclooxygenase-2 gene. Here we extended our study to the repression of another antifibrotic gene, the potent angiostatic chemokine gamma interferon (IFN-gamma)-inducible protein of 10 kDa (IP-10), in lung fibroblasts from patients with IPF. We revealed that this involved not only histone deacetylation, as with cyclooxygenase-2 repression, but also histone H3 hypermethylation, as a result of decreased recruitment of histone acetyltransferases and increased presence of histone deacetylase (HDAC)-containing repressor complexes, histone methyltransferases G9a and SUV39H1, and heterochromatin protein 1 at the IP-10 promoter, leading to reduced transcription factor binding. More importantly, treatment of diseased cells with HDAC or G9a inhibitors similarly reversed the repressive histone deacetylation and hypermethylation and restored IP-10 expression. These findings strongly suggest that epigenetic dysregulation involving interactions between histone deacetylation and hypermethylation is responsible for targeted repression of IP-10 and potentially other antifibrotic genes in fibrotic lung disease and that this is amenable to therapeutic targeting.
Collapse
|
33
|
Khanna S, Park HA, Sen CK, Golakoti T, Sengupta K, Venkateswarlu S, Roy S. Neuroprotective and antiinflammatory properties of a novel demethylated curcuminoid. Antioxid Redox Signal 2009; 11:449-68. [PMID: 18724833 PMCID: PMC2787730 DOI: 10.1089/ars.2008.2230] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2008] [Revised: 08/25/2008] [Accepted: 08/26/2008] [Indexed: 12/24/2022]
Abstract
A demethylated derivative of curcumin (DC; 67.8% bisdemethylcurcumin, 20.7% demethylmonodemethoxycurcumin, 5.86% bisdemethoxycurcumin, 2.58% demethylcurcumin) was prepared by using a 95% extract of curcumin (C(95); 72.2% curcumin, 18.8% monodemethoxycurcumin, 4.5% bisdemethoxycurcumin). DC increased glutathione and reduced reactive oxygen species (ROS) in HT4 neuronal cells. In a model of glutamate-induced death of HT4, DC was more effective than C(95) in neuroprotection. The protective effects of DC were retained even when DC was withdrawn from culture media after pretreatment. DC treatment, unlike an equal dose of C(95), completely spared glutamate-induced loss of cellular GSH. Both DC and C(95) prevented glutamate-induced elevation of cellular ROS but failed to attenuate glutamate-induced elevation of intracellular calcium. In human microvascular endothelial cells (HMECs) challenged with TNF-alpha, GeneChip analysis revealed that only a subcluster of 23 TNF-alpha-inducible genes were uniquely sensitive to C(95). In sharp contrast, 1,065 TNF-alpha-inducible genes were sensitive to DC but not to C(95), suggesting that DC was more effective in antagonizing the effects of TNF-alpha on HMECs. Functional analysis identified that the genes uniquely sensitive to DC belonged in four functional categories: cytokine-receptor interaction, focal adhesion, cell adhesion, and apoptosis. Real-time PCR as well as ELISA studies demonstrated that TNF-alpha-inducible CXCL10 and CXCL11 expression was sensitive to DC but not to C(95). Flow-cytometry studies recognized ICAM-1 and VCAM-1 as TNF-alpha-inducible adhesion molecules that were uniquely sensitive to DC. Taken together, DC exhibited promising neuroprotective and antiinflammatory properties that must be characterized in vivo.
Collapse
Affiliation(s)
- Savita Khanna
- Departments of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio
| | - Han-A Park
- Departments of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio
| | - Chandan K. Sen
- Departments of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio
| | | | | | | | - Sashwati Roy
- Departments of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio
| |
Collapse
|
34
|
Deletion of nonstructural proteins NS1 and NS2 from pneumonia virus of mice attenuates viral replication and reduces pulmonary cytokine expression and disease. J Virol 2008; 83:1969-80. [PMID: 19052095 DOI: 10.1128/jvi.02041-08] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Pneumonia virus of mice (PVM) strain 15 causes fatal pneumonia in mice and provides a convenient model for human respiratory syncytial virus pathogenesis and immunobiology. We prepared PVM mutants lacking the genes for nonstructural proteins NS1 and/or NS2. In Vero cells, which lack type I interferon (IFN), deletion of these proteins had no effect on the efficiency of virus growth. In IFN-competent mouse embryo fibroblasts, wild-type (wt) PVM and the DeltaNS1 virus grew efficiently and strongly inhibited the IFN response, whereas virus lacking NS2 was highly attenuated and induced high levels of IFN and IFN-inducible genes. In BALB/c mice, intranasal infection with wt PVM caused overt disease that began on day 6 and was lethal by day 9 postinoculation. In comparison, DeltaNS1 induced transient, reduced disease, and DeltaNS2 and DeltaNS12 caused no disease. Thus, NS1 and NS2 are virulence factors, with NS2 being a major antagonist of the type I IFN system. The pulmonary titers of wt PVM and DeltaNS1 were high on day 3 and increased further by day 6; in addition, expression of IFN and representative proinflammatory cytokines/chemokines and T lymphocyte-related cytokines was undetectable on day 3 but increased dramatically by day 6 coincident with the onset of disease. The titers of DeltaNS2 and DeltaNS12 were somewhat lower on day 3 and decreased further by day 6; in addition, these viruses induced a more circumscribed set of cytokines/chemokines (IFN, interleukin-6 [IL-6], and CXCL10) that were detected on day 3 and had largely subsided by day 6. Lung immunohistology revealed abundant PVM-positive pneumocytes and bronchial and bronchiolar epithelial cells in wt PVM- and DeltaNS1-infected mice on day 6 compared to few PVM-positive foci with DeltaNS2 and DeltaNS12. These results indicate that severe PVM disease is associated with high, poorly controlled virus replication driving the expression of high levels of pulmonary IFN and a broad array of cytokines/chemokines. In contrast, in the absence of NS2, there was an early, transient innate response involving moderate levels of IFN, IL-6, and CXCL10 that restricted virus replication and prevented disease.
Collapse
|
35
|
Upregulation of the TLR3 pathway by Kaposi's sarcoma-associated herpesvirus during primary infection. J Virol 2008; 82:5440-9. [PMID: 18367536 DOI: 10.1128/jvi.02590-07] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is associated with several different human malignancies, including Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. KSHV establishes lifelong latency in the host and modulates the host immune response. Innate immunity is critical for controlling de novo viral infection. Toll-like receptors (TLRs) are key components of the innate immune system, and they serve as pathogen recognition receptors that stimulate the host antiviral response. In particular, TLR3 has been implicated in RNA virus recognition. Currently, there is no information regarding how KSHV infection modulates any TLR pathway. We report the first evidence that KSHV upregulates TLR3 expression in human monocytes during primary infection. This is also the first demonstration of a human DNA tumor virus upregulating TLR3, a TLR that thus far has been associated with the recognition of RNA viruses. We found that KSHV upregulates the TLR3 pathway and induces TLR3-specific cytokines and chemokines, including beta 1 interferon (IFN-beta1) and CXCL10 (IP-10). Small interfering RNAs directed against TLR3 greatly reduced the ability of KSHV to upregulate IFN-beta1 and CXCL10 upon infection.
Collapse
|
36
|
Eskan MA, Rose BG, Benakanakere MR, Lee MJ, Kinane DF. Sphingosine 1-phosphate 1 and TLR4 mediate IFN-beta expression in human gingival epithelial cells. THE JOURNAL OF IMMUNOLOGY 2008; 180:1818-25. [PMID: 18209079 DOI: 10.4049/jimmunol.180.3.1818] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IFN-beta production is a critical step in human innate immune responses and is primarily controlled at the transcription level by highly ordered mechanisms. IFN-beta can be induced by pattern-recognition receptors such as the TLR4. S1P1 is a G protein-coupled receptor, which has a high affinity for sphingosine 1-phosphate (S1P). Although many of the receptors and signaling pathways leading to the expression of IFN-beta have been identified and characterized, it is still unclear how IFN-beta is regulated in primary human gingival epithelial cells (HGECs). In this study, we demonstrate that S1P1 and TLR4, acting in unison, play an important role in IFN-beta expression at the protein and mRNA level in HGECs. We demonstrate that the expression of both IFN-beta and IFN-inducible protein-10 (CXCL-10) is significantly up-regulated by LPS and S1P or LPS and a specific S1P1 agonist. This enhanced innate immune response is attenuated in HGECs by small interfering RNA knockdown of either TLR4 or S1P1. Moreover, we show that triggering of TLR4 results in the increased expression of S1P1 receptors. Furthermore, we found that IFN-regulatory factor 3 activation was maximized by LPS and S1P through PI3K. Our data show that triggering TLR4 increases S1P1, such that both TLR4 and S1P1 acting through PI3K enhancement of IFN-regulatory factor 3 activation increase IFN-beta expression in epithelial cells. The functional association between TLR4 and the S1P1 receptor demonstrates a novel mechanism in the regulation of IFN-beta and CXCL-10 in human primary gingival epithelial cells.
Collapse
Affiliation(s)
- Mehmet A Eskan
- Center for Oral Health and Systemic Disease, Department of Periodontics, Endodontics, and Dental Hygiene, University of Louisville School of Dentistry, 501 South Preston Street, Louisville, KY 40202, USA
| | | | | | | | | |
Collapse
|
37
|
Roberts ZJ, Ching LM, Vogel SN. IFN-β-Dependent Inhibition of Tumor Growth by the Vascular Disrupting Agent 5,6-Dimethylxanthenone-4-Acetic Acid (DMXAA). J Interferon Cytokine Res 2008; 28:133-9. [DOI: 10.1089/jir.2007.0992] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Zachary J. Roberts
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD 21201
| | - Lai-Ming Ching
- Auckland Cancer Society Research Center, School of Medicine, University of Auckland, New Zealand
| | - Stefanie N. Vogel
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD 21201
| |
Collapse
|
38
|
Buttmann M, Merzyn C, Hofstetter HH, Rieckmann P. TRAIL, CXCL10 and CCL2 plasma levels during long-term Interferon-β treatment of patients with multiple sclerosis correlate with flu-like adverse effects but do not predict therapeutic response. J Neuroimmunol 2007; 190:170-6. [PMID: 17884184 DOI: 10.1016/j.jneuroim.2007.08.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2006] [Revised: 07/31/2007] [Accepted: 08/29/2007] [Indexed: 01/05/2023]
Abstract
High serum levels of soluble TRAIL (sTRAIL) before or during the first year of Interferon-beta (IFN-beta) therapy were shown to predict an individual therapeutic response of patients with relapsing-remitting multiple sclerosis (RRMS). Here, we investigated whether sTRAIL plasma levels during long-term IFN-beta treatment correlate with future therapeutic response or adverse effects of treatment. Postinjection short-time bursts of sTRAIL were associated with flu-like symptoms and IP-10/CXCL10 as well as MCP-1/CCL2 induction, and were detected after up to 6 years of continuous IFN-beta therapy. However, neither sTRAIL nor chemokine levels allowed prediction of one- and two-year clinical treatment response in 30 RRMS patients, prospectively followed by blinded investigators.
Collapse
Affiliation(s)
- Mathias Buttmann
- Department of Neurology, Julius-Maximilians-University, Josef-Schneider-Strasse 11, Würzburg, Germany.
| | | | | | | |
Collapse
|