1
|
Aliaga-Gaspar P, Brichette-Mieg I, Fernández-Arjona M, Rodríguez-Bada JL, López-Moreno Y, Serrano-Castro P, Fernández-Fernández O, Ciano-Petersen NL, Oliver-Martos B. Recombinant soluble type I interferon receptor exerts antiviral activity by inducing proteins related to autophagy. Biomed Pharmacother 2024; 181:117678. [PMID: 39577364 DOI: 10.1016/j.biopha.2024.117678] [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: 07/25/2024] [Revised: 10/25/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
The soluble type I IFN receptor (sIFNAR2) is produced by alternative splicing and is present in body fluids. Although it can modulate IFN-ß activity, its biological role remains unknown. METHODS An in-silico study was conducted to compare the structure of recombinant human soluble IFNAR2 (r-sIFNAR2) with its native form. The antiviral activity of r-sIFNAR2, produced in BL21-bacteria and CHO cells, was tested using a cytopathic effect assay including appropriate controls. Viability and toxicity were assessed by MTT assays. Proteomic analysis using mass spectrometry was conducted in the A549/EMCV bioassay to elucidate the mechanism of action, and then it was validated by Western blot. RESULTS The BL21-sIFNAR2 had a sequence identity of 83.6 % with the native form, showing variations only in terminal regions. BL21-sIFNAR2 and CHO-sIFNAR2 showed significantly higher percentage of cell viability compared to the viral control, similar to IFN-ß. Cell viability with BL21-sIFNAR2 was comparable to the cell control across all tested concentrations. Proteomic analysis revealed an up regulation of pathways related with autophagy (macroautophagy, autophagy, pexophagy, and mitophagy) with an SQSTM1 overexpression that was then confirmed by Western Blot, especially after virus infection. However, pathways related to interferon signaling, and antiviral mechanisms mediated by IFN-stimulated genes were down-regulated. CONCLUSION r-sIFNAR2 exhibits significant antiviral activity regardless of the expression system used for its production and good safety profile, suggesting its use as a potential antiviral drug. Proteins related to autophagy are involved in the protection from the virus. This study highlights the biological relevance of soluble cytokine receptors as effectors so far overlooked.
Collapse
Affiliation(s)
- Pablo Aliaga-Gaspar
- Neuroimmunology and Neuroinflammation group. Biomedical Research Institute of Málaga-IBIMA Plataforma Bionand Hospital Regional Universitario de Málaga, Málaga, Spain; Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Isabel Brichette-Mieg
- Neuroimmunology and Neuroinflammation group. Biomedical Research Institute of Málaga-IBIMA Plataforma Bionand Hospital Regional Universitario de Málaga, Málaga, Spain
| | - MdM Fernández-Arjona
- Neuroimmunology and Neuroinflammation group. Biomedical Research Institute of Málaga-IBIMA Plataforma Bionand Hospital Regional Universitario de Málaga, Málaga, Spain
| | - José Luis Rodríguez-Bada
- Neuroimmunology and Neuroinflammation group. Biomedical Research Institute of Málaga-IBIMA Plataforma Bionand Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Yolanda López-Moreno
- Neuroimmunology and Neuroinflammation group. Biomedical Research Institute of Málaga-IBIMA Plataforma Bionand Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Pedro Serrano-Castro
- Neuroimmunology and Neuroinflammation group. Biomedical Research Institute of Málaga-IBIMA Plataforma Bionand Hospital Regional Universitario de Málaga, Málaga, Spain; Red Andaluza de Investigación Clínica y Traslacional en Neurología (Neuro-RECA), Málaga, Spain; Department of Medicine and Dermatology, Faculty of Medicine, University of Málaga, Málaga, Spain
| | | | - Nicolás Lundahl Ciano-Petersen
- Neuroimmunology and Neuroinflammation group. Biomedical Research Institute of Málaga-IBIMA Plataforma Bionand Hospital Regional Universitario de Málaga, Málaga, Spain; Red Andaluza de Investigación Clínica y Traslacional en Neurología (Neuro-RECA), Málaga, Spain.
| | - Begoña Oliver-Martos
- Neuroimmunology and Neuroinflammation group. Biomedical Research Institute of Málaga-IBIMA Plataforma Bionand Hospital Regional Universitario de Málaga, Málaga, Spain; Facultad de Medicina, Universidad de Málaga, Málaga, Spain; Department of Cell Biology, Genetics and Physiology, Physiology Area, Faculty of Science University of Malaga, Málaga, Spain.
| |
Collapse
|
2
|
Gao L, Li L, Zhang D, Qiu J, Qian J, Liu H. TAPI-1 Exhibits Anti-tumor Efficacy in Human Esophageal Squamous Cell Carcinoma Cells via Suppression of NF-κB Signaling Pathway. Dig Dis Sci 2024; 69:81-94. [PMID: 38007701 PMCID: PMC10787672 DOI: 10.1007/s10620-023-08181-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 11/02/2023] [Indexed: 11/28/2023]
Abstract
BACKGROUND TNF-α processing inhibitor-1 (TAPI-1) is a known metalloproteinase inhibitor with potential anti-inflammatory effects. However, its anti-cancer effects on esophageal squamous cell carcinoma (ESCC) have not been uncovered. AIM In the present study, the effects of TAPI-1 on ESCC cell viability, migration, invasion, and cisplatin resistance and the underlying molecular mechanisms were investigated in TE-1 and Eca109 cells. METHODS To this end, TE-1 and Eca109 cells were exposed to TAPI-1 for indicated time intervals. Cell viability was assessed using cell counting kit-8 assay and apoptosis was evaluated using flow cytometry assay. Migration and invasion were assessed using Transwell assays. Gene expressions were analyzed using quantitative reverse transcription polymerase chain reaction. The activation of NF-κB signaling pathway was elucidated via Western blot and chromatin immunoprecipitation assay. RESULTS We observed that higher doses (10, 20 μM) of TAPI-1 inhibited ESCC cell viability, while a lower dose (5 μM) of TAPI-1 inhibited ESCC cell migration and invasion and enhanced the chemosensitivity of ESCC cells to cisplatin. Moreover, TAPI-1 suppressed the activation of NF-κB signaling and the target genes expression in the stage of transcription initiation. Furthermore, blocking NF-κB signaling in advance could abolish all the effects of TAPI-1 on ESCC cells. CONCLUSION Overall, these results indicated that TAPI-1 impairs ESCC cell viability, migration, and invasion and facilitates cisplatin-induced apoptosis via suppression of NF-κB signaling pathway. TAPI-1 may serve as a potential adjuvant agent with cisplatin for ESCC therapy.
Collapse
Affiliation(s)
- Lin Gao
- Medical Research Center, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, People's Republic of China
| | - Li Li
- Department of Pathology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Shengli Road No. 666, Nantong, 226001, Jiangsu, People's Republic of China
| | - Dongmei Zhang
- Medical Research Center, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, People's Republic of China
| | - Jianwei Qiu
- Department of Gastroenterology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, People's Republic of China
| | - Junbo Qian
- Department of Gastroenterology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, People's Republic of China
| | - Hongbin Liu
- Department of Pathology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Shengli Road No. 666, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
3
|
Amsden H, Kourko O, Roth M, Gee K. Antiviral Activities of Interleukin-27: A Partner for Interferons? Front Immunol 2022; 13:902853. [PMID: 35634328 PMCID: PMC9134790 DOI: 10.3389/fimmu.2022.902853] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/14/2022] [Indexed: 12/17/2022] Open
Abstract
Emergence of new, pandemic-level viral threats has brought to the forefront the importance of viral immunology and continued improvement of antiviral therapies. Interleukin-27 (IL-27) is a pleiotropic cytokine that regulates both innate and adaptive immune responses. Accumulating evidence has revealed potent antiviral activities of IL-27 against numerous viruses, including HIV, influenza, HBV and more. IL-27 contributes to the immune response against viruses indirectly by increasing production of interferons (IFNs) which have various antiviral effects. Additionally, IL-27 can directly interfere with viral infection both by acting similarly to an IFN itself and by modulating the differentiation and function of various immune cells. This review discusses the IFN-dependent and IFN-independent antiviral mechanisms of IL-27 and highlights the potential of IL-27 as a therapeutic cytokine for viral infection.
Collapse
Affiliation(s)
| | | | | | - Katrina Gee
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| |
Collapse
|
4
|
García-Juárez M, Camacho-Morales A. Defining the role of anti- and pro-inflammatory outcomes of Interleukin-6 in mental health. Neuroscience 2022; 492:32-46. [DOI: 10.1016/j.neuroscience.2022.03.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/03/2022] [Accepted: 03/16/2022] [Indexed: 01/03/2023]
|
5
|
Villazana-Kretzer DL, Wuertz KM, Newhouse D, Damicis JR, Dornisch EM, Voss KM, Muruato AE, Paymaster JA, Schmiedecke SS, Edwards SM, Napolitano PG, Tisoncik-Go J, Ieronimakis N, Gale M. ZIKV can infect human term placentas in the absence of maternal factors. Commun Biol 2022; 5:243. [PMID: 35304593 PMCID: PMC8933440 DOI: 10.1038/s42003-022-03158-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 02/09/2022] [Indexed: 12/05/2022] Open
Abstract
Zika virus infection can result in devastating pregnancy outcomes when it crosses the placental barrier. For human pregnancies, the mechanisms of vertical transmission remain enigmatic. Utilizing a human placenta-cotyledon perfusion model, we examined Zika virus exposure in the absence of maternal factors. To distinguish responses related to viral infection vs. recognition, we evaluated cotyledons perfused with either active or inactivated Zika virus. Active Zika virus exposure resulted in infection, cell death and syncytium injury. Pathology corresponded with transcriptional changes related to inflammation and innate immunity. Inactive Zika virus exposure also led to syncytium injury and related changes in gene expression but not cell death. Our observations reveal pathologies and innate immune responses that are dependent on infection or virus placenta interactions independent of productive infection. Importantly, our findings indicate that Zika virus can infect and compromise placentas in the absence of maternal humoral factors that may be protective. Villazana-Kretzer et al. compare histology, physiology and gene expression in cotyledons from term placentas perfused with either active or UV-inactivated Zika virus. They show that ZIKV can infect human term placentas in the absence of maternal factors and identify unique transcriptional responses to active ZIKA virus.
Collapse
Affiliation(s)
| | - Kathryn McGuckin Wuertz
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Daniel Newhouse
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Jennifer R Damicis
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA, USA
| | - Elisabeth M Dornisch
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA, USA
| | - Kathleen M Voss
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Antonio E Muruato
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Jennifer A Paymaster
- Division of Maternal Fetal Medicine, Madigan Army Medical Center, Tacoma, WA, USA
| | - Stacey S Schmiedecke
- Division of Maternal Fetal Medicine, Madigan Army Medical Center, Tacoma, WA, USA
| | - Sarah M Edwards
- Division of Maternal Fetal Medicine, Madigan Army Medical Center, Tacoma, WA, USA
| | - Peter G Napolitano
- Department of OB/GYN, University of Washington Medical Center, Seattle, WA, USA
| | - Jennifer Tisoncik-Go
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Nicholas Ieronimakis
- Division of Maternal Fetal Medicine, Madigan Army Medical Center, Tacoma, WA, USA. .,Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA, USA.
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
6
|
Zuo Q, Cheng Z, Zhang G, Xia Y, Xu G, Cao W, Yang X, Fu Y, He R, Fang P, Guo Y, Nie L, Huang Y, Liu L, Zhan J, Liu S, Zhu Y. Role of IL-6-IL-27 Complex in Host Antiviral Immune Response. THE JOURNAL OF IMMUNOLOGY 2021; 207:577-589. [PMID: 34145061 DOI: 10.4049/jimmunol.2100179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/10/2021] [Indexed: 11/19/2022]
Abstract
The IL family of cytokines participates in immune response and regulation. We previously found that soluble IL-6 receptor plays an important role in the host antiviral response. In this study, we detected the IL-6-IL-27 complex in serum and throat swab samples from patients infected with influenza A virus. A plasmid expressing the IL-6-IL-27 complex was constructed to explore its biological function. The results indicated that the IL-6-IL-27 complex has a stronger antiviral effect than the individual subunits of IL-6, IL-27A, and EBV-induced gene 3. Furthermore, the activity of the IL-6-IL-27 complex is mainly mediated by the IL-27A subunit and the IL-27 receptor α. The IL-6-IL-27 complex can positively regulate virus-triggered expression of IFN and IFN-stimulated genes by interacting with adaptor protein mitochondrial antiviral signaling protein, potentiating the ubiquitination of TNF receptor-associated factors 3 and 6 and NF-κB nuclear translocation. The secreted IL-6-IL-27 complex can induce the phosphorylation of STAT1 and STAT3 and shows antiviral activity. Our results demonstrate a previously unrecognized mechanism by which IL-6, IL-27A, and EBV-induced gene 3 form a large complex both intracellularly and extracellularly, and this complex acts in the host antiviral response.
Collapse
Affiliation(s)
- Qi Zuo
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China; and
| | - Zhikui Cheng
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China; and
| | - Guoqing Zhang
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China; and
| | - Yongfang Xia
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China; and
| | - Gang Xu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China; and
| | - Wei Cao
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China; and
| | - Xiaodan Yang
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China; and
| | - Yundong Fu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China; and
| | - Rui He
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China; and
| | - Peining Fang
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China; and
| | - Yifei Guo
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China; and
| | - Longyu Nie
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China; and
| | - Yu Huang
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China; and
| | - Lin Liu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China; and
| | - Jianbo Zhan
- Institute of Health Inspection and Testing, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Shi Liu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China; and
| | - Ying Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China; and
| |
Collapse
|
7
|
Birzer A, Krawczyk A, Draßner C, Kuhnt C, Mühl-Zürbes P, Heilingloh CS, Steinkasserer A, Popella L. HSV-1 Modulates IL-6 Receptor Expression on Human Dendritic Cells. Front Immunol 2020; 11:1970. [PMID: 32983130 PMCID: PMC7479228 DOI: 10.3389/fimmu.2020.01970] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022] Open
Abstract
Dendritic cells (DCs) are the guardians of the immune system since they are located in the majority of peripheral tissues. In addition, they are crucial for the induction of an effective immune response based on their unique capacity to stimulate naive T cells. During co-evolution, the human pathogen herpes simplex virus type 1 (HSV-1) has evolved several immune evasion mechanisms in order to subvert the host's immune system especially by targeting DC biology and function. Here we demonstrate that HSV-1 infection influences the IL-6 receptor (IL6R) expression both on protein and mRNA levels in/on human monocyte-derived mature DCs (mDCs). Surprisingly, reduced IL6R expression levels were also observed on uninfected bystander mDCs. Mechanistically, we clearly show that HSV-1-derived non-infectious light (L-) particles are sufficient to trigger IL6R regulation on uninfected bystander mDCs. These L-particles lack the viral DNA-loaded capsid and are predominantly produced during infection of mDCs. Our results show that the deletion of the HSV-1 tegument protein vhs partially rescued the reduced IL6R surface expression levels on/in bystander mDCs. Using a neutralizing antibody, which perturbs the transfer of L-particles to bystander mDCs, was sufficient to rescue the modulation of IL6R surface expression on uninfected bystander mDCs. This study provides evidence that L-particles transfer specific viral proteins to uninfected bystander mDCs, thereby negatively interfering with their IL6R expression levels, however, to a lesser extend compared to H-particles. Due to their immune-modulatory capacity, L-particles represent an elaborated approach of HSV-1-mediated immune evasion.
Collapse
Affiliation(s)
- Alexandra Birzer
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Adalbert Krawczyk
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christina Draßner
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christine Kuhnt
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Petra Mühl-Zürbes
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christiane Silke Heilingloh
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | - Linda Popella
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
8
|
Eribo OA, Leqheka MS, Malherbe ST, McAnda S, Stanley K, van der Spuy GD, Walzl G, Chegou NN. Host urine immunological biomarkers as potential candidates for the diagnosis of tuberculosis. Int J Infect Dis 2020; 99:473-481. [PMID: 32800854 DOI: 10.1016/j.ijid.2020.08.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/29/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To investigate the potential of host urinary biomarkers as diagnostic candidates for tuberculosis (TB). METHODS Adults self-presenting with symptoms requiring further investigation for TB were enrolled in Cape Town, South Africa. Participants were later classified as having TB or other respiratory diseases (ORD) using results from TB confirmatory tests. The concentrations of 29 analytes were evaluated in urine samples from participants using the Luminex platform, and their diagnostic potential was assessed using standard statistical approaches. RESULTS Of the 151 study participants, 34 (22.5%) were diagnosed with TB and 26 (17.2%) were HIV-positive. Seven biomarkers showed potential as TB diagnostic candidates, with accuracy improving (in HIV-positives) when stratified according to HIV status (area under the receiver operating characteristics curve; AUC ≥0.80). In HIV-positive participants, a four-marker biosignature (sIL6R, MMP-9, IL-2Ra, IFN-γ) diagnosed TB with AUC of 0.96, sensitivity of 85.7% (95% confidence interval (CI) 42.1-99.6%), and specificity of 94.7% (95% CI 74.0-99.9%). In HIV-negatives, the most promising was a two-marker biosignature (sIL6R and sIL-2Ra), which diagnosed TB with AUC of 0.76, sensitivity of 53.9% (95% CI 33.4-73.4%), and specificity of 79.6% (95% CI 70.3-87.1%). CONCLUSIONS Urinary host inflammatory biomarkers possess TB diagnostic potential but may be influenced by HIV infection. The results of this study require validation in larger studies.
Collapse
Affiliation(s)
- Osagie A Eribo
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O. Box 241, Cape Town 8000, South Africa.
| | - Monkoe S Leqheka
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O. Box 241, Cape Town 8000, South Africa.
| | - Stephanus T Malherbe
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O. Box 241, Cape Town 8000, South Africa.
| | - Shirley McAnda
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O. Box 241, Cape Town 8000, South Africa.
| | - Kim Stanley
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O. Box 241, Cape Town 8000, South Africa.
| | - Gian D van der Spuy
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O. Box 241, Cape Town 8000, South Africa.
| | - Gerhard Walzl
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O. Box 241, Cape Town 8000, South Africa.
| | - Novel N Chegou
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O. Box 241, Cape Town 8000, South Africa.
| |
Collapse
|
9
|
Hurtado-Guerrero I, Hernáez B, Pinto-Medel MJ, Calonge E, Rodriguez-Bada JL, Urbaneja P, Alonso A, Mena-Vázquez N, Aliaga P, Issazadeh-Navikas S, Pavia J, Leyva L, Alcamí J, Alcamí A, Fernández Ó, Oliver-Martos B. Antiviral, Immunomodulatory and Antiproliferative Activities of Recombinant Soluble IFNAR2 without IFN-ß Mediation. J Clin Med 2020; 9:jcm9040959. [PMID: 32244308 PMCID: PMC7230527 DOI: 10.3390/jcm9040959] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/26/2020] [Accepted: 03/29/2020] [Indexed: 12/21/2022] Open
Abstract
Soluble receptors of cytokines are able to modify cytokine activities and therefore the immune system, and some have intrinsic biological activities without mediation from their cytokines. The soluble interferon beta (IFN-ß) receptor is generated through alternative splicing of IFNAR2 and has both agonist and antagonist properties for IFN-ß, but its role is unknown. We previously demonstrated that a recombinant human soluble IFN-ß receptor showed intrinsic therapeutic efficacy in a mouse model of multiple sclerosis. Here we evaluate the potential biological activities of recombinant sIFNAR2 without the mediation of IFN-ß in human cells. Recombinant sIFNAR2 down-regulated the production of IL-17 and IFN-ɣ and reduced the cell proliferation rate. Moreover, it showed a strong antiviral activity, fully protecting the cell monolayer after being infected by the virus. Specific inhibitors completely abrogated the antiviral activity of IFN-ß, but not that of the recombinant sIFNAR2, and there was no activation of the JAK-STAT signaling pathway. Consequently, r-sIFNAR2 exerts immunomodulatory, antiproliferative and antiviral activities without IFN-ß mediation, and could be a promising treatment against viral infections and immune-mediated diseases.
Collapse
Affiliation(s)
- Isaac Hurtado-Guerrero
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
- Neuroinflammation Unit, Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, Copenhagen Biocentre, University of Copenhagen, 2200 Copenhagen, Denmark;
| | - Bruno Hernáez
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain; (B.H.); (A.A.)
| | - María J. Pinto-Medel
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
| | - Esther Calonge
- AIDS Immunopathology Unit, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda 28220 Madrid, Spain; (E.C.); (J.A.)
| | - José L. Rodriguez-Bada
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
| | - Patricia Urbaneja
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
| | - Ana Alonso
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
| | - Natalia Mena-Vázquez
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC de Reumatología, Hospital Regional Universitario de Málaga, 29009 Málaga, Spain
| | - Pablo Aliaga
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Shohreh Issazadeh-Navikas
- Neuroinflammation Unit, Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, Copenhagen Biocentre, University of Copenhagen, 2200 Copenhagen, Denmark;
| | - José Pavia
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- Departamento de Farmacología y Pediatría, Facultad de Medicina. Universidad de Málaga, 29010 Málaga, Spain
| | - Laura Leyva
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
| | - José Alcamí
- AIDS Immunopathology Unit, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda 28220 Madrid, Spain; (E.C.); (J.A.)
- HIV Unit, Infectious Disease Service, Hospital Universitari de Bellvitge, 08907 Barcelona, Spain
| | - Antonio Alcamí
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain; (B.H.); (A.A.)
| | - Óscar Fernández
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- Departamento de Farmacología y Pediatría, Facultad de Medicina. Universidad de Málaga, 29010 Málaga, Spain
| | - Begoña Oliver-Martos
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
- Correspondence: ; Tel.: +34-951-290-223
| |
Collapse
|
10
|
Sultana S, Bishayi B. Potential anti-arthritic and anti-inflammatory effects of TNF-α processing inhibitor-1 (TAPI-1): A new approach to the treatment of S. aureus arthritis. Immunobiology 2019; 225:151887. [PMID: 31822434 DOI: 10.1016/j.imbio.2019.11.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/26/2019] [Indexed: 12/22/2022]
Abstract
Treatment of septic arthritis has become more challenging due to the rise of multidrug resistant strains of Staphylococcus aureus (S. aureus) in recent years. Failure of antibiotic therapies has compelled to initiate the search for new alternatives. This study aimed to unveil the potential anti-arthritic effects of TAPI-1 (TNF-α processing inhibitor-1), an inhibitor that inhibits TACE (TNF-α converting enzyme) mediated release of soluble TNF-α and its receptors along with attenuation of other inflammatory and joint destructive factors responsible for the progression of arthritis. Male Swiss albino mice were inoculated with live S. aureus (5 × 106 cells/mouse) for the development of septic arthritis. TAPI-1 was administered intraperitoneally (10 mg/kg body weight) post S. aureus infection at regular intervals. Throughout the experiment, the severity of arthritis was obtained to be significantly low after TAPI-1 administration. Arthritis index and histopathology confirmed effectiveness of TAPI-1 in mitigating inflammation induced paw swelling and less bone-cartilage destruction in the arthritic knee joints. Lower levels of soluble tumor necrosis factor alpha (sTNF-α) and soluble tumor necrosis factor alpha receptor-1 (sTNFR-1) were detected in the TAPI-1 treated group suggesting TAPI-1 mediated blocking of TACE with subsequent inhibition of TNF-α signalling. Treatment with TAPI-1 lowered the levels of reactive species; matrix metalloproteinase-2 (MMP-2), receptor activator of nuclear factor kappa-B ligand (RANKL) and osteopontin (OPN) denoting less matrix degradation and less osteoclastic bone resorption. Together, this experimental work authenticates TAPI-1 as an alternative therapeutic intervention for the treatment of S. aureus arthritis.
Collapse
Affiliation(s)
- Sahin Sultana
- Department of Physiology, Immunology and Microbiology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700 009, West Bengal, India
| | - Biswadev Bishayi
- Department of Physiology, Immunology and Microbiology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700 009, West Bengal, India.
| |
Collapse
|
11
|
Zhou Y, Xia Z, Cheng Z, Xu G, Yang X, Liu S, Zhu Y. Inducible microRNA-590-5p inhibits host antiviral response by targeting the soluble interleukin-6 (IL6) receptor. J Biol Chem 2018; 293:18168-18179. [PMID: 30291142 DOI: 10.1074/jbc.ra118.005057] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/21/2018] [Indexed: 01/09/2023] Open
Abstract
MicroRNA (miR)-590-5p has been identified as an important regulator of some signaling pathways such as cell proliferation and tumorigenesis. However, little is known about its role during viral infection. Here, we report that miR-590-5p was significantly induced by various viruses and effectively potentiated virus replication in different viral infection systems. Furthermore, miR-590-5p substantially attenuated the virus-induced expression of type I and type III interferons (IFNs) and inflammatory cytokines, resulting in impaired downstream antiviral signaling. Interleukin-6 receptor (IL6R) was identified as a target of miR-590-5p. Interestingly, the role of miR-590-5p in virus-triggered signaling was abolished in IL6R knockout cells, and this could be rescued by restoring the expression of the soluble IL6R (sIL6R) but not the membrane-bound IL6R (mIL6R), suggesting that sIL6R is indispensable for miR-590-5p in modulating the host antiviral response. Furthermore, miR-590-5p down-regulated endogenous sIL6R and mIL6R expression through a translational repression mechanism. These findings thus uncover a previously uncharacterized role and the underlying mechanism of miR-590-5p in the innate immune response to viral infection.
Collapse
Affiliation(s)
- Yaqin Zhou
- From the State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Zhangchuan Xia
- From the State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Zhikui Cheng
- From the State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Gang Xu
- From the State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Xiaodan Yang
- From the State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Shi Liu
- From the State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Ying Zhu
- From the State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China.
| |
Collapse
|
12
|
Fischer T, Spohn M, Olearo F, Zinser ME, Kasonta R, Stubbe HC, Rechtien A, Ly ML, Schmiedel S, Lohse AW, Grundhoff A, Addo MM, Dahlke C. Dynamic changes of circulating miRNAs induced by the Ebola virus vaccine VSV-EBOV. Vaccine 2018; 36:7083-7094. [PMID: 30244872 DOI: 10.1016/j.vaccine.2018.09.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/02/2018] [Accepted: 09/08/2018] [Indexed: 12/18/2022]
Abstract
VSV-EBOV is a replication-competent Ebola virus (EBOV) vaccine, which was tested in clinical trials as response to the Ebola virus disease (EVD) outbreak 2013-2016. It is the most advanced EBOV candidate currently in the licensure process. The experimental vaccine was again administered as response to outbreaks in the Democratic Republic of Congo. However, underlying molecular mechanisms that convey protection remain incompletely understood. MicroRNAs (miRNAs) are known key regulators that influence gene expression on a post-transcriptional level. The miRNA-mediated control has emerged as a critical regulatory principle in the immune system, which strongly influences the balance of innate and adaptive immune responses by modulation of signaling pathways critical for differentiation of immune cells. We investigated expression levels of circulating miRNAs (c-miRNAs) in plasma from healthy vaccinees, as they may reflect cellular dynamics following VSV-EBOV immunization and additionally may serve as potential biomarkers for vaccine efficacy. As part of the WHO-led VEBCON consortium, we investigated safety and immunogenicity of VSV-EBOV in a phase I trial. A comprehensive analysis of expression levels on c-miRNAs from plasma samples following VSV-EBOV immunization (day 0, 1, 3 post vaccination) was conducted using RT-qPCR assays. Potential biological relevance was assessed using in silico analyses. Additionally, we correlated dynamics of miRNA expressions with our previously reported data on vaccine-induced antibody and cytokine responses and finally evaluated the prognostic power by generating ROC curves. We identified four promising miRNAs (hsa-miR-146a, hsa-miR-126, hsa-miR-199a, hsa-miR-484), showing a strong association with adaptive immune responses, exhibited favourable prognostic performance and are implicated in immunology-related functions. Our results provide evidence that miRNAs may serve as useful biomarkers for prediction of vaccine-induced immunogenicity. Furthermore, our unique data set provides insight into molecular mechanisms that underlie VSV-EBOV-mediated protective immune responses, which may help to decipher VSV-EBOV immune signature and accelerate strategic vaccine design or personalized approaches.
Collapse
Affiliation(s)
- T Fischer
- University Medical Center Hamburg-Eppendorf (UKE), 1st Department of Medicine, Hamburg, Germany; German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems, Germany
| | - M Spohn
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - F Olearo
- University Medical Center Hamburg-Eppendorf (UKE), 1st Department of Medicine, Hamburg, Germany
| | - M E Zinser
- University Medical Center Hamburg-Eppendorf (UKE), 1st Department of Medicine, Hamburg, Germany; German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems, Germany
| | - Rahel Kasonta
- University Medical Center Hamburg-Eppendorf (UKE), 1st Department of Medicine, Hamburg, Germany; German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems, Germany
| | - H C Stubbe
- Division of Infectious Diseases, Department of Medicine II, LMU, Munich, Germany
| | - A Rechtien
- University Medical Center Hamburg-Eppendorf (UKE), 1st Department of Medicine, Hamburg, Germany; German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems, Germany; Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - M L Ly
- University Medical Center Hamburg-Eppendorf (UKE), 1st Department of Medicine, Hamburg, Germany; German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems, Germany; University Medical Center Hamburg-Eppendorf (UKE), Division of Infectious Diseases, Hamburg, Germany
| | - S Schmiedel
- University Medical Center Hamburg-Eppendorf (UKE), 1st Department of Medicine, Hamburg, Germany; University Medical Center Hamburg-Eppendorf (UKE), Division of Infectious Diseases, Hamburg, Germany
| | - A W Lohse
- University Medical Center Hamburg-Eppendorf (UKE), 1st Department of Medicine, Hamburg, Germany; German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems, Germany
| | - A Grundhoff
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | | | - M M Addo
- University Medical Center Hamburg-Eppendorf (UKE), 1st Department of Medicine, Hamburg, Germany; German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems, Germany; University Medical Center Hamburg-Eppendorf (UKE), Division of Infectious Diseases, Hamburg, Germany.
| | - C Dahlke
- University Medical Center Hamburg-Eppendorf (UKE), 1st Department of Medicine, Hamburg, Germany; German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems, Germany; University Medical Center Hamburg-Eppendorf (UKE), Division of Infectious Diseases, Hamburg, Germany.
| |
Collapse
|
13
|
Chang Z, Wang Y, Bian L, Liu Q, Long JE. Enterovirus 71 antagonizes the antiviral activity of host STAT3 and IL-6R with partial dependence on virus-induced miR-124. J Gen Virol 2017; 98:3008-3025. [PMID: 29120300 DOI: 10.1099/jgv.0.000967] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Enterovirus 71 (EV71) has caused major outbreaks of hand, foot and mouth disease. EV71 infections increase the production of many host cytokines and pro-inflammatory factors, including interleukin (IL)-6, IL-10 and COX-2. Some of these molecules could stimulate the signal transducer and activator of transcription 3 (STAT3), which plays a key role in regulating host immune responses and several viral diseases. However, the role of STAT3 in EV71 infection remains unknown. This study found that the phosphorylation levels of STAT3 (pY705-STAT3) are closely related to EV71 infection. Further experiments revealed that STAT3 exerts an anti-EV71 activity. However, the antiviral activity of STAT3 is partially antagonized by EV71-induced miR-124, which directly targets STAT3 mRNA. Similarly, IL-6R, the α-subunit of the IL-6 receptor complex, exhibits anti-EV71 activity and is directly targeted by the virus-induced miR-124. These results indicate that EV71 can evade host IL-6R- and STAT3-mediated antiviral activities by EV71-induced miR-124. This suggests that controlling miR-124 and the downstream targets, IL-6R and STAT3, might benefit the antiviral treatment of EV71 infection.
Collapse
Affiliation(s)
- Zhangmei Chang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| | - Yan Wang
- Department of Medical Microbiology and Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China
| | - Liang Bian
- Department of Medical Microbiology and Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China
| | - Qingqing Liu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| | - Jian-Er Long
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China.,Department of Medical Microbiology and Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China
| |
Collapse
|
14
|
Shiels MS, Shu XO, Chaturvedi AK, Gao YT, Xiang YB, Cai Q, Hu W, Shelton G, Ji BT, Pinto LA, Kemp TJ, Rothman N, Zheng W, Hildesheim A, Lan Q. A prospective study of immune and inflammation markers and risk of lung cancer among female never smokers in Shanghai. Carcinogenesis 2017; 38:1004-1010. [PMID: 28981818 DOI: 10.1093/carcin/bgx075] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 07/15/2017] [Indexed: 12/17/2022] Open
Abstract
There is a paucity of data on risk factors for lung cancer among never smokers. Here, we have carried out the first large study of circulating inflammation markers and lung cancer risk among female never smokers in Shanghai. A study of 248 lung cancer cases in female never smokers and 263 controls was nested within the Shanghai Women's Health Study (n = 75221), matched by dates of birth and blood collection (mean follow-up time = 7.5 years). Prediagnostic plasma levels of 65 inflammation markers were measured using a Luminex bead-based assay. Odds ratios (ORs) were estimated with multivariable logistic regression. Nine of 61 evaluable markers were statistically significantly associated with lung cancer risk among never smoking Chinese women (P-trend across categories <0.05). Soluble interleukin-6 receptor [sIL-6R; highest versus lowest category OR = 2.37; 95% confidence interval (CI) 1.40-4.02) and chemokine (C-C motif) ligand 2/monocyte chemotactic protein 1; (OR = 1.62; 95% CI 0.94-2.80) were associated with an increased risk of lung cancer, whereas interleukin (IL)-21 (OR = 0.53; 95%CI 0.31-0.93), chemokine (C-X3-C motif) ligand 1/fractalkine (OR = 0.54; 95% CI 0.30-0.96), soluble vascular endothelial growth factor receptor 2 (sVEGFR2, OR = 0.45; 95% CI 0.26-0.76), sVEGFR3 (OR = 0.53; 95% CI 0.32-0.90), soluble tumor necrosis factor receptor I (OR = 0.49; 95% CI 0.29-0.83), IL-10 (OR = 0.60; 95% CI 0.34-1.05) and C-reactive protein (OR = 0.63; 95% CI 0.37-1.06) were associated with a decreased risk. sIL-6R remained significantly associated with lung cancer risk >7.5 years prior to diagnosis. Markers involved in various aspects of the immune response were associated with subsequent lung cancer risk, implicating inflammation in the etiology of lung cancer among female never smokers.
Collapse
Affiliation(s)
- Meredith S Shiels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Anil K Chaturvedi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Yu-Tang Gao
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China
| | - Yong-Bing Xiang
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China
| | - Qiuyin Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Wei Hu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Gloriana Shelton
- HPV Immunology Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc, Frederick, MD, USA
| | - Bu-Tian Ji
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Ligia A Pinto
- HPV Immunology Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc, Frederick, MD, USA
| | - Troy J Kemp
- HPV Immunology Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc, Frederick, MD, USA
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Allan Hildesheim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Qing Lan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| |
Collapse
|
15
|
Xi Y, Troy NM, Anderson D, Pena OM, Lynch JP, Phipps S, Bosco A, Upham JW. Critical Role of Plasmacytoid Dendritic Cells in Regulating Gene Expression and Innate Immune Responses to Human Rhinovirus-16. Front Immunol 2017; 8:1351. [PMID: 29118754 PMCID: PMC5660993 DOI: 10.3389/fimmu.2017.01351] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/03/2017] [Indexed: 11/16/2022] Open
Abstract
Though human rhinoviruses (HRVs) are usually innocuous viruses, they can trigger serious consequences in certain individuals, especially in the setting of impaired interferon (IFN) synthesis. Plasmacytoid dendritic cells (pDCs) are key IFN producing cells, though we know little about the role of pDC in HRV-induced immune responses. Herein, we used gene expression microarrays to examine HRV-activated peripheral blood mononuclear cells (PBMCs) from healthy people, in combination with pDC depletion, to assess whether observed gene expression patterns were pDC dependent. As expected, pDC depletion led to a major reduction in IFN-α release. This was associated with profound differences in gene expression between intact PBMC and pDC-depleted PBMC, and major changes in upstream regulators: 70–80% of the HRV activated genes appeared to be pDC dependent. Real-time PCR confirmed key changes in gene expression, in which the following selected genes were shown to be highly pDC dependent: the transcription factor IRF7, both IL-27 chains (IL-27p28 and EBI3), the alpha chain of the IL-15 receptor (IL-15RA) and the IFN-related gene IFI27. HRV-induced IL-6, IFN-γ, and IL-27 protein synthesis were also highly pDC dependent. Supplementing pDC-depleted cultures with recombinant IL-15, IFN-γ, IL-27, or IL-6 was able to restore the IFN-α response, thereby compensating for the absence of pDC. Though pDC comprise only a minority population of migratory leukocytes, our findings highlight the profound extent to which these cells contribute to the immune response to HRV.
Collapse
Affiliation(s)
- Yang Xi
- Lung and Allergy Research Center, Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Niamh M Troy
- Systems Immunology, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Denise Anderson
- Systems Immunology, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Olga M Pena
- Lung and Allergy Research Center, Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Jason P Lynch
- Respiratory Immunology Group, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Simon Phipps
- Respiratory Immunology Group, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Anthony Bosco
- Systems Immunology, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - John W Upham
- Lung and Allergy Research Center, Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia.,Department of Respiratory Medicine, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| |
Collapse
|
16
|
Yücel G, Zhao Z, El-Battrawy I, Lan H, Lang S, Li X, Buljubasic F, Zimmermann WH, Cyganek L, Utikal J, Ravens U, Wieland T, Borggrefe M, Zhou XB, Akin I. Lipopolysaccharides induced inflammatory responses and electrophysiological dysfunctions in human-induced pluripotent stem cell derived cardiomyocytes. Sci Rep 2017; 7:2935. [PMID: 28592841 PMCID: PMC5462745 DOI: 10.1038/s41598-017-03147-4] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/24/2017] [Indexed: 12/19/2022] Open
Abstract
Severe infections like sepsis lead frequently to cardiomyopathy. The mechanisms are unclear and an optimal therapy for septic cardiomyopathy still lacks. The aim of this study is to establish an endotoxin-induced inflammatory model using human induced pluripotent stem cell (hiPSC) derived cardiomyocytes (hiPSC-CMs) for mechanistic and therapeutic studies. hiPSC-CMs were treated by lipopolysaccharide (LPS) in different concentrations for different times. ELISA, FACS, qPCR, and patch-clamp techniques were used for the study. TLR4 (Toll-like receptor 4) and its associated proteins, CD14, LBP (lipopolysaccharide binding protein), TIRAP (toll-interleukin 1 receptor domain containing adaptor protein), Ly96 (lymphocyte antigen 96) and nuclear factor kappa B as well as some pro-and anti-inflammatory factors are expressed in hiPSC-CMs. LPS-treatment for 6 hours increased the expression levels of pro-inflammatory and chemotactic cytokines (TNF-a, IL-1ß, IL-6, CCL2, CCL5, IL-8), whereas 48 hour-treatment elevated the expression of anti-inflammatory factors (IL-10 and IL-6). LPS led to cell injury resulting from exaggerated cell apoptosis and necrosis. Finally, LPS inhibited small conductance Ca2+-activated K+ channel currents, enhanced Na+/Ca2+-exchanger currents, prolonged action potential duration, suggesting cellular electrical dysfunctions. Our data demonstrate that hiPSC-CMs possess the functional reaction system involved in endotoxin-induced inflammation and can model some bacterium-induced inflammatory responses in cardiac myocytes.
Collapse
Affiliation(s)
- Gökhan Yücel
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim, Göttingen, Germany
| | - Zhihan Zhao
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim, Göttingen, Germany
| | - Ibrahim El-Battrawy
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim, Göttingen, Germany
| | - Huan Lan
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Siegfried Lang
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim, Göttingen, Germany
| | - Xin Li
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Fanis Buljubasic
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim, Göttingen, Germany
| | - Wolfram-Hubertus Zimmermann
- DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim, Göttingen, Germany.,Institute of Pharmacology and Toxicology, University of Göttingen, Göttingen, Germany
| | - Lukas Cyganek
- DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim, Göttingen, Germany.,Stem Cell Unit, Heart Research Center Göttingen, Göttingen, Germany
| | - Jochen Utikal
- DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim, Göttingen, Germany.,Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ursula Ravens
- Institute of Experimental Cardiovascular Medicine, University Heart Centre Freiburg∙Bad Krozingen, Freiburg, Germany
| | - Thomas Wieland
- DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim, Göttingen, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Martin Borggrefe
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim, Göttingen, Germany
| | - Xiao-Bo Zhou
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany. .,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim, Göttingen, Germany. .,Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China.
| | - Ibrahim Akin
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim, Göttingen, Germany
| |
Collapse
|
17
|
Feng J, Cao Z, Wang L, Wan Y, Peng N, Wang Q, Chen X, Zhou Y, Zhu Y. Inducible GBP5 Mediates the Antiviral Response via Interferon-Related Pathways during Influenza A Virus Infection. J Innate Immun 2017; 9:419-435. [PMID: 28376501 DOI: 10.1159/000460294] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 02/08/2017] [Indexed: 12/18/2022] Open
Abstract
Guanylate binding protein (GBP) 5 belongs to the GBP family, which is involved in important cellular processes, including signal transduction, translation, vesicle trafficking, and exocytosis. Structurally, GBPs display a high degree of homology and share highly conserved GTP-binding or hydrolysis domains. GBP5 was reported to be a critical cellular factor in inflammasome assembly. However, little is known about its role in the host antiviral innate immune response. In this study, we found that GBP5 expression was significantly elevated in influenza patients and influenza A virus-infected A549 human lung epithelial cells. The overexpression of GBP5 inhibited virus replication by enhancing the expression of virus-induced interferon (IFN) and IFN-related effectors. Knockdown of GBP5 had the opposite effect. Moreover, GBP5 enhanced endogenous IFN expression by interacting with the NF-κB-essential modulator complex and stimulating NF-κB signaling. Additionally, the expression of proinflammatory factors, such as IL-6, IL-8, tumor necrosis factor-α, cyclooxygenase-2, and inducible nitric oxide synthase, was also activated by GBP5. Taken together, our results reveal that GBP5 inhibited virus replication through the activation of IFN signaling and proinflammatory factors.
Collapse
Affiliation(s)
- Jian Feng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Xia Z, Xu G, Yang X, Peng N, Zuo Q, Zhu S, Hao H, Liu S, Zhu Y. Inducible TAP1 Negatively Regulates the Antiviral Innate Immune Response by Targeting the TAK1 Complex. THE JOURNAL OF IMMUNOLOGY 2017; 198:3690-3704. [PMID: 28356387 DOI: 10.4049/jimmunol.1601588] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 03/03/2017] [Indexed: 12/17/2022]
Abstract
The innate immune response is critical for host defense and must be tightly controlled, but the molecular mechanisms responsible for its negative regulation are not yet completely understood. In this study, we report that transporter 1, ATP-binding cassette, subfamily B (TAP1), a virus-inducible endoplasmic reticulum-associated protein, negatively regulated the virus-triggered immune response. In this study, we observed upregulated expression of TAP1 following virus infection in human lung epithelial cells (A549), THP-1 monocytes, HeLa cells, and Vero cells. The overexpression of TAP1 enhanced virus replication by inhibiting the virus-triggered activation of NF-κB signaling and the production of IFNs, IFN-stimulated genes, and proinflammatory cytokines. TAP1 depletion had the opposite effect. In response to virus infection, TAP1 interacted with the TGF-β-activated kinase (TAK)1 complex and impaired the phosphorylation of TAK1, subsequently suppressing the phosphorylation of the IκB kinase complex and NF-κB inhibitor α (IκBα) as well as NF-κB nuclear translocation. Our findings collectively suggest that TAP1 plays a novel role in the negative regulation of virus-triggered NF-κB signaling and the innate immune response by targeting the TAK1 complex.
Collapse
Affiliation(s)
- Zhangchuan Xia
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Gang Xu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Xiaodan Yang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Nanfang Peng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Qi Zuo
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Shengli Zhu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Hua Hao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Shi Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Ying Zhu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
19
|
Mirhafez SR, Avan A, Tajfard M, Mohammadi S, Moohebati M, Fallah A, Ghazavi H, Savadi H, Ghayour Mobarhan M. Relationship between serum cytokines receptors and matrix metalloproteinase 9 levels and coronary artery disease. J Clin Lab Anal 2016; 31. [PMID: 27910999 DOI: 10.1002/jcla.22100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 10/29/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The prevalence of coronary artery disease (CAD) is increasing globally, supporting the need for the identification of novel biomarkers. Therefore in the present study, we have explored the association of SIL2A, SIL6R, STNFRI, STNFRII, and MMP9 in CAD patients. METHODS Twenty one patients with angiographically defined CAD with more than 50% occlusion, at least, in one coronary artery and twenty healthy subjects (n=20) without the history of cardiovascular symptoms were enrolled. Demographic and biochemical analysis (e.g. Total Cholesterol (TC), Triglyceride (TG), and HDL-C) were measured in all the subjects. The level of cytokines receptor (SIL2A, SIL6R, SIL6R, STNFRI, STNFRII, and matrix metallopeptidase 9 (MMP9) were evaluated. RESULTS Our results showed the higher level of MMP9 in patients group compared to the control subjects, while no significant differences were detected for other cytokines. In particular the level of MMP9 was significantly (P=.015) increased from 181.16 ng/mL (95%CI: 112.1-199.2) to 192.0 ng/mL (95%CI: 181.5-265.2). Moreover, the sensitivity and specificity of MMP9 were 95.45% and 45%, respectively, as detected by receiver operating characteristic (ROC) curves. CONCLUSION We demonstrate the significant correlation of MMP-9 with CAD with sensitivity of 95.45%, suggesting its role as a biomarker in CAD patients. Further studies in larger population - preferably multicenter setting - are warranted to explore the functional role of this marker in coronary artery disease.
Collapse
Affiliation(s)
- Seyed Reza Mirhafez
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran.,Cardiovascular Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Tajfard
- Department of Health Education and Health Promotion, School of Health, Mashhad University of Medical Science, Mashhad, Iran.,Management and Social Determinants of Health Research Center, School of Health, Mashhad University of Medical Science, Mashhad, Iran
| | - Shabnam Mohammadi
- Department of Basic Sciences, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.,Neurogenic Inflammation Research Centre, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Moohebati
- Cardiovascular Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arash Fallah
- Cardiovascular Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Ghazavi
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Savadi
- Department of Medicine, Mashhad Medical Sciences Branch, Islamic Azad University, Mashhad, Iran
| | - Majid Ghayour Mobarhan
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Biochemistry and Nutrition Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Systematic Identification and Bioinformatic Analysis of MicroRNAs in Response to Infections of Coxsackievirus A16 and Enterovirus 71. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4302470. [PMID: 27843944 PMCID: PMC5098103 DOI: 10.1155/2016/4302470] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 09/29/2016] [Indexed: 12/18/2022]
Abstract
Hand, foot, and mouth disease (HFMD), mainly caused by coxsackievirus A16 (CVA16) and enterovirus 71 (EV71) infections, remains a serious public health issue with thousands of newly diagnostic cases each year since 2008 in China. The mechanisms underlying viral infection, however, are elusive to date. In the present study, we systematically investigated the host cellular microRNA (miRNA) expression patterns in response to CVA16 and EV71 infections. Through microarray examination, 27 miRNAs (15 upregulated and 12 downregulated) were found to be coassociated with the replication process of two viruses, while the expression levels of 15 and 5 miRNAs were significantly changed in CVA16- and EV71-infected cells, respectively. A great number of target genes of 27 common differentially expressed miRNAs were predicted by combined use of two computational target prediction algorithms, TargetScan and MiRanda. Comprehensive bioinformatic analysis of target genes in GO categories and KEGG pathways indicated the involvement of diverse biological functions and signaling pathways during viral infection. These results provide an overview of the roles of miRNAs in virus-host interaction, which will contribute to further understanding of HFMD pathological mechanisms.
Collapse
|
21
|
Yang P, Lou X, Zheng Y, Shi W, Li Y, Dong Y, Han Y, Pang X, Liu S, Wang Q. Cytokines and chemokines in mild/asymptomatic cases infected with avian influenza A (H7N9) virus. J Med Microbiol 2016; 65:1232-1235. [DOI: 10.1099/jmm.0.000333] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Peng Yang
- Beijing Center for Disease Prevention and Control, Beijing Research Center for Preventive Medicine, School of Public Health, Capital Medical University, Beijing, PR China
| | - Xiaomin Lou
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, PR China
| | - Yang Zheng
- Beijing Center for Disease Prevention and Control, Beijing Research Center for Preventive Medicine, School of Public Health, Capital Medical University, Beijing, PR China
| | - Weixian Shi
- Beijing Center for Disease Prevention and Control, Beijing Research Center for Preventive Medicine, School of Public Health, Capital Medical University, Beijing, PR China
| | - Yang Li
- Beijing Protein Innovation, Beijing, PR China
| | | | - Yuning Han
- Beijing Protein Innovation, Beijing, PR China
| | - Xinghuo Pang
- Beijing Center for Disease Prevention and Control, Beijing Research Center for Preventive Medicine, School of Public Health, Capital Medical University, Beijing, PR China
| | - Siqi Liu
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, PR China
| | - Quanyi Wang
- Beijing Center for Disease Prevention and Control, Beijing Research Center for Preventive Medicine, School of Public Health, Capital Medical University, Beijing, PR China
| |
Collapse
|
22
|
Cao Z, Xia Z, Zhou Y, Yang X, Hao H, Peng N, Liu S, Zhu Y. Methylcrotonoyl-CoA carboxylase 1 potentiates RLR-induced NF-κB signaling by targeting MAVS complex. Sci Rep 2016; 6:33557. [PMID: 27629939 PMCID: PMC5024325 DOI: 10.1038/srep33557] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/30/2016] [Indexed: 02/07/2023] Open
Abstract
RNA virus infections are detected by the RIG-I family of receptors, which signal through the adaptor molecule mitochondrial antiviral signaling (MAVS). MAVS then recruits the adaptor’s tumor necrosis factor receptor-associated factor (TRAF) 3 and TRAF6, which in turn activate IRF3 and NF-κB, respectively, to induce interferons (IFNs) and inflammatory responses. Here we show that the biotin-containing enzyme methylcrotonoyl-CoA carboxylase 1 (MCCC1) enhances virus-induced, MAVS-mediated IFN and inflammatory cytokine expression through the NF-κB signaling pathway. MCCC1 knockdown strongly inhibits induction of IFNs and inflammatory cytokines. Furthermore, MCCC1 shows extensive antiviral activity toward RNA viruses, including influenza A virus, human enterovirus 71, and vesicular stomatitis virus. Here, we have elucidated the mechanism underlying MCCC1-mediated inhibition of viral replication. MCCC1 interacts with MAVS and components of the MAVS signalosome and contributes to enhanced production of type I IFNs and pro-inflammatory cytokines by promoting phosphorylation of the IκB kinase (IKK) complex and NF-κB inhibitor-α (IκBα), as well as NF-κB nuclear translocation. This process leads to activation of IFNs and cytokine expression and subsequent activation of IFN-stimulated genes, including double-stranded RNA-dependent protein kinase PKR and myxovirus resistance protein 1. These findings demonstrate that MCCC1 plays an essential role in virus-triggered, MAVS-mediated activation of NF-κB signaling.
Collapse
Affiliation(s)
- Zhongying Cao
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zhangchuan Xia
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yaqin Zhou
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Xiaodan Yang
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Hua Hao
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Nanfang Peng
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Shi Liu
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Ying Zhu
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
23
|
Yang X, Hao H, Xia Z, Xu G, Cao Z, Chen X, Liu S, Zhu Y. Soluble IL-6 Receptor and IL-27 Subunit p28 Protein Complex Mediate the Antiviral Response through the Type III IFN Pathway. THE JOURNAL OF IMMUNOLOGY 2016; 197:2369-81. [PMID: 27527594 DOI: 10.4049/jimmunol.1600627] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/13/2016] [Indexed: 02/07/2023]
Abstract
Previously, we demonstrated that the soluble IL-6R (sIL-6R) plays an important role in the host antiviral response through induction of type I IFN and sIL-6R-mediated antiviral action via the IL-27 subunit p28; however, the mechanism that underlies sIL-6R and p28 antiviral action and whether type III IFN is involved remain unknown. In this study, we constructed a sIL-6R and p28 fusion protein (sIL-6R/p28 FP) and demonstrated that the fusion protein has stronger antiviral activity than sIL-6R alone. Consequently, knockout of sIL-6R inhibited virus-triggered IFN-λ1 expression. In addition, sIL-6R/p28 FP associated with mitochondrial antiviral signaling protein and TNFR-associated factor 6, the retinoic acid-inducible gene I adapter complex, and the antiviral activity mediated by sIL-6R/p28 FP was dependent on mitochondrial antiviral signaling protein. Furthermore, significantly reduced binding of p50/p65 and IFN regulatory factor 3 to the IFN-λ1 promoter was observed in sIL-6R knockout cells compared with the control cells. Interestingly, a novel heterodimer of c-Fos and activating transcription factor 1 was identified as a crucial transcriptional activator of IFN-λ1 The sIL-6R/p28 FP upregulated IFN-λ1 expression by increasing the binding abilities of c-Fos and activating transcription factor 1 to the IFN-λ1 promoter via the p38 MAPK signaling pathway. In conclusion, these results demonstrate the important role of sIL-6R/p28 FP in mediating virus-induced type III IFN production.
Collapse
Affiliation(s)
- Xiaodan Yang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Hua Hao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zhangchuan Xia
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Gang Xu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zhongying Cao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Xueyuan Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Shi Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Ying Zhu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
24
|
Wang L, Zhu S, Xu G, Feng J, Han T, Zhao F, She YL, Liu S, Ye L, Zhu Y. Gene Expression and Antiviral Activity of Interleukin-35 in Response to Influenza A Virus Infection. J Biol Chem 2016; 291:16863-76. [PMID: 27307042 DOI: 10.1074/jbc.m115.693101] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Indexed: 01/22/2023] Open
Abstract
Interleukin-35 (IL-35) is a newly described member of the IL-12 family. It has been reported to inhibit inflammation and autoimmune inflammatory disease and can increase apoptotic sensitivity. Little is known about the role of IL-35 during viral infection. Herein, high levels of IL-35 were found in peripheral blood mononuclear cells and throat swabs from patients with seasonal influenza A virus (IAV) relative to healthy individuals. IAV infection of human lung epithelial and primary cells increased levels of IL-35 mRNA and protein. Further studies demonstrated that IAV-induced IL-35 transcription is regulated by NF-κB. IL-35 expression was significantly suppressed by selective inhibitors of cyclooxygenase-2 (COX-2) and inducible nitric-oxide synthase, indicating their involvement in IL-35 expression. Interestingly, IL-35 production may have suppressed IAV RNA replication and viral protein synthesis via induction of type I and III interferons (IFN), leading to activation of downstream IFN effectors, including double-stranded RNA-dependent protein kinase, 2',5'-oligoadenylate synthetase, and myxovirus resistance protein. IL-35 exhibited extensive antiviral activity against the hepatitis B virus, enterovirus 71, and vesicular stomatitis virus. Our results demonstrate that IL-35 is a novel IAV-inducible cytokine, and its production elicits antiviral activity.
Collapse
Affiliation(s)
- Li Wang
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Shengli Zhu
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Gang Xu
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Jian Feng
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Tao Han
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Fanpeng Zhao
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Ying-Long She
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Shi Liu
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Linbai Ye
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Ying Zhu
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| |
Collapse
|
25
|
Wang SR, Min YQ, Wang JQ, Liu CX, Fu BS, Wu F, Wu LY, Qiao ZX, Song YY, Xu GH, Wu ZG, Huang G, Peng NF, Huang R, Mao WX, Peng S, Chen YQ, Zhu Y, Tian T, Zhang XL, Zhou X. A highly conserved G-rich consensus sequence in hepatitis C virus core gene represents a new anti-hepatitis C target. SCIENCE ADVANCES 2016; 2:e1501535. [PMID: 27051880 PMCID: PMC4820367 DOI: 10.1126/sciadv.1501535] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/18/2016] [Indexed: 05/24/2023]
Abstract
G-quadruplex (G4) is one of the most important secondary structures in nucleic acids. Until recently, G4 RNAs have not been reported in any ribovirus, such as the hepatitis C virus. Our bioinformatics analysis reveals highly conserved guanine-rich consensus sequences within the core gene of hepatitis C despite the high genetic variability of this ribovirus; we further show using various methods that such consensus sequences can fold into unimolecular G4 RNA structures, both in vitro and under physiological conditions. Furthermore, we provide direct evidences that small molecules specifically targeting G4 can stabilize this structure to reduce RNA replication and inhibit protein translation of intracellular hepatitis C. Ultimately, the stabilization of G4 RNA in the genome of hepatitis C represents a promising new strategy for anti-hepatitis C drug development.
Collapse
Affiliation(s)
- Shao-Ru Wang
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Wuhan University, Wuhan 430072, Hubei, China
| | - Yuan-Qin Min
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, School of Medicine, Wuhan University, Wuhan 430071, Hubei, China
| | - Jia-Qi Wang
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Wuhan University, Wuhan 430072, Hubei, China
| | - Chao-Xing Liu
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Wuhan University, Wuhan 430072, Hubei, China
| | - Bo-Shi Fu
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Wuhan University, Wuhan 430072, Hubei, China
| | - Fan Wu
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Wuhan University, Wuhan 430072, Hubei, China
| | - Ling-Yu Wu
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Wuhan University, Wuhan 430072, Hubei, China
| | - Zhi-Xian Qiao
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, Hubei, China
| | - Yan-Yan Song
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Wuhan University, Wuhan 430072, Hubei, China
| | - Guo-Hua Xu
- Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, Hubei, China
| | - Zhi-Guo Wu
- College of Life Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Gai Huang
- College of Life Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Nan-Fang Peng
- College of Life Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Rong Huang
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Wuhan University, Wuhan 430072, Hubei, China
| | - Wu-Xiang Mao
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Wuhan University, Wuhan 430072, Hubei, China
| | - Shuang Peng
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Wuhan University, Wuhan 430072, Hubei, China
| | - Yu-Qi Chen
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Wuhan University, Wuhan 430072, Hubei, China
| | - Ying Zhu
- College of Life Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Tian Tian
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Wuhan University, Wuhan 430072, Hubei, China
| | - Xiao-Lian Zhang
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, School of Medicine, Wuhan University, Wuhan 430071, Hubei, China
| | - Xiang Zhou
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Wuhan University, Wuhan 430072, Hubei, China
| |
Collapse
|
26
|
Cao Z, Zhou Y, Zhu S, Feng J, Chen X, Liu S, Peng N, Yang X, Xu G, Zhu Y. Pyruvate Carboxylase Activates the RIG-I-like Receptor-Mediated Antiviral Immune Response by Targeting the MAVS signalosome. Sci Rep 2016; 6:22002. [PMID: 26906558 PMCID: PMC4764940 DOI: 10.1038/srep22002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 02/05/2016] [Indexed: 02/07/2023] Open
Abstract
When retinoic acid-inducible gene 1 protein (RIG-I)-like receptors sense viral dsRNA in the cytosol, RIG-I and melanoma differentiation-associated gene 5 (MDA5) are recruited to the mitochondria to interact with mitochondrial antiviral signaling protein (MAVS) and initiate antiviral immune responses. In this study, we demonstrate that the biotin-containing enzyme pyruvate carboxylase (PC) plays an essential role in the virus-triggered activation of nuclear factor kappa B (NF-κB) signaling mediated by MAVS. PC contributes to the enhanced production of type I interferons (IFNs) and pro-inflammatory cytokines, and PC knockdown inhibits the virus-triggered innate immune response. In addition, PC shows extensive antiviral activity against RNA viruses, including influenza A virus (IAV), human enterovirus 71 (EV71), and vesicular stomatitis virus (VSV). Furthermore, PC mediates antiviral action by targeting the MAVS signalosome and induces IFNs and pro-inflammatory cytokines by promoting phosphorylation of NF-κB inhibitor-α (IκBα) and the IκB kinase (IKK) complex, as well as NF-κB nuclear translocation, which leads to activation of interferon-stimulated genes (ISGs), including double-stranded RNA-dependent protein kinase (PKR) and myxovirus resistance protein 1 (Mx1). Our findings suggest that PC is an important player in host antiviral signaling.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/immunology
- Animals
- Cell Line, Tumor
- Cytokines/genetics
- Cytokines/immunology
- DEAD Box Protein 58/genetics
- DEAD Box Protein 58/immunology
- Enterovirus A, Human/genetics
- Enterovirus A, Human/immunology
- Gene Expression Regulation
- Genes, Reporter
- HEK293 Cells
- Hepatocytes/immunology
- Hepatocytes/virology
- Humans
- Immunity, Innate
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/immunology
- Interferon Type I/genetics
- Interferon Type I/immunology
- Interferon-Induced Helicase, IFIH1/genetics
- Interferon-Induced Helicase, IFIH1/immunology
- Luciferases/genetics
- Luciferases/immunology
- NF-KappaB Inhibitor alpha/genetics
- NF-KappaB Inhibitor alpha/immunology
- NF-kappa B/genetics
- NF-kappa B/immunology
- Pyruvate Carboxylase/antagonists & inhibitors
- Pyruvate Carboxylase/genetics
- Pyruvate Carboxylase/immunology
- RNA, Small Interfering/genetics
- RNA, Small Interfering/immunology
- RNA, Viral/genetics
- RNA, Viral/immunology
- Receptors, Immunologic
- Signal Transduction
- Vesiculovirus/genetics
- Vesiculovirus/immunology
- eIF-2 Kinase/genetics
- eIF-2 Kinase/immunology
Collapse
Affiliation(s)
- Zhongying Cao
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yaqin Zhou
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Shengli Zhu
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Jian Feng
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Xueyuan Chen
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Shi Liu
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Nanfang Peng
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Xiaodan Yang
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Gang Xu
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Ying Zhu
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
27
|
Arshad MI, Guihard P, Danger Y, Noel G, Le Seyec J, Boutet MA, Richards CD, L'Helgoualc'h A, Genet V, Lucas-Clerc C, Gascan H, Blanchard F, Piquet-Pellorce C, Samson M. Oncostatin M induces IL-33 expression in liver endothelial cells in mice and expands ST2+CD4+ lymphocytes. Am J Physiol Gastrointest Liver Physiol 2015; 309:G542-53. [PMID: 26251474 DOI: 10.1152/ajpgi.00398.2014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 06/22/2015] [Indexed: 01/31/2023]
Abstract
Interleukin (IL)-33 is crucially involved in liver pathology and drives hepatoprotective functions. However, the regulation of IL-33 by cytokines of the IL-6 family, including oncostatin M (OSM) and IL-6, is not well studied. The aim of the present study was to determine whether OSM mediates regulation of IL-33 expression in liver cells. Intramuscular administration in mice of an adenovirus encoding OSM (AdOSM) leads to increase in expression of OSM in muscles, liver, and serum of AdOSM-infected mice compared with control mice. The increase of circulating OSM markedly regulated mRNA of genes associated with blood vessel biology, chemotaxis, cellular death, induction of cell adhesion molecules, and the alarmin cytokine IL-33 in liver. Steady-state IL-33 mRNA was upregulated by OSM at an early phase (8 h) following AdOSM infection. At the protein level, the expression of IL-33 was significantly induced in liver endothelial cells [liver sinusoidal endothelial cells (LSEC) and vascular endothelial cells] with a peak at 8 days post-AdOSM infection in mice. In addition, we found OSM-stimulated human microvascular endothelial HMEC-1 cells and human LSEC/TRP3 cells showed a significant increase in expression of IL-33 mRNA in a dose-dependent manner in cell culture. The OSM-mediated overexpression of IL-33 was associated with the activation/enrichment of CD4(+)ST2(+) cells in liver of AdOSM-infected mice compared with adenovirus encoding green fluorescent protein-treated control mice. In summary, these data suggest that the cytokine OSM regulates the IL-33 expression in liver endothelial cells in vivo and in HMEC-1/TRP3 cells in vitro and may specifically expand the target CD4(+)ST2(+) cells in liver.
Collapse
Affiliation(s)
- Muhammad Imran Arshad
- Institut National de la Santé et de la Recherche Médicale, U1085, Institut de Recherche Santé Environnement et Travail, Rennes, France; Université de Rennes 1, Rennes, France; Structure Fédérative BioSit UMS 3480 Centre National de la Recherche Scientifique-US18 Institut National de la Santé et de la Recherche Médicale, Rennes, France
| | - Pierre Guihard
- Institut National de la Santé et de la Recherche Médicale, UMR 957, Equipe Labellisée LIGUE 2012, Nantes, France
| | - Yannic Danger
- Structure Fédérative BioSit UMS 3480 Centre National de la Recherche Scientifique-US18 Institut National de la Santé et de la Recherche Médicale, Rennes, France; EFS, Rennes, France
| | - Gregory Noel
- Institut National de la Santé et de la Recherche Médicale, U1085, Institut de Recherche Santé Environnement et Travail, Rennes, France; Université de Rennes 1, Rennes, France; Structure Fédérative BioSit UMS 3480 Centre National de la Recherche Scientifique-US18 Institut National de la Santé et de la Recherche Médicale, Rennes, France
| | - Jacques Le Seyec
- Institut National de la Santé et de la Recherche Médicale, U1085, Institut de Recherche Santé Environnement et Travail, Rennes, France; Université de Rennes 1, Rennes, France; Structure Fédérative BioSit UMS 3480 Centre National de la Recherche Scientifique-US18 Institut National de la Santé et de la Recherche Médicale, Rennes, France
| | - Marie-Astrid Boutet
- Institut National de la Santé et de la Recherche Médicale, UMR 957, Equipe Labellisée LIGUE 2012, Nantes, France
| | - Carl D Richards
- McMaster Immunology Research Center, McMaster University, Hamilton, Ontario, Canada
| | - Annie L'Helgoualc'h
- Institut National de la Santé et de la Recherche Médicale, U1085, Institut de Recherche Santé Environnement et Travail, Rennes, France; Université de Rennes 1, Rennes, France; Structure Fédérative BioSit UMS 3480 Centre National de la Recherche Scientifique-US18 Institut National de la Santé et de la Recherche Médicale, Rennes, France
| | - Valentine Genet
- Institut National de la Santé et de la Recherche Médicale, U1085, Institut de Recherche Santé Environnement et Travail, Rennes, France; Université de Rennes 1, Rennes, France; Structure Fédérative BioSit UMS 3480 Centre National de la Recherche Scientifique-US18 Institut National de la Santé et de la Recherche Médicale, Rennes, France
| | - Catherine Lucas-Clerc
- Université de Rennes 1, Rennes, France; Service de Biochimie Centre Hospitalier Universitaire Rennes, Université de Rennes 1, Rennes, France
| | - Hugues Gascan
- Centre National de la Recherche Scientifique, UMR 6290, Institute of Genetics and Development of Rennes, Rennes, France; and
| | - Frédéric Blanchard
- Institut National de la Santé et de la Recherche Médicale, UMR 957, Equipe Labellisée LIGUE 2012, Nantes, France
| | - Claire Piquet-Pellorce
- Institut National de la Santé et de la Recherche Médicale, U1085, Institut de Recherche Santé Environnement et Travail, Rennes, France; Université de Rennes 1, Rennes, France; Structure Fédérative BioSit UMS 3480 Centre National de la Recherche Scientifique-US18 Institut National de la Santé et de la Recherche Médicale, Rennes, France
| | - Michel Samson
- Institut National de la Santé et de la Recherche Médicale, U1085, Institut de Recherche Santé Environnement et Travail, Rennes, France; Université de Rennes 1, Rennes, France; Structure Fédérative BioSit UMS 3480 Centre National de la Recherche Scientifique-US18 Institut National de la Santé et de la Recherche Médicale, Rennes, France;
| |
Collapse
|
28
|
Segarra NG, Ballhausen D, Crawford H, Perreau M, Campos-Xavier B, van Spaendonck-Zwarts K, Vermeer C, Russo M, Zambelli PY, Stevenson B, Royer-Bertrand B, Rivolta C, Candotti F, Unger S, Munier FL, Superti-Furga A, Bonafé L. NBAS mutations cause a multisystem disorder involving bone, connective tissue, liver, immune system, and retina. Am J Med Genet A 2015; 167A:2902-12. [PMID: 26286438 DOI: 10.1002/ajmg.a.37338] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 08/10/2015] [Indexed: 11/09/2022]
Abstract
We report two unrelated patients with a multisystem disease involving liver, eye, immune system, connective tissue, and bone, caused by biallelic mutations in the neuroblastoma amplified sequence (NBAS) gene. Both presented as infants with recurrent episodes triggered by fever with vomiting, dehydration, and elevated transaminases. They had frequent infections, hypogammaglobulinemia, reduced natural killer cells, and the Pelger-Huët anomaly of their granulocytes. Their facial features were similar with a pointed chin and proptosis; loose skin and reduced subcutaneous fat gave them a progeroid appearance. Skeletal features included short stature, slender bones, epiphyseal dysplasia with multiple phalangeal pseudo-epiphyses, and small C1-C2 vertebrae causing cervical instability and myelopathy. Retinal dystrophy and optic atrophy were present in one patient. NBAS is a component of the synthaxin-18 complex and is involved in nonsense-mediated mRNA decay control. Putative loss-of-function mutations in NBAS are already known to cause disease in humans. A specific founder mutation has been associated with short stature, optic nerve atrophy and Pelger-Huët anomaly of granulocytes (SOPH) in the Siberian Yakut population. A more recent report associates NBAS mutations with recurrent acute liver failure in infancy in a group of patients of European descent. Our observations indicate that the phenotypic spectrum of NBAS deficiency is wider than previously known and includes skeletal, hepatic, metabolic, and immunologic aspects. Early recognition of the skeletal phenotype is important for preventive management of cervical instability.
Collapse
Affiliation(s)
- Nuria Garcia Segarra
- Center for Molecular Diseases, Lausanne University Hospital, Lausanne, Switzerland
| | - Diana Ballhausen
- Center for Molecular Diseases, Lausanne University Hospital, Lausanne, Switzerland
| | - Heather Crawford
- Clinical Metabolic Genetics, Cook Children's Health Care System, Fort Worth, Texas, USA
| | - Matthieu Perreau
- Division of Immunology and Allergy, Lausanne University Hospital, Lausanne, Switzerland
| | | | | | - Cees Vermeer
- VitaK and Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Michel Russo
- Pediatric Service, Centre Hospitalier du Centre Valais, Sion, Switzerland
| | - Pierre-Yves Zambelli
- Service of Pediatric Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Brian Stevenson
- Vital-IT group, Swiss Institute of Bioinformatics, University of Lausanne, Switzerland
| | - Beryl Royer-Bertrand
- Department of Medical Genetics, Lausanne University Hospital, Lausanne, Switzerland
| | - Carlo Rivolta
- Department of Medical Genetics, Lausanne University Hospital, Lausanne, Switzerland
| | - Fabio Candotti
- Division of Immunology and Allergy, Lausanne University Hospital, Lausanne, Switzerland
| | - Sheila Unger
- Department of Medical Genetics, Lausanne University Hospital, Lausanne, Switzerland
| | | | | | - Luisa Bonafé
- Center for Molecular Diseases, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
29
|
Important Role of the IL-32 Inflammatory Network in the Host Response against Viral Infection. Viruses 2015; 7:3116-29. [PMID: 26087456 PMCID: PMC4488729 DOI: 10.3390/v7062762] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 06/11/2015] [Indexed: 12/11/2022] Open
Abstract
The pro-inflammatory cytokine interleukin (IL)-32 has gained much attention recently because of its important role in the inflammatory network. Since the discovery of IL-32 in 2005, our appreciation for its diverse roles continues to grow. Recent studies have discovered the antiviral effects induced by IL-32 and its associated regulatory mechanisms. The interactions between IL-32 and various cytokines including cyclooxygenase 2 (COX-2), inducible nitric oxide synthase (iNOS), interferon (IFN)-λ1, interleukin (IL)-6, and soluble IL-6 receptor have been described. This review aims to integrate these new findings into explicit concepts and raises the intriguing possibility of IL-32 as a therapeutic target.
Collapse
|
30
|
Soluble interleukin-6 receptor is elevated during influenza A virus infection and mediates the IL-6 and IL-32 inflammatory cytokine burst. Cell Mol Immunol 2014; 12:633-44. [PMID: 25176527 DOI: 10.1038/cmi.2014.80] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 07/29/2014] [Accepted: 07/30/2014] [Indexed: 02/07/2023] Open
Abstract
Influenza A virus (IAV) infection is a major worldwide public health problem. However, the factors involved in mediating the inflammatory response to this infection and their relationships remain poorly understood. Here, we show that IAV infection stimulates the expression of the soluble IL-6 receptor (sIL-6R), a multifunctional protein involved in IL-6 signaling. Interestingly, sIL-6R expression upregulated the levels of its own ligand, IL-6 and those of the pro-inflammatory cytokine IL-32. shRNA-mediated knockdown of sIL-6R suppressed IL-6 and IL-32, indicating that this regulation is dependent on sIL-6R during IAV infection. Furthermore, our results demonstrate that IL-32 participates in a negative feedback loop that inhibits sIL-6R while upregulating IL-6 expression during IAV infection. Therefore, we show that sIL-6R is a critical cellular factor involved in the acute inflammatory response to viral infection.
Collapse
|