1
|
Albertini S, Martuscelli L, Borgogna C, Virdi S, Indenbirken D, Lo Cigno I, Griffante G, Calati F, Boldorini R, Fischer N, Gariglio M. Cancer-Associated Fibroblasts Exert Proangiogenic Activity in Merkel Cell Carcinoma. J Invest Dermatol 2022; 143:965-976.e15. [PMID: 36572089 DOI: 10.1016/j.jid.2022.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/01/2022] [Accepted: 12/05/2022] [Indexed: 12/25/2022]
Abstract
The tumor microenvironment is a complex niche enveloping a tumor formed by extracellular matrix, blood vessels, immune cells, and fibroblasts constantly interacting with cancer cells. Although tumor microenvironment is increasingly recognized as a major player in cancer initiation and progression in many tumor types, its involvement in Merkel cell carcinoma (MCC) pathogenesis is currently unknown. In this study, we provide a molecular and functional characterization of cancer-associated fibroblasts (CAFs), the major tumor microenvironment component, in patient-derived xenografts of patients with MCC. We show that subcutaneous coinjection of patient-derived CAFs and human MCC MKL-1 cells into severe combined immunodeficient mice significantly promotes tumor growth and metastasis. These fast-growing xenografts are characterized by areas densely populated with human CAFs, mainly localized around blood vessels. We provide evidence that the growth-promoting activity of MCC-derived CAFs is mediated by the aminopeptidase A/angiotensin II and III/angiotensin II type 1 receptor axis, with the expression of aminopeptidase A in CAFs being a triggering event. Together, our findings point to aminopeptidase A as a potential marker for MCC prognostic stratification and as a candidate for therapeutic intervention.
Collapse
Affiliation(s)
- Silvia Albertini
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Virology Unit, Department of Translational Medicine, Novara Medical School, Novara, Italy
| | - Licia Martuscelli
- Virology Unit, Department of Translational Medicine, Novara Medical School, Novara, Italy
| | - Cinzia Borgogna
- Virology Unit, Department of Translational Medicine, Novara Medical School, Novara, Italy
| | - Sanamjeet Virdi
- Technology Platform Next Generation Sequencing, Leibniz Institute for Virology, Hamburg, Germany
| | - Daniela Indenbirken
- Technology Platform Next Generation Sequencing, Leibniz Institute for Virology, Hamburg, Germany
| | - Irene Lo Cigno
- Virology Unit, Department of Translational Medicine, Novara Medical School, Novara, Italy
| | - Gloria Griffante
- Virology Unit, Department of Translational Medicine, Novara Medical School, Novara, Italy
| | - Federica Calati
- Virology Unit, Department of Translational Medicine, Novara Medical School, Novara, Italy
| | - Renzo Boldorini
- Pathology Unit, Department of Health Sciences, Novara Medical School, Novara, Italy
| | - Nicole Fischer
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marisa Gariglio
- Virology Unit, Department of Translational Medicine, Novara Medical School, Novara, Italy.
| |
Collapse
|
2
|
Fu Q, He Q, Dong Q, Xie J, Geng Y, Han H, Huang Y, Lu J, Zeng Z, Wang W, Chen K, Zhan X. The role of cyclic GMP-AMP synthase and Interferon-I-inducible protein 16 as candidatebiomarkers of systemic lupus erythematosus. Clin Chim Acta 2022; 524:69-77. [PMID: 34742679 DOI: 10.1016/j.cca.2021.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 10/24/2021] [Accepted: 11/01/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Diverse clinical and serological manifestations of systemic lupus erythematosus (SLE) compromise its diagnosis and treatment. A more reliable biomarker for SLE, which can play a critical role in either diagnosis, monitoring the disease progress or evaluating the response to treatment for individualized therapeutic, is necessary. DNA sensor is an important mediator of inflammation in systemic autoimmune diseases. However, the potential role for DNA sensor as disease activity biomarkers for SLE remained obscure. We detected the aberrant activation of DNA sensors and the corresponding IFN-β response in SLE patients, and to evaluate their potential role as disease biomarkers for SLE. METHODS We quantified the expressions of IFN-I and DNA sensor, such as cGAS, IFI16, DDX41, DAI and their down-stream adaptor STING in PBMC derived from patients with SLE (n = 100), healthy controls (HCs) (n = 62) by real-time PCR. The relationships between the expression of cGAS or IFI16 and clinical features in SLE patients were investigated. ROC curve analysis was performed to examine the predictive value of cGAS and IFI16 in SLE diagnosis, disease activity monitoring, specific organ manifestation and therapeutic response. RNA interference-mediated depletion of IFI16 or cGAS was conducted to evaluate their impact on IFN-I response. RESULTS The expressions of cGAS and IFI16 were significantly higher in PBMC from SLE patients, closely correlated with the SLEDAI scores and high anti-dsDNA antibody titers. While the AUC for cGAS (0.767) was less than that of IFI16 and IFN-β, the AUC for IFI16 (0.856) and IFN-β (0.856) were similar. Expression of cGAS and IFI16 combine with IFN-β in PBMC showed high sensitivity (89.2%) and specificity (89.1%) for discrimination between mild and moderate/severe disease activity in SLE. Higher expression of IFI16 was association with ocular disorder in SLE patients. Neither IFI16 nor cGAS was a reliable indicator of therapeutic response. RNA interference-mediated depletion of IFI16 or cGAS prevented active SLE serum-induced upregulating in both IFN-α and IFN-β. CONCLUSIONS High expression levels of cGAS and IFI16 in PBMC from SLE patients correlated strongly with disease activity. Both cGAS and IFI16 mediated signaling pathway were account for the robust production of IFN-β. Expression of cGAS and IFI16 combined with IFN-β in PBMC might serve as potential biomarkers for early diagnosis and monitoring disease activity in SLE.
Collapse
Affiliation(s)
- Qiang Fu
- Department of Laboratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, 2 Zhongshan Road, Guangzhou 510080, Guangdong, China
| | - Qiuying He
- Department of Laboratory Medicine, Zhongshan Hospital of Sun Yat-sen University, 2 Sunwendong Road, Zhongshan 528403, Guangdong, China
| | - Qian Dong
- Department of Laboratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, 2 Zhongshan Road, Guangzhou 510080, Guangdong, China
| | - Jinye Xie
- Department of Laboratory Medicine, Zhongshan Hospital of Sun Yat-sen University, 2 Sunwendong Road, Zhongshan 528403, Guangdong, China
| | - Yiyun Geng
- Department of Laboratory Medicine, Zhongshan Hospital of Sun Yat-sen University, 2 Sunwendong Road, Zhongshan 528403, Guangdong, China
| | - Hui Han
- Department of Laboratory Medicine, Zhongshan Hospital of Sun Yat-sen University, 2 Sunwendong Road, Zhongshan 528403, Guangdong, China
| | - Yanhua Huang
- Department of Laboratory Medicine, Zhongshan Hospital of Sun Yat-sen University, 2 Sunwendong Road, Zhongshan 528403, Guangdong, China
| | - Jianqiang Lu
- Department of Laboratory Medicine, Zhongshan Hospital of Sun Yat-sen University, 2 Sunwendong Road, Zhongshan 528403, Guangdong, China
| | - Zhijie Zeng
- Department of Laboratory Medicine, Zhongshan Hospital of Sun Yat-sen University, 2 Sunwendong Road, Zhongshan 528403, Guangdong, China
| | - Weijia Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, 2 Zhongshan Road, Guangzhou 510080, Guangdong, China.
| | - Kang Chen
- Department of Laboratory Medicine, Zhongshan Hospital of Sun Yat-sen University, 2 Sunwendong Road, Zhongshan 528403, Guangdong, China.
| | - Xiaoxia Zhan
- Department of Laboratory Medicine, Zhongshan Hospital of Sun Yat-sen University, 2 Sunwendong Road, Zhongshan 528403, Guangdong, China.
| |
Collapse
|
3
|
Lo Cigno I, Calati F, Borgogna C, Zevini A, Albertini S, Martuscelli L, De Andrea M, Hiscott J, Landolfo S, Gariglio M. Human Papillomavirus E7 Oncoprotein Subverts Host Innate Immunity via SUV39H1-Mediated Epigenetic Silencing of Immune Sensor Genes. J Virol 2020; 94:e01812-19. [PMID: 31776268 PMCID: PMC6997746 DOI: 10.1128/jvi.01812-19] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 11/20/2019] [Indexed: 12/23/2022] Open
Abstract
Subversion of innate immunity by oncoviruses, such as human papillomavirus (HPV), favors carcinogenesis because the mechanism(s) of viral immune evasion can also hamper cancer immunosurveillance. Previously, we demonstrated that high-risk (hr) HPVs trigger simultaneous epigenetic silencing of multiple effectors of innate immunity to promote viral persistence. Here, we expand on those observations and show that the HPV E7 oncoprotein upregulates the H3K9-specific methyltransferase, whose action shuts down the host innate immune response. Specifically, we demonstrate that SUV39H1 contributes to chromatin repression at the promoter regions of the viral nucleic acid sensors RIG-I and cGAS and the adaptor molecule STING in HPV-transformed cells. Inhibition of SUV39H1 leads to transcriptional activation of these genes, especially RIG-I, followed by increased beta interferon (IFN-β) and IFN-λ1 production after poly(dA·dT) or RIG-I agonist M8 transfection. Collectively, our findings provide new evidence that the E7 oncoprotein plays a central role in dampening host innate immunity and raise the possibility that targeting the downstream effector SUV39H1 or the RIG-I pathway is a viable strategy to treat viral and neoplastic disease.IMPORTANCE High-risk HPVs are major viral human carcinogens responsible for approximately 5% of all human cancers. The growth of HPV-transformed cells depends on the ability of viral oncoproteins to manipulate a variety of cellular circuits, including those involved in innate immunity. Here, we show that one of these strategies relies on E7-mediated transcriptional activation of the chromatin repressor SUV39H1, which then promotes epigenetic silencing of RIG-I, cGAS, and STING genes, thereby shutting down interferon secretion in HPV-transformed cells. Pharmacological or genetic inhibition of SUV39H1 restored the innate response in HPV-transformed cells, mostly through activation of RIG-I signaling. We also show that IFN production upon transfection of poly(dA·dT) or the RIG-I agonist M8 predominantly occurs through RIG-I signaling. Altogether, the reversible nature of the modifications associated with E7-mediated SUV39H1 upregulation provides a rationale for the design of novel anticancer and antiviral therapies targeting these molecules.
Collapse
Affiliation(s)
- Irene Lo Cigno
- University of Piemonte Orientale Medical School, Department of Translational Medicine, Molecular Virology Unit, Novara, Italy
| | - Federica Calati
- University of Piemonte Orientale Medical School, Department of Translational Medicine, Molecular Virology Unit, Novara, Italy
| | - Cinzia Borgogna
- University of Piemonte Orientale Medical School, Department of Translational Medicine, Molecular Virology Unit, Novara, Italy
| | | | - Silvia Albertini
- University of Piemonte Orientale Medical School, Department of Translational Medicine, Molecular Virology Unit, Novara, Italy
| | - Licia Martuscelli
- University of Piemonte Orientale Medical School, Department of Translational Medicine, Molecular Virology Unit, Novara, Italy
| | - Marco De Andrea
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Novara, Italy
- University of Turin Medical School, Department of Public Health and Pediatric Sciences, Viral Pathogenesis Unit, Turin, Italy
| | - John Hiscott
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | - Santo Landolfo
- University of Turin Medical School, Department of Public Health and Pediatric Sciences, Viral Pathogenesis Unit, Turin, Italy
| | - Marisa Gariglio
- University of Piemonte Orientale Medical School, Department of Translational Medicine, Molecular Virology Unit, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Novara, Italy
| |
Collapse
|
4
|
Piccaluga PP, Navari M, Visani A, Rigotti F, Agostinelli C, Righi S, Diani E, Ligozzi M, Carelli M, Ponti C, Bon I, Zipeto D, Landolfo S, Gibellini D. Interferon gamma inducible protein 16 (IFI16) expression is reduced in mantle cell lymphoma. Heliyon 2019; 5:e02643. [PMID: 31840115 PMCID: PMC6893061 DOI: 10.1016/j.heliyon.2019.e02643] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 07/12/2019] [Accepted: 10/09/2019] [Indexed: 11/05/2022] Open
Abstract
IFI16, member of the IFN-inducible PYHIN-200 gene family, modulates proliferation, survival and differentiation of different cell lineages. In particular, IFI16 expression, which is regulated during the differentiation of B cells, was recently studied in B-CLL as well. Here, we compared IFI16 expression in several lymphomas including Burkitt lymphoma, diffuse large B-cell lymphoma, follicular lymphoma, marginal zone lymphoma and mantle cell lymphoma with respect to normal cell counterparts. We observed that IFI16 expression was significantly deregulated only in mantle cell lymphoma (p < 0.05). Notably, IFI16 was associated with the expression of genes involved in interferon response, cell cycle, cell death and proliferation and, interestingly, lipid and glucose metabolism, suggesting that IFI16 deregulation might be associated with relevant changes in cell biology. In our group of mantle cell lymphoma samples a correlation between patient survival and IFI16 expression was not detected even though mantle cell lymphoma prognosis is known to be associated with cell proliferation. Altogether, these results suggest a complex relationship between IFI16 expression and MCL which needs to be analyzed in further studies.
Collapse
Affiliation(s)
- Pier Paolo Piccaluga
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy.,Istituto Euro-Mediterraneo di Scienza e Tecnologia (IEMEST) Palermo, Italy.,Department of Pathology, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Mohsen Navari
- Department of Medical Biotechnology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.,Research Center of Advanced Technologies in Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.,Bioinformatics Research Group, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Axel Visani
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Flavia Rigotti
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Claudio Agostinelli
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Simona Righi
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Erica Diani
- Department of Diagnostic and Public Health, Unit of Microbiology, University of Verona, Verona, Italy
| | - Marco Ligozzi
- Department of Diagnostic and Public Health, Unit of Microbiology, University of Verona, Verona, Italy
| | - Maria Carelli
- Department of Diagnostic and Public Health, Unit of Microbiology, University of Verona, Verona, Italy
| | - Cristina Ponti
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Isabella Bon
- Department of Experimental, Diagnostic, and Specialty Medicine, Microbiology Unit, University of Bologna, Bologna, Italy
| | - Donato Zipeto
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Santo Landolfo
- Department of Public Health and Microbiology, University of Turin, Turin, Italy
| | - Davide Gibellini
- Department of Diagnostic and Public Health, Unit of Microbiology, University of Verona, Verona, Italy
| |
Collapse
|
5
|
Strategy of Human Cytomegalovirus To Escape Interferon Beta-Induced APOBEC3G Editing Activity. J Virol 2018; 92:JVI.01224-18. [PMID: 30045985 DOI: 10.1128/jvi.01224-18] [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] [Received: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 01/26/2023] Open
Abstract
The apolipoprotein B editing enzyme catalytic subunit 3 (APOBEC3) is a family of DNA cytosine deaminases that mutate and inactivate viral genomes by single-strand DNA editing, thus providing an innate immune response against a wide range of DNA and RNA viruses. In particular, APOBEC3A (A3A), a member of the APOBEC3 family, is induced by human cytomegalovirus (HCMV) in decidual tissues where it efficiently restricts HCMV replication, thereby acting as an intrinsic innate immune effector at the maternal-fetal interface. However, the widespread incidence of congenital HCMV infection implies that HCMV has evolved to counteract APOBEC3-induced mutagenesis through mechanisms that still remain to be fully established. Here, we have assessed gene expression and deaminase activity of various APOBEC3 gene family members in HCMV-infected primary human foreskin fibroblasts (HFFs). Specifically, we show that APOBEC3G (A3G) gene products and, to a lesser degree, those of A3F but not of A3A, are upregulated in HCMV-infected HFFs. We also show that HCMV-mediated induction of A3G expression is mediated by interferon beta (IFN-β), which is produced early during HCMV infection. However, knockout or overexpression of A3G does not affect HCMV replication, indicating that A3G is not a restriction factor for HCMV. Finally, through a bioinformatics approach, we show that HCMV has evolved mutational robustness against IFN-β by limiting the presence of A3G hot spots in essential open reading frames (ORFs) of its genome. Overall, our findings uncover a novel immune evasion strategy by HCMV with profound implications for HCMV infections.IMPORTANCE APOBEC3 family of proteins plays a pivotal role in intrinsic immunity defense mechanisms against multiple viral infections, including retroviruses, through the deamination activity. However, the currently available data on APOBEC3 editing mechanisms upon HCMV infection remain unclear. In the present study, we show that particularly the APOBEC3G (A3G) member of the deaminase family is strongly induced upon infection with HCMV in fibroblasts and that its upregulation is mediated by IFN-β. Furthermore, we were able to demonstrate that neither A3G knockout nor A3G overexpression appears to modulate HCMV replication, indicating that A3G does not inhibit HCMV replication. This may be explained by HCMV escape strategy from A3G activity through depletion of the preferred nucleotide motifs (hot spots) from its genome. The results may shed light on antiviral potential of APOBEC3 activity during HCMV infection, as well as the viral counteracting mechanisms under A3G-mediated selective pressure.
Collapse
|
6
|
Human Cytomegalovirus Tegument Protein pp65 (pUL83) Dampens Type I Interferon Production by Inactivating the DNA Sensor cGAS without Affecting STING. J Virol 2018; 92:JVI.01774-17. [PMID: 29263269 DOI: 10.1128/jvi.01774-17] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/14/2017] [Indexed: 01/06/2023] Open
Abstract
The innate immune response plays a pivotal role during human cytomegalovirus (HCMV) primary infection. Indeed, HCMV infection of primary fibroblasts rapidly triggers strong induction of type I interferons (IFN-I), accompanied by proinflammatory cytokine release. Here, we show that primary human foreskin fibroblasts (HFFs) infected with a mutant HCMV TB40/E strain unable to express UL83-encoded pp65 (v65Stop) produce significantly higher IFN-β levels than HFFs infected with the wild-type TB40/E strain or the pp65 revertant (v65Rev), suggesting that the tegument protein pp65 may dampen IFN-β production. To clarify the mechanisms through which pp65 inhibits IFN-β production, we analyzed the activation of the cGAS/STING/IRF3 axis in HFFs infected with either the wild type, the revertant v65Rev, or the pp65-deficient mutant v65Stop. We found that pp65 selectively binds to cGAS and prevents its interaction with STING, thus inactivating the signaling pathway through the cGAS/STING/IRF3 axis. Consistently, addition of exogenous cGAMP to v65Rev-infected cells triggered the production of IFN-β levels similar to those observed with v65Stop-infected cells, confirming that pp65 inactivation of IFN-β production occurs at the cGAS level. Notably, within the first 24 h of HCMV infection, STING undergoes proteasome degradation independently of the presence or absence of pp65. Collectively, our data provide mechanistic insights into the interplay between HCMV pp65 and cGAS, leading to subsequent immune evasion by this prominent DNA virus.IMPORTANCE Primary human foreskin fibroblasts (HFFs) produce type I IFN (IFN-I) when infected with HCMV. However, we observed significantly higher IFN-β levels when HFFs were infected with HCMV that was unable to express UL83-encoded pp65 (v65Stop), suggesting that pp65 (pUL83) may constitute a viral evasion factor. This study demonstrates that the HCMV tegument protein pp65 inhibits IFN-β production by binding and inactivating cGAS early during infection. In addition, this inhibitory activity specifically targets cGAS, since it can be bypassed via the addition of exogenous cGAMP, even in the presence of pp65. Notably, STING proteasome-mediated degradation was observed in both the presence and absence of pp65. Collectively, our data underscore the important role of the tegument protein pp65 as a critical molecular hub in HCMV's evasion strategy against the innate immune response.
Collapse
|
7
|
Albertini S, Lo Cigno I, Calati F, De Andrea M, Borgogna C, Dell'Oste V, Landolfo S, Gariglio M. HPV18 Persistence Impairs Basal and DNA Ligand-Mediated IFN-β and IFN-λ 1 Production through Transcriptional Repression of Multiple Downstream Effectors of Pattern Recognition Receptor Signaling. THE JOURNAL OF IMMUNOLOGY 2018; 200:2076-2089. [PMID: 29386255 DOI: 10.4049/jimmunol.1701536] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/05/2018] [Indexed: 12/17/2022]
Abstract
Although it is clear that high-risk human papillomaviruses (HPVs) can selectively infect keratinocytes and persist in the host, it still remains to be unequivocally determined whether they can escape antiviral innate immunity by interfering with pattern recognition receptor (PRR) signaling. In this study, we have assessed the innate immune response in monolayer and organotypic raft cultures of NIKS cells harboring multiple copies of episomal HPV18 (NIKSmcHPV18), which fully recapitulates the persistent state of infection. We show for the first time, to our knowledge, that NIKSmcHPV18, as well as HeLa cells (a cervical carcinoma-derived cell line harboring integrated HPV18 DNA), display marked downregulation of several PRRs, as well as other PRR downstream effectors, such as the adaptor protein stimulator of IFN genes and the transcription factors IRF1 and 7. Importantly, we provide evidence that downregulation of stimulator of IFN genes, cyclic GMP-AMP synthase, and retinoic acid-inducible gene I mRNA levels occurs at the transcriptional level through a novel epigenetic silencing mechanism, as documented by the accumulation of repressive heterochromatin markers seen at the promoter region of these genes. Furthermore, stimulation of NIKSmcHPV18 cells with salmon sperm DNA or poly(deoxyadenylic-deoxythymidylic) acid, two potent inducers of PRR signaling, only partially restored PRR protein expression. Accordingly, the production of IFN-β and IFN-λ1 was significantly reduced in comparison with the parental NIKS cells, indicating that HPV18 exerts its immunosuppressive activity through downregulation of PRR signaling. Altogether, our findings indicate that high-risk human papillomaviruses have evolved broad-spectrum mechanisms that allow simultaneous depletion of multiple effectors of the innate immunity network, thereby creating an unreactive cellular milieu suitable for viral persistence.
Collapse
Affiliation(s)
- Silvia Albertini
- Virology Unit, Department of Translational Medicine, Novara Medical School, Novara 28100, Italy; and
| | - Irene Lo Cigno
- Virology Unit, Department of Translational Medicine, Novara Medical School, Novara 28100, Italy; and
| | - Federica Calati
- Virology Unit, Department of Translational Medicine, Novara Medical School, Novara 28100, Italy; and
| | - Marco De Andrea
- Virology Unit, Department of Translational Medicine, Novara Medical School, Novara 28100, Italy; and.,Viral Pathogenesis Unit, Department of Public Health and Pediatric Sciences, Turin Medical School, Turin 10126, Italy
| | - Cinzia Borgogna
- Virology Unit, Department of Translational Medicine, Novara Medical School, Novara 28100, Italy; and
| | - Valentina Dell'Oste
- Viral Pathogenesis Unit, Department of Public Health and Pediatric Sciences, Turin Medical School, Turin 10126, Italy
| | - Santo Landolfo
- Viral Pathogenesis Unit, Department of Public Health and Pediatric Sciences, Turin Medical School, Turin 10126, Italy
| | - Marisa Gariglio
- Virology Unit, Department of Translational Medicine, Novara Medical School, Novara 28100, Italy; and
| |
Collapse
|
8
|
Pisano G, Roy A, Ahmed Ansari M, Kumar B, Chikoti L, Chandran B. Interferon-γ-inducible protein 16 (IFI16) is required for the maintenance of Epstein-Barr virus latency. Virol J 2017; 14:221. [PMID: 29132393 PMCID: PMC5683537 DOI: 10.1186/s12985-017-0891-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 11/02/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Epstein-Barr virus (EBV) exhibits both lytic and latent (Lat. I, II, and III) phases in an infected individual. It's during the latent phase of EBV that all EBV-associated cancers, including Burkitt's lymphoma, nasopharyngeal carcinoma and lymphoproliferative disease arise. Interferon-γ-inducible protein 16 (IFI16) is a well-established innate immune sensor and viral transcriptional regulator involved in response to invading DNA viruses. During latency, IFI16 remains in the nucleus, in part bound to the EBV genome; however, neither its role in EBV lytic cycle or latency has been established. METHODS Short interfering RNA against IFI16 and IFI16 overexpression were used to identify the role of IFI16 in the maintenance of EBV latency I. We also studied how induction of the lytic cycle affected IFI16 using the EBV positive, latently infected Akata or MUTU-1 cell lines. Akata cells were induced with TPA and MUTU-1 cells with TGF-β up to 96 h and changes in IFI16 protein were analyzed by Western blotting and immunofluorescence microscopy. To assess the mechanism of IFI16 decrease, EBV DNA replication and late lytic transcripts were blocked using the viral DNA polymerase inhibitor phosphonoacetic acid. RESULTS Knockdown of IFI16 mRNA by siRNA resulted in enhanced levels of EBV lytic gene expression from all temporal gene classes, as well as an increase in the total EBV genome abundance, whereas overexpression of exogenous IFI16 reversed these effects. Furthermore, 96 h after induction of the lytic cycle with either TPA (Akata) or TGF-β (MUTU-1), IFI16 protein levels decreased up to 80% as compared to the EBV-negative cell line BJAB. Reduction in IFI16 was observed in cells expressing EBV lytic envelope glycoprotein. The decreased levels of IFI16 protein do not appear to be dependent on late lytic transcripts of EBV but suggest involvement of the immediate early, early, or a combination of both gene classes. CONCLUSIONS Reduction of IFI16 protein levels following lytic cycle induction, as well as reactivation from latency after IFI16 mRNA knockdown suggests that IFI16 is crucial for the maintenance of EBV latency. More importantly, these results identify IFI16 as a unique host factor protein involved in the EBV lifecycle, making it a potential therapeutic target to combat EBV-related malignancies.
Collapse
Affiliation(s)
- Gina Pisano
- H.M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA.
| | - Arunava Roy
- H.M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Mairaj Ahmed Ansari
- H.M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Binod Kumar
- H.M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Leela Chikoti
- H.M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Bala Chandran
- H.M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| |
Collapse
|
9
|
McMahan ZH, Shah AA, Vaidya D, Wigley FM, Rosen A, Casciola-Rosen L. Anti-Interferon-Inducible Protein 16 Antibodies Associate With Digital Gangrene in Patients With Scleroderma. Arthritis Rheumatol 2017; 68:1262-71. [PMID: 26714268 DOI: 10.1002/art.39558] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 12/15/2015] [Indexed: 11/07/2022]
Abstract
OBJECTIVE To examine the association between anti-interferon-inducible protein 16 (anti-IFI-16) antibodies and clinical features of scleroderma. METHODS Sera from a discovery sample of 94 patients with scleroderma and 47 healthy controls were assayed for anti-IFI-16 antibodies by enzyme-linked immunosorbent assay, and associations were examined using regression analyses. Since anti-IFI-16 autoantibodies were found to be strongly associated with digital gangrene in the discovery sample, a subsequent case-control study (with subjects matched 1:1 on disease duration) was designed for further exploration. Cases were patients with scleroderma and digital gangrene, while controls were patients with scleroderma and Raynaud's phenomenon alone (n = 39 matched pairs). Nonparametric, unadjusted matched pairs analysis as well as univariate and multivariable conditional logistic regression analyses were performed. RESULTS In the discovery sample, anti-IFI-16 antibodies were more prevalent in patients with scleroderma than in healthy controls (18% versus 2%; P = 0.01). Patients with anti-IFI-16 antibodies, compared to anti-IFI-16 antibody-negative patients, were more likely to have limited scleroderma (77% versus 46%; P = 0.03), a longer disease duration (median 15.2 years [interquartile range 10.6-18.3] versus 6.0 years [interquartile range 3.4-13.8]; P < 0.01), digital gangrene (24% versus 4%; P = 0.02), and a low diffusing capacity for carbon monoxide (DLco) (P < 0.01). In the case-control study, 35 (45%) of 78 patients were anti-IFI-16 antibody positive. Anti-IFI-16 antibody levels were significantly higher in cases with digital gangrene than in matched controls (P = 0.02). In analyses adjusted for age, cutaneous scleroderma subtype, smoking, and DLco, high anti-IFI-16 antibody levels were associated with the presence of digital gangrene (adjusted odds ratio 2.3, 95% confidence interval 1.0-5.6, P = 0.05). The odds of having digital gangrene increased with higher anti-IFI-16 antibody titers, in a dose-dependent manner. CONCLUSION Anti-IFI-16 antibodies are associated with digital gangrene in patients with scleroderma. Longitudinal prospective studies exploring anti-IFI-16 antibodies as a disease biomarker, and biologic studies investigating the pathogenicity of these antibodies, are warranted.
Collapse
Affiliation(s)
| | - Ami A Shah
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dhananjay Vaidya
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Antony Rosen
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | |
Collapse
|
10
|
McMahan ZH, Cottrell TR, Wigley FM, Antiochos B, Zambidis ET, Park TS, Halushka MK, Gutierrez-Alamillo L, Cimbro R, Rosen A, Casciola-Rosen L. Enrichment of Scleroderma Vascular Disease-Associated Autoantigens in Endothelial Lineage Cells. Arthritis Rheumatol 2016; 68:2540-9. [PMID: 27159521 PMCID: PMC5042822 DOI: 10.1002/art.39743] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 04/28/2016] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Scleroderma patients with autoantibodies to CENPs and/or interferon-inducible protein 16 (IFI-16) are at increased risk of severe vascular complications. This study was undertaken to determine whether these autoantigens are enriched in cells of the vasculature. METHODS Successive stages of embryoid bodies (EBs) as well as vascular progenitors were used to evaluate the expression of scleroderma autoantigens IFI-16 and CENP by immunoblotting. CD31 was included to mark early blood vessels. IFI-16 and CD31 expression were defined in paraffin-embedded skin sections from scleroderma patients and from healthy controls. IFI-16 expression was determined by flow cytometric analysis in circulating endothelial cells (CECs) and circulating hematopoietic progenitor cells. RESULTS Expression of CENP-A, IFI-16, and CD31 was enriched in EBs on days 10 and 12 of differentiation, and particularly in cultures enriched in vascular progenitors (IFI-16, CD31, and CENPs A and B). This pattern was distinct from that of comparator autoantigens. Immunohistochemical staining of paraffin-embedded skin sections showed enrichment of IFI-16 in CD31-positive vascular endothelial cells in biopsy specimens from scleroderma patients and normal controls. Flow cytometric analysis revealed IFI-16 expression in circulating hematopoietic progenitor cells but minimal expression in CECs. CONCLUSION Our findings indicate that expression of the scleroderma autoantigens IFI-16 and CENPs, which are associated with severe vascular disease, is increased in vascular progenitors and mature endothelial cells. High level, lineage-enriched expression of autoantigens may explain the striking association between clinical phenotypes and the immune targeting of specific autoantigens.
Collapse
Affiliation(s)
| | | | | | | | - Elias T Zambidis
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Tea Soon Park
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marc K Halushka
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Raffaello Cimbro
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Antony Rosen
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | |
Collapse
|
11
|
Nuclear Innate Immune DNA Sensor IFI16 Is Degraded during Lytic Reactivation of Kaposi's Sarcoma-Associated Herpesvirus (KSHV): Role of IFI16 in Maintenance of KSHV Latency. J Virol 2016; 90:8822-41. [PMID: 27466416 PMCID: PMC5021400 DOI: 10.1128/jvi.01003-16] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/13/2016] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED IFI16 (interferon gamma-inducible protein 16) recognizes nuclear episomal herpesvirus (Kaposi's sarcoma-associated herpesvirus [KSHV], Epstein-Barr virus [EBV], and herpes simplex virus 1 [HSV-1]) genomes and induces the inflammasome and interferon beta responses. It also acts as a lytic replication restriction factor and inhibits viral DNA replication (human cytomegalovirus [HCMV] and human papillomavirus [HPV]) and transcription (HSV-1, HCMV, and HPV) through epigenetic modifications of the viral genomes. To date, the role of IFI16 in the biology of latent viruses is not known. Here, we demonstrate that knockdown of IFI16 in the latently KSHV-infected B-lymphoma BCBL-1 and BC-3 cell lines results in lytic reactivation and increases in levels of KSHV lytic transcripts, proteins, and viral genome replication. Similar results were also observed during KSHV lytic cycle induction in TREX-BCBL-1 cells with the doxycycline-inducible lytic cycle switch replication and transcription activator (RTA) gene. Overexpression of IFI16 reduced lytic gene induction by the chemical agent 12-O-tetradecoylphorbol-13-acetate (TPA). IFI16 protein levels were significantly reduced or absent in TPA- or doxycycline-induced cells expressing lytic KSHV proteins. IFI16 is polyubiquitinated and degraded via the proteasomal pathway. The degradation of IFI16 was absent in phosphonoacetic acid-treated cells, which blocks KSHV DNA replication and, consequently, late lytic gene expression. Chromatin immunoprecipitation assays of BCBL-1 and BC-3 cells demonstrated that IFI16 binds to KSHV gene promoters. Uninfected epithelial SLK and osteosarcoma U2OS cells transfected with KSHV luciferase promoter constructs confirmed that IFI16 functions as a transcriptional repressor. These results reveal that KSHV utilizes the innate immune nuclear DNA sensor IFI16 to maintain its latency and repression of lytic transcripts, and a late lytic KSHV gene product(s) targets IFI16 for degradation during lytic reactivation. IMPORTANCE Like all herpesviruses, latency is an integral part of the life cycle of Kaposi's sarcoma-associated herpesvirus (KSHV), an etiological agent for many human cancers. Herpesviruses utilize viral and host factors to successfully evade the host immune system to maintain latency. Reactivation is a complex event where the latent episomal viral genome springs back to active transcription of lytic cycle genes. Our studies reveal that KSHV has evolved to utilize the innate immune sensor IFI16 to keep lytic cycle transcription in dormancy. We demonstrate that IFI16 binds to the lytic gene promoter, acts as a transcriptional repressor, and thereby helps to maintain latency. We also discovered that during the late stage of lytic replication, KSHV selectively degrades IFI16, thus relieving transcriptional repression. This is the first report to demonstrate the role of IFI16 in latency maintenance of a herpesvirus, and further understanding will lead to the development of strategies to eliminate latent infection.
Collapse
|
12
|
Regulatory Interaction between the Cellular Restriction Factor IFI16 and Viral pp65 (pUL83) Modulates Viral Gene Expression and IFI16 Protein Stability. J Virol 2016; 90:8238-50. [PMID: 27384655 DOI: 10.1128/jvi.00923-16] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 06/28/2016] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED A key player in the intrinsic resistance against human cytomegalovirus (HCMV) is the interferon-γ-inducible protein 16 (IFI16), which behaves as a viral DNA sensor in the first hours postinfection and as a repressor of viral gene transcription in the later stages. Previous studies on HCMV replication demonstrated that IFI16 binds to the viral protein kinase pUL97, undergoes phosphorylation, and relocalizes to the cytoplasm of infected cells. In this study, we demonstrate that the tegument protein pp65 (pUL83) recruits IFI16 to the promoter of the UL54 gene and downregulates viral replication, as shown by use of the HCMV mutant v65Stop, which lacks pp65 expression. Interestingly, at late time points of HCMV infection, IFI16 is stabilized by its interaction with pp65, which stood in contrast to IFI16 degradation, observed in herpes simplex virus 1 (HSV-1)-infected cells. Moreover, we found that its translocation to the cytoplasm, in addition to pUL97, strictly depends on pp65, as demonstrated with the HCMV mutant RV-VM1, which expresses a form of pp65 unable to translocate into the cytoplasm. Thus, these data reveal a dual role for pp65: during early infection, it modulates IFI16 activity at the promoter of immediate-early and early genes; subsequently, it delocalizes IFI16 from the nucleus into the cytoplasm, thereby stabilizing and protecting it from degradation. Overall, these data identify a novel activity of the pp65/IFI16 interactome involved in the regulation of UL54 gene expression and IFI16 stability during early and late phases of HCMV replication. IMPORTANCE The DNA sensor IFI16, a member of the PYHIN proteins, restricts HCMV replication by impairing viral DNA synthesis. Using a mutant virus lacking the tegument protein pp65 (v65Stop), we demonstrate that pp65 recruits IFI16 to the early UL54 gene promoter. As a putative counteraction to its restriction activity, pp65 supports the nucleocytoplasmic export of IFI16, which was demonstrated with the viral mutant RV-VM1 expressing a nuclearly retained pp65. These data reveal a dual role of pp65 in IFI16 regulation: in the early phase of HCMV infection, it contributes to viral evasion from IFI16 restriction activity, while at later time points, it promotes the nuclear delocalization of IFI16, thereby stabilizing and protecting it from degradation. In the present work, we further clarify the mechanisms HCMV relies on to overcome intracellular innate immune restriction and provide new insights into the relevance of DNA-sensing restriction factor IFI16 during HCMV infection.
Collapse
|
13
|
Landolfo S, De Andrea M, Dell’Oste V, Gugliesi F. Intrinsic host restriction factors of human cytomegalovirus replication and mechanisms of viral escape. World J Virol 2016; 5:87-96. [PMID: 27563536 PMCID: PMC4981826 DOI: 10.5501/wjv.v5.i3.87] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/03/2016] [Accepted: 07/13/2016] [Indexed: 02/05/2023] Open
Abstract
Before a pathogen even enters a cell, intrinsic immune defenses are active. This first-line defense is mediated by a variety of constitutively expressed cell proteins collectively termed “restriction factors” (RFs), and they form a vital element of the immune response to virus infections. Over time, however, viruses have evolved in a variety ways so that they are able to overcome these RF defenses via mechanisms that are specific for each virus. This review provides a summary of the universal characteristics of RFs, and goes on to focus on the strategies employed by some of the most important RFs in their attempt to control human cytomegalovirus (HCMV) infection. This is followed by a discussion of the counter-restriction mechanisms evolved by viruses to circumvent the host cell’s intrinsic immune defenses. RFs include nuclear proteins IFN-γ inducible protein 16 (IFI16) (a Pyrin/HIN domain protein), Sp100, promyelocytic leukemia, and hDaxx; the latter three being the keys elements of nuclear domain 10 (ND10). IFI16 inhibits the synthesis of virus DNA by down-regulating UL54 transcription - a gene encoding a CMV DNA polymerase; in response, the virus antagonizes IFI16 via a process involving viral proteins UL97 and pp65 (pUL83), which results in the mislocalizing of IFI16 into the cytoplasm. In contrast, viral regulatory proteins, including pp71 and IE1, seek to modify or disrupt the ND10 proteins and thus block or reverse their inhibitory effects upon virus replication. All in all, detailed knowledge of these HCMV counter-restriction mechanisms will be fundamental for the future development of new strategies for combating HCMV infection and for identifying novel therapeutic agents.
Collapse
|
14
|
Choubey D, Panchanathan R. IFI16, an amplifier of DNA-damage response: Role in cellular senescence and aging-associated inflammatory diseases. Ageing Res Rev 2016; 28:27-36. [PMID: 27063514 DOI: 10.1016/j.arr.2016.04.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 03/30/2016] [Accepted: 04/04/2016] [Indexed: 12/22/2022]
Abstract
DNA-damage induces a DNA-damage response (DDR) in mammalian cells. The response, depending upon the cell-type and the extent of DNA-damage, ultimately results in cell death or cellular senescence. DDR-induced signaling in cells activates the ATM-p53 and ATM-IKKα/β-interferon (IFN)-β signaling pathways, thus leading to an induction of the p53 and IFN-inducible IFI16 gene. Further, upon DNA-damage, DNA accumulates in the cytoplasm, thereby inducing the IFI16 protein and STING-dependent IFN-β production and activation of the IFI16 inflammasome, resulting in the production of proinflammatory cytokines (e.g., IL-1β and IL-18). Increased expression of IFI16 protein in a variety of cell-types promotes cellular senescence. However, reduced expression of IFI16 in cells promotes cell proliferation. Because expression of the IFI16 gene is induced by activation of DNA-damage response in cells and increased levels of IFI16 protein in cells potentiate the p53-mediated transcriptional activation of genes and p53 and pRb-mediated cell cycle arrest, we discuss how an improved understanding of the role of IFI16 protein in cellular senescence and associated inflammatory secretory phenotype is likely to identify the molecular mechanisms that contribute to the development of aging-associated human inflammatory diseases and a failure to cancer therapy.
Collapse
Affiliation(s)
- Divaker Choubey
- Cincinnati VA Medical Center, 3200 Vine Street, Cincinnati, OH 45220, United States; Department of Environmental Health, University of Cincinnati, 160 Panzeca Way, P.O. Box-670056, Cincinnati, OH 45267, United States.
| | - Ravichandran Panchanathan
- Cincinnati VA Medical Center, 3200 Vine Street, Cincinnati, OH 45220, United States; Department of Environmental Health, University of Cincinnati, 160 Panzeca Way, P.O. Box-670056, Cincinnati, OH 45267, United States
| |
Collapse
|
15
|
IFI16 Expression Is Related to Selected Transcription Factors during B-Cell Differentiation. J Immunol Res 2015; 2015:747645. [PMID: 26185770 PMCID: PMC4491573 DOI: 10.1155/2015/747645] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 04/27/2015] [Accepted: 05/14/2015] [Indexed: 01/21/2023] Open
Abstract
The interferon-inducible DNA sensor IFI16 is involved in the modulation of cellular survival, proliferation, and differentiation. In the hematopoietic system, IFI16 is consistently expressed in the CD34+ stem cells and in peripheral blood lymphocytes; however, little is known regarding its regulation during maturation of B- and T-cells. We explored the role of IFI16 in normal B-cell subsets by analysing its expression and relationship with the major transcription factors involved in germinal center (GC) development and plasma-cell (PC) maturation. IFI16 mRNA was differentially expressed in B-cell subsets with significant decrease in IFI16 mRNA in GC and PCs with respect to naïve and memory subsets. IFI16 mRNA expression is inversely correlated with a few master regulators of B-cell differentiation such as BCL6, XBP1, POU2AF1, and BLIMP1. In contrast, IFI16 expression positively correlated with STAT3, REL, SPIB, RELA, RELB, IRF4, STAT5B, and STAT5A. ARACNE algorithm indicated a direct regulation of IFI16 by BCL6, STAT5B, and RELB, whereas the relationship between IFI16 and the other factors is modulated by intermediate factors. In addition, analysis of the CD40 signaling pathway showed that IFI16 gene expression directly correlated with NF-κB activation, indicating that IFI16 could be considered an upstream modulator of NF-κB in human B-cells.
Collapse
|
16
|
The Nuclear DNA Sensor IFI16 Acts as a Restriction Factor for Human Papillomavirus Replication through Epigenetic Modifications of the Viral Promoters. J Virol 2015; 89:7506-20. [PMID: 25972554 DOI: 10.1128/jvi.00013-15] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 04/30/2015] [Indexed: 01/01/2023] Open
Abstract
UNLABELLED The human interferon-inducible IFI16 protein, an innate immune sensor of intracellular DNA, was recently demonstrated to act as a restriction factor for human cytomegalovirus (HCMV) and herpes simplex virus 1 (HSV-1) infection by inhibiting both viral-DNA replication and transcription. Through the use of two distinct cellular models, this study provides strong evidence in support of the notion that IFI16 can also restrict human papillomavirus 18 (HPV18) replication. In the first model, an immortalized keratinocyte cell line (NIKS) was used, in which the IFI16 protein was knocked down through the use of small interfering RNA (siRNA) technology and overexpressed following transduction with the adenovirus IFI16 (AdVIFI16) vector. The second model consisted of U2OS cells transfected by electroporation with HPV18 minicircles. In differentiated IFI16-silenced NIKS-HPV18 cells, viral-load values were significantly increased compared with differentiated control cells. Consistent with this, IFI16 overexpression severely impaired HPV18 replication in both NIKS and U2OS cells, thus confirming its antiviral restriction activity. In addition to the inhibition of viral replication, IFI16 was also able to reduce viral transcription, as demonstrated by viral-gene expression analysis in U2OS cells carrying episomal HPV18 minicircles and HeLa cells. We also provide evidence that IFI16 promotes the addition of heterochromatin marks and the reduction of euchromatin marks on viral chromatin at both early and late promoters, thus reducing both viral replication and transcription. Altogether, these results argue that IFI16 restricts chromatinized HPV DNA through epigenetic modifications and plays a broad surveillance role against viral DNA in the nucleus that is not restricted to herpesviruses. IMPORTANCE Intrinsic immunity is mediated by cellular restriction factors that are constitutively expressed and active even before a pathogen enters the cell. The host nuclear factor IFI16 acts as a sensor of foreign DNA and an antiviral restriction factor, as recently demonstrated by our group for human cytomegalovirus (HCMV) and herpes simplex virus 1 (HSV-1). Here, we provide the first evidence that IFI16 inhibits HPV18 replication by repressing viral-gene expression and replication. This antiviral restriction activity was observed in immortalized keratinocytes transfected with the religated genomes and in U2OS cells transfected with HPV18 minicircles, suggesting that it is not cell type specific. We also show that IFI16 promotes the assembly of heterochromatin on HPV DNA. These changes in viral chromatin structure lead to the generation of a repressive state at both early and late HPV18 promoters, thus implicating the protein in the epigenetic regulation of HPV gene expression and replication.
Collapse
|
17
|
Chaum E, Winborn CS, Bhattacharya S. Genomic regulation of senescence and innate immunity signaling in the retinal pigment epithelium. Mamm Genome 2015; 26:210-21. [PMID: 25963977 DOI: 10.1007/s00335-015-9568-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/02/2015] [Indexed: 01/04/2023]
Abstract
The tumor suppressor p53 is a major regulator of genes important for cell cycle arrest, senescence, apoptosis, and innate immunity, and has recently been implicated in retinal aging. In this study we sought to identify the genetic networks that regulate p53 function in the retina using quantitative trait locus (QTL) analysis. First we examined age-associated changes in the activation and expression levels of p53; known p53 target proteins and markers of innate immune system activation in primary retinal pigment epithelial (RPE) cells that were harvested from young and aged human donors. We observed increased expression of p53, activated caspase-1, CDKN1A, CDKN2A (p16INK4a), TLR4, and IFNα in aged primary RPE cell lines. We used the Hamilton Eye Institute (HEI) retinal dataset ( www.genenetwork.org ) to identify genomic loci that modulate expression of genes in the p53 pathway in recombinant inbred BXD mouse strains using a QTL systems biology-based approach. We identified a significant trans-QTL on chromosome 1 (region 172-177 Mb) that regulates the expression of Cdkn1a. Many of the genes in this QTL locus are involved in innate immune responses, including Fc receptors, interferon-inducible family genes, and formin 2. Importantly, we found an age-related increase in FCGR3A and FMN2 and a decrease in IFI16 levels in RPE cultures. There is a complex multigenic innate immunity locus that controls expression of genes in the p53 pathway in the RPE, which may play an important role in modulating age-related changes in the retina.
Collapse
Affiliation(s)
- Edward Chaum
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA,
| | | | | |
Collapse
|
18
|
Rusetskaya NY, Borodulin VB. Biological activity of organoselenium compounds in heavy metal intoxication. BIOCHEMISTRY MOSCOW-SUPPLEMENT SERIES B-BIOMEDICAL CHEMISTRY 2015. [DOI: 10.1134/s1990750815010072] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Bawadekar M, De Andrea M, Lo Cigno I, Baldanzi G, Caneparo V, Graziani A, Landolfo S, Gariglio M. The Extracellular IFI16 Protein Propagates Inflammation in Endothelial Cells Via p38 MAPK and NF-κB p65 Activation. J Interferon Cytokine Res 2015; 35:441-53. [PMID: 25715050 DOI: 10.1089/jir.2014.0168] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The nuclear interferon-inducible-16 (IFI16) protein acts as DNA sensor in inflammasome signaling and as viral restriction factor. Following Herpesvirus infection or UV-B treatment, IFI16 delocalizes from the nucleus to the cytoplasm and is eventually released into the extracellular milieu. Recently, our group has demonstrated the occurrence of IFI16 in sera of systemic-autoimmune patients that hampers biological activity of endothelia through high-affinity membrane binding. As a continuation, we studied the activity of endotoxin-free recombinant IFI16 (rIFI16) protein on primary endothelial cells. rIFI16 caused dose/time-dependent upregulation of IL-6, IL-8, CCL2, CCL5, CCL20, ICAM1, VCAM1, and TLR4, while secretion of IL-6 and IL-8 was amplified with lipopolysaccharide synergy. Overall, cytokine secretion was completely inhibited in MyD88-silenced cells and partially by TLR4-neutralizing antibodies. By screening downstream signaling pathways, we found that IFI16 activates p38, p44/42 MAP kinases, and NF-kB. In particular, activation of p38 is an early event required for subsequent p44/42 MAP kinases activity and cytokine induction indicating a key role of this kinase in IFI16 signaling. Altogether, our data conclude that extracellular IFI16 protein alone or by synergy with lipopolysaccharide acts like Damage-associated molecular patterns propagating "Danger Signal" through MyD88-dependent TLR-pathway.
Collapse
Affiliation(s)
- Mandar Bawadekar
- 1 Department of Translational Medicine, University of Eastern Piedmont , Novara, Italy .,2 Interdisciplinary Research Center of Autoimmune Diseases (IRCAD) , Novara, Italy .,3 Division of Rheumatology, Department of Medicine, University of Wisconsin-Madison , Wisconsin
| | - Marco De Andrea
- 4 Department of Public Health and Pediatric Sciences, University of Turin , Medical School, Turin, Italy
| | - Irene Lo Cigno
- 1 Department of Translational Medicine, University of Eastern Piedmont , Novara, Italy .,2 Interdisciplinary Research Center of Autoimmune Diseases (IRCAD) , Novara, Italy
| | - Gianluca Baldanzi
- 1 Department of Translational Medicine, University of Eastern Piedmont , Novara, Italy
| | - Valeria Caneparo
- 1 Department of Translational Medicine, University of Eastern Piedmont , Novara, Italy .,2 Interdisciplinary Research Center of Autoimmune Diseases (IRCAD) , Novara, Italy
| | - Andrea Graziani
- 1 Department of Translational Medicine, University of Eastern Piedmont , Novara, Italy
| | - Santo Landolfo
- 4 Department of Public Health and Pediatric Sciences, University of Turin , Medical School, Turin, Italy
| | - Marisa Gariglio
- 1 Department of Translational Medicine, University of Eastern Piedmont , Novara, Italy .,2 Interdisciplinary Research Center of Autoimmune Diseases (IRCAD) , Novara, Italy
| |
Collapse
|
20
|
Rusetskaya N, Borodulin V. Biological activity of selenorganic compounds at heavy metal salts intoxication. ACTA ACUST UNITED AC 2015; 61:449-61. [DOI: 10.18097/pbmc20156104449] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Possible mechanisms of the antitoxic action of organoselenium compounds in heavy metal poisoning have been considered. Heavy metal toxicity associated with intensification of free radical oxidation, suppression of the antioxidant system, damage to macromolecules, mitochondria and the genetic material can cause apoptotic cell death or the development of carcinogenesis. Organic selenium compounds are effective antioxidants during heavy metal poisoning; they exhibit higher bioavailability in mammals than inorganic ones and they are able to activate antioxidant defense, bind heavy metal ions and reactive oxygen species formed during metal-induced oxidative stress. One of promising organoselenium compounds is diacetophenonyl selenide (DAPS-25), which is characterized by antioxidant and antitoxic activity, under conditions including heavy metal intoxication
Collapse
Affiliation(s)
- N.Y. Rusetskaya
- Razumovskiy Saratov State Medical University, Saratov, Russia
| | - V.B. Borodulin
- Razumovskiy Saratov State Medical University, Saratov, Russia
| |
Collapse
|
21
|
Landolfo S, Andrea MD, Gariglio M. Restriction factors against human CMV. Future Virol 2014. [DOI: 10.2217/fvl.14.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cellular proteins called 'restriction factors' (RFs) form an important component of the innate immune response to viral replication. However, viruses have learned how to antagonize RFs through mechanisms that are specific for each virus. Here, we summarize the general hallmarks of RFs before going on to discuss the specific strategies recruited by some key RFs that strive to hold human CMV (HCMV) infection back, as well as the counter-restriction mechanisms employed by the virus to overcome this innate defense. Such RFs include the cellular constituents of nuclear domain 10 (ND10), and IFI16, a nuclear member of the PYHIN protein family. Viral regulatory proteins, such as IE1 or pp71, try to oppose the ND10-induced blockade of virus replication by either modifying or disrupting this RF. IFI16, on the other hand, inhibits virus DNA synthesis by downregulating the transcription of viral gene UL54; the intruding virus attempts to antagonize IFI16 by mislocalizing it from the nucleus to the cytoplasm via the action of viral protein UL97. Finally, we consider how Viperin, a RF initially thought to inhibit HCMV maturation late during infection, has actually been demonstrated to enhance virus maturation by increasing lipid metabolism and enhancing virus envelopment.
Collapse
Affiliation(s)
- Santo Landolfo
- Viral Pathogenesis Unit, Department of Public Health & Pediatric Sciences, Medical School, University of Turin, Italy
| | - Marco De Andrea
- Viral Pathogenesis Unit, Department of Public Health & Pediatric Sciences, Medical School, University of Turin, Italy
- Virology Unit, Department of Translational Medicine, Medical School of Novara, Italy
| | - Marisa Gariglio
- Virology Unit, Department of Translational Medicine, Medical School of Novara, Italy
| |
Collapse
|
22
|
Innate nuclear sensor IFI16 translocates into the cytoplasm during the early stage of in vitro human cytomegalovirus infection and is entrapped in the egressing virions during the late stage. J Virol 2014; 88:6970-82. [PMID: 24696486 DOI: 10.1128/jvi.00384-14] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Intrinsic immune mechanisms mediated by constitutively expressed proteins termed "restriction factors" provide frontline antiviral defense. We recently demonstrated that the DNA sensor IFI16 restricts human cytomegalovirus (HCMV) replication by downregulating viral early and late but not immediate-early mRNAs and their protein expression. We show here that at an early time point during the in vitro infection of low-passage-number human embryonic lung fibroblasts, IFI16 binds to HCMV DNA. However, during a later phase following infection, IFI16 is mislocalized to the cytoplasmic virus assembly complex (AC), where it colocalizes with viral structural proteins. Indeed, upon its binding to pUL97, IFI16 undergoes phosphorylation and relocalizes to the cytoplasm of HCMV-infected cells. ESCRT (endosomal sorting complex required for transport) machinery regulates the translocation of IFI16 into the virus AC by sorting and trafficking IFI16 into multivesicular bodies (MVB), as demonstrated by the interaction of IFI16 with two MVB markers: Vps4 and TGN46. Finally, IFI16 becomes incorporated into the newly assembled virions as demonstrated by Western blotting of purified virions and electron microscopy. Together, these results suggest that HCMV has evolved mechanisms to mislocalize and hijack IFI16, trapping it within mature virions. However, the significance of this IFI16 trapping following nuclear mislocalization remains to be established. IMPORTANCE Intracellular viral DNA sensors and restriction factors are critical components of host defense, which alarm and sensitize immune system against intruding pathogens. We have recently demonstrated that the DNA sensor IFI16 restricts human cytomegalovirus (HCMV) replication by downregulating viral early and late but not immediate-early mRNAs and their protein expression. However, viruses are known to evolve numerous strategies to cope and counteract such restriction factors and neutralize the first line of host defense mechanisms. Our findings describe that during early stages of infection, IFI16 successfully recognizes HCMV DNA. However, in late stages HCMV mislocalizes IFI16 into the cytoplasmic viral assembly complex and finally entraps the protein into mature virions. We clarify here the mechanisms HCMV relies to overcome intracellular viral restriction, which provides new insights about the relevance of DNA sensors during HCMV infection.
Collapse
|
23
|
Marinho HS, Real C, Cyrne L, Soares H, Antunes F. Hydrogen peroxide sensing, signaling and regulation of transcription factors. Redox Biol 2014; 2:535-62. [PMID: 24634836 PMCID: PMC3953959 DOI: 10.1016/j.redox.2014.02.006] [Citation(s) in RCA: 585] [Impact Index Per Article: 53.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 02/19/2014] [Accepted: 02/21/2014] [Indexed: 12/12/2022] Open
Abstract
The regulatory mechanisms by which hydrogen peroxide (H2O2) modulates the activity of transcription factors in bacteria (OxyR and PerR), lower eukaryotes (Yap1, Maf1, Hsf1 and Msn2/4) and mammalian cells (AP-1, NRF2, CREB, HSF1, HIF-1, TP53, NF-κB, NOTCH, SP1 and SCREB-1) are reviewed. The complexity of regulatory networks increases throughout the phylogenetic tree, reaching a high level of complexity in mammalians. Multiple H2O2 sensors and pathways are triggered converging in the regulation of transcription factors at several levels: (1) synthesis of the transcription factor by upregulating transcription or increasing both mRNA stability and translation; (ii) stability of the transcription factor by decreasing its association with the ubiquitin E3 ligase complex or by inhibiting this complex; (iii) cytoplasm–nuclear traffic by exposing/masking nuclear localization signals, or by releasing the transcription factor from partners or from membrane anchors; and (iv) DNA binding and nuclear transactivation by modulating transcription factor affinity towards DNA, co-activators or repressors, and by targeting specific regions of chromatin to activate individual genes. We also discuss how H2O2 biological specificity results from diverse thiol protein sensors, with different reactivity of their sulfhydryl groups towards H2O2, being activated by different concentrations and times of exposure to H2O2. The specific regulation of local H2O2 concentrations is also crucial and results from H2O2 localized production and removal controlled by signals. Finally, we formulate equations to extract from typical experiments quantitative data concerning H2O2 reactivity with sensor molecules. Rate constants of 140 M−1 s−1 and ≥1.3 × 103 M−1 s−1 were estimated, respectively, for the reaction of H2O2 with KEAP1 and with an unknown target that mediates NRF2 protein synthesis. In conclusion, the multitude of H2O2 targets and mechanisms provides an opportunity for highly specific effects on gene regulation that depend on the cell type and on signals received from the cellular microenvironment. Complexity of redox regulation increases along the phylogenetic tree. Complex regulatory networks allow for a high degree of H2O2 biological plasticity. H2O2 modulates gene expression at all steps from transcription to protein synthesis. Fast response (s) is mediated by sensors with high H2O2 reactivity. Low reactivity H2O2 sensors may mediate slow (h) or localized H2O2 responses.
Collapse
Affiliation(s)
- H. Susana Marinho
- Departamento de Química e Bioquímica, Centro de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Carla Real
- Departamento de Química e Bioquímica, Centro de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Luísa Cyrne
- Departamento de Química e Bioquímica, Centro de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Helena Soares
- Departamento de Química e Bioquímica, Centro de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
- Escola Superior de Tecnologia da Saúde de Lisboa, IPL, Lisboa, Portugal
| | - Fernando Antunes
- Departamento de Química e Bioquímica, Centro de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- Corresponding author.
| |
Collapse
|
24
|
Alabdali A, Al-Ayadhi L, El-Ansary A. Association of social and cognitive impairment and biomarkers in autism spectrum disorders. J Neuroinflammation 2014; 11:4. [PMID: 24400970 PMCID: PMC3896747 DOI: 10.1186/1742-2094-11-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 12/17/2013] [Indexed: 11/21/2022] Open
Abstract
Objectives The neurological basis for autism is still not fully understood, and the role of the interaction between neuro-inflammation and neurotransmission impairment needs to be clearer. This study aims to test the possible association between impaired levels of gamma aminobutyric acid (GABA), serotonin, dopamine, oxytocin, and interferon-γ-induced protein-16 (IFI16) and the severity of social and cognitive dysfunctions in individuals with autism spectrum disorders. Materials and methods GABA, serotonin, dopamine, oxytocin, and IFI16 as biochemical parameters related to neurochemistry and inflammation were determined in the plasma of 52 Saudi autistic male patients, categorized as mild-moderate and severe as indicated by their Childhood Autism Rating Scale (CARS) or social responsiveness scale (SRS), and compared to 30 age- and gender-matched control samples. Results The data indicated that Saudi patients with autism have remarkably impaired plasma levels of the measured parameters compared to age and gender-matched controls. While serotonin in platelet-free plasma and dopamine did not correlated with the severity in social and cognitive dysfunction, GABA, oxytocin, and IFI16 were remarkably associated with the severity of both tested scores (SRS and CARS). Conclusions The relationship between the selected parameters confirms the role of impaired neurochemistry and neuro-inflammation in the etiology of autism spectrum disorders and the possibility of using GABA, oxytocin, and IFI16 as markers of autism severity. Receiver operating characteristic analysis together with predictiveness diagrams proved that the measured parameters could be used as predictive biomarkers of clinical symptoms and provide significant guidance for future therapeutic strategy to re-establish physiological homeostasis.
Collapse
Affiliation(s)
| | | | - Afaf El-Ansary
- Biochemistry Department, Science College, King Saud University, P,O box 22452, Zip code 11495 Riyadh, Saudi Arabia.
| |
Collapse
|
25
|
Nuclear DNA sensor IFI16 as circulating protein in autoimmune diseases is a signal of damage that impairs endothelial cells through high-affinity membrane binding. PLoS One 2013; 8:e63045. [PMID: 23690979 PMCID: PMC3653904 DOI: 10.1371/journal.pone.0063045] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 03/28/2013] [Indexed: 01/30/2023] Open
Abstract
IFI16, a nuclear pathogenic DNA sensor induced by several pro-inflammatory cytokines, is a multifaceted protein with various functions. It is also a target for autoantibodies as specific antibodies have been demonstrated in the sera of patients affected by systemic autoimmune diseases. Following transfection of virus-derived DNA, or treatment with UVB, IFI16 delocalizes from the nucleus to the cytoplasm and is then eventually released into the extracellular milieu. In this study, using an in-house capture enzyme-linked immunsorbent assay we demonstrate that significant levels of IFI16 protein can also exist as circulating form in the sera of autoimmune patients. We also show that the rIFI16 protein, when added in-vitro to endothelial cells, does not affect cell viability, but severely limits their tubulogenesis and transwell migration activities. These inhibitory effects are fully reversed in the presence of anti-IFI16 N-terminal antibodies, indicating that its extracellular activity resides within the N-terminus. It was further demonstrated that endogenous IFI16 released by apoptotic cells bind neighboring cells in a co-culture. Immunofluorescence assays revealed existence of high-affinity binding sites on the plasma membrane of endothelial cells. Free recombinant IFI16 binds these sites on HUVEC with dissociation constant of 2.7 nM, radioiodinated and unlabeled IFI16 compete for binding sites, with inhibition constant (Ki) of 14.43 nM and half maximal inhibitory concentration (IC50) of 67.88 nM; these data allow us to estimate the presence of 250,000 to 450,000 specific binding sites per cell. Corroborating the results from functional assays, this binding could be completely inhibited using anti-IFI16 N-terminal antibody, but not with an antibody raised against the IFI16 C-terminal. Altogether, these data demonstrate that IFI16 may exist as circulating protein in the sera of autoimmune patients which binds endothelial cells causing damage, suggesting a new pathogenic and alarmin function through which this protein triggers the development of autoimmunity.
Collapse
|
26
|
The intracellular DNA sensor IFI16 gene acts as restriction factor for human cytomegalovirus replication. PLoS Pathog 2012; 8:e1002498. [PMID: 22291595 PMCID: PMC3266931 DOI: 10.1371/journal.ppat.1002498] [Citation(s) in RCA: 193] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 12/07/2011] [Indexed: 02/08/2023] Open
Abstract
Human interferon (IFN)-inducible IFI16 protein, an innate immune sensor of intracellular DNA, modulates various cell functions, however, its role in regulating virus growth remains unresolved. Here, we adopt two approaches to investigate whether IFI16 exerts pro- and/or anti-viral actions. First, the IFI16 gene was silenced using specific small interfering RNAs (siRNA) in human embryo lung fibroblasts (HELF) and replication of DNA and RNA viruses evaluated. IFI16-knockdown resulted in enhanced replication of Herpesviruses, in particular, Human Cytomegalovirus (HCMV). Consistent with this, HELF transduction with a dominant negative form of IFI16 lacking the PYRIN domain (PYD) enhanced the replication of HCMV. Second, HCMV replication was compared between HELFs overexpressing either the IFI16 gene or the LacZ gene. IFI16 overexpression decreased both virus yield and viral DNA copy number. Early and late, but not immediate-early, mRNAs and proteins were strongly down-regulated, thus IFI16 may exert its antiviral effect by impairing viral DNA synthesis. Constructs with the luciferase reporter gene driven by deleted or site-specific mutated forms of the HCMV DNA polymerase (UL54) promoter demonstrated that the inverted repeat element 1 (IR-1), located between −54 and −43 relative to the transcription start site, is the target of IFI16 suppression. Indeed, electrophoretic mobility shift assays and chromatin immunoprecipitation demonstrated that suppression of the UL54 promoter is mediated by IFI16-induced blocking of Sp1-like factors. Consistent with these results, deletion of the putative Sp1 responsive element from the HCMV UL44 promoter also relieved IFI16 suppression. Together, these data implicate IFI16 as a novel restriction factor against HCMV replication and provide new insight into the physiological functions of the IFN-inducible gene IFI16 as a viral restriction factor. Only recently, intrinsic cellular-based defense mechanisms which give cells the capacity to resist pathogens have been discovered as an essential component of immunity. However, unlike the innate and adaptive branches of the immune system, intrinsic immune defenses are mediated by cellular restriction factors that are constitutively expressed and active even before a pathogen enters the cell. The protein family HIN-200 may act as sensors of foreign DNA and modulate various functions such as growth, apoptosis, and senescence. Here we show that, in the absence of functional IFI16, the replication of some Herpesviruses and in particular of Human Cytomegalovirus (HCMV) is significantly enhanced. Accordingly, IFI16 overexpression strongly inhibited HCMV replication. Accumulation of viral DNA copies was down-regulated along with expression of early and late viral gene expression suggesting that IFI16 inhibits viral DNA synthesis. Using transient transfection, luciferase, gel shift assay, and chromatin immunoprecipitation, we demonstrate that IFI16 suppresses the transcriptional activity of the viral DNA polymerase gene (UL54) and the UL44 gene, also required for viral DNA synthesis. The finding that the nuclear DNA sensor IFI16 controls virus growth represents an important step forward in understanding the intrinsic mechanisms that drive viral infections sustained by DNA viruses such as Herpesviruses.
Collapse
|
27
|
Veeranki S, Choubey D. Interferon-inducible p200-family protein IFI16, an innate immune sensor for cytosolic and nuclear double-stranded DNA: regulation of subcellular localization. Mol Immunol 2011; 49:567-71. [PMID: 22137500 DOI: 10.1016/j.molimm.2011.11.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Revised: 11/06/2011] [Accepted: 11/09/2011] [Indexed: 01/09/2023]
Abstract
The interferon (IFN)-inducible p200-protein family includes structurally related murine (for example, p202a, p202b, p204, and Aim2) and human (for example, AIM2 and IFI16) proteins. All proteins in the family share a partially conserved repeat of 200-amino acid residues (also called HIN-200 domain) in the C-terminus. Additionally, most proteins (except the p202a and p202b proteins) also share a protein-protein interaction pyrin domain (PYD) in the N-terminus. The HIN-200 domain contains two consecutive oligosaccharide/oligonucleotide binding folds (OB-folds) to bind double stranded DNA (dsDNA). The PYD domain in proteins allows interactions with the family members and an adaptor protein ASC. Upon sensing cytosolic dsDNA, Aim2, p204, and AIM2 proteins recruit ASC protein to form an inflammasome, resulting in increased production of proinflammatory cytokines. However, IFI16 protein can sense cytosolic as well as nuclear dsDNA. Interestingly, the IFI16 protein contains a nuclear localization signal (NLS). Accordingly, the initial studies had indicated that the endogenous IFI16 protein is detected in the nucleus and within the nucleus in the nucleolus. However, several recent reports suggest that subcellular localization of IFI16 protein in nuclear versus cytoplasmic (or both) compartment depends on cell type. Given that the IFI16 protein can sense cytosolic as well as nuclear dsDNA and can initiate different innate immune responses (production of IFN-β versus proinflammatory cytokines), here we evaluate the experimental evidence for the regulation of subcellular localization of IFI16 protein in various cell types. We conclude that further studies are needed to understand the molecular mechanisms that regulate the subcellular localization of IFI16 protein.
Collapse
Affiliation(s)
- Sudhakar Veeranki
- Department of Environmental Health, University of Cincinnati, 3223 Eden Avenue, P. O. Box-670056, Cincinnati, OH 45267, USA
| | | |
Collapse
|
28
|
Reynier F, Petit F, Paye M, Turrel-Davin F, Imbert PE, Hot A, Mougin B, Miossec P. Importance of correlation between gene expression levels: application to the type I interferon signature in rheumatoid arthritis. PLoS One 2011; 6:e24828. [PMID: 22043277 PMCID: PMC3197194 DOI: 10.1371/journal.pone.0024828] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Accepted: 08/19/2011] [Indexed: 11/24/2022] Open
Abstract
Background The analysis of gene expression data shows that many genes display similarity in their expression profiles suggesting some co-regulation. Here, we investigated the co-expression patterns in gene expression data and proposed a correlation-based research method to stratify individuals. Methodology/Principal Findings Using blood from rheumatoid arthritis (RA) patients, we investigated the gene expression profiles from whole blood using Affymetrix microarray technology. Co-expressed genes were analyzed by a biclustering method, followed by gene ontology analysis of the relevant biclusters. Taking the type I interferon (IFN) pathway as an example, a classification algorithm was developed from the 102 RA patients and extended to 10 systemic lupus erythematosus (SLE) patients and 100 healthy volunteers to further characterize individuals. We developed a correlation-based algorithm referred to as Classification Algorithm Based on a Biological Signature (CABS), an alternative to other approaches focused specifically on the expression levels. This algorithm applied to the expression of 35 IFN-related genes showed that the IFN signature presented a heterogeneous expression between RA, SLE and healthy controls which could reflect the level of global IFN signature activation. Moreover, the monitoring of the IFN-related genes during the anti-TNF treatment identified changes in type I IFN gene activity induced in RA patients. Conclusions In conclusion, we have proposed an original method to analyze genes sharing an expression pattern and a biological function showing that the activation levels of a biological signature could be characterized by its overall state of correlation.
Collapse
Affiliation(s)
- Frédéric Reynier
- Joint Unit Hospices Civils de Lyon - bioMérieux, Hôpital Edouard Herriot, Lyon, France
| | - Fabien Petit
- Joint Unit Hospices Civils de Lyon - bioMérieux, Hôpital Edouard Herriot, Lyon, France
| | - Malick Paye
- Joint Unit Hospices Civils de Lyon - bioMérieux, Hôpital Edouard Herriot, Lyon, France
| | - Fanny Turrel-Davin
- Joint Unit Hospices Civils de Lyon - bioMérieux, Hôpital Edouard Herriot, Lyon, France
| | | | - Arnaud Hot
- Joint Unit Hospices Civils de Lyon - bioMérieux, Hôpital Edouard Herriot, Lyon, France
| | - Bruno Mougin
- Joint Unit Hospices Civils de Lyon - bioMérieux, Hôpital Edouard Herriot, Lyon, France
| | - Pierre Miossec
- Joint Unit Hospices Civils de Lyon - bioMérieux, Hôpital Edouard Herriot, Lyon, France
- Department of Clinical Immunology and Rheumatology, and immunogenomics and Inflammation Research Unit EA 4130, University of Lyon, Hôpital Edouard Herriot, Lyon, France
- * E-mail:
| |
Collapse
|
29
|
Differential macrophage polarization promotes tissue remodeling and repair in a model of ischemic retinopathy. Sci Rep 2011; 1:76. [PMID: 22355595 PMCID: PMC3216563 DOI: 10.1038/srep00076] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 08/08/2011] [Indexed: 11/24/2022] Open
Abstract
Diabetic retinopathy is the leading cause of visual loss in individuals under the age of 55. Umbilical cord blood (UCB)–derived myeloid progenitor cells have been shown to decrease neuronal damage associated with ischemia in the central nervous system. In this study we show that UCB-derived CD14+ progenitor cells provide rescue effects in a mouse model of ischemic retinopathy by promoting physiological angiogenesis and reducing associated inflammation. We use confocal microscopy to trace the fate of injected human UCB-derived CD14+ cells and PCR with species-specific probes to investigate their gene expression profile before and after injection. Metabolomic analysis measures changes induced by CD14+ cells. Our results demonstrate that human cells differentiate in vivo into M2 macrophages and induce the polarization of resident M2 macrophages. This leads to stabilization of the ischemia-injured retinal vasculature by modulating the inflammatory response, reducing oxidative stress and apoptosis and promoting tissue repair.
Collapse
|
30
|
Gugliesi F, Dell'Oste V, De Andrea M, Baggetta R, Mondini M, Zannetti C, Bussolati B, Camussi G, Gariglio M, Landolfo S. Tumor-Derived Endothelial Cells Evade Apoptotic Activity of the Interferon-Inducible IFI16 Gene. J Interferon Cytokine Res 2011; 31:609-18. [DOI: 10.1089/jir.2011.0001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Francesca Gugliesi
- Department of Public Health and Microbiology, Medical School, University of Turin, Turin, Italy
| | - Valentina Dell'Oste
- Department of Public Health and Microbiology, Medical School, University of Turin, Turin, Italy
| | - Marco De Andrea
- Department of Public Health and Microbiology, Medical School, University of Turin, Turin, Italy
- Department of Clinical and Experimental Medicine, Medical School, University of Eastern Piedmont “A. Avogadro,” Novara, Italy
| | - Rossella Baggetta
- Department of Public Health and Microbiology, Medical School, University of Turin, Turin, Italy
| | - Michele Mondini
- Department of Clinical and Experimental Medicine, Medical School, University of Eastern Piedmont “A. Avogadro,” Novara, Italy
| | - Claudia Zannetti
- Infection and Cancer Biology Group, International Agency for Research on Cancer, Lyon, France
| | - Benedetta Bussolati
- Department of Internal Medicine, Center for Experimental Research and Medical Studies, San Giovanni Battista Hospital, Turin, Italy
| | - Gianni Camussi
- Department of Internal Medicine, Center for Experimental Research and Medical Studies, San Giovanni Battista Hospital, Turin, Italy
| | - Marisa Gariglio
- Department of Clinical and Experimental Medicine, Medical School, University of Eastern Piedmont “A. Avogadro,” Novara, Italy
| | - Santo Landolfo
- Department of Public Health and Microbiology, Medical School, University of Turin, Turin, Italy
| |
Collapse
|
31
|
Costa S, Borgogna C, Mondini M, De Andrea M, Meroni PL, Berti E, Gariglio M, Landolfo S. Redistribution of the nuclear protein IFI16 into the cytoplasm of ultraviolet B-exposed keratinocytes as a mechanism of autoantigen processing. Br J Dermatol 2011; 164:282-90. [PMID: 20973769 DOI: 10.1111/j.1365-2133.2010.10097.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND The skin has long been recognized as a prominent target tissue in systemic lupus erythematosus (SLE) which plays a crucial role in the initiation and perpetuation of the autoimmune reaction cascade as a consequence of ultraviolet (UV)-induced keratinocyte apoptosis. Antibodies against IFI16 (interferon-inducible protein 16) have been detected in the sera of patients with SLE. OBJECTIVES To verify whether the induction of autoimmunity against IFI16 involves redistribution of this nuclear protein in keratinocytes during UVB-induced cell death. METHODS An in vitro epidermal model was developed to investigate the fate of the IFI16 protein in keratinocytes after irradiation with UVB; both keratinocyte monolayers and human skin explants were used. IFI16 expression and localization were also analysed in diseased skin sections of patients with SLE. RESULTS We demonstrated that IFI16, normally restricted to the nucleus, can be induced to appear in the cytoplasm under conditions of UVB-induced cell injury. This nucleus to cytoplasm translocation was also observed in skin explants exposed to UVB and in the diseased skin sections from patients with SLE. In addition, IFI16 was found in the supernatants of UVB-exposed keratinocytes. CONCLUSIONS The finding that IFI16 is present in the cytoplasm of diseased skin cells from patients with SLE and the demonstration of IFI16 in the supernatants of UVB-exposed keratinocytes, suggest that UVB irradiation or other stimuli may favour an abnormal IFI16 presentation to the afferent limb of the immune system and potentially an autoimmune response against the protein itself.
Collapse
Affiliation(s)
- S Costa
- Department of Clinical and Experimental Medicine, University of Piemonte Orientale A. Avogadro, Via Generale Solaroli 17, 28100 Novara, Italy
| | | | | | | | | | | | | | | |
Collapse
|
32
|
IFI16 induction by glucose restriction in human fibroblasts contributes to autophagy through activation of the ATM/AMPK/p53 pathway. PLoS One 2011; 6:e19532. [PMID: 21573174 PMCID: PMC3088686 DOI: 10.1371/journal.pone.0019532] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2010] [Accepted: 04/06/2011] [Indexed: 11/28/2022] Open
Abstract
Background Glucose restriction in cells increases the AMP/ATP ratio (energetic stress), which activates the AMPK/p53 pathway. Depending upon the energetic stress levels, cells undergo either autophagy or cell death. Given that the activated p53 induces the expression of IFI16 protein, we investigated the potential role of the IFI16 protein in glucose restriction-induced responses. Methodology/Principal Findings We found that glucose restriction or treatment of human diploid fibroblasts (HDFs) with the activators of the AMPK/p53 pathway induced the expression of IFI16 protein. The induced levels of IFI16 protein were associated with the induction of autophagy and reduced cell survival. Moreover, the increase in the IFI16 protein levels was dependent upon the expression of the functional ATM protein kinase. Importantly, the knockdown of the IFI16 expression in HDFs inhibited the activation of the ATM/AMPK/p53 pathway in response to glucose restriction and also increased the survival of HDFs. Conclusions/Significance Our observations demonstrate a role for the IFI16 protein in the energetic stress-induced regulation of autophagy and cell survival. Additionally, our findings also indicate that the loss of IFI16 expression, as found in certain cancers, may provide a survival advantage to cancer cells in microenvironments with low glucose levels.
Collapse
|
33
|
Gariglio M, Mondini M, De Andrea M, Landolfo S. The multifaceted interferon-inducible p200 family proteins: from cell biology to human pathology. J Interferon Cytokine Res 2011; 31:159-72. [PMID: 21198352 DOI: 10.1089/jir.2010.0106] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The interferon-inducible p200 family proteins consist of a group of homologous human and mouse proteins that have an N-terminal Pyrin domain and 1 or 2 partially conserved 200 amino acid long C-terminal domains (designated the HIN domain or p200 X domain). These proteins display multifaceted activity due to their ability to bind to various target proteins (eg, transcription factors, signaling proteins, and tumor suppressor proteins) and modulate different cell functions. In addition to a role in interferon biology, increasing evidence supports a role for these proteins as regulators of various cell functions, including proliferation, differentiation, apoptosis, senescence, inflammasome assembly, and control of organ transplants. As a consequence, alterations in their expression and function may be of relevance in the pathogenesis of human diseases, such as systemic autoimmune syndromes, tumors, and degenerative diseases. This review summarizes the literature describing these data, highlights some of the important findings derived from recent studies, and speculates about future perspectives.
Collapse
Affiliation(s)
- Marisa Gariglio
- Department of Clinical and Experimental Medicine, Medical School of Novara, Novara, Italy
| | | | | | | |
Collapse
|
34
|
Gugliesi F, De Andrea M, Mondini M, Cappello P, Giovarelli M, Shoenfeld Y, Meroni P, Gariglio M, Landolfo S. The proapoptotic activity of the Interferon-inducible gene IFI16 provides new insights into its etiopathogenetic role in autoimmunity. J Autoimmun 2010; 35:114-23. [DOI: 10.1016/j.jaut.2010.04.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Revised: 04/16/2010] [Accepted: 04/19/2010] [Indexed: 12/22/2022]
|
35
|
Mondini M, Costa S, Sponza S, Gugliesi F, Gariglio M, Landolfo S. The interferon-inducible HIN-200 gene family in apoptosis and inflammation: implication for autoimmunity. Autoimmunity 2010; 43:226-31. [PMID: 20187706 DOI: 10.3109/08916930903510922] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The Ifi-200/HIN-200 gene family encodes highly homologous human (IFI16, myeloid cell nuclear differentiation antigen, absent in melanoma 2, and IFIX) and murine proteins (Ifi202a, Ifi202b, Ifi203, Ifi204, Ifi205, and Ifi206), which are induced by type I and II interferons (IFN). These proteins have been described as regulators of cell proliferation and differentiation and, more recently, several reports have suggested their involvement in both apoptotic and inflammatory processes. The relevance of HIN-200 proteins in human disease is beginning to be clarified, and emerging experimental data indicate their role in autoimmunity. Autoimmune disorders are sustained by perpetual activation of inflammatory process and a link between autoimmunity and apoptosis has been clearly established. Moreover, the interferon system is now considered as a key player in autoimmune disorders such as systemic lupus erythemathosus, systemic sclerosis, and Sjögren's syndrome, and it is therefore conceivable to hypothesize that HIN-200 may be among the pivotal mediators of IFN activity in autoimmune disease. In particular, the participation of HIN-200 proteins in apoptosis and inflammation could support their potential role in autoimmunity.
Collapse
|
36
|
Human cytomegalovirus pUL83 stimulates activity of the viral immediate-early promoter through its interaction with the cellular IFI16 protein. J Virol 2010; 84:7803-14. [PMID: 20504932 DOI: 10.1128/jvi.00139-10] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The human cytomegalovirus (HCMV) virion protein pUL83 (also termed pp65) inhibits the expression of interferon-inducible cellular genes. In this work we demonstrate that pUL83 is also important for efficient induction of transcription from the viral major immediate-early promoter. Infection with a mutant virus containing a premature translation termination codon in the UL83 open reading frame (ORF) (UL83Stop) resulted in decreased transcription from the major immediate-early promoter in a time- and multiplicity-dependent manner. Expression of pUL83 alone is capable of transactivating the promoter in a reporter assay, and pUL83 associates with the promoter in infected cells. To investigate the mechanism by which the protein regulates the major immediate-early promoter, we utilized a mutant virus expressing an epitope-tagged pUL83 from its own promoter to identify protein binding partners for pUL83 during infection. We identified and confirmed the interaction of pUL83 with cellular IFI16 family members throughout the course of HCMV infection. pUL83 recruits IFI16 to the major immediate-early promoter, and IFI16 binding at the promoter is dependent upon the presence of pUL83. Consistent with the results obtained with the UL83Stop virus, infection of IFI16 knockdown cells with wild-type virus resulted in decreased levels of immediate-early transcripts compared to those of control cells. These data identify a previously unknown role for pUL83 in the initiation of the human cytomegalovirus gene expression cascade.
Collapse
|
37
|
The interferon-inducible gene IFI16 secretome of endothelial cells drives the early steps of the inflammatory response. Eur J Immunol 2010; 40:2182-9. [DOI: 10.1002/eji.200939995] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
38
|
Berry A, Matthews L, Jangani M, Plumb J, Farrow S, Buchan N, Wilson PA, Singh D, Ray DW, Donn RP. Interferon-inducible factor 16 is a novel modulator of glucocorticoid action. FASEB J 2010; 24:1700-13. [PMID: 20086048 PMCID: PMC3000051 DOI: 10.1096/fj.09-139998] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Previously, we used cDNA expression profiling to identify genes associated with glucocorticoid (Gc) sensitivity. We now identify which of these directly influence Gc action. Interferon-inducible protein 16 (IFI16), bone morphogenetic protein receptor type II (BMPRII), and regulator of G-protein signaling 14 (RGS14) increased Gc transactivation, whereas sialyltransferase 4B (SIAT4B) had a negative effect. Amyloid β (A4) precursor-protein binding, family B, member 1 (APBB1/Fe65) and neural cell expressed developmentally down-regulated 9 (NEDD9) were without effect. Only IFI16 potentiated Gc repression of NF-κB. In addition, IFI16 affected basal expression, and Gc induction of endogenous target genes. IFI16 did not affect glucocorticoid receptor (GR) expression, ligand-dependent repression of GR expression, or the ligand-dependent induction of GR phosphorylation on Ser-211 or Ser-203. Coimmunoprecipitation revealed an interaction, suggesting that IFI16 modulation of GR function is mediated by protein crosstalk. Transfection analysis with GR mutants showed that the ligand-binding domain of GR binds IFI16 and is the target domain for IFI16 regulation. Analysis of human lung sections identified colocalization of GR and IFI16, suggesting a physiologically relevant interaction. We demonstrate that IFI16 is a novel modulator of GR function and show the importance of analyzing variation in Gc sensitivity in humans, using appropriate technology, to drive discovery.—Berry, A., Matthews, L. Jangani, M., Plumb, J., Farrow, S., Buchan, N., Wilson, P. A., Singh, D., Ray, D., W., Donn, R. P. Interferon-inducible factor 16 is a novel modulator of glucocorticoid action.
Collapse
Affiliation(s)
- A Berry
- Arthritis Research Campaign Epidemiology Unit, University of Manchester, Manchester, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
In vivo growth inhibition of head and neck squamous cell carcinoma by the Interferon-inducible gene IFI16. Cancer Lett 2010; 287:33-43. [DOI: 10.1016/j.canlet.2009.05.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Revised: 05/14/2009] [Accepted: 05/29/2009] [Indexed: 12/30/2022]
|
40
|
Tang X, Yang X, Peng Y, Lin J. Protective Effects of Lycopene against H2O2-Induced Oxidative Injury and Apoptosis in Human Endothelial Cells. Cardiovasc Drugs Ther 2009; 23:439-48. [DOI: 10.1007/s10557-009-6206-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
41
|
Borgogna C, Toniutto P, Smirne C, Azzimonti B, Rittà M, Avellini C, Fabris C, Landolfo S, Gariglio M, Pirisi M. Expression of the interferon-inducible proteins MxA and IFI16 in liver allografts. Histopathology 2009; 54:837-46. [PMID: 19635103 DOI: 10.1111/j.1365-2559.2009.03311.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
AIMS To test the hypothesis that the activation of the interferon (IFN) system pathways might link hepatitis C virus (HCV) recurrence in the liver allograft with acute cellular rejection. METHODS AND RESULTS In this retrospective study, allograft biopsy specimens from 28 adult patients (14 HCV+ and 14 HCV-) who had undergone their first liver transplantation were analysed. Eleven biopsy specimens showed acute cellular rejection (Banff rejection activity index score > or =3). Specimens were immunostained for two IFN-inducible proteins, MxA and IFI16, and for CD45. The predominant MxA reactivity pattern was hepatocytic, whereas IFI16 was expressed in both the hepatocellular and inflammatory compartments. Moderate to strong MxA expression in hepatocytes was associated positively with rejection score (P < 0.01), donor's age < or =45 years (P < 0.05) and aspartate aminotransferase levels >40 U/l on the day of biopsy (P < 0.05), and inversely with infiltration of portal triads by IFI16+/CD45+ cells (P < 0.005) and time to progression beyond Ishak stage 2 of recurrent hepatitis C (P < 0.01). On multivariate analysis, MxA expression in hepatocytes was independently associated with allograft rejection and donor's age. CONCLUSIONS Acute allograft rejection and recurrence of HCV infection in the liver allograft appear to intersect in the IFN system pathways.
Collapse
Affiliation(s)
- Cinzia Borgogna
- DPMSC, Medical Liver Transplantation Unit, University of Udine, Udine, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Role of the interferon-inducible IFI16 gene in the induction of ICAM-1 by TNF-alpha. Cell Immunol 2009; 257:55-60. [PMID: 19338980 DOI: 10.1016/j.cellimm.2009.02.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Revised: 02/25/2009] [Accepted: 02/25/2009] [Indexed: 11/23/2022]
Abstract
The Interferon-inducible gene IFI16, a member of the HIN200 family, is activated by oxidative stress and cell density, in addition to Interferons, and it is implicated in the regulation of endothelial cell proliferation and vessel formation in vitro. We have previously shown that IFI16 is required for proinflammatory gene stimulation by IFN-gamma through the NF-kappaB complex. To examine whether IFI16 induction might be extended to other proinflammatory cytokines such as tumor necrosis factor (TNF)-alpha, we used the strategy of the RNA interference to knock down IFI16 expression, and analyze the capability of TNF-alpha to stimulate intercellular adhesion molecule-1 (ICAM-1 or CD54) expression in the absence of functional IFI16. Our studies demonstrate that IFI16 mediates ICAM-1 stimulation by TNF-alpha through the NF-kappaB pathway, thus reinforcing the role of the IFI16 molecule in the inflammation process.
Collapse
|
43
|
De Andrea M, Gioia D, Mondini M, Azzimonti B, Renò F, Pecorari G, Landolfo V, Tommasino M, Accardi R, Herold-Mende C, Landolfo S, Gariglio M. Effects of IFI16 overexpression on the growth and doxorubicin sensitivity of head and neck squamous cell carcinoma–derived cell lines. Head Neck 2007; 29:835-44. [PMID: 17510972 DOI: 10.1002/hed.20611] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND In a previous analysis of head and neck squamous cell carcinomas (HNSCCs), we showed that the levels of the interferon-inducible protein IFI16 inversely correlate with cancer grade. In this study, we further evaluate the molecular role of IFI16 in the development of HNSCCs. METHODS The effect of IFI16 expression was evaluated by its retroviral restoration in an IFI16-negative HNSCC-derived cell line, HNO136. Growth rate and soft agar colony formation were evaluated. The effect of IFI16 restoration in cells exposed to doxorubicin was also analyzed. RESULTS IFI16 restoration resulted in the inhibition of both cell growth and in vitro transforming activity and increased doxorubicin-induced cell death by accumulating the cells at the G2/M phase. CONCLUSION In agreement with our previous in vivo data, IFI16 appears to be involved in maintaining the normal growth of epithelial cells, whereas its downregulation may contribute to uncontrolled cell proliferation and tumorigenesis.
Collapse
Affiliation(s)
- Marco De Andrea
- Department of Public Health and Microbiology, Medical School of Torino, Via Santena 9, 10126 Torino, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mondini M, Vidali M, Airò P, De Andrea M, Riboldi P, Meroni PL, Gariglio M, Landolfo S. Role of the Interferon-Inducible Gene IFI16 in the Etiopathogenesis of Systemic Autoimmune Disorders. Ann N Y Acad Sci 2007; 1110:47-56. [PMID: 17911419 DOI: 10.1196/annals.1423.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Interferons (IFNs) are now known to exert a multitude of immunological functions on both the innate and adaptive immunity. Given their pleiotropic effects on the immune system, it is conceivable that excess type I IFN or aberrant regulation of its signaling could contribute to autoimmunity. Several lines of evidence link IFNs to autoimmune disorders, in particular to systemic lupus erythematosus (SLE) and systemic sclerosis (SSc). Expression of a spectrum of genes that constitutes an "IFN signature" is the most significant observation indicating that IFNs may be dominant among the pathogenic mediators involved in some autoimmune diseases. A family of IFN-inducible genes, designated HIN-200 in the human and IFI-200 in the murine species, encodes evolutionary related human (IFI16, MNDA, AIM2, IFIX) and murine proteins (Ifi202 a, Ifi202b, Ifi203, Ifi204, Ifi205/D3). Physiological IFI16 expression was found in cells of the immune system, in endothelial cells, and in stratified squamous epithelia, such as skin. The presence of anti-IFI16 antibodies was reported in SLE and primary/secondary Sjögren's syndrome. More recently, we reported that anti-IFI16 autoantibodies differentiate limited cutaneous systemic sclerosis (lcSSc) from diffuse systemic sclerosis (dcSSc). Molecular studies performed in primary endothelial cells overexpressing IFI16 demonstrated that it may be involved in the early steps of inflammation by modulating endothelial cell function, such as expression of adhesion molecules and chemokine production, cell growth, and apoptosis. Moreover, here we report that IFI16 expression is induced by proinflammatory cytokines. In this article the role of the IFI16 protein and its corresponding autoantibodies in the etiopathogenesis of systemic autoimmune diseases, in which chronic inflammation is involved, are discussed.
Collapse
Affiliation(s)
- Michele Mondini
- Department of Public Health and Microbiology, Medical School, University of Turin, V. Santena 9, 10126, Turin, Italy
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Caposio P, Gugliesi F, Zannetti C, Sponza S, Mondini M, Medico E, Hiscott J, Young HA, Gribaudo G, Gariglio M, Landolfo S. A novel role of the interferon-inducible protein IFI16 as inducer of proinflammatory molecules in endothelial cells. J Biol Chem 2007; 282:33515-33529. [PMID: 17699163 DOI: 10.1074/jbc.m701846200] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The human IFI16 gene is an interferon-inducible gene implicated in the regulation of endothelial cell proliferation and tube morphogenesis. Immunohistochemical analysis has demonstrated that this gene is highly expressed in endothelial cells in addition to hematopoietic tissues. In this study, gene array analysis of human umbilical vein endothelial cells overexpressing IFI16 revealed an increased expression of genes involved in immunomodulation, cell growth, and apoptosis. Consistent with these observations, IFI16 triggered expression of adhesion molecules such as ICAM-1 and E-selectin or chemokines such as interleukin-8 or MCP-1. Treatment of cells with short hairpin RNA targeting IFI16 significantly inhibited ICAM-1 induction by interferon (IFN)-gamma demonstrating that IFI16 is required for proinflammatory gene stimulation. Moreover, functional analysis of the ICAM-1 promoter by deletion- or site-specific mutation demonstrated that NF-kappaB is the main mediator of IFI16-driven gene induction. NF-kappaB activation appears to be triggered by IFI16 through a novel mechanism involving suppression of IkappaBalpha mRNA and protein expression. Support for this finding comes from the observation that IFI16 targeting with specific short hairpin RNA down-regulates NF-kappaB binding activity to its cognate DNA and inhibits ICAM-1 expression induced by IFN-gamma. Using transient transfection and luciferase assay, electrophoretic mobility shift assay, and chromatin immunoprecipitation, we demonstrate indeed that activation of the NF-kappaB response is mediated by IFI16-induced block of Sp1-like factor recruitment to the promoter of the IkappaBalpha gene, encoding the main NF-kappaB inhibitor. Activation of NF-kappaB accompanied by induction of proinflammatory molecules was also observed when IkappaBalpha expression was down-regulated by specific small interfering RNA, resulting in an outcome similar to that observed with IFI16 overexpression. Taken together, these data implicate IFI16 as a novel regulator of endothelial proinflammatory activity and provide new insights into the physiological functions of the IFN-inducible gene IFI16.
Collapse
Affiliation(s)
- Patrizia Caposio
- Department of Public Health and Microbiology, University of Turin, Turin, 10126, Italy
| | - Francesca Gugliesi
- Department of Public Health and Microbiology, University of Turin, Turin, 10126, Italy
| | - Claudia Zannetti
- Department of Public Health and Microbiology, University of Turin, Turin, 10126, Italy
| | - Simone Sponza
- Department of Public Health and Microbiology, University of Turin, Turin, 10126, Italy
| | - Michele Mondini
- Department of Public Health and Microbiology, University of Turin, Turin, 10126, Italy; Department of Clinical and Experimental Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Enzo Medico
- Institute for Cancer Research and Treatment, University of Turin, Turin 10126, Italy
| | - John Hiscott
- Lady Davis Institute, McGill University, Montreal H3T 1E2, Canada
| | - Howard A Young
- Laboratory of Experimental Immunology, Center for Cancer Research, NCI-Frederick, National Institutes of Health, Frederick, Maryland 21702
| | - Giorgio Gribaudo
- Department of Public Health and Microbiology, University of Turin, Turin, 10126, Italy
| | - Marisa Gariglio
- Department of Clinical and Experimental Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Santo Landolfo
- Department of Public Health and Microbiology, University of Turin, Turin, 10126, Italy.
| |
Collapse
|
46
|
Drygin D, Koo S, Perera R, Barone S, Bennett CF. Induction of toll-like receptors and NALP/PAN/PYPAF family members by modified oligonucleotides in lung epithelial carcinoma cells. Oligonucleotides 2006; 15:105-18. [PMID: 15989425 DOI: 10.1089/oli.2005.15.105] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
ISIS 199044 is a chimeric 2'-O-methyl-containing oligonucleotide that produces toxicity in several cultured cell lines. Upon investigation into the mechanism of cytotoxicity, we discovered that treatment of lung epithelial carcinoma cells, A549, with ISIS 199044 and several other cytotoxic oligonucleotides induces a group of genes that are not normally expressed in these cells. These genes are involved in host response to foreign materials. Among them were toll-like receptor 7 (TLR7) and TLR9, members of the toll-like receptor family, responsible for immune response to nucleic acids and cryopyrin, a member of NALP/PAN/PYPAF family, which is known to assemble with ASC and regulate NF-kappaB activation and to modulate apoptosis. Maximal induction occurred 12-24 hours posttreatment with 500 nM oligonucleotide in the presence of Lipofectin reagent. Furthermore, we have shown that this induction is chemistry dependent; it can be negated by certain modifications, such as replacement of 2'-O-methyl with 2'-O-methoxyethyl groups or substitution of phosphorothioates with phosphodiester linkages. DNA microarray analysis identified additional genes modulated by ISIS 199044, particularly genes involved in DNA damage/repair.
Collapse
|
47
|
Xie J, Shaikh ZA. Cadmium induces cell cycle arrest in rat kidney epithelial cells in G2/M phase. Toxicology 2006; 224:56-65. [PMID: 16730872 DOI: 10.1016/j.tox.2006.04.026] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2005] [Revised: 04/01/2006] [Accepted: 04/07/2006] [Indexed: 12/31/2022]
Abstract
Cadmium (Cd) has been reported to cause cell cycle arrest in various cell types by p53-dependent and -independent mechanisms. This study was designed to investigate cell cycle progression in kidney cells that are the target of chronic Cd toxicity. Rat renal proximal tubular epithelial cells, NRK-52E, were treated with up to 20 microM CdCl2 in DMEM containing 10% calf serum for up to 24 h. Flow cytometric analysis revealed time- and concentration-dependent increases in cells in G2/M phase of the cell cycle. As compared to the control cells, the cells exposed to 20 microM Cd showed a doubling of the number of cells in this phase after 24 h. The cell cycle arrest was associated with a decrease in protein levels of both cyclins A and B. Further investigation into the mechanism revealed that Cd treatment led to down-modulation of cyclin-dependent kinases, Cdk1 and Cdk2, apparently by elevating the expression of cyclin kinase inhibitors, KIP1/p27 and WAF1/p21. Furthermore, the wild-type p53 DNA-binding activity was up-regulated. Based on these observations, it appears that Cd causes G2/M phase arrest in NRK-52E cells via elevation of p53 activity, increasing the expression of cyclin kinase inhibitors p27 and p21, and decreasing the expression of cyclin-dependent kinases Cdk1 and 2, and of cyclins A and B.
Collapse
Affiliation(s)
- Jianxun Xie
- Department of Biomedical and Pharmaceutical Sciences and Center for Molecular Toxicology, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | | |
Collapse
|
48
|
Mondini M, Vidali M, De Andrea M, Azzimonti B, Airò P, D'Ambrosio R, Riboldi P, Meroni PL, Albano E, Shoenfeld Y, Gariglio M, Landolfo S. A novel autoantigen to differentiate limited cutaneous systemic sclerosis from diffuse cutaneous systemic sclerosis: The interferon-inducible gene IFI16. ACTA ACUST UNITED AC 2006; 54:3939-44. [PMID: 17133607 DOI: 10.1002/art.22266] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To investigate the presence and clinical significance of autoantibodies against the interferon-inducible gene IFI16 in systemic sclerosis (SSc), systemic lupus erythematosus (SLE), and other autoimmune diseases. METHODS Immunohistochemical analysis was used to evaluate the expression of IFI16 in skin biopsy specimens obtained from patients with SSc and patients with SLE. Levels of antibodies against IFI16 in sera from 82 patients with SSc and 100 patients with SLE were determined by enzyme-linked immunosorbent assay. Other autoimmune diseases such as primary Sjögren's syndrome (SS), rheumatoid arthritis (RA), chronic urticaria, and hepatitis C virus (HCV) infection were also examined. RESULTS Expression of IFI16 was greatly increased and was ubiquitous in all layers of the epidermis and in the dermal inflammatory infiltrates of lesional skin from both patients with SLE and patients with SSc. Patients with SLE, those with primary SS, and those with SSc exhibited significantly higher anti-IFI16 IgG antibody levels compared with normal controls (for SLE, P < 0.002; for primary SS, P < 0.001; for SSc, P < 0.0005). Anti-IFI16 titers above the ninety-fifth percentile for control subjects were observed in 26% of the patients with SLE, 50% of those with primary SS, and 21% of those with SSc (28% of patients with limited cutaneous SSc [lcSSc] versus 4% of patients with diffuse cutaneous SSc [dcSSc]). In contrast, the prevalence of anti-IFI16 was 4% in patients with RA, 5% in those with chronic urticaria, and 13% in those with HCV infection. CONCLUSION The results of this study provide evidence that an IFN-inducible gene, IFI16, may be involved in the pathophysiologic mechanisms of connective tissue disorders such as SSc. Moreover, a strict correlation with lcSSc was also demonstrated, thus providing a novel tool in the differential diagnosis of lcSSc from dcSSc.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Arthritis, Rheumatoid/blood
- Arthritis, Rheumatoid/immunology
- Autoantibodies/blood
- Autoantigens
- Diagnosis, Differential
- Enzyme-Linked Immunosorbent Assay
- Female
- Hepatitis C/blood
- Hepatitis C/immunology
- Humans
- Immunoglobulin G/blood
- Male
- Middle Aged
- Nuclear Proteins/immunology
- Nuclear Proteins/metabolism
- Phosphoproteins/immunology
- Phosphoproteins/metabolism
- Scleroderma, Diffuse/blood
- Scleroderma, Diffuse/diagnosis
- Scleroderma, Diffuse/immunology
- Scleroderma, Limited/blood
- Scleroderma, Limited/diagnosis
- Scleroderma, Limited/immunology
- Sjogren's Syndrome/blood
- Sjogren's Syndrome/immunology
- Skin/metabolism
- Skin/pathology
Collapse
Affiliation(s)
- Michele Mondini
- Medical School of Turin, Turin, and Medical School of Piemonte Orientale, Novara, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|