1
|
Basavaraja R, Zhang H, Holczbauer Á, Lu Z, Radaelli E, Assenmacher CA, George SS, Nallamala VC, Beiting DP, Meyer-Ficca ML, Meyer RG, Guo W, Fan Y, Modzelewski AJ, Spiegelman VS, Cohen MS, Fuchs SY. PARP11 inhibition inactivates tumor-infiltrating regulatory T cells and improves the efficacy of immunotherapies. Cell Rep Med 2024; 5:101649. [PMID: 39019005 PMCID: PMC11293321 DOI: 10.1016/j.xcrm.2024.101649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/18/2024] [Accepted: 06/17/2024] [Indexed: 07/19/2024]
Abstract
Tumor-infiltrating regulatory T cells (TI-Tregs) elicit immunosuppressive effects in the tumor microenvironment (TME) leading to accelerated tumor growth and resistance to immunotherapies against solid tumors. Here, we demonstrate that poly-(ADP-ribose)-polymerase-11 (PARP11) is an essential regulator of immunosuppressive activities of TI-Tregs. Expression of PARP11 correlates with TI-Treg cell numbers and poor responses to immune checkpoint blockade (ICB) in human patients with cancer. Tumor-derived factors including adenosine and prostaglandin E2 induce PARP11 in TI-Tregs. Knockout of PARP11 in the cells of the TME or treatment of tumor-bearing mice with selective PARP11 inhibitor ITK7 inactivates TI-Tregs and reinvigorates anti-tumor immune responses. Accordingly, ITK7 decelerates tumor growth and significantly increases the efficacy of anti-tumor immunotherapies including ICB and adoptive transfer of chimeric antigen receptor (CAR) T cells. These results characterize PARP11 as a key driver of TI-Treg activities and a major regulator of immunosuppressive TME and argue for targeting PARP11 to augment anti-cancer immunotherapies.
Collapse
Affiliation(s)
- Raghavendra Basavaraja
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongru Zhang
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ágnes Holczbauer
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhen Lu
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Enrico Radaelli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Charles-Antoine Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Subin S George
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vamshidhar C Nallamala
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel P Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mirella L Meyer-Ficca
- Department of Veterinary Clinical and Life Sciences, College of Veterinary Medicine, Utah State University, Logan, UT 84332, USA
| | - Ralph G Meyer
- Department of Veterinary Clinical and Life Sciences, College of Veterinary Medicine, Utah State University, Logan, UT 84332, USA
| | - Wei Guo
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Fan
- Departments of Radiation Oncology and of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew J Modzelewski
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vladimir S Spiegelman
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Michael S Cohen
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
2
|
NAGATA S. Cloning of human Type I interferon cDNAs. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2024; 100:1-14. [PMID: 37648466 PMCID: PMC10864172 DOI: 10.2183/pjab.100.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/29/2023] [Indexed: 09/01/2023]
Abstract
In the late 1970s, crude interferon samples were found to exhibit anti-tumour activity. This discovery led to the interferon as a "magic drug" for cancer patients. Many groups, including those in Tokyo, Zürich, and San Francisco, attempted to identify human interferon cDNAs. Tadatsugu Taniguchi was the first to announce the cloning of human interferon-β cDNA in the December 1979 issue of Proc. Jpn. Acad. Ser. B. This was followed by the cloning of human interferon-α by a Zürich group and interferon-γ by a group in Genentech in San Francisco. Recombinant interferon proteins were produced on a large scale, and interferon-α was widely used to treat C-type hepatitis patients. The biological functions of interferons were quickly elucidated with the purified recombinant interferons. The molecular mechanisms underlying virus-induced interferon gene expression were also examined using cloned chromosomal genes. The background that led to interferon gene cloning and its impact on cytokine gene hunting is described herein.
Collapse
Affiliation(s)
- Shigekazu NAGATA
- Biochemistry & Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
3
|
Hou J, Zheng Y, Gao C. Regulation of cellular senescence by innate immunity. BIOPHYSICS REPORTS 2023; 9:338-351. [PMID: 38524701 PMCID: PMC10960571 DOI: 10.52601/bpr.2023.230032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/12/2024] [Indexed: 03/26/2024] Open
Abstract
During the COVID-19 pandemic, the interplay between the processes of immunity and senescence is drawing more and more intensive attention. SARS-CoV-2 infection induces senescence in lung cells, failure to clear infected cells and increased presence of inflammatory factors could lead to a cytokine storm and acute respiratory disease syndrome (ARDS), which together with aging and age-associated disease lead to 70% of COVID-19-related deaths. Studies on how senescence initiates upon viral infection and how to restrict excessive accumulation of senescent cells to avoid harmful inflammation are crucially important. Senescence can induce innate immune signaling, and innate immunity can engage cell senescence. Here, we mainly review the innate immune pathways, such as cGAS-STING, TLRs, NF-κB, and NLRP3 inflammasome, participating in the senescence process. In these pathways, IFN-I and inflammatory factors play key roles. At the end of the review, we propose the strategies by which we can improve the immune function and reduce inflammation based on these findings.
Collapse
Affiliation(s)
- Jinxiu Hou
- Key Laboratory of Infection and Immunity, Shandong Province & Key Laboratory for Experimental Teratology, Ministry of Education, Shandong University, Jinan 250012, China
- Department of Immunology, the School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Yi Zheng
- Key Laboratory of Infection and Immunity, Shandong Province & Key Laboratory for Experimental Teratology, Ministry of Education, Shandong University, Jinan 250012, China
- Department of Immunology, the School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity, Shandong Province & Key Laboratory for Experimental Teratology, Ministry of Education, Shandong University, Jinan 250012, China
- Department of Immunology, the School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| |
Collapse
|
4
|
Patasova K, Lundberg IE, Holmqvist M. Genetic Influences in Cancer-Associated Myositis. Arthritis Rheumatol 2023; 75:153-163. [PMID: 36053262 PMCID: PMC10107284 DOI: 10.1002/art.42345] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/28/2022] [Accepted: 08/31/2022] [Indexed: 02/02/2023]
Abstract
Idiopathic inflammatory myopathies (IIMs) comprise a heterogeneous group of rare immune-mediated disorders that primarily affect muscles but also lead to dysfunction in other organs. Five different clinical subphenotypes of IIM have been distinguished: dermatomyositis, polymyositis, inclusion body myositis, antisynthetase syndrome, and immune-mediated necrotizing myopathy. Excess mortality and morbidity associated with IIM are largely attributed to comorbidities, particularly cancer. The risk of malignancy is not equally distributed among IIM groups and is particularly high among patients with dermatomyositis. The cancer risk peaks around 3 years on either side of the IIM diagnosis and remains elevated even 10 years after the onset of the disease. Lung, colorectal, and ovarian neoplasms typically arise before the onset of IIM, whereas melanoma, cervical, oropharyngeal, and nonmelanoma skin cancers usually develop after IIM diagnosis. Given the close temporal proximity between IIM diagnosis and the emergence of malignancy, it has been proposed that IIM could be a consequence rather than a cause of cancer, a process known as a paramalignant phenomenon. Thus, a separate group of IIMs related to paramalignant phenomenon has been distinguished, known as cancer-associated myositis (CAM). Although the relationship between IIM and cancer is widely recognized, the pathophysiology of CAM remains elusive. Given that genetic factors play a role in the development of IIM, dissection of the molecular mechanisms shared between IIM and cancer presents an opportunity to examine the role of autoimmunity in cancer development and progression. In this review, the evidence supporting the contribution of genetics to CAM will be discussed.
Collapse
Affiliation(s)
- Karina Patasova
- Clinical Epidemiology Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Ingrid E Lundberg
- Rheumatology Division, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Marie Holmqvist
- Clinical Epidemiology Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
5
|
Zhang H, Tomar VS, Li J, Basavaraja R, Yan F, Gui J, McBrearty N, Costich TL, Beiting DP, Blanco MA, Conejo-Garcia JR, Saggu G, Berger A, Nefedova Y, Gabrilovich DI, Fuchs SY. Protection of Regulatory T Cells from Fragility and Inactivation in the Tumor Microenvironment. Cancer Immunol Res 2022; 10:1490-1505. [PMID: 36255418 PMCID: PMC9722544 DOI: 10.1158/2326-6066.cir-22-0295] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/01/2022] [Accepted: 10/12/2022] [Indexed: 01/10/2023]
Abstract
Fragility of regulatory T (Treg) cells manifested by the loss of neuropilin-1 (NRP1) and expression of IFNγ undermines the immune suppressive functions of Treg cells and contributes to the success of immune therapies against cancers. Intratumoral Treg cells somehow avoid fragility; however, the mechanisms by which Treg cells are protected from fragility in the tumor microenvironment are not well understood. Here, we demonstrate that the IFNAR1 chain of the type I IFN (IFN1) receptor was downregulated on intratumoral Treg cells. Downregulation of IFNAR1 mediated by p38α kinase protected Treg cells from fragility and maintained NRP1 levels, which were decreased in response to IFN1. Genetic or pharmacologic inactivation of p38α and stabilization of IFNAR1 in Treg cells induced fragility and inhibited their immune suppressive and protumorigenic activities. The inhibitor of sumoylation TAK981 (Subasumstat) upregulated IFNAR1, eliciting Treg fragility and inhibiting tumor growth in an IFNAR1-dependent manner. These findings describe a mechanism by which intratumoral Treg cells retain immunosuppressive activities and suggest therapeutic approaches for inducing Treg fragility and increasing the efficacy of immunotherapies.
Collapse
Affiliation(s)
- Hongru Zhang
- Department of Biomedical Sciences, School of Veterinary
Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vivek S. Tomar
- Department of Biomedical Sciences, School of Veterinary
Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jinyang Li
- Department of Pathology and Laboratory Medicine, Perelman
School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Raghavendra Basavaraja
- Department of Biomedical Sciences, School of Veterinary
Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fangxue Yan
- Department of Biomedical Sciences, School of Veterinary
Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jun Gui
- Department of Biomedical Sciences, School of Veterinary
Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Noreen McBrearty
- Department of Biomedical Sciences, School of Veterinary
Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tara Lee Costich
- Department of Immunology, H. Lee Moffitt Cancer Center and
Research Institute, Tampa, FL, USA
| | - Daniel P. Beiting
- Department of Pathobiology, School of Veterinary Medicine,
University of Pennsylvania, Philadelphia, PA 19104, USA
| | - M. Andres Blanco
- Department of Biomedical Sciences, School of Veterinary
Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jose R. Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center and
Research Institute, Tampa, FL, USA
| | - Gurpanna Saggu
- Takeda Development Center Americas, Inc., Lexington, MA,
02421, USA
| | - Allison Berger
- Takeda Development Center Americas, Inc., Lexington, MA,
02421, USA
| | | | | | - Serge Y. Fuchs
- Department of Biomedical Sciences, School of Veterinary
Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Correspondence to: Serge Y.
Fuchs, Dept. of Biomedical Sciences, School of Veterinary Medicine, University
of Pennsylvania, 380 S. University Ave, Hill 316, Philadelphia, PA 19104; USA.
Tel: 1-215-573-6949;
| |
Collapse
|
6
|
Perego M, Fu S, Cao Y, Kossenkov A, Yao M, Bonner E, Alicea-Torres K, Liu W, Jiang Z, Chen Z, Fuchs SY, Zhou J, Gabrilovich DI. Mechanisms regulating transitory suppressive activity of neutrophils in newborns: PMNs-MDSCs in newborns. J Leukoc Biol 2022; 112:955-968. [PMID: 35726818 PMCID: PMC9794389 DOI: 10.1002/jlb.4hi0921-514rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 12/30/2022] Open
Abstract
Transitory appearance of immune suppressive polymorphonuclear neutrophils (PMNs) defined as myeloid-derived suppressor cells (PMNs-MDSCs) in newborns is important for their protection from inflammation associated with newly established gut microbiota. Here, we report that inhibition of the type I IFN (IFN1) pathway played a major role in regulation of PMNs-MDSCs-suppressive activity during first weeks of life. Expression of the IFN1 receptor IFNAR1 was markedly lower in PMNs-MDSCs. However, in newborn mice, down-regulation of IFNAR1 was not sufficient to render PMNs immune suppressive. That also required the presence of a positive signal from lactoferrin via its receptor low-density lipoprotein receptor-related protein 2. The latter effect was mediated via NF-κB activation, which was tempered by IFN1 in a manner that involved suppressor of cytokine signaling 3. Thus, we discovered a mechanism of tight regulation of immune suppressive PMNs-MDSCs in newborns, which may be used in the development of therapies of neonatal pathologies.
Collapse
Affiliation(s)
| | - Shuyu Fu
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Yingjiao Cao
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | | | - Meng Yao
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Erin Bonner
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Kevin Alicea-Torres
- The Wistar Institute, Philadelphia, Pennsylvania, USA
- Biology Department, University of Puerto Rico-Humacao, Humacao, Puerto Rico, USA
| | - Wangkai Liu
- Department of Pediatrics, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhilong Jiang
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital; Shanghai Institute of Respiratory Disease, Fudan University, Shanghai, China
| | - Zhihong Chen
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital; Shanghai Institute of Respiratory Disease, Fudan University, Shanghai, China
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jie Zhou
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | | |
Collapse
|
7
|
Luo TY, Shi Y, Wang G, Spaner DE. Enhanced IFN Sensing by Aggressive Chronic Lymphocytic Leukemia Cells. THE JOURNAL OF IMMUNOLOGY 2022; 209:1662-1673. [DOI: 10.4049/jimmunol.2200199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/18/2022] [Indexed: 01/04/2023]
Abstract
Abstract
Type I IFN is made by cells in response to stress. Cancer cells exist in a state of stress, but their IFN response is complex and not completely understood. This study investigated the role of autocrine IFN in human chronic lymphocytic leukemia (CLL) cells. CLL cells were found to make low amounts of IFN via TANK-binding kinase 1 pathways, but p-STAT1 and -STAT2 proteins along with IFN-stimulated genes that reflect IFN activation were variably downregulated in cultured CLL cells by the neutralizing IFNAR1 Ab anifrolumab. Patients with CLL were segregated into two groups based on the response of their leukemia cells to anifrolumab. Samples associated with more aggressive clinical behavior indicated by unmutated IGHV genes along with high CD38 and p-Bruton’s tyrosine kinase expression exhibited responses to low amounts of IFN that were blocked by anifrolumab. Samples with more indolent behavior were unaffected by anifrolumab. Hypersensitivity to IFN was associated with higher expression of IFNAR1, MX1, STAT1, and STAT2 proteins and lower activity of negative regulatory tyrosine phosphatases. Autocrine IFN protected responsive CLL cells from stressful tissue culture environments and therapeutic drugs such as ibrutinib and venetoclax in vitro, in part by upregulating Mcl-1 expression. These findings suggest hypersensitivity to IFN may promote aggressive clinical behavior. Specific blockade of IFN signaling may improve outcomes for patients with CLL with higher-risk disease.
Collapse
Affiliation(s)
- Tina YuXuan Luo
- *Biology Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- †Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Yonghong Shi
- *Biology Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Guizhi Wang
- *Biology Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - David E. Spaner
- *Biology Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- †Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- ‡Biology Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- §Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; and
- ¶Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Xiong J, Jiang Y, Zhang J, Chen Y, Hu Y. CK1α upregulates the IFNAR1 expression to prompt the anti-HBV effect of type I IFN in hepatoma carcinoma cells. Virol Sin 2022; 37:894-903. [PMID: 35985475 PMCID: PMC9797371 DOI: 10.1016/j.virs.2022.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 08/08/2022] [Indexed: 01/01/2023] Open
Abstract
Casein kinase 1α (CK1α) mediates the phosphorylation and degradation of interferon-α/β receptor 1 (IFNAR1) in response to viral infection. However, how CK1α regulates hepatitis B virus (HBV) replication and the anti-HBV effects of IFN-α are less reported. Here we show that CK1α can interact with IFNAR1 in hepatoma carcinoma cells and increased the abundance of IFNAR1 by reducing the ubiquitination levels in the presence of HBV. Furthermore, CK1α promotes the IFN-α triggered JAK-STAT signaling pathway and consequently enhances the antiviral effects of IFN-α against HBV replication. Our results collectively provide evidence that CK1α positively regulates the anti-HBV activity of IFN-α in hepatoma carcinoma cells, which would be a promising therapeutic target to improve the effectiveness of IFN-α therapy to cure CHB.
Collapse
|
9
|
Targeting PARP11 to avert immunosuppression and improve CAR T therapy in solid tumors. NATURE CANCER 2022; 3:808-820. [PMID: 35637402 PMCID: PMC9339499 DOI: 10.1038/s43018-022-00383-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 04/22/2022] [Indexed: 02/05/2023]
Abstract
Evasion of antitumor immunity and resistance to therapies in solid tumors are aided by an immunosuppressive tumor microenvironment (TME). We found that TME factors, such as regulatory T cells and adenosine, downregulated type I interferon receptor IFNAR1 on CD8+ cytotoxic T lymphocytes (CTLs). These events relied upon poly-ADP ribose polymerase-11 (PARP11), which was induced in intratumoral CTLs and acted as a key regulator of the immunosuppressive TME. Ablation of PARP11 prevented loss of IFNAR1, increased CTL tumoricidal activity and inhibited tumor growth in an IFNAR1-dependent manner. Accordingly, genetic or pharmacologic inactivation of PARP11 augmented the therapeutic benefits of chimeric antigen receptor T cells. Chimeric antigen receptor CTLs engineered to inactivate PARP11 demonstrated a superior efficacy against solid tumors. These findings highlight the role of PARP11 in the immunosuppressive TME and provide a proof of principle for targeting this pathway to optimize immune therapies.
Collapse
|
10
|
Matsushita M. Novel Treatment Strategies Utilizing Immune Reactions against Chronic Myelogenous Leukemia Stem Cells. Cancers (Basel) 2021; 13:cancers13215435. [PMID: 34771599 PMCID: PMC8582551 DOI: 10.3390/cancers13215435] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 01/21/2023] Open
Abstract
Simple Summary Although tyrosine kinase inhibitors (TKIs) are highly effective in the treatment of patients with chronic myelogenous leukemia (CML), leukemic stem cells (LSCs) are known to be resistant to TKIs. As a result, the application of immunotherapies against LSCs may cure CML. Abstract Introduction of tyrosine kinase inhibitors (TKIs) has improved the prognosis of patients with chronic myelogenous leukemia (CML), and treatment-free remission (TFR) is now a treatment goal. However, about half of the patients experience molecular relapse after cessation of TKIs, suggesting that leukemic stem cells (LSCs) are resistant to TKIs. Eradication of the remaining LSCs using immunotherapies including interferon-alpha, vaccinations, CAR-T cells, and other drugs would be a key strategy to achieve TFR.
Collapse
Affiliation(s)
- Maiko Matsushita
- Division of Clinical Physiology and Therapeutics, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
| |
Collapse
|
11
|
Lightcap ES, Yu P, Grossman S, Song K, Khattar M, Xega K, He X, Gavin JM, Imaichi H, Garnsey JJ, Koenig E, Zhang H, Lu Z, Shah P, Fu Y, Milhollen MA, Hatton BA, Riceberg J, Shinde V, Li C, Minissale J, Yang X, England D, Klinghoffer RA, Langston S, Galvin K, Shapiro G, Pulukuri SM, Fuchs SY, Huszar D. A small-molecule SUMOylation inhibitor activates antitumor immune responses and potentiates immune therapies in preclinical models. Sci Transl Med 2021; 13:eaba7791. [PMID: 34524860 DOI: 10.1126/scitranslmed.aba7791] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Eric S Lightcap
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Pengfei Yu
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen Grossman
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Keli Song
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Mithun Khattar
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Kristina Xega
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Xingyue He
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - James M Gavin
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Hisashi Imaichi
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - James J Garnsey
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Erik Koenig
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Hongru Zhang
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhen Lu
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pooja Shah
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Yu Fu
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Michael A Milhollen
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | | | - Jessica Riceberg
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Vaishali Shinde
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Cong Li
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - James Minissale
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Xiaofeng Yang
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Dylan England
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | | | - Steve Langston
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Katherine Galvin
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Gary Shapiro
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Sai M Pulukuri
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dennis Huszar
- Millennium Pharmaceuticals Inc., Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA
| |
Collapse
|
12
|
Alicea-Torres K, Sanseviero E, Gui J, Chen J, Veglia F, Yu Q, Donthireddy L, Kossenkov A, Lin C, Fu S, Mulligan C, Nam B, Masters G, Denstman F, Bennett J, Hockstein N, Rynda-Apple A, Nefedova Y, Fuchs SY, Gabrilovich DI. Immune suppressive activity of myeloid-derived suppressor cells in cancer requires inactivation of the type I interferon pathway. Nat Commun 2021; 12:1717. [PMID: 33741967 PMCID: PMC7979850 DOI: 10.1038/s41467-021-22033-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSC) are pathologically activated neutrophils and monocytes with potent immune suppressive activity. These cells play an important role in accelerating tumor progression and undermining the efficacy of anti-cancer therapies. The natural mechanisms limiting MDSC activity are not well understood. Here, we present evidence that type I interferons (IFN1) receptor signaling serves as a universal mechanism that restricts acquisition of suppressive activity by these cells. Downregulation of the IFNAR1 chain of this receptor is found in MDSC from cancer patients and mouse tumor models. The decrease in IFNAR1 depends on the activation of the p38 protein kinase and is required for activation of the immune suppressive phenotype. Whereas deletion of IFNAR1 is not sufficient to convert neutrophils and monocytes to MDSC, genetic stabilization of IFNAR1 in tumor bearing mice undermines suppressive activity of MDSC and has potent antitumor effect. Stabilizing IFNAR1 using inhibitor of p38 combined with the interferon induction therapy elicits a robust anti-tumor effect. Thus, negative regulatory mechanisms of MDSC function can be exploited therapeutically.
Collapse
Affiliation(s)
| | - Emilio Sanseviero
- The Wistar Institute, Philadelphia, PA, USA
- AstraZeneca, Gaithersburg, MD, USA
| | - Jun Gui
- Department of Biomedical Sciences, School of Veterinary Medicine University of Pennsylvania, Philadelphia, PA, USA
- State Key Laboratory of Oncogenes and Related Genes, Stem Cell Research Center, Renji Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, China
| | - Jinyun Chen
- Department of Biomedical Sciences, School of Veterinary Medicine University of Pennsylvania, Philadelphia, PA, USA
| | - Filippo Veglia
- The Wistar Institute, Philadelphia, PA, USA
- H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Qiujin Yu
- Department of Biomedical Sciences, School of Veterinary Medicine University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Cindy Lin
- The Wistar Institute, Philadelphia, PA, USA
| | - Shuyu Fu
- The Wistar Institute, Philadelphia, PA, USA
| | - Charles Mulligan
- Helen F. Graham Cancer Center and Research Institute, Newark, DE, USA
| | - Brian Nam
- Helen F. Graham Cancer Center and Research Institute, Newark, DE, USA
| | - Gregory Masters
- Helen F. Graham Cancer Center and Research Institute, Newark, DE, USA
| | - Fred Denstman
- Helen F. Graham Cancer Center and Research Institute, Newark, DE, USA
| | - Joseph Bennett
- Helen F. Graham Cancer Center and Research Institute, Newark, DE, USA
| | - Neil Hockstein
- Helen F. Graham Cancer Center and Research Institute, Newark, DE, USA
| | - Agnieszka Rynda-Apple
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | | | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
13
|
Zanker DJ, Owen KL, Baschuk N, Spurling AJ, Parker BS. Loss of type I IFN responsiveness impairs natural killer cell antitumor activity in breast cancer. Cancer Immunol Immunother 2021; 70:2125-2138. [PMID: 33449132 DOI: 10.1007/s00262-021-02857-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 01/06/2021] [Indexed: 12/21/2022]
Abstract
Competent type I IFN signaling is the lynchpin of most immune surveillance mechanisms and has recently proven critical to the efficacy of several anticancer agents. Expression of the type I IFN receptor, IFNAR, underpins type I IFN responsiveness in all cells and facilitates the activation and cytotoxic potential of lymphocytes, while loss of IFNAR on lymphocytes has previously been associated with tumor progression and poor patient survival. This study underscores the importance of intact type I IFN signaling to NK cells in the regulation of tumorigenesis and metastasis, whereby ablation of NK cell IFNAR1 impairs antitumor activity and tumor clearance. Using a preclinical model of triple negative breast cancer, we identified that intact IFNAR on NK cells is required for an effective response to type I IFN-inducing immunotherapeutics that may be mediated by pathways associated with NK cell degranulation. Taken together, these data provide a rationale for considering the IFNAR status on NK cells when devising therapeutic strategies aimed at inducing systemic type I IFN signaling in breast cancer.
Collapse
Affiliation(s)
- Damien J Zanker
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Katie L Owen
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Nikola Baschuk
- Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Alex J Spurling
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Belinda S Parker
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia.
- Department of Biochemistry and Genetics, La Trobe Institute from Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
14
|
Rabl J. BRCA1-A and BRISC: Multifunctional Molecular Machines for Ubiquitin Signaling. Biomolecules 2020; 10:biom10111503. [PMID: 33142801 PMCID: PMC7692841 DOI: 10.3390/biom10111503] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022] Open
Abstract
The K63-linkage specific deubiquitinase BRCC36 forms the core of two multi-subunit deubiquitination complexes: BRCA1-A and BRISC. BRCA1-A is recruited to DNA repair foci, edits ubiquitin signals on chromatin, and sequesters BRCA1 away from the site of damage, suppressing homologous recombination by limiting resection. BRISC forms a complex with metabolic enzyme SHMT2 and regulates the immune response, mitosis, and hematopoiesis. Almost two decades of research have revealed how BRCA1-A and BRISC use the same core of subunits to perform very distinct biological tasks.
Collapse
Affiliation(s)
- Julius Rabl
- Cryo-EM Knowledge Hub, ETH Zürich, Otto-Stern-Weg 3, HPM C51, 8093 Zürich, Switzerland
| |
Collapse
|
15
|
Frisch SM, MacFawn IP. Type I interferons and related pathways in cell senescence. Aging Cell 2020; 19:e13234. [PMID: 32918364 PMCID: PMC7576263 DOI: 10.1111/acel.13234] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/18/2020] [Accepted: 08/07/2020] [Indexed: 01/10/2023] Open
Abstract
This review article addresses the largely unanticipated convergence of two landmark discoveries. The first is the discovery of interferons, critical signaling molecules for all aspects of both innate and adaptive immunity, discovered originally by Isaacs and Lindenmann at the National Institute for Medical Research, London, in 1957 (Proceedings of the Royal Society of London. Series B: Biological Sciences, 1957, 147, 258). The second, formerly unrelated discovery, by Leonard Hayflick and Paul Moorhead (Wistar Institute, Philadelphia) is that cultured cells undergo an irreversible but viable growth arrest, termed senescence, after a finite and predictable number of cell divisions (Experimental Cell Research, 1961, 25, 585). This phenomenon was suspected to relate to organismal aging, which was confirmed subsequently (Nature, 2011, 479, 232). Cell senescence has broad‐ranging implications for normal homeostasis, including immunity, and for diverse disease states, including cancer progression and response to therapy (Nature Medicine, 2015, 21, 1424; Cell, 2019, 179, 813; Cell, 2017, 169, 1000; Trends in Cell Biology, 2018, 28, 436; Journal of Cell Biology, 2018, 217, 65). Here, we critically address the bidirectional interplay between interferons (focusing on type I) and cell senescence, with important implications for health and healthspan.
Collapse
Affiliation(s)
- Steven M. Frisch
- Department of Biochemistry and WVU Cancer Institute West Virginia University Morgantown West Virginia USA
| | - Ian P. MacFawn
- Department of Biochemistry and WVU Cancer Institute West Virginia University Morgantown West Virginia USA
| |
Collapse
|
16
|
Cho C, Mukherjee R, Peck AR, Sun Y, McBrearty N, Katlinski KV, Gui J, Govindaraju PK, Puré E, Rui H, Fuchs SY. Cancer-associated fibroblasts downregulate type I interferon receptor to stimulate intratumoral stromagenesis. Oncogene 2020; 39:6129-6137. [PMID: 32807917 PMCID: PMC7502515 DOI: 10.1038/s41388-020-01424-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/30/2020] [Accepted: 08/07/2020] [Indexed: 12/20/2022]
Abstract
Activation of cancer-associated fibroblasts (CAFs) and ensuing desmoplasia play an important role in the growth and progression of solid tumors. Here we demonstrate that, within colon and pancreatic ductal adenocarcinoma tumors, efficient stromagenesis relies on downregulation of the IFNAR1 chain of the type I interferon (IFN1) receptor. Expression of the fibroblast activation protein (FAP) and accumulation of the extracellular matrix (ECM) was notably impaired in tumors grown in the Ifnar1S526A (SA) knock-in mice, which are deficient in IFNAR1 downregulation. Primary fibroblasts from these mice exhibited elevated levels of Smad7, a negative regulator of the transforming growth factor-β (TGFβ) pathway. Knockdown of Smad7 alleviated deficient ECM production in SA fibroblasts in response to TGFβ. Analysis of human colorectal cancers revealed an inverse correlation between IFNAR1 and FAP levels. Whereas growth of tumors in SA mice was stimulated by co-injection of wild type but not SA fibroblasts, genetic ablation of IFNAR1 in fibroblasts also accelerated tumor growth. We discuss how inactivation of IFNAR1 in CAFs acts to stimulate stromagenesis and tumor growth.
Collapse
Affiliation(s)
- Christina Cho
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Riddhita Mukherjee
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Amy R Peck
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Yunguang Sun
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Noreen McBrearty
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kanstantsin V Katlinski
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jun Gui
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Priya K Govindaraju
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ellen Puré
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
17
|
Odnokoz O, Yu P, Peck AR, Sun Y, Kovatich AJ, Hooke JA, Hu H, Mitchell EP, Rui H, Fuchs SY. Malignant cell-specific pro-tumorigenic role of type I interferon receptor in breast cancers. Cancer Biol Ther 2020; 21:629-636. [PMID: 32378445 DOI: 10.1080/15384047.2020.1750297] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Within the microenvironment of solid tumors, stress associated with deficit of nutrients and oxygen as well as tumor-derived factors triggers the phosphorylation-dependent degradation of the IFNAR1 chain of type I interferon (IFN1) receptor and ensuing suppression of the IFN1 pathway. Here we sought to examine the importance of these events in malignant mammary cells. Expression of non-degradable IFNAR1S526A mutant in mouse mammary adenocarcinoma cells stimulated the IFN1 pathway yet did not affect growth of these cells in vitro or ability to form subcutaneous tumors in the syngeneic mice. Remarkably, these cells exhibited a notably accelerated growth when transplanted orthotopically into mammary glands. Importantly, in human patients with either ER+ or ER- breast cancers, high levels of IFNAR1 were associated with poor prognosis. We discuss the putative mechanisms underlying the pro-tumorigenic role of IFNAR1 in malignant breast cells.
Collapse
Affiliation(s)
- Olena Odnokoz
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania , Philadelphia, PA, USA
| | - Pengfei Yu
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania , Philadelphia, PA, USA
| | - Amy R Peck
- Department of Pathology, Medical College of Wisconsin , Milwaukee, WI, USA
| | - Yunguang Sun
- Department of Pathology, Medical College of Wisconsin , Milwaukee, WI, USA
| | - Albert J Kovatich
- John P. Murtha Cancer Center Research Program, Uniformed Services University and Walter Reed National Military Medical Center , Bethesda, MD, USA
| | - Jeffrey A Hooke
- John P. Murtha Cancer Center Research Program, Uniformed Services University and Walter Reed National Military Medical Center , Bethesda, MD, USA
| | - Hai Hu
- Chan Soon-Shiong Institute of Molecular Medicine , Windber, PA, USA
| | - Edith P Mitchell
- Department of Medical Oncology, Thomas Jefferson University , Philadelphia, PA, USA
| | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin , Milwaukee, WI, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania , Philadelphia, PA, USA
| |
Collapse
|
18
|
PARP1 Enhances Influenza A Virus Propagation by Facilitating Degradation of Host Type I Interferon Receptor. J Virol 2020; 94:JVI.01572-19. [PMID: 31915279 DOI: 10.1128/jvi.01572-19] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/18/2019] [Indexed: 02/06/2023] Open
Abstract
Influenza A virus (IAV) utilizes multiple strategies to confront or evade host type I interferon (IFN)-mediated antiviral responses in order to enhance its own propagation within the host. One such strategy is to induce the degradation of type I IFN receptor 1 (IFNAR1) by utilizing viral hemagglutinin (HA). However, the molecular mechanism behind this process is poorly understood. Here, we report that a cellular protein, poly(ADP-ribose) polymerase 1 (PARP1), plays a critical role in mediating IAV HA-induced degradation of IFNAR1. We identified PARP1 as an interacting partner for IAV HA through mass spectrometry analysis. This interaction was confirmed by coimmunoprecipitation analyses. Furthermore, confocal fluorescence microscopy showed altered localization of endogenous PARP1 upon transient IAV HA expression or during IAV infection. Knockdown or inhibition of PARP1 rescued IFNAR1 levels upon IAV infection or HA expression, exemplifying the importance of PARP1 for IAV-induced reduction of IFNAR1. Notably, PARP1 was crucial for the robust replication of IAV, which was associated with regulation of the type I IFN receptor signaling pathway. These results indicate that PARP1 promotes IAV replication by controlling viral HA-induced degradation of host type I IFN receptor. Altogether, these findings provide novel insight into interactions between influenza virus and the host innate immune response and reveal a new function for PARP1 during influenza virus infection.IMPORTANCE Influenza A virus (IAV) infections cause seasonal and pandemic influenza outbreaks, which pose a devastating global health concern. Despite the availability of antivirals against influenza, new IAV strains continue to persist by overcoming the therapeutics. Therefore, much emphasis in the field is placed on identifying new therapeutic targets that can more effectively control influenza. IAV utilizes several tactics to evade host innate immunity, which include the evasion of antiviral type I interferon (IFN) responses. Degradation of type I IFN receptor (IFNAR) is one known method of subversion, but the molecular mechanism for IFNAR downregulation during IAV infection remains unclear. Here, we have found that a host protein, poly(ADP-ribose) polymerase 1 (PARP1), facilitates IFNAR degradation and accelerates IAV replication. The findings reveal a novel cellular target for the potential development of antivirals against influenza, as well as expand our base of knowledge regarding interactions between influenza and the host innate immunity.
Collapse
|
19
|
Yuan Y, Miao Y, Qian L, Zhang Y, Liu C, Liu J, Zuo Y, Feng Q, Guo T, Zhang L, Chen X, Jin L, Huang F, Zhang H, Zhang W, Li W, Xu G, Zheng H. Targeting UBE4A Revives Viperin Protein in Epithelium to Enhance Host Antiviral Defense. Mol Cell 2020; 77:734-747.e7. [DOI: 10.1016/j.molcel.2019.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/26/2019] [Accepted: 10/30/2019] [Indexed: 01/26/2023]
|
20
|
Ickler J, Francois S, Widera M, Santiago ML, Dittmer U, Sutter K. HIV infection does not alter interferon α/β receptor 2 expression on mucosal immune cells. PLoS One 2020; 15:e0218905. [PMID: 31935222 PMCID: PMC6959566 DOI: 10.1371/journal.pone.0218905] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 12/23/2019] [Indexed: 01/12/2023] Open
Abstract
The innate immune response induced by type I interferons (IFNs) plays a critical role in the establishment of HIV infection. IFNs are induced early in HIV infection and trigger an antiviral defense program by signaling through the IFNα/β receptor (IFNAR), which consists of two subunits, IFNAR1 and IFNAR2. Changes in IFNAR expression in HIV target cells, as well as other immune cells, could therefore have important consequences for initial HIV spread. It was previously reported that IFNAR2 expression is increased in peripheral blood CD4+ CXCR4+ T cells of HIV+ patients compared to HIV uninfected controls, suggesting that HIV infection may alter the IFN responsiveness of target cells. However, the earliest immune cells affected by HIV in vivo reside in the gut-associated lymphoid tissue (GALT). To date, it remains unknown if IFNAR expression is altered in GALT immune cells in the context of HIV infection and exposure to IFNs, including the 12 IFNα subtypes. Here, we analyzed the expression of surface bound and soluble IFNAR2 on Lamina propria mononuclear cells (LPMCs) isolated from the GALT of HIV- individuals and in plasma samples of HIV+ patients. IFNAR2 expression varied between different T cells, B cells and natural killer cells, but was not altered following HIV infection. Furthermore, expression of the soluble IFNAR2a isoform was not changed in HIV+ patients compared to healthy donors, nor in LPMCs after HIV-1 infection ex vivo. Even though the 12 human IFNα subtypes trigger different biological responses and vary in their affinity to both receptor subunits, stimulation of LPMCs with different recombinant IFNα subtypes did not result in any significant changes in IFNAR2 surface expression. Our data suggests that potential changes in the IFN responsiveness of mucosal immune cells during HIV infection are unlikely dictated by changes in IFNAR2 expression.
Collapse
Affiliation(s)
- Julia Ickler
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sandra Francois
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Marek Widera
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Mario L. Santiago
- Department of Medicine, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Kathrin Sutter
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
21
|
Cellular Caspase-3 Contributes to EV-A71 2A pro-Mediated Down-Regulation of IFNAR1 at the Translation Level. Virol Sin 2019; 35:64-72. [PMID: 31512106 DOI: 10.1007/s12250-019-00151-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 05/20/2019] [Indexed: 01/12/2023] Open
Abstract
Enterovirus A71 (EV-A71) is the major pathogen responsible for the severe hand, foot and mouth disease worldwide, for which few effective antiviral drugs are presently available. Interferon-α (IFN-α) has been used in antiviral therapy for decades; it has been reported that EV-A71 antagonizes the antiviral activity of IFN-α based on viral 2Apro-mediated reduction of the interferon-alpha receptor 1 (IFNAR1); however, the mechanism remains unknown. Here, we showed a significant increase in IFNAR1 protein induced by IFN-α in RD cells, whereas EV-A71 infection caused obvious down-regulation of the IFNAR1 protein and blockage of IFN-α signaling. Subsequently, we observed that EV-A71 2Apro inhibited IFNAR1 translation by cleavage of the eukaryotic initiation factor 4GI (eIF4GI), without affecting IFNAR1 mRNA levels induced by IFN-α. The inhibition of IFNAR1 translation also occurred in puromycin-induced apoptotic cells when caspase-3 cleaved eIF4GI. Importantly, we verified that 2Apro could activate cellular caspase-3, which was subsequently involved in eIF4GI cleavage mediated by 2Apro. Furthermore, inhibition of caspase-3 activation resulted in the partial restoration of IFNAR1 in cells transfected with 2A or infected with EV-A71, suggesting the pivotal role of both viral 2Apro and caspase-3 activation in the disturbance of IFN-α signaling. Collectively, we elucidate a novel mechanism by which cellular caspase-3 contributes to viral 2Apro-mediated down-regulation of IFNAR1 at the translation level during EV-A71 infection, indicating that caspase-3 inhibition could be a potential complementary strategy to improve clinical anti-EV-A71 therapy with IFN-α.
Collapse
|
22
|
Fang C, Li J, Qi S, Lei Y, Zeng Y, Yu P, Hu Z, Zhou Y, Wang Y, Dai R, Li J, Huang S, Xu P, Chen K, Ding C, Yu FX. An alternatively transcribed TAZ variant negatively regulates JAK-STAT signaling. EMBO Rep 2019; 20:embr.201847227. [PMID: 30979708 DOI: 10.15252/embr.201847227] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 03/25/2019] [Accepted: 03/25/2019] [Indexed: 12/11/2022] Open
Abstract
Type I interferon (IFN)-induced Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling drives the expression of IFN-stimulated genes (ISGs) to mediate antiviral response. The strength and duration of JAK-STAT signaling are tightly regulated to ensure effective antiviral defense while avoiding pathological inflammation and autoimmunity. Here, we report that cTAZ, an isoform of the Hippo pathway effector TAZ, is transcribed by an alternative promoter. Although majority of C-terminal sequences of TAZ is retained, cTAZ is not regulated by the Hippo signaling and does not mediate its growth-inhibitory functions. Instead, cTAZ negatively regulates JAK-STAT signaling by inhibiting STAT1/2 nuclear localization and ISG expression, and its expression is induced by type I IFN Thus, cTAZ functions as a modulator of JAK-STAT signaling and may play a role in fine-tuning cellular antiviral response.
Collapse
Affiliation(s)
- Chuantao Fang
- Children's Hospital and Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Fudan University, Shanghai, China
| | - Jian Li
- Children's Hospital and Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Fudan University, Shanghai, China
| | - Sixian Qi
- Children's Hospital and Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Fudan University, Shanghai, China
| | - Yubin Lei
- Children's Hospital and Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Fudan University, Shanghai, China
| | - Yan Zeng
- Children's Hospital and Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Fudan University, Shanghai, China
| | - Pengcheng Yu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhaolan Hu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yufeng Zhou
- Children's Hospital and Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Fudan University, Shanghai, China
| | - Yulong Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ruping Dai
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jin Li
- Children's Hospital and Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Fudan University, Shanghai, China
| | - Shenglin Huang
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Pinglong Xu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, China
| | - Kang Chen
- Perinatology Research Branch, Eunice Kennedy Shriver NICHD, National Institutes of Health, Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA.,Jiangsu Province Hospital of TCM, The Affiliated Hospital of Nanjing University of TCM, Nanjing, China
| | - Chen Ding
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institutes of Biomedical Sciences, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fa-Xing Yu
- Children's Hospital and Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Fudan University, Shanghai, China
| |
Collapse
|
23
|
Gui J, Katlinski KV, Koumenis C, Diehl JA, Fuchs SY. The PKR-Like Endoplasmic Reticulum Kinase Promotes the Dissemination of Myc-Induced Leukemic Cells. Mol Cancer Res 2019; 17:1450-1458. [PMID: 30902831 DOI: 10.1158/1541-7786.mcr-19-0002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/14/2019] [Accepted: 03/19/2019] [Indexed: 02/07/2023]
Abstract
Hyperactive oncogenic Myc stimulates protein synthesis that induces the unfolded protein response, which requires the function of the eukaryotic translation initiation factor 2-alpha kinase 3, also known as protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK). Activated PERK acts to limit mRNA translation, enable proper protein folding, and restore the homeostasis in the endoplasmic reticulum. Given that Myc activation contributes to many types of lymphoid and myeloid human leukemias, we used a mouse model to examine the importance of PERK in development and progression of Myc-induced leukemias. We found that genetic ablation of Perk does not suppress the generation of the leukemic cells in the bone marrow. However, the cell-autonomous Perk deficiency restricts the dissemination of leukemic cells into peripheral blood, lymph nodes, and vital peripheral organs. Whereas the loss of the IFNAR1 chain of type I IFN receptor stimulated leukemia, Perk ablation did not stabilize IFNAR1, suggesting that PERK stimulates the leukemic cells' dissemination in an IFNAR1-independent manner. We discuss the rationale for using PERK inhibitors against Myc-driven leukemias. IMPLICATIONS: The role of PERK in dissemination of Myc-induced leukemic cells demonstrated in this study argues for the use of PERK inhibitors against leukemia progression.
Collapse
Affiliation(s)
- Jun Gui
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kanstantsin V Katlinski
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - J Alan Diehl
- Department of Biochemistry, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
24
|
Stanifer ML, Pervolaraki K, Boulant S. Differential Regulation of Type I and Type III Interferon Signaling. Int J Mol Sci 2019; 20:E1445. [PMID: 30901970 PMCID: PMC6471306 DOI: 10.3390/ijms20061445] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022] Open
Abstract
Interferons (IFNs) are very powerful cytokines, which play a key role in combatting pathogen infections by controlling inflammation and immune response by directly inducing anti-pathogen molecular countermeasures. There are three classes of IFNs: type I, type II and type III. While type II IFN is specific for immune cells, type I and III IFNs are expressed by both immune and tissue specific cells. Unlike type I IFNs, type III IFNs have a unique tropism where their signaling and functions are mostly restricted to epithelial cells. As such, this class of IFN has recently emerged as a key player in mucosal immunity. Since the discovery of type III IFNs, the last 15 years of research in the IFN field has focused on understanding whether the induction, the signaling and the function of these powerful cytokines are regulated differently compared to type I IFN-mediated immune response. This review will cover the current state of the knowledge of the similarities and differences in the signaling pathways emanating from type I and type III IFN stimulation.
Collapse
Affiliation(s)
- Megan L Stanifer
- Schaller research group at CellNetworks, Department of Infectious Diseases, Heidelberg University Hospital, 69120 Heidelberg, Germany.
- Research Group "Cellular polarity and viral infection" (F140), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Kalliopi Pervolaraki
- Schaller research group at CellNetworks, Department of Infectious Diseases, Heidelberg University Hospital, 69120 Heidelberg, Germany.
- Research Group "Cellular polarity and viral infection" (F140), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Steeve Boulant
- Schaller research group at CellNetworks, Department of Infectious Diseases, Heidelberg University Hospital, 69120 Heidelberg, Germany.
- Research Group "Cellular polarity and viral infection" (F140), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
25
|
Ortiz A, Gui J, Zahedi F, Yu P, Cho C, Bhattacharya S, Carbone CJ, Yu Q, Katlinski KV, Katlinskaya YV, Handa S, Haas V, Volk SW, Brice AK, Wals K, Matheson NJ, Antrobus R, Ludwig S, Whiteside TL, Sander C, Tarhini AA, Kirkwood JM, Lehner PJ, Guo W, Rui H, Minn AJ, Koumenis C, Diehl JA, Fuchs SY. An Interferon-Driven Oxysterol-Based Defense against Tumor-Derived Extracellular Vesicles. Cancer Cell 2019; 35:33-45.e6. [PMID: 30645975 PMCID: PMC6336114 DOI: 10.1016/j.ccell.2018.12.001] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 09/04/2018] [Accepted: 11/29/2018] [Indexed: 12/26/2022]
Abstract
Tumor-derived extracellular vesicles (TEV) "educate" healthy cells to promote metastases. We found that melanoma TEV downregulated type I interferon (IFN) receptor and expression of IFN-inducible cholesterol 25-hydroxylase (CH25H). CH25H produces 25-hydroxycholesterol, which inhibited TEV uptake. Low CH25H levels in leukocytes from melanoma patients correlated with poor prognosis. Mice incapable of downregulating the IFN receptor and Ch25h were resistant to TEV uptake, TEV-induced pre-metastatic niche, and melanoma lung metastases; however, ablation of Ch25h reversed these phenotypes. An anti-hypertensive drug, reserpine, suppressed TEV uptake and disrupted TEV-induced formation of the pre-metastatic niche and melanoma lung metastases. These results suggest the importance of CH25H in defense against education of normal cells by TEV and argue for the use of reserpine in adjuvant melanoma therapy.
Collapse
Affiliation(s)
- Angelica Ortiz
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jun Gui
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Farima Zahedi
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pengfei Yu
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christina Cho
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sabyasachi Bhattacharya
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher J Carbone
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Qiujing Yu
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kanstantsin V Katlinski
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuliya V Katlinskaya
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Simran Handa
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Victor Haas
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Susan W Volk
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Angela K Brice
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kim Wals
- Department of Medicine, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Nicholas J Matheson
- Department of Medicine, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Robin Antrobus
- Department of Medicine, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Sonja Ludwig
- Departments of Pathology, Immunology, and Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Otorhinolaryngology, University of Duisburg-Essen, Duisburg, Germany
| | - Theresa L Whiteside
- Departments of Pathology, Immunology, and Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Cindy Sander
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Ahmad A Tarhini
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - John M Kirkwood
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Paul J Lehner
- Department of Medicine, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Wei Guo
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Andy J Minn
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - J Alan Diehl
- Department of Biochemistry, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
26
|
Winkler R, Gillis E, Lasman L, Safra M, Geula S, Soyris C, Nachshon A, Tai-Schmiedel J, Friedman N, Le-Trilling VTK, Trilling M, Mandelboim M, Hanna JH, Schwartz S, Stern-Ginossar N. m 6A modification controls the innate immune response to infection by targeting type I interferons. Nat Immunol 2018; 20:173-182. [PMID: 30559377 DOI: 10.1038/s41590-018-0275-z] [Citation(s) in RCA: 306] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 11/07/2018] [Indexed: 01/07/2023]
Abstract
N6-methyladenosine (m6A) is the most common mRNA modification. Recent studies have revealed that depletion of m6A machinery leads to alterations in the propagation of diverse viruses. These effects were proposed to be mediated through dysregulated methylation of viral RNA. Here we show that following viral infection or stimulation of cells with an inactivated virus, deletion of the m6A 'writer' METTL3 or 'reader' YTHDF2 led to an increase in the induction of interferon-stimulated genes. Consequently, propagation of different viruses was suppressed in an interferon-signaling-dependent manner. Significantly, the mRNA of IFNB, the gene encoding the main cytokine that drives the type I interferon response, was m6A modified and was stabilized following repression of METTL3 or YTHDF2. Furthermore, we show that m6A-mediated regulation of interferon genes was conserved in mice. Together, our findings uncover the role m6A serves as a negative regulator of interferon response by dictating the fast turnover of interferon mRNAs and consequently facilitating viral propagation.
Collapse
Affiliation(s)
- Roni Winkler
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ella Gillis
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Lior Lasman
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Modi Safra
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Shay Geula
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Clara Soyris
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Aharon Nachshon
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Julie Tai-Schmiedel
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Nehemya Friedman
- Central Virology Laboratory, Ministry of Health, Chaim Sheba Medical Center, Ramat-Gan, Israel.,Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Mirko Trilling
- Institut für Virologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Michal Mandelboim
- Central Virology Laboratory, Ministry of Health, Chaim Sheba Medical Center, Ramat-Gan, Israel.,Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Jacob H Hanna
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Schraga Schwartz
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Noam Stern-Ginossar
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
27
|
Xia C, Anderson P, Hahm B. Viral dedication to vigorous destruction of interferon receptors. Virology 2018; 522:19-26. [PMID: 30014854 PMCID: PMC6087481 DOI: 10.1016/j.virol.2018.06.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/27/2018] [Accepted: 06/28/2018] [Indexed: 01/12/2023]
Abstract
Interferons (IFNs) exhibit forceful inhibitory activities against numerous viruses by inducing synthesis of anti-viral proteins or promoting immune cell functions, which help eradicate the vicious microbes. Consequently, the degree to which viruses evade or counterattack IFN responses influences viral pathogenicity. Viruses have developed many strategies to interfere with the synthesis of IFNs or IFN receptor signaling pathway. Furthermore, multiple viruses decrease levels of IFN receptors via diverse tactics, which include decreasing type I IFN receptor mRNA expression, blocking post-translational modification of the receptor, and degrading IFN receptors. Recently, influenza virus was found to induce CK1α-induced phosphorylation and subsequent degradation of the receptor for type I and II IFNs. In this review, viral mechanisms that remove IFN receptors are summarized with an emphasis on the mechanisms for virus-induced degradation of IFN receptors.
Collapse
Affiliation(s)
- Chuan Xia
- Departments of Surgery and Molecular Microbiology & Immunology, University of Missouri, Columbia, MO 65212, USA
| | - Paul Anderson
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA; Laboratory for Infectious Disease Research, University of Missouri, Columbia, MO 65211, USA
| | - Bumsuk Hahm
- Departments of Surgery and Molecular Microbiology & Immunology, University of Missouri, Columbia, MO 65212, USA.
| |
Collapse
|
28
|
Klinkhammer J, Schnepf D, Ye L, Schwaderlapp M, Gad HH, Hartmann R, Garcin D, Mahlakõiv T, Staeheli P. IFN-λ prevents influenza virus spread from the upper airways to the lungs and limits virus transmission. eLife 2018; 7:33354. [PMID: 29651984 PMCID: PMC5953542 DOI: 10.7554/elife.33354] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 04/11/2018] [Indexed: 12/20/2022] Open
Abstract
Host factors restricting the transmission of respiratory viruses are poorly characterized. We analyzed the contribution of type I and type III interferon (IFN) using a mouse model in which the virus is selectively administered to the upper airways, mimicking a natural respiratory virus infection. Mice lacking functional IFN-λ receptors (Ifnlr1−/−) no longer restricted virus dissemination from the upper airways to the lungs. Ifnlr1−/− mice shed significantly more infectious virus particles via the nostrils and transmitted the virus much more efficiently to naïve contacts compared with wild-type mice or mice lacking functional type I IFN receptors. Prophylactic treatment with IFN-α or IFN-λ inhibited initial virus replication in all parts of the respiratory tract, but only IFN-λ conferred long-lasting antiviral protection in the upper airways and blocked virus transmission. Thus, IFN-λ has a decisive and non-redundant function in the upper airways that greatly limits transmission of respiratory viruses to naïve contacts. Influenza (‘the flu’) and other respiratory viruses make millions of people ill every year, placing a large burden on the healthcare system and the economy. Unfortunately, few options for preventing or treating these infections currently exist. The flu virus spreads from infected individuals, enters a new host through the nose and establishes an infection in the upper airways. If the infection stays restricted to this region of the respiratory tract – which consists of the nasal cavity, sinuses, throat and larynx – it causes a rather mild disease. However, if it spreads to the lungs it can cause potentially life-threatening viral pneumonia. Epithelial cells line the upper respiratory tract, forming a physical border between the outside world and the human body. These cells are therefore the first to face the incoming virus. In response, the epithelial cells release messenger molecules termed interferons that warn nearby cells to increase their antiviral defenses. There are several subtypes of interferons, such as IFN-α, IFN-β and IFN-λ, but it was not known how each subtype helps to combat respiratory viruses. To investigate, Klinkhammer, Schnepf et al. exposed mice to flu viruses in a way that mimicked how an infection would naturally start in the upper airways in humans. Some of the mice were genetically engineered so that they could not respond to either IFN-α/β or IFN-λ. The virus spread most effectively from the nasal cavity to the lungs in mice whose IFN-λ system was defective. Infections in mice that lacked IFN-λ were also more likely to spread to other individuals. Furthermore, treating mice with IFN-λ, but not IFN-α, gave their upper respiratory tract long-lasting protection against flu infections and prevented the spread of the virus. IFN-λ therefore has a specific and significant role in protecting the upper airways against viruses, and could potentially be used as a drug to block the spread of infections between humans. Currently, IFN-λ is in clinical trials as a potential treatment for hepatitis D. To repurpose it for upper respiratory tract infections, its effectiveness against specific respiratory viruses will first have to be evaluated.
Collapse
Affiliation(s)
- Jonas Klinkhammer
- Institute of Virology, Medical Center, University of Freiburg, Freiburg, Germany.,MOTI-VATE Graduate School, Medical Center, University of Freiburg, Freiburg, Germany
| | - Daniel Schnepf
- Institute of Virology, Medical Center, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Liang Ye
- Institute of Virology, Medical Center, University of Freiburg, Freiburg, Germany
| | | | - Hans Henrik Gad
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Rune Hartmann
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Dominique Garcin
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Tanel Mahlakõiv
- Institute of Virology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Peter Staeheli
- Institute of Virology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
29
|
Xia C, Wolf JJ, Vijayan M, Studstill CJ, Ma W, Hahm B. Casein Kinase 1α Mediates the Degradation of Receptors for Type I and Type II Interferons Caused by Hemagglutinin of Influenza A Virus. J Virol 2018; 92:e00006-18. [PMID: 29343571 PMCID: PMC5972889 DOI: 10.1128/jvi.00006-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 01/08/2018] [Indexed: 01/24/2023] Open
Abstract
Although influenza A virus (IAV) evades cellular defense systems to effectively propagate in the host, the viral immune-evasive mechanisms are incompletely understood. Our recent data showed that hemagglutinin (HA) of IAV induces degradation of type I IFN receptor 1 (IFNAR1). Here, we demonstrate that IAV HA induces degradation of type II IFN (IFN-γ) receptor 1 (IFNGR1), as well as IFNAR1, via casein kinase 1α (CK1α), resulting in the impairment of cellular responsiveness to both type I and II IFNs. IAV infection or transient HA expression induced degradation of both IFNGR1 and IFNAR1, whereas HA gene-deficient IAV failed to downregulate the receptors. IAV HA caused the phosphorylation and ubiquitination of IFNGR1, leading to the lysosome-dependent degradation of IFNGR1. Influenza viral HA strongly decreased cellular sensitivity to type II IFNs, as it suppressed the activation of STAT1 and the induction of IFN-γ-stimulated genes in response to exogenously supplied recombinant IFN-γ. Importantly, CK1α, but not p38 MAP kinase or protein kinase D2, was proven to be critical for HA-induced degradation of both IFNGR1 and IFNAR1. Pharmacologic inhibition of CK1α or small interfering RNA (siRNA)-based knockdown of CK1α repressed the degradation processes of both IFNGR1 and IFNAR1 triggered by IAV infection. Further, CK1α was shown to be pivotal for proficient replication of IAV. Collectively, the results suggest that IAV HA induces degradation of IFN receptors via CK1α, creating conditions favorable for viral propagation. Therefore, the study uncovers a new immune-evasive pathway of influenza virus.IMPORTANCE Influenza A virus (IAV) remains a grave threat to humans, causing seasonal and pandemic influenza. Upon infection, innate and adaptive immunity, such as the interferon (IFN) response, is induced to protect hosts against IAV infection. However, IAV seems to be equipped with tactics to evade the IFN-mediated antiviral responses, although the detailed mechanisms need to be elucidated. In the present study, we show that IAV HA induces the degradation of the type II IFN receptor IFNGR1 and thereby substantially attenuates cellular responses to IFN-γ. Of note, a cellular kinase, casein kinase 1α (CK1α), is crucial for IAV HA-induced degradation of both IFNGR1 and IFNAR1. Accordingly, CK1α is proven to positively regulate IAV propagation. Thus, this study unveils a novel strategy employed by IAV to evade IFN-mediated antiviral activities. These findings may provide new insights into the interplay between IAV and host immunity to impact influenza virus pathogenicity.
Collapse
MESH Headings
- A549 Cells
- Animals
- Casein Kinase I/genetics
- Casein Kinase I/immunology
- Chlorocebus aethiops
- Dogs
- HEK293 Cells
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Immune Evasion
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza, Human/genetics
- Influenza, Human/immunology
- Influenza, Human/pathology
- Madin Darby Canine Kidney Cells
- Protein Kinase D2
- Protein Kinases/genetics
- Protein Kinases/immunology
- Proteolysis
- Receptor, Interferon alpha-beta/genetics
- Receptor, Interferon alpha-beta/immunology
- Receptors, Interferon/genetics
- Receptors, Interferon/immunology
- STAT1 Transcription Factor/genetics
- STAT1 Transcription Factor/immunology
- Vero Cells
- p38 Mitogen-Activated Protein Kinases/genetics
- p38 Mitogen-Activated Protein Kinases/immunology
- Interferon gamma Receptor
Collapse
Affiliation(s)
- Chuan Xia
- Department of Surgery, University of Missouri, Columbia, Missouri, USA
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Jennifer J Wolf
- Department of Surgery, University of Missouri, Columbia, Missouri, USA
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Madhuvanthi Vijayan
- Department of Surgery, University of Missouri, Columbia, Missouri, USA
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Caleb J Studstill
- Department of Surgery, University of Missouri, Columbia, Missouri, USA
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Wenjun Ma
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Bumsuk Hahm
- Department of Surgery, University of Missouri, Columbia, Missouri, USA
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
30
|
Rotavirus Degrades Multiple Interferon (IFN) Type Receptors To Inhibit IFN Signaling and Protects against Mortality from Endotoxin in Suckling Mice. J Virol 2017; 92:JVI.01394-17. [PMID: 29070687 DOI: 10.1128/jvi.01394-17] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/17/2017] [Indexed: 02/07/2023] Open
Abstract
STAT1 phosphorylation in response to exogenous interferon (IFN) administration can be inhibited by rotaviral replication both in vitro and in vivo In addition many rotavirus strains are resistant to the actions of different IFN types. The regulation by rotaviruses (RVs) of antiviral pathways mediated by multiple IFN types is not well understood. In this study, we find that during infection in vitro and in vivo, RVs significantly deplete IFN type I, II, and III receptors (IFNRs). Regulation of IFNRs occurred exclusively within RV-infected cells and could be abrogated by inhibiting the lysosomal-endosomal degradation pathway. In vitro, IFNR degradation was conserved across multiple RV strains that differ in their modes of regulating IFN induction. In suckling mice, exogenously administered type I, II, or III IFN induced phosphorylation of STAT1-Y701 within intestinal epithelial cells (IECs) of suckling mice. Murine EW strain RV infection transiently activated intestinal STAT1 at 1 day postinfection (dpi) but not subsequently at 2 to 3 dpi. In response to injection of purified IFN-α/β or -λ, IECs in EW-infected mice exhibited impaired STAT1-Y701 phosphorylation, correlating with depletion of different intestinal IFNRs and impaired IFN-mediated transcription. The ability of EW murine RV to inhibit multiple IFN types led us to test protection of suckling mice from endotoxin-mediated shock, an outcome that is dependent on the host IFN response. Compared to mortality in controls, mice infected with EW murine RV were substantially protected against mortality following parenteral endotoxin administration. These studies identify a novel mechanism of IFN subversion by RV and reveal an unexpected protective effect of RV infection on endotoxin-mediated shock in suckling mice.IMPORTANCE Antiviral functions of types I, II, and III IFNs are mediated by receptor-dependent activation of STAT1. Here, we find that RV degrades the types I, II, and III IFN receptors (IFNRs) in vitro In a suckling mouse model, RV effectively blocked STAT1 activation and transcription following injection of different purified IFNs. This correlated with significantly decreased protein expression of intestinal types I and II IFNRs. Recent studies demonstrate that in mice lipopolysaccharide (LPS)-induced lethality is prevented by genetic ablation of IFN signaling genes such as IFNAR1 and STAT1. When suckling mice were infected with RV, they were substantially protected from lethal exposure to endotoxin. These findings provide novel insights into the mechanisms underlying rotavirus regulation of different interferons and are likely to stimulate new research into both rotavirus pathogenesis and endotoxemia.
Collapse
|
31
|
Gui J, Zhao B, Lyu K, Tong W, Fuchs SY. Downregulation of the IFNAR1 chain of type 1 interferon receptor contributes to the maintenance of the haematopoietic stem cells. Cancer Biol Ther 2017; 18:534-543. [PMID: 28678581 DOI: 10.1080/15384047.2017.1345395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Recent studies demonstrated that prolonged exposure of haematopoietic stem cells (HSCs) to type I interferons (IFN) stimulates HSCs entrance into cell cycle, continuous proliferation and eventual exhaustion, which could be prevented by ablation of the Ifnar1 chain of IFN receptor. Given that levels IFNAR1 expression can be robustly affected by IFN-independent ubiquitination and downregulation of IFNAR1 in response to activation of protein kinases such as protein kinase R-like endoplasmic reticulum kinase (PERK) and casein kinase 1α (CK1α), we aimed to determine the role of IFNAR1 downregulation in the maintenance of HSCs. Mice harboring the ubiquitination-deficient Ifnar1S526A allele displayed greater levels of haematopoietic cell progenitors but reduced numbers of the long-term HSCs compared with wild type mice and animals lacking Ifnar1. Studies using competitive bone marrow repopulation assays showed that CK1α (but not PERK) is essential for the long-term HSCs function. Concurrent ablation of Ifnar1 led to a modest attenuation of the CK1α-null phenotype indicating that, although other CK1α targets are likely to be important, IFNAR1 downregulation can contribute to the maintenance of the HSCs function.
Collapse
Affiliation(s)
- Jun Gui
- a Department of Biomedical Sciences and Mari Lowe Center for Comparative Oncology , School of Veterinary Medicine, University of Pennsylvania , Philadelphia , PA , USA
| | - Bin Zhao
- a Department of Biomedical Sciences and Mari Lowe Center for Comparative Oncology , School of Veterinary Medicine, University of Pennsylvania , Philadelphia , PA , USA
| | - Kaosheng Lyu
- b Division of Hematology , Children's Hospital of Philadelphia, Perelman School of Medicine , Philadelphia , PA , USA.,c Department of Pediatrics , Perelman School of Medicine , Philadelphia , PA , USA
| | - Wei Tong
- b Division of Hematology , Children's Hospital of Philadelphia, Perelman School of Medicine , Philadelphia , PA , USA.,c Department of Pediatrics , Perelman School of Medicine , Philadelphia , PA , USA
| | - Serge Y Fuchs
- a Department of Biomedical Sciences and Mari Lowe Center for Comparative Oncology , School of Veterinary Medicine, University of Pennsylvania , Philadelphia , PA , USA
| |
Collapse
|
32
|
Brief Report: Increased Expression of the Type I Interferon Receptor on CD4+ T Lymphocytes in HIV-1-Infected Individuals. J Acquir Immune Defic Syndr 2017; 74:473-478. [PMID: 28009639 DOI: 10.1097/qai.0000000000001280] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Type I interferons (IFN1s; eg, interferon-alpha and interferon-beta) are potent cytokines that inhibit the replication of human immunodeficiency virus-1 (HIV-1) and other viruses. The antiviral and immunoregulatory activities of IFN1 are mediated through ligand-receptor interactions with the IFN1 receptor complex (IFNAR). Variation in the cell-surface density of IFNAR could play a role in HIV-1 pathogenesis. METHODS In this cross-sectional study of fresh whole blood, we used flow cytometry to evaluate the expression of IFNAR2 on lymphocyte subsets from HIV-1-infected (n = 33) and HIV-1-uninfected (n = 22) individuals. RESULTS In comparison with healthy blood bank donors, we observed that the HIV-1-infected individuals, particularly those having advanced to disease, exhibited the increased expression of IFNAR2 on CD4 T cells (relative fluorescence intensity 6.9 vs. 9.0; P = 0.027). The CD4:CD4 T-cell IFNAR2 expression-level ratio provides an internally standardized measure of this alteration. The observed increased expression of IFNAR2 was largely restricted to CD4 T cells that expressed the chemokine receptor CXCR4 and lacked the expression of CCR5. CONCLUSIONS HIV-1-infected individuals exhibit an increased expression of the IFN1 receptor on CD4 T cells. The level of IFNAR2 expression seems to increase with disease progression. These findings provide insight for the immunologic alterations associated with HIV-1 infection and possibly new therapeutic approaches.
Collapse
|
33
|
Zhao B, Bhattacharya S, Yu Q, Fuchs SY. Expression of the IFNAR1 chain of type 1 interferon receptor in benign cells protects against progression of acute leukemia. Leuk Lymphoma 2017; 59:171-177. [PMID: 28503979 DOI: 10.1080/10428194.2017.1319053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Type I interferons (IFN) were widely used for leukemia treatment. These cytokines act on cell surface receptor consisting of the IFNAR1/2 chains to induce anti-tumorigenic effects. Given that levels of IFNAR1 can be regulated by phosphorylation-driven ubiquitination and degradation that undermines IFN signaling and anti-tumorigenic effects, we sought to determine the importance of IFNAR1 downregulation in progression of acute leukemia. Using knock-in mice deficient in downregulation of IFNAR1, we uncovered that IFNAR1 expression in stromal benign cells functions to protect against progression of leukemia. We discuss putative mechanisms of this regulation and potential of therapeutic targeting of IFNAR1 downregulation to treat leukemia.
Collapse
Affiliation(s)
- Bin Zhao
- a Department of Biomedical Sciences , Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania , Philadelphia , PA , USA
| | - Sabyasachi Bhattacharya
- a Department of Biomedical Sciences , Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania , Philadelphia , PA , USA
| | - Qiujing Yu
- a Department of Biomedical Sciences , Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania , Philadelphia , PA , USA
| | - Serge Y Fuchs
- a Department of Biomedical Sciences , Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania , Philadelphia , PA , USA
| |
Collapse
|
34
|
Abstract
Type I interferons (IFN-1) are cytokines that affect the expression of thousands of genes, resulting in profound cellular changes. IFN-1 activates the cell by dimerizing its two-receptor chains, IFNAR1 and IFNAR2, which are expressed on all nucleated cells. Despite a similar mode of binding, the different IFN-1s activate a spectrum of activities. The causes for differential activation may stem from differences in IFN-1-binding affinity, duration of binding, number of surface receptors, induction of feedbacks, and cell type-specific variations. All together these will alter the signal that is transmitted from the extracellular domain inward. The intracellular domain binds, directly or indirectly, different effector proteins that transmit signals. The composition of effector molecules deviates between different cell types and tissues, inserting an additional level of complexity to the system. Moreover, IFN-1s do not act on their own, and clearly there is much cross-talk between the activated effector molecules by IFN-1 and other cytokines. The outcome generated by all of these factors (processing step) is an observed phenotype, which can be the transformation of the cell to an antiviral state, differentiation of the cell to a specific immune cell, senescence, apoptosis, and many more. IFN-1 activities can be divided into robust and tunable. Antiviral activity, which is stimulated by minute amounts of IFN-1 and is common to all cells, is termed robust. The other activities, which we term tunable, are cell type-specific and often require more stringent modes of activation. In this review, I summarize the current knowledge on the mode of activation and processing that is initiated by IFN-1, in perspective of the resulting phenotypes.
Collapse
Affiliation(s)
- Gideon Schreiber
- From the Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
35
|
Zhang KJ, Yin XF, Yang YQ, Li HL, Xu YN, Chen LY, Liu XJ, Yuan SJ, Fang XL, Xiao J, Wu S, Xu HN, Chu L, Katlinski KV, Katlinskaya YV, Guo RB, Wei GW, Wang DC, Liu XY, Fuchs SY. A Potent In Vivo Antitumor Efficacy of Novel Recombinant Type I Interferon. Clin Cancer Res 2017; 23:2038-2049. [PMID: 27683179 PMCID: PMC5373932 DOI: 10.1158/1078-0432.ccr-16-1386] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/28/2016] [Accepted: 09/11/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Antiproliferative, antiviral, and immunomodulatory activities of endogenous type I IFNs (IFN1) prompt the design of recombinant IFN1 for therapeutic purposes. However, most of the designed IFNs exhibited suboptimal therapeutic efficacies against solid tumors. Here, we report evaluation of the in vitro and in vivo antitumorigenic activities of a novel recombinant IFN termed sIFN-I.Experimental Design: We compared primary and tertiary structures of sIFN-I with its parental human IFNα-2b, as well as affinities of these ligands for IFN1 receptor chains and pharmacokinetics. These IFN1 species were also compared for their ability to induce JAK-STAT signaling and expression of the IFN1-stimulated genes and to elicit antitumorigenic effects. Effects of sIFN-I on tumor angiogenesis and immune infiltration were also tested in transplanted and genetically engineered immunocompetent mouse models.Results: sIFN-I displayed greater affinity for IFNAR1 (over IFNAR2) chain of the IFN1 receptor and elicited a greater extent of IFN1 signaling and expression of IFN-inducible genes in human cells. Unlike IFNα-2b, sIFN-I induced JAK-STAT signaling in mouse cells and exhibited an extended half-life in mice. Treatment with sIFN-I inhibited intratumoral angiogenesis, increased CD8+ T-cell infiltration, and robustly suppressed growth of transplantable and genetically engineered tumors in immunodeficient and immunocompetent mice.Conclusions: These findings define sIFN-I as a novel recombinant IFN1 with potent preclinical antitumorigenic effects against solid tumor, thereby prompting the assessment of sIFN-I clinical efficacy in humans. Clin Cancer Res; 23(8); 2038-49. ©2016 AACR.
Collapse
Affiliation(s)
- Kang-Jian Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
- Sichuan Huiyang Life Science and Technology Corp., Chengdu, Sichuan, China
| | - Xiao-Fei Yin
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuan-Qin Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, China
| | - Hui-Ling Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan-Ni Xu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Lie-Yang Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xi-Jun Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Su-Jing Yuan
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xian-Long Fang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jing Xiao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shuai Wu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hai-Neng Xu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Liang Chu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | - Yuliya V Katlinskaya
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rong-Bing Guo
- Sichuan Huiyang Life Science and Technology Corp., Chengdu, Sichuan, China
| | - Guang-Wen Wei
- Sichuan Huiyang Life Science and Technology Corp., Chengdu, Sichuan, China
| | - Da-Cheng Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xin-Yuan Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, China
| | - Serge Y Fuchs
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
36
|
Sanlorenzo M, Vujic I, Carnevale-Schianca F, Quaglino P, Gammaitoni L, Fierro MT, Aglietta M, Sangiolo D. Role of interferon in melanoma: old hopes and new perspectives. Expert Opin Biol Ther 2017; 17:475-483. [PMID: 28274138 DOI: 10.1080/14712598.2017.1289169] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Interferons (IFNs) play a key role in modulating anti-microbial and antitumor immune responses. In oncology, past attempts to exploit IFNs therapeutically did not fulfill expectations, and had only modest clinical results, mostly limited to adjuvant melanoma treatment. The recent successes of immunotherapy in oncology have brought new attention to the potential of immune-modulatory agents like the IFNs. Areas covered: The authors review the biological effects of IFN on melanoma and immune cells. Then, the authors summarize the clinical results of adjuvant and therapeutic IFN in melanoma, giving focus to possible prognostic factors and new on-going clinical trials. Expert opinion: IFNs offer intriguing opportunities for synergism between conventional treatments and recently introduced molecular-targeted and immunotherapy approaches. However, the full comprehension of all IFN effects and their multiple biologic links is challenging. A strong commitment toward parallel translational research is needed to facilitate the interpretation of IFN's expected and unexpected effects, guiding the rational design of informative clinical studies.
Collapse
Affiliation(s)
- Martina Sanlorenzo
- a Department of Oncology , University of Torino , Candiolo , Torino , Italy.,b Department of Medical Sciences, Section of Dermatology , University of Turin , Torino , Italy.,c Division of Medical Oncology, Experimental Cell Therapy , Candiolo Cancer Institute , Candiolo , Torino , Italy
| | - Igor Vujic
- d School of Medicine , Sigmund Freud University , Vienna , Austria.,e Department of Dermatology , The Rudolfstiftung Hospital, Academic Teaching Hospital, Medical University Vienna , Vienna , Austria
| | - Fabrizio Carnevale-Schianca
- c Division of Medical Oncology, Experimental Cell Therapy , Candiolo Cancer Institute , Candiolo , Torino , Italy
| | - Pietro Quaglino
- b Department of Medical Sciences, Section of Dermatology , University of Turin , Torino , Italy
| | - Loretta Gammaitoni
- c Division of Medical Oncology, Experimental Cell Therapy , Candiolo Cancer Institute , Candiolo , Torino , Italy
| | - Maria Teresa Fierro
- b Department of Medical Sciences, Section of Dermatology , University of Turin , Torino , Italy
| | - Massimo Aglietta
- a Department of Oncology , University of Torino , Candiolo , Torino , Italy.,c Division of Medical Oncology, Experimental Cell Therapy , Candiolo Cancer Institute , Candiolo , Torino , Italy
| | - Dario Sangiolo
- a Department of Oncology , University of Torino , Candiolo , Torino , Italy.,c Division of Medical Oncology, Experimental Cell Therapy , Candiolo Cancer Institute , Candiolo , Torino , Italy
| |
Collapse
|
37
|
Katlinski KV, Gui J, Katlinskaya YV, Ortiz A, Chakraborty R, Bhattacharya S, Carbone CJ, Beiting DP, Girondo MA, Peck AR, Puré E, Chatterji P, Rustgi AK, Diehl JA, Koumenis C, Rui H, Fuchs SY. Inactivation of Interferon Receptor Promotes the Establishment of Immune Privileged Tumor Microenvironment. Cancer Cell 2017; 31:194-207. [PMID: 28196594 PMCID: PMC5313042 DOI: 10.1016/j.ccell.2017.01.004] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/15/2016] [Accepted: 01/09/2017] [Indexed: 12/12/2022]
Abstract
Refractoriness of solid tumors, including colorectal cancers (CRCs), to immunotherapies is attributed to the immunosuppressive tumor microenvironment that protects malignant cells from cytotoxic T lymphocytes (CTLs). We found that downregulation of the type I interferon receptor chain IFNAR1 occurs in human CRC and mouse models of CRC. Downregulation of IFNAR1 in tumor stroma stimulated CRC development and growth, played a key role in formation of the immune-privileged niche, and predicted poor prognosis in human CRC patients. Genetic stabilization of IFNAR1 improved CTL survival and increased the efficacy of the chimeric antigen receptor T cell transfer and PD-1 inhibition. Likewise, pharmacologic stabilization of IFNAR1 suppressed tumor growth providing the rationale for upregulating IFNAR1 to improve anti-cancer therapies.
Collapse
Affiliation(s)
- Kanstantsin V Katlinski
- Department of Biomedical Sciences, Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jun Gui
- Department of Biomedical Sciences, Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuliya V Katlinskaya
- Department of Biomedical Sciences, Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Angelíca Ortiz
- Department of Biomedical Sciences, Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Riddhita Chakraborty
- Department of Biomedical Sciences, Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sabyasachi Bhattacharya
- Department of Biomedical Sciences, Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher J Carbone
- Department of Biomedical Sciences, Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel P Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Melanie A Girondo
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Amy R Peck
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ellen Puré
- Department of Biomedical Sciences, Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Priya Chatterji
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anil K Rustgi
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - J Alan Diehl
- Department of Biochemistry, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
38
|
The Many Faces of the Flavivirus NS5 Protein in Antagonism of Type I Interferon Signaling. J Virol 2017; 91:JVI.01970-16. [PMID: 27881649 DOI: 10.1128/jvi.01970-16] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The vector-borne flaviviruses cause severe disease in humans on every inhabited continent on earth. Their transmission by arthropods, particularly mosquitoes, facilitates large emergence events such as witnessed with Zika virus (ZIKV) or West Nile virus in the Americas. Every vector-borne flavivirus examined thus far that causes disease in humans, from dengue virus to ZIKV, antagonizes the host type I interferon (IFN-I) response by preventing JAK-STAT signaling, suggesting that suppression of this pathway is an important determinant of infection. The most direct and potent viral inhibitor of this pathway is the nonstructural protein NS5. However, the mechanisms utilized by NS5 from different flaviviruses are often quite different, sometimes despite close evolutionary relationships between viruses. The varied mechanisms of NS5 as an IFN-I antagonist are also surprising given that the evolution of NS5 is restrained by the requirement to maintain function of two enzymatic activities critical for virus replication, the methyltransferase and RNA-dependent RNA polymerase. This review discusses the different strategies used by flavivirus NS5 to evade the antiviral effects of IFN-I and how this information can be used to better model disease and develop antiviral countermeasures.
Collapse
|
39
|
Ziegler SM, Beisel C, Sutter K, Griesbeck M, Hildebrandt H, Hagen SH, Dittmer U, Altfeld M. Human pDCs display sex-specific differences in type I interferon subtypes and interferon α/β receptor expression. Eur J Immunol 2017; 47:251-256. [DOI: 10.1002/eji.201646725] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/31/2016] [Accepted: 11/24/2016] [Indexed: 11/06/2022]
Affiliation(s)
- Susanne M. Ziegler
- Department of Virus Immunology; Heinrich Pette Institute; Leibniz Institute for Experimental Virology; Hamburg Germany
| | - Claudia Beisel
- Department of Virus Immunology; Heinrich Pette Institute; Leibniz Institute for Experimental Virology; Hamburg Germany
- Section Infectious Diseases; I. Department of Internal Medicine; University Medical Center Hamburg-Eppendorf, and DZIF partner site (German Center for Infection Research); Hamburg Germany
| | - Kathrin Sutter
- Institute for Virology University Hospital in Essen; University of Duisburg-Essen; Essen Germany
| | - Morgane Griesbeck
- CIMI; Université Pierre et Marie Curie/INSERM U1135; Hospital Pitié Salpêtrière; Paris France
| | - Heike Hildebrandt
- Department of Virus Immunology; Heinrich Pette Institute; Leibniz Institute for Experimental Virology; Hamburg Germany
| | - Sven H. Hagen
- Department of Virus Immunology; Heinrich Pette Institute; Leibniz Institute for Experimental Virology; Hamburg Germany
| | - Ulf Dittmer
- Institute for Virology University Hospital in Essen; University of Duisburg-Essen; Essen Germany
| | - Marcus Altfeld
- Department of Virus Immunology; Heinrich Pette Institute; Leibniz Institute for Experimental Virology; Hamburg Germany
| |
Collapse
|
40
|
Abstract
Cancer, like many others, is a disease due to dysfunction of cytokine-regulated networks. In this background, the 1st Aegean conference meeting focused on the Cytokine Signaling in Cancer was organized and held at Chania, Crete, Greece in May of 2015. A number of novel aspects of the cytokine signaling and their relevance to oncogenic processes were presented at that meeting. This special issue of Cytokine captures snap shots of this meeting in the form of reviews. In addition to these reviews, several other original research articles on the roles of cytokines and chemokines in various cancers are also present in this issue.
Collapse
Affiliation(s)
- Dhan V Kalvakolanu
- Department of Microbiology & Immunology, and Greenebaum NCI-Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America.
| |
Collapse
|
41
|
He Y, Huang J, Wang P, Shen X, Li S, Yang L, Liu W, Suksamrarn A, Zhang G, Wang F. Emodin potentiates the antiproliferative effect of interferon α/β by activation of JAK/STAT pathway signaling through inhibition of the 26S proteasome. Oncotarget 2016; 7:4664-79. [PMID: 26683360 PMCID: PMC4826234 DOI: 10.18632/oncotarget.6616] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 11/29/2015] [Indexed: 12/12/2022] Open
Abstract
The 26S proteasome is a negative regulator of type I interferon (IFN-α/β) signaling. Inhibition of the 26S proteasome by small molecules may be a new strategy to enhance the efficacy of type I IFNs and reduce their side effects. Using cell-based screening assay for new 26S proteasome inhibitors, we found that emodin, a natural anthraquinone, was a potent inhibitor of the human 26S proteasome. Emodin preferably inhibited the caspase-like and chymotrypsin-like activities of the human 26S proteasome and increased the ubiquitination of endogenous proteins in cells. Computational modeling showed that emodin exhibited an orientation/conformation favorable to nucleophilic attack in the active pocket of the β1, β2, and β5 subunits of the 26S proteasome. Emodin increased phosphorylation of STAT1, decreased phosphorylation of STAT3 and increased endogenous gene expression stimulated by IFN-α. Emodin inhibited IFN-α-stimulated ubiquitination and degradation of type I interferon receptor 1 (IFNAR1). Emodin also sensitized the antiproliferative effect of IFN-α in HeLa cervical carcinoma cells and reduced tumor growth in Huh7 hepatocellular carcinoma-bearing mice. These results suggest that emodin potentiates the antiproliferative effect of IFN-α by activation of JAK/STAT pathway signaling through inhibition of 26S proteasome-stimulated IFNAR1 degradation. Therefore, emodin warrants further investigation as a new means to enhance the efficacy of IFN-α/β.
Collapse
Affiliation(s)
- Yujiao He
- Key Laboratory of Natural Medicine and Clinical Translation, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Junmei Huang
- Key Laboratory of Natural Medicine and Clinical Translation, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China.,School of Chinese Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ping Wang
- School of Chinese Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofei Shen
- Key Laboratory of Natural Medicine and Clinical Translation, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Sheng Li
- Key Laboratory of Natural Medicine and Clinical Translation, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Lijuan Yang
- Key Laboratory of Natural Medicine and Clinical Translation, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Wanli Liu
- MOE Key Laboratory of Protein Science, School of Life Sciences, Tsinghua University, Beijing, China
| | - Apichart Suksamrarn
- Department of Chemistry and Center for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Guolin Zhang
- Key Laboratory of Natural Medicine and Clinical Translation, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China.,Sichuan Translational Medicine Research Hospital, Chinese Academy of Sciences, Chengdu, China
| | - Fei Wang
- Key Laboratory of Natural Medicine and Clinical Translation, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China.,Sichuan Translational Medicine Research Hospital, Chinese Academy of Sciences, Chengdu, China
| |
Collapse
|
42
|
Gui J, Gober M, Yang X, Katlinski KV, Marshall CM, Sharma M, Werth VP, Baker DP, Rui H, Seykora JT, Fuchs SY. Therapeutic Elimination of the Type 1 Interferon Receptor for Treating Psoriatic Skin Inflammation. J Invest Dermatol 2016; 136:1990-2002. [PMID: 27369778 PMCID: PMC5035634 DOI: 10.1016/j.jid.2016.06.608] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 05/25/2016] [Accepted: 06/09/2016] [Indexed: 12/27/2022]
Abstract
Phototherapy with UV light is a standard treatment for psoriasis, yet the mechanisms underlying the therapeutic effects are not well understood. Studies in human and mouse keratinocytes and in the skin tissues from human patients and mice showed that UV treatment triggers ubiquitination and downregulation of the type I IFN receptor chain IFNAR1, leading to suppression of IFN signaling and an ensuing decrease in the expression of inflammatory cytokines and chemokines. The severity of imiquimod-induced psoriasiform inflammation was greatly exacerbated in skin of mice deficient in IFNAR1 ubiquitination (Ifnar1(SA)). Furthermore, these mice did not benefit from UV phototherapy. Pharmacologic induction of IFNAR1 ubiquitination and degradation by an antiprotozoal agent halofuginone also relieved psoriasiform inflammation in wild-type but not in Ifnar1(SA) mice. These data identify downregulation of IFNAR1 by UV as a major mechanism of the UV therapeutic effects against the psoriatic inflammation and provide a proof of principle for future development of agents capable of inducing IFNAR1 ubiquitination and downregulation for the treatment of psoriasis.
Collapse
Affiliation(s)
- Jun Gui
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael Gober
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xiaoping Yang
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kanstantsin V Katlinski
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christine M Marshall
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Meena Sharma
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Victoria P Werth
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - John T Seykora
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
43
|
Kavrochorianou N, Markogiannaki M, Haralambous S. IFN-β differentially regulates the function of T cell subsets in MS and EAE. Cytokine Growth Factor Rev 2016; 30:47-54. [DOI: 10.1016/j.cytogfr.2016.03.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 03/21/2016] [Indexed: 12/30/2022]
|
44
|
Li S, Yang LJ, Wang P, He YJ, Huang JM, Liu HW, Shen XF, Wang F. Dietary apigenin potentiates the inhibitory effect of interferon-α on cancer cell viability through inhibition of 26S proteasome-mediated interferon receptor degradation. Food Nutr Res 2016; 60:31288. [PMID: 27356910 PMCID: PMC4928072 DOI: 10.3402/fnr.v60.31288] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/29/2016] [Accepted: 04/30/2016] [Indexed: 01/09/2023] Open
Abstract
Background Type I interferons (IFN-α/β) have broad and potent immunoregulatory and antiproliferative activities. However, it is still known whether the dietary flavonoids exhibit their antiviral and anticancer properties by modulating the function of type I IFNs.
Objective This study aimed at determining the role of apigenin, a dietary plant flavonoid abundant in common fruits and vegetables, on the type I IFN-mediated inhibition of cancer cell viability. Design Inhibitory effect of apigenin on human 26S proteasome, a known negative regulator of type I IFN signaling, was evaluated in vitro. Molecular docking was conducted to know the interaction between apigenin and subunits of 26S proteasome. Effects of apigenin on JAK/STAT pathway, 26S proteasome-mediated interferon receptor stability, and cancer cells viability were also investigated. Results Apigenin was identified to be a potent inhibitor of human 26S proteasome in a cell-based assay. Apigenin inhibited the chymotrypsin-like, caspase-like, and trypsin-like activities of the human 26S proteasome and increased the ubiquitination of endogenous proteins in cells. Results from computational modeling of the potential interactions of apigenin with the chymotrypsin site (β5 subunit), caspase site (β1 subunit), and trypsin site (β2 subunit) of the proteasome were consistent with the observed proteasome inhibitory activity. Apigenin enhanced the phosphorylation of signal transducer and activator of transcription proteins (STAT1 and STAT2) and promoted the endogenous IFN-α-regulated gene expression. Apigenin inhibited the IFN-α-stimulated ubiquitination and degradation of type I interferon receptor 1 (IFNAR1). Apigenin also sensitized the inhibitory effect of IFN-α on viability of cervical carcinoma HeLa cells. Conclusion These results suggest that apigenin potentiates the inhibitory effect of IFN-α on cancer cell viability by activating JAK/STAT signaling pathway through inhibition of 26S proteasome-mediated IFNAR1 degradation. This may provide a novel mechanism for increasing the efficacy of IFN-α/β.
Collapse
Affiliation(s)
- Sheng Li
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Li-Juan Yang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Ping Wang
- School of Chinese Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-Jiao He
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Jun-Mei Huang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China.,School of Chinese Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Han-Wei Liu
- Ningbo Entry-Exit Inspection and Quarantine Bureau Technical Center, Ningbo, China
| | - Xiao-Fei Shen
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Fei Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China.,Sichuan Translational Medicine Research Hospital, Chinese Academy of Sciences, Chengdu, China;
| |
Collapse
|
45
|
The Type I Interferon Response Determines Differences in Choroid Plexus Susceptibility between Newborns and Adults in Herpes Simplex Virus Encephalitis. mBio 2016; 7:e00437-16. [PMID: 27073094 PMCID: PMC4959527 DOI: 10.1128/mbio.00437-16] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Newborns are significantly more susceptible to severe viral encephalitis than adults, with differences in the host response to infection implicated as a major factor. However, the specific host signaling pathways responsible for differences in susceptibility and neurologic morbidity have remained unknown. In a murine model of HSV encephalitis, we demonstrated that the choroid plexus (CP) is susceptible to herpes simplex virus 1 (HSV-1) early in infection of the newborn but not the adult brain. We confirmed susceptibility of the CP to HSV infection in a human case of newborn HSV encephalitis. We investigated components of the type I interferon (IFN) response in the murine brain that might account for differences in cell susceptibility and found that newborns have a dampened interferon response and significantly lower basal levels of the alpha/beta interferon (IFN-α/β) receptor (IFNAR) than do adults. To test the contribution of IFNAR to restricting infection from the CP, we infected IFNAR knockout (KO) adult mice, which showed restored CP susceptibility to HSV-1 infection in the adult. Furthermore, reduced IFNAR levels did not account for differences we found in the basal levels of several other innate signaling proteins in the wild-type newborn and the adult, including protein kinase R (PKR), that suggested specific regulation of innate immunity in the developing brain. Viral targeting of the CP, a region of the brain that plays a critical role in neurodevelopment, provides a link between newborn susceptibility to HSV and long-term neurologic morbidity among survivors of newborn HSV encephalitis. Compared to adults, newborns are significantly more susceptible to severe disease following HSV infection. Over half of newborn HSV infections result in disseminated disease or encephalitis, with long-term neurologic morbidity in 2/3 of encephalitis survivors. We investigated differences in host cell susceptibility between newborns and adults that contribute to severe central nervous system disease in the newborn. We found that, unlike the adult brain, the newborn choroid plexus (CP) was susceptible early in HSV-1 infection. We demonstrated that IFN-α/β receptor levels are lower in the newborn brain than in the adult brain and that deletion of this receptor restores susceptibility of the CP in the adult brain. The CP serves as a barrier between the blood and the cerebrospinal fluid and plays a role in proper neurodevelopment. Susceptibility of the newborn choroid plexus to HSV-1 has important implications in viral spread to the brain and, also, in the neurologic morbidity following HSV encephalitis.
Collapse
|
46
|
Lubick KJ, Robertson SJ, McNally KL, Freedman BA, Rasmussen AL, Taylor RT, Walts AD, Tsuruda S, Sakai M, Ishizuka M, Boer EF, Foster EC, Chiramel AI, Addison CB, Green R, Kastner DL, Katze MG, Holland SM, Forlino A, Freeman AF, Boehm M, Yoshii K, Best SM. Flavivirus Antagonism of Type I Interferon Signaling Reveals Prolidase as a Regulator of IFNAR1 Surface Expression. Cell Host Microbe 2016; 18:61-74. [PMID: 26159719 DOI: 10.1016/j.chom.2015.06.007] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 04/08/2015] [Accepted: 06/16/2015] [Indexed: 11/25/2022]
Abstract
Type I interferon (IFN-α/β or IFN-I) signals through two receptor subunits, IFNAR1 and IFNAR2, to orchestrate sterile and infectious immunity. Cellular pathways that regulate IFNAR1 are often targeted by viruses to suppress the antiviral effects of IFN-I. Here we report that encephalitic flaviviruses, including tick-borne encephalitis virus and West Nile virus, antagonize IFN-I signaling by inhibiting IFNAR1 surface expression. Loss of IFNAR1 was associated with binding of the viral IFN-I antagonist, NS5, to prolidase (PEPD), a cellular dipeptidase implicated in primary immune deficiencies in humans. Prolidase was required for IFNAR1 maturation and accumulation, activation of IFNβ-stimulated gene induction, and IFN-I-dependent viral control. Human fibroblasts derived from patients with genetic prolidase deficiency exhibited decreased IFNAR1 surface expression and reduced IFNβ-stimulated signaling. Thus, by understanding flavivirus IFN-I antagonism, prolidase is revealed as a central regulator of IFN-I responses.
Collapse
Affiliation(s)
- Kirk J Lubick
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, MT 59840, USA
| | - Shelly J Robertson
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, MT 59840, USA
| | - Kristin L McNally
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, MT 59840, USA
| | - Brett A Freedman
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, MT 59840, USA
| | - Angela L Rasmussen
- Department of Microbiology, University of Washington, Seattle, WA 98109, USA
| | - R Travis Taylor
- Department of Medical Microbiology and Immunology, College of Medicine, University of Toledo Health Science Campus, Toledo, OH 43614, USA
| | - Avram D Walts
- Translational Medicine Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Seitaro Tsuruda
- Laboratory of Public Health, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
| | - Mizuki Sakai
- Laboratory of Public Health, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
| | - Mariko Ishizuka
- Laboratory of Public Health, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
| | - Elena F Boer
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, MT 59840, USA
| | - Erin C Foster
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, MT 59840, USA
| | - Abhilash I Chiramel
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, MT 59840, USA
| | - Conrad B Addison
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, MT 59840, USA
| | - Richard Green
- Department of Microbiology, University of Washington, Seattle, WA 98109, USA
| | - Daniel L Kastner
- Metabolic, Cardiovascular and Inflammatory Disease Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Michael G Katze
- Department of Microbiology, University of Washington, Seattle, WA 98109, USA
| | - Steven M Holland
- Immunopathogenesis Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20814, USA
| | - Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, 27100 Pavia, Italy
| | - Alexandra F Freeman
- Immunopathogenesis Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20814, USA
| | - Manfred Boehm
- Translational Medicine Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Kentaro Yoshii
- Laboratory of Public Health, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
| | - Sonja M Best
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, MT 59840, USA.
| |
Collapse
|
47
|
Katlinskaya YV, Katlinski KV, Yu Q, Ortiz A, Beiting DP, Brice A, Davar D, Sanders C, Kirkwood JM, Rui H, Xu X, Koumenis C, Diehl JA, Fuchs SY. Suppression of Type I Interferon Signaling Overcomes Oncogene-Induced Senescence and Mediates Melanoma Development and Progression. Cell Rep 2016; 15:171-180. [PMID: 27052162 DOI: 10.1016/j.celrep.2016.03.006] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/01/2016] [Accepted: 02/26/2016] [Indexed: 12/31/2022] Open
Abstract
Oncogene activation induces DNA damage responses and cell senescence. We report a key role of type I interferons (IFNs) in oncogene-induced senescence. IFN signaling-deficient melanocytes expressing activated Braf do not exhibit senescence and develop aggressive melanomas. Restoration of IFN signaling in IFN-deficient melanoma cells induces senescence and suppresses melanoma progression. Additional data from human melanoma patients and mouse transplanted tumor models suggest the importance of non-cell-autonomous IFN signaling. Inactivation of the IFN pathway is mediated by the IFN receptor IFNAR1 downregulation that invariably occurs during melanoma development. Mice harboring an IFNAR1 mutant, which is partially resistant to downregulation, delay melanoma development, suppress metastatic disease, and better respond to BRAF or PD-1 inhibitors. These results suggest that IFN signaling is an important tumor-suppressive pathway that inhibits melanoma development and progression and argue for targeting IFNAR1 downregulation to prevent metastatic disease and improve the efficacy of molecularly target and immune-targeted melanoma therapies.
Collapse
Affiliation(s)
- Yuliya V Katlinskaya
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kanstantsin V Katlinski
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Qiujing Yu
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Angelica Ortiz
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel P Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Angela Brice
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Diwakar Davar
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
| | - Cindy Sanders
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
| | - John M Kirkwood
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
| | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - J Alan Diehl
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
48
|
Kabilova TO, Sen’kova AV, Nikolin VP, Popova NA, Zenkova MA, Vlassov VV, Chernolovskaya EL. Antitumor and Antimetastatic Effect of Small Immunostimulatory RNA against B16 Melanoma in Mice. PLoS One 2016; 11:e0150751. [PMID: 26981617 PMCID: PMC4794162 DOI: 10.1371/journal.pone.0150751] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 02/17/2016] [Indexed: 12/23/2022] Open
Abstract
Small interfering RNAs, depending on their structure, delivery system and sequence, can stimulate innate and adaptive immunity. The aim of this study was to investigate the antitumor and antimetastatic effects of immunostimulatory 19-bp dsRNA with 3'- trinucleotide overhangs (isRNA) on melanoma B16 in C57Bl/6 mice. Recently developed novel cationic liposomes 2X3-DOPE were used for the in vivo delivery of isRNA. Administration of isRNA/2X3-DOPE complexes significantly inhibits melanoma tumor growth and metastasis. Histopathological analysis of spleen cross sections showed hyperplasia of the lymphoid white pulp and formation of large germinal centers after isRNA/2X3-DOPE administration, indicating activation of the immune system. The treatment of melanoma-bearing mice with isRNA/2X3-DOPE decreases the destructive changes in the liver parenchyma. Thus, the developed isRNA displays pronounced immunostimulatory, antitumor and antimetastatic properties against melanoma B16 and may be considered a potential agent in the immunotherapy of melanoma.
Collapse
Affiliation(s)
- Tatyana O. Kabilova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 8, Lavrentiev Avenue, Novosibirsk, Russia, 630090
| | - Aleksandra V. Sen’kova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 8, Lavrentiev Avenue, Novosibirsk, Russia, 630090
| | - Valeriy P. Nikolin
- Institute of Cytology and Genetics SB RAS, 10, Lavrentiev Avenue, Novosibirsk, Russia, 630090
| | - Nelly A. Popova
- Institute of Cytology and Genetics SB RAS, 10, Lavrentiev Avenue, Novosibirsk, Russia, 630090
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 8, Lavrentiev Avenue, Novosibirsk, Russia, 630090
| | - Valentin V. Vlassov
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 8, Lavrentiev Avenue, Novosibirsk, Russia, 630090
| | - Elena L. Chernolovskaya
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 8, Lavrentiev Avenue, Novosibirsk, Russia, 630090
- * E-mail:
| |
Collapse
|
49
|
Abstract
The interferons (IFNs) are a family of cytokines that protect against disease by direct effects on target cells and by activating immune responses. The production and actions of IFNs are finely tuned to achieve maximal protection and avoid the potential toxicity associated with excessive responses. IFNs are back in the spotlight owing to mounting evidence that is reshaping how we can exploit this pathway therapeutically. As IFNs can be produced by, and act on, both tumour cells and immune cells, understanding this reciprocal interaction will enable the development of improved single-agent or combination therapies that exploit IFN pathways and new 'omics'-based biomarkers to indicate responsive patients.
Collapse
Affiliation(s)
- Belinda S Parker
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Jai Rautela
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Paul J Hertzog
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
50
|
Type I Interferons Control Proliferation and Function of the Intestinal Epithelium. Mol Cell Biol 2016; 36:1124-35. [PMID: 26811327 DOI: 10.1128/mcb.00988-15] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 01/15/2016] [Indexed: 01/08/2023] Open
Abstract
Wnt pathway-driven proliferation and renewal of the intestinal epithelium must be tightly controlled to prevent development of cancer and barrier dysfunction. Although type I interferons (IFN) produced in the gut under the influence of microbiota are known for their antiproliferative effects, the role of these cytokines in regulating intestinal epithelial cell renewal is largely unknown. Here we report a novel role for IFN in the context of intestinal knockout of casein kinase 1α (CK1α), which controls the ubiquitination and degradation of both β-catenin and the IFNAR1 chain of the IFN receptor. Ablation of CK1α leads to the activation of both β-catenin and IFN pathways and prevents the unlimited proliferation of intestinal epithelial cells despite constitutive β-catenin activity. IFN signaling contributes to the activation of the p53 pathway and the appearance of apoptotic and senescence markers in the CK1α-deficient gut. Concurrent genetic ablation of CK1α and IFNAR1 leads to intestinal hyperplasia, robust attenuation of apoptosis, and rapid and lethal loss of barrier function. These data indicate that IFN play an important role in controlling the proliferation and function of the intestinal epithelium in the context of β-catenin activation.
Collapse
|