1
|
Becher B, Derfuss T, Liblau R. Targeting cytokine networks in neuroinflammatory diseases. Nat Rev Drug Discov 2024; 23:862-879. [PMID: 39261632 DOI: 10.1038/s41573-024-01026-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/13/2024]
Abstract
In neuroinflammatory diseases, systemic (blood-borne) leukocytes invade the central nervous system (CNS) and lead to tissue damage. A causal relationship between neuroinflammatory diseases and dysregulated cytokine networks is well established across several preclinical models. Cytokine dysregulation is also observed as an inadvertent effect of cancer immunotherapy, where it often leads to neuroinflammation. Neuroinflammatory diseases can be separated into those in which a pathogen is at the centre of the immune response and those of largely unknown aetiology. Here, we discuss the pathophysiology, cytokine networks and therapeutic landscape of 'sterile' neuroinflammatory diseases such as multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), neurosarcoidosis and immune effector cell-associated neurotoxicity syndrome (ICANS) triggered by cancer immunotherapy. Despite successes in targeting cytokine networks in preclinical models of neuroinflammation, the clinical translation of targeting cytokines and their receptors has shown mixed and often paradoxical responses.
Collapse
Affiliation(s)
- Burkhard Becher
- Institute of experimental Immunology, University of Zurich, Zurich, Switzerland.
| | - Tobias Derfuss
- Department of Neurology and Biomedicine, Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Roland Liblau
- Institute for inflammatory and infectious diseases, INSERM UMR1291 - CNRS UMR505, Toulouse, France.
| |
Collapse
|
2
|
Farhangian M, Azarafrouz F, Valian N, Dargahi L. The role of interferon beta in neurological diseases and its potential therapeutic relevance. Eur J Pharmacol 2024; 981:176882. [PMID: 39128808 DOI: 10.1016/j.ejphar.2024.176882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 07/14/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Interferon beta (IFNβ) is a member of the type-1 interferon family and has various immunomodulatory functions in neuropathological conditions. Although the level of IFNβ is low under healthy conditions, it is increased during inflammatory processes to protect the central nervous system (CNS). In particular, microglia and astrocytes are the main sources of IFNβ upon inflammatory insult in the CNS. The protective effects of IFNβ are well characterized in reducing the progression of multiple sclerosis (MS); however, little is understood about its effects in other neurological/neurodegenerative diseases. In this review, different types of IFNs and their signaling pathways will be described. Then we will focus on the potential role and therapeutic effect of IFNβ in several CNS-related diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, stroke, spinal cord injury, prion disease and spinocerebellar ataxia 7.
Collapse
Affiliation(s)
- Mohsen Farhangian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Forouzan Azarafrouz
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Valian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Harrer DC, Eder M, Barden M, Pan H, Herr W, Abken H. Ectopic PU.1 Expression Provides Chimeric Antigen Receptor (CAR) T Cells with Innate Cell Capacities Including IFN-β Release. Cancers (Basel) 2024; 16:2737. [PMID: 39123467 PMCID: PMC11311516 DOI: 10.3390/cancers16152737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has achieved extraordinary success in eliminating B cell malignancies; however, so far, it has shown limited efficacy in the treatment of solid tumors, which is thought to be due to insufficient CAR T cell activation. We hypothesized that the transcription factor PU.1, a master regulator of innate cell functionality, may augment pro-inflammatory CAR T cell activation. T cells were engineered with a CEA-specific CAR together with the constitutive expression of PU.1. CAR-redirected T cell activation was recorded for canonical functionality in vitro under conditions of prolonged repetitive antigen exposure. Ectopic PU.1 expression in CAR T cells upregulated the costimulatory receptors CD40, CD80, CD86, and CD70, which, unexpectedly, did not augment effector functions but hampered the upregulation of 4-1BB, decreased IL-2 production, reduced CAR T cell proliferation, and impaired their cytotoxic capacities. Under "stress" conditions of repetitive engagement of cognate tumor cells, CAR T cells with ectopic PU.1 showed reduced persistence, and finally failed to control the growth of cancer cells. Mechanistically, PU.1 caused CAR T cells to secrete IFN-β, a cytokine known to promote CAR T cell attrition and apoptosis. Collectively, PU.1 can polarize the functional capacities of CAR T cells towards innate cells.
Collapse
Affiliation(s)
- Dennis Christoph Harrer
- Department of Hematology and Internal Oncology, University Hospital Regensburg, 93053 Regensburg, Germany;
- Leibniz Institute for Immunotherapy, Division of Genetic Immunotherapy, University Regensburg, 93053 Regensburg, Germany; (M.E.); (M.B.); (H.P.); (H.A.)
| | - Matthias Eder
- Leibniz Institute for Immunotherapy, Division of Genetic Immunotherapy, University Regensburg, 93053 Regensburg, Germany; (M.E.); (M.B.); (H.P.); (H.A.)
| | - Markus Barden
- Leibniz Institute for Immunotherapy, Division of Genetic Immunotherapy, University Regensburg, 93053 Regensburg, Germany; (M.E.); (M.B.); (H.P.); (H.A.)
| | - Hong Pan
- Leibniz Institute for Immunotherapy, Division of Genetic Immunotherapy, University Regensburg, 93053 Regensburg, Germany; (M.E.); (M.B.); (H.P.); (H.A.)
| | - Wolfgang Herr
- Department of Hematology and Internal Oncology, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Hinrich Abken
- Leibniz Institute for Immunotherapy, Division of Genetic Immunotherapy, University Regensburg, 93053 Regensburg, Germany; (M.E.); (M.B.); (H.P.); (H.A.)
| |
Collapse
|
4
|
Huang S, Kang Y, Zheng R, Yang L, Gao J, Tang W, Jiang J, He J, Xie J. Two cytokine receptor family B (CRFB) members in orange-spotted grouper Epinephelus coioides, EcCRFB3 and EcCRFB4, negatively regulate interferon immune responses to assist nervous necrosis virus replication. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109718. [PMID: 38909635 DOI: 10.1016/j.fsi.2024.109718] [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: 05/17/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 06/25/2024]
Abstract
Receptors of type I interferon (IFNR) play a vital role in the antiviral immune response. However, little is known about the negative regulatory role of the IFNR. Nervous necrosis virus (NNV) is one of the most significant viruses in cultured fish, resulting in great economic losses for the aquaculture industry. In this study, two orange-spotted grouper (Epinephelus coioides) cytokine receptor family B (CRFB) members, EcCRFB3 and EcCRFB4 were cloned and characterized from NNV infected grouper brain (GB) cells. The open reading frame (ORF) of EcCRFB3 consists of 852 bp encoding 283 amino acids, while EcCRFB4 has an ORF of 990 bp encoding 329 amino acids. The mRNA levels of EcCRFB3 or EcCRFB4 were significantly upregulated after NNV infection and the stimulation of poly (I:C) or NNV-encoded Protein A. In addition, EcCRFB3 or EcCRFB4 overexpression facilitated NNV replication, whereas EcCRFB3 or EcCRFB4 silencing resisted NNV replication. Overexpressed EcCRFB3 or EcCRFB4 inhibited the expression of IFN-I-induced ISGs. Taken together, our research provides the first evidence in fish demonstrating the role of IFNRs to regulate the IFN signaling pathway negatively. Our findings enrich the understanding of the functions of IFNRs and reveal a novel escape mechanism of NNV.
Collapse
Affiliation(s)
- Siyou Huang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Yiling Kang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Rui Zheng
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Linwei Yang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Jie Gao
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Wanting Tang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Jing Jiang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Jianguo He
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Junfeng Xie
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China.
| |
Collapse
|
5
|
Yasmeen F, Pirzada RH, Ahmad B, Choi B, Choi S. Understanding Autoimmunity: Mechanisms, Predisposing Factors, and Cytokine Therapies. Int J Mol Sci 2024; 25:7666. [PMID: 39062908 PMCID: PMC11277571 DOI: 10.3390/ijms25147666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
Autoimmunity refers to an organism's immune response against its own healthy cells, tissues, or components, potentially leading to irreversible damage to vital organs. Central and peripheral tolerance mechanisms play crucial roles in preventing autoimmunity by eliminating self-reactive T and B cells. The disruption of immunological tolerance, characterized by the failure of these mechanisms, results in the aberrant activation of autoreactive lymphocytes that target self-tissues, culminating in the pathogenesis of autoimmune disorders. Genetic predispositions, environmental exposures, and immunoregulatory disturbances synergistically contribute to the susceptibility and initiation of autoimmune pathologies. Within the realm of immune therapies for autoimmune diseases, cytokine therapies have emerged as a specialized strategy, targeting cytokine-mediated regulatory pathways to rectify immunological imbalances. Proinflammatory cytokines are key players in inducing and propagating autoimmune inflammation, highlighting the potential of cytokine therapies in managing autoimmune conditions. This review discusses the etiology of autoimmune diseases, current therapeutic approaches, and prospects for future drug design.
Collapse
Affiliation(s)
- Farzana Yasmeen
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea; (F.Y.); (B.C.)
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongtong-gu, Suwon 16502, Republic of Korea
| | - Rameez Hassan Pirzada
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea; (F.Y.); (B.C.)
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongtong-gu, Suwon 16502, Republic of Korea
| | - Bilal Ahmad
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongtong-gu, Suwon 16502, Republic of Korea
| | - Bogeum Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea; (F.Y.); (B.C.)
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea; (F.Y.); (B.C.)
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongtong-gu, Suwon 16502, Republic of Korea
| |
Collapse
|
6
|
Karakoese Z, Ingola M, Sitek B, Dittmer U, Sutter K. IFNα Subtypes in HIV Infection and Immunity. Viruses 2024; 16:364. [PMID: 38543729 PMCID: PMC10975235 DOI: 10.3390/v16030364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/23/2024] [Accepted: 02/24/2024] [Indexed: 05/23/2024] Open
Abstract
Type I interferons (IFN), immediately triggered following most viral infections, play a pivotal role in direct antiviral immunity and act as a bridge between innate and adaptive immune responses. However, numerous viruses have evolved evasion strategies against IFN responses, prompting the exploration of therapeutic alternatives for viral infections. Within the type I IFN family, 12 IFNα subtypes exist, all binding to the same receptor but displaying significant variations in their biological activities. Currently, clinical treatments for chronic virus infections predominantly rely on a single IFNα subtype (IFNα2a/b). However, the efficacy of this therapeutic treatment is relatively limited, particularly in the context of Human Immunodeficiency Virus (HIV) infection. Recent investigations have delved into alternative IFNα subtypes, identifying certain subtypes as highly potent, and their antiviral and immunomodulatory properties have been extensively characterized. This review consolidates recent findings on the roles of individual IFNα subtypes during HIV and Simian Immunodeficiency Virus (SIV) infections. It encompasses their induction in the context of HIV/SIV infection, their antiretroviral activity, and the diverse regulation of the immune response against HIV by distinct IFNα subtypes. These insights may pave the way for innovative strategies in HIV cure or functional cure studies.
Collapse
Affiliation(s)
- Zehra Karakoese
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (Z.K.); (U.D.)
- Institute for the Research on HIV and AIDS-Associated Diseases, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Martha Ingola
- Medical Proteome Center, Ruhr University Bochum, 44801 Bochum, Germany; (M.I.); (B.S.)
| | - Barbara Sitek
- Medical Proteome Center, Ruhr University Bochum, 44801 Bochum, Germany; (M.I.); (B.S.)
- Department of Anesthesia, Intensive Care Medicine and Pain Therapy, University Hospital Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (Z.K.); (U.D.)
- Institute for the Research on HIV and AIDS-Associated Diseases, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Kathrin Sutter
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (Z.K.); (U.D.)
- Institute for the Research on HIV and AIDS-Associated Diseases, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| |
Collapse
|
7
|
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
|
8
|
Qin A, Wu CR, Ho MC, Tsai CY, Chen PJ. Sequential Therapy with Ropeginterferon Alfa-2b and Anti-Programmed Cell Death 1 Antibody for Inhibiting the Recurrence of Hepatitis B-Related Hepatocellular Carcinoma: From Animal Modeling to Phase I Clinical Results. Int J Mol Sci 2023; 25:433. [PMID: 38203603 PMCID: PMC10778875 DOI: 10.3390/ijms25010433] [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: 12/06/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Hepatocellular carcinoma (HCC) usually recurs after curative surgical resection. Currently, no approved adjuvant therapy has been shown to reduce HCC recurrence rates. In this study, the in vivo effect of sequential combination treatment with recombinant mouse interferon-alpha (rmIFN-α) and an anti-mouse-PD1 antibody on hepatitis B virus (HBV) clearance in mice was evaluated. A Phase I clinical trial was then conducted to assess the safety, tolerability, and inhibitory activity of sequential therapy with ropeginterferon alfa-2b and nivolumab in patients with HCC recurrence who underwent curative surgery for HBV-related HCC. The animal modeling study showed that HBV suppression was significantly greater with the rmIFN-α and anti-PD1 sequential combination treatment in comparison with sole treatment with rmIFN-α or anti-PD1. In the Phase I study, eleven patients completed the sequential therapy with ropeginterferon alfa-2b every two weeks for six doses at 450 µg, followed by three doses of nivolumab every two weeks up to 0.75 mg/kg. A notable decrease in or clearance of HBV surface antigen was observed in two patients. The dose-limiting toxicity of grade 3 alanine transaminase and aspartate aminotransferase increases was observed in one patient. The maximum tolerated dose was then determined. To date, no HCC recurrence has been observed. The treatment modality was well tolerated. These data support the further clinical development of sequential combination therapy as a post-surgery prophylactic measure against the recurrence of HBV-related HCC.
Collapse
Affiliation(s)
- Albert Qin
- Medical Research & Clinical Operations, PharmaEssentia Corporation, Taipei 115, Taiwan
| | - Chang-Ru Wu
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Ming-Chih Ho
- Department of Surgery, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chan-Yen Tsai
- Medical Research & Clinical Operations, PharmaEssentia Corporation, Taipei 115, Taiwan
| | - Pei-Jer Chen
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Hepatitis Research Center, National Taiwan University Hospital, Taipei 100, Taiwan
| |
Collapse
|
9
|
Karakoese Z, Le-Trilling VTK, Schuhenn J, Francois S, Lu M, Liu J, Trilling M, Hoffmann D, Dittmer U, Sutter K. Targeted mutations in IFNα2 improve its antiviral activity against various viruses. mBio 2023; 14:e0235723. [PMID: 37874130 PMCID: PMC10746204 DOI: 10.1128/mbio.02357-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 10/25/2023] Open
Abstract
IMPORTANCE The potency of interferon (IFN)α to restrict viruses was already discovered in 1957. However, until today, only IFNα2 out of the 12 distinct human IFNα subtypes has been therapeutically used against chronic viral infections. There is convincing evidence that other IFNα subtypes are far more efficient than IFNα2 against many viruses. In order to identify critical antiviral residues within the IFNα subtype sequence, we designed hybrid molecules based on the IFNα2 backbone with individual sequence motifs from the more potent subtypes IFNα6 and IFNα14. In different antiviral assays with HIV or HBV, residues binding to IFNAR1 as well as combinations of residues in the IFNAR1 binding region, the putative tunable anchor, and residues outside these regions were identified to be crucial for the antiviral activity of IFNα. Thus, we designed artificial IFNα molecules, based on the clinically approved IFNα2 backbone, but with highly improved antiviral activity against several viruses.
Collapse
Affiliation(s)
- Zehra Karakoese
- University Hospital Essen, University of Duisburg-Essen, Institute for Virology, Essen, Germany
- University Hospital Essen, University of Duisburg-Essen, Institute for Translational HIV Research, Essen, Germany
| | | | - Jonas Schuhenn
- University Hospital Essen, University of Duisburg-Essen, Institute for Virology, Essen, Germany
| | - Sandra Francois
- University Hospital Essen, University of Duisburg-Essen, Institute for Virology, Essen, Germany
| | - Mengji Lu
- University Hospital Essen, University of Duisburg-Essen, Institute for Virology, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Liu
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, China
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mirko Trilling
- University Hospital Essen, University of Duisburg-Essen, Institute for Virology, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, China
| | - Daniel Hoffmann
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, China
- Research Group Bioinformatics, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Ulf Dittmer
- University Hospital Essen, University of Duisburg-Essen, Institute for Virology, Essen, Germany
- University Hospital Essen, University of Duisburg-Essen, Institute for Translational HIV Research, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, China
| | - Kathrin Sutter
- University Hospital Essen, University of Duisburg-Essen, Institute for Virology, Essen, Germany
- University Hospital Essen, University of Duisburg-Essen, Institute for Translational HIV Research, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Tanaka A, Hayano S, Nagata M, Kosami T, Wang Z, Kamioka H. Ruxolitinib altered IFN-β induced necroptosis of human dental pulp stem cells during osteoblast differentiation. Arch Oral Biol 2023; 155:105797. [PMID: 37633030 DOI: 10.1016/j.archoralbio.2023.105797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/01/2023] [Accepted: 08/21/2023] [Indexed: 08/28/2023]
Abstract
OBJECTIVE This study aimed to evaluate the role of ruxolitinib in the interferon beta (IFN-β) mediated osteoblast differentiation using human dental pulp stem cells (hDPSCs). DESIGN hDPSCs from five deciduous teeth of healthy patients were stimulated by adding human recombinant IFN-β protein (1 or 2 ng/ml) to the osteogenic differentiation induction medium. Substrate formation was determined using Alizarin Red staining, calcium concentration, and osteoblast marker expression levels. Ruxolitinib was used to inhibit the Janus kinase/signal transducers and activators of transcription (JAK-STAT) pathway. Apoptosis was detected using terminal deoxynucleotidyl nick-end labeling (TUNEL) staining, and necroptosis was detected using propidium iodide staining and phosphorylated mixed lineage kinase domain-like protein (pMLKL) expression. RESULTS In the IFN-β-treated group, substrate formation was inhibited by a reduction in alkaline phosphatase (ALP) expression in a concentration-dependent manner. Although the proliferation potency was unchanged between the IFN-β-treated and control groups, the cell number was significantly reduced in the experimental group. TUNEL-positive cell number was not significantly different; however, the protein level of necroptosis markers, interleukin-6 (IL-6) and pMLKL were significantly increased in the substrate formation. Cell number and ALP expression level were improved in the group administered ruxolitinib, a JAK-STAT inhibitor. Additionally, ruxolitinib significantly suppressed IL-6 and pMLKL levels. CONCLUSION Ruxolitinib interfered with the IFN-β-mediated necroptosis and osteogenic differentiation via the JAK-STAT pathway.
Collapse
Affiliation(s)
- Atsuko Tanaka
- Department of Orthodontics, Okayama University Hospital, Okayama, Japan
| | - Satoru Hayano
- Department of Orthodontics, Okayama University Hospital, Okayama, Japan.
| | - Masayo Nagata
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takahiro Kosami
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ziyi Wang
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan; Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Hiroshi Kamioka
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
11
|
Ramirez D, B Chuong E, D Dowell R. Nascent transcription upon interferon-α2 stimulation on human and rhesus macaque lymphoblastoid cell lines. BMC Res Notes 2023; 16:292. [PMID: 37885027 PMCID: PMC10604760 DOI: 10.1186/s13104-023-06465-1] [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: 10/04/2022] [Accepted: 08/18/2023] [Indexed: 10/28/2023] Open
Abstract
OBJECTIVES The interferon-triggered innate immune response has been observed to be under strong diversifying selection to counteract the many pathogens hosts have to defend against. In particular, rewiring of gene transcription regulation allows organisms to rapidly acquire new phenotypes by removing and adding genes into the innate immune gene network. Dissecting the molecular processes by which this rewiring takes place, either by changing the DNA regulatory elements or by changing the activity of the regulators across species, is key to better understand this evolutionary process. DATA DESCRIPTION To better comprehend the evolutionary dynamics that have occurred in the initial transcriptional response to interferon in primates, we present Precision Run-On (PRO-seq) datasets made after 1 h of interferon-α2 stimulation on human and rhesus macaque lymphoblastoid cell lines. Further, we tested the difference between using either species' cognate interferon versus using the other orthologous interferon to account for any potential impacts in the interaction of the orthologous interferons with their cellular membrane receptors. This data provides insights into the regulatory mechanisms that drive species-specific responses to environmental perturbations, such as the one driven by the interactions of pathogens and their hosts.
Collapse
Affiliation(s)
- Daniel Ramirez
- Department of Molecular, Cellular, and Developmental Biology; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Edward B Chuong
- Department of Molecular, Cellular, and Developmental Biology; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Robin D Dowell
- Department of Molecular, Cellular, and Developmental Biology; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA.
| |
Collapse
|
12
|
Noe P, Wang JH, Chung K, Cheng Z, Field JJ, Shen X, Cortesio CL, Pastuskovas CV, Phee H, Tarbell KV, Egen JG, Casbon AJ. Therapeutically targeting type I interferon directly to XCR1+ dendritic cells reveals the role of cDC1s in anti-drug antibodies. Front Immunol 2023; 14:1272055. [PMID: 37942313 PMCID: PMC10628189 DOI: 10.3389/fimmu.2023.1272055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023] Open
Abstract
Conventional type 1 dendritic cells (cDC1s) are superior in antigen cross-presentation and priming CD8+ T cell anti-tumor immunity and thus, are a target of high interest for cancer immunotherapy. Type I interferon (IFN) is a potent inducer of antigen cross-presentation, but, unfortunately, shows only modest results in the clinic given the short half-life and high toxicity of current type I IFN therapies, which limit IFN exposure in the tumor. CD8+ T cell immunity is dependent on IFN signaling in cDC1s and preclinical studies suggest targeting IFN directly to cDC1s may be sufficient to drive anti-tumor immunity. Here, we engineered an anti-XCR1 antibody (Ab) and IFN mutein (IFNmut) fusion protein (XCR1Ab-IFNmut) to determine whether systemic delivery could drive selective and sustained type I IFN signaling in cDC1s leading to anti-tumor activity and, in parallel, reduced systemic toxicity. We found that the XCR1Ab-IFNmut fusion specifically enhanced cDC1 activation in the tumor and spleen compared to an untargeted control IFN. However, multiple treatments with the XCR1Ab-IFNmut fusion resulted in robust anti-drug antibodies (ADA) and loss of drug exposure. Using other cDC1-targeting Ab-IFNmut fusions, we found that localizing IFN directly to cDC1s activates their ability to promote ADA responses, regardless of the cDC1 targeting antigen. The development of ADA remains a major hurdle in immunotherapy drug development and the cellular and molecular mechanisms governing the development of ADA responses in humans is not well understood. Our results reveal a role of cDC1s in ADA generation and highlight the potential ADA challenges with targeting immunostimulatory agents to this cellular compartment.
Collapse
Affiliation(s)
- Paul Noe
- Oncology Research, Amgen Research, South San Francisco, CA, United States
| | - Joy H. Wang
- Oncology Research, Amgen Research, South San Francisco, CA, United States
| | - Kyu Chung
- Oncology Research, Amgen Research, South San Francisco, CA, United States
| | - Zhiyong Cheng
- Oncology Research, Amgen Research, South San Francisco, CA, United States
| | - Jessica J. Field
- Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, CA, United States
| | - Xiaomeng Shen
- Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, CA, United States
| | - Christa L. Cortesio
- Therapeutics Discovery, Amgen Research, South San Francisco, CA, United States
| | - Cinthia V. Pastuskovas
- Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, CA, United States
| | - Hyewon Phee
- Oncology Research, Amgen Research, South San Francisco, CA, United States
| | - Kristin V. Tarbell
- Oncology Research, Amgen Research, South San Francisco, CA, United States
| | - Jackson G. Egen
- Oncology Research, Amgen Research, South San Francisco, CA, United States
| | - Amy-Jo Casbon
- Oncology Research, Amgen Research, South San Francisco, CA, United States
| |
Collapse
|
13
|
Yan L, Zhang Y, Wang P, Zhao C, Zhang B, Qiu L. Antiviral functions of IFNd against ISKNV and interaction analysis of IFNd and its receptors in spotted seabass (Lateolabrax maculatus). FISH & SHELLFISH IMMUNOLOGY 2023; 140:108935. [PMID: 37454880 DOI: 10.1016/j.fsi.2023.108935] [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: 05/18/2023] [Revised: 07/03/2023] [Accepted: 07/03/2023] [Indexed: 07/18/2023]
Abstract
Type I interferons (IFNs) play a significant role in antiviral innate immunity. But, the antiviral function of IFNd is controversial in teleosts. Here, we identified three IFNd receptors belonging to cytokine receptor family B (LmCRFB1, LmCRFB2, and LmCRFB5) in spotted seabass (Lateolabrax maculatus). LmIFNd and its receptors were highly expressed in gill, spleen and head kidney tissues. Additionally, LmIFNd, its receptors, and their downstream signal genes (LmTYK2, LmJAK1, LmSTAT1, and LmSTAT2) were induced by infectious spleen and kidney necrosis virus (ISKNV) infection. Injection of recombinant protein (LmIFNd-His) in vivo and incubation with the LmIFNd-His in vitro both induced expressions of IFN-stimulated genes (LmISGs). IFNd-His had a dose-dependent protective effect on the activity of brain cells infected by ISKNV and reduced the number of ISNKV copies. LmIFNd-His also bound to extracellular domains of the three receptors in vitro in the pull-down assay. LmIFNd-His preferentially induced ISG expression through receptor complex LmCRFB1 and LmCRFB5, followed by LmCRFB2 and LmCRFB5, to induce the expressions of LmISGs. Our results show that LmIFNd can enhance the antiviral immune response of spotted seabass, and it uses receptor complex LmCRFB1 and LmCRFB5 as well as LmCRFB2 and LmCRFB5 to induce LmISG expression. It is the first study about the antiviral function of LmIFNd and its receptor complex in spotted seabass, and it provides a reference for further studies of the controversial anti-viral function of IFNd in teleosts.
Collapse
Affiliation(s)
- Lulu Yan
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Sanya Tropical Fisheries Research Institute, Sanya, China
| | - Yaqing Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Pengfei Wang
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Sanya Tropical Fisheries Research Institute, Sanya, China
| | - Chao Zhao
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Sanya Tropical Fisheries Research Institute, Sanya, China
| | - Bo Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Sanya Tropical Fisheries Research Institute, Sanya, China
| | - Lihua Qiu
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Sanya Tropical Fisheries Research Institute, Sanya, China; Key Laboratory of Aquatic Genomics, Ministry of Agriculture and Rural Affairs, Chinese Academy of Fishery Science, Beijing, China.
| |
Collapse
|
14
|
Wang C, Du Z, Li R, Luo Y, Zhu C, Ding N, Lei A. Interferons as negative regulators of ILC2s in allergic lung inflammation and respiratory viral infections. J Mol Med (Berl) 2023; 101:947-959. [PMID: 37414870 DOI: 10.1007/s00109-023-02345-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/08/2023]
Abstract
Group 2 innate lymphoid cells (ILC2s), characterized by a lack of antigen receptors, have been regarded as an important component of type 2 pulmonary immunity. Analogous to Th2 cells, ILC2s are capable of releasing type 2 cytokines and amphiregulin, thus playing an essential role in a variety of diseases, such as allergic diseases and virus-induced respiratory diseases. Interferons (IFNs), an important family of cytokines with potent antiviral effects, can be triggered by microbial products, microbial exposure, and pathogen infections. Interestingly, the past few years have witnessed encouraging progress in revealing the important role of IFNs and IFN-producing cells in modulating ILC2 responses in allergic lung inflammation and respiratory viral infections. This review underscores recent progress in understanding the role of IFNs and IFN-producing cells in shaping ILC2 responses and discusses disease phenotypes, mechanisms, and therapeutic targets in the context of allergic lung inflammation and infections with viruses, including influenza virus, rhinovirus (RV), respiratory syncytial virus (RSV), and severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2).
Collapse
Affiliation(s)
- Cui Wang
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Zhaoxiang Du
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Ranhui Li
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Ying Luo
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Cuiming Zhu
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Nan Ding
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Aihua Lei
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China.
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China.
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China.
| |
Collapse
|
15
|
Valdés-López JF, Urcuqui-Inchima S. Antiviral response and immunopathogenesis of interleukin 27 in COVID-19. Arch Virol 2023; 168:178. [PMID: 37310504 DOI: 10.1007/s00705-023-05792-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 04/11/2023] [Indexed: 06/14/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is associated with a high mortality rate. The clinical course is attributed to the severity of pneumonia and systemic complications. In COVID-19 patients and murine models of SARS-CoV-2 infection, the disease may be accompanied by excessive production of cytokines, leading to an accumulation of immune cells in affected organs such as lungs. Previous reports have shown that SARS-CoV-2 infection antagonizes interferon (IFN)-dependent antiviral response, thereby preventing the expression of IFN-stimulated genes (ISGs). Lower IFN levels have been linked to more-severe COVID-19. Interleukin 27 (IL27) is a heterodimeric cytokine composed of IL27p28 and EBI3 subunits, which induce both pro- and anti-inflammatory responses. Recently, we and others have reported that IL27 also induces a strong antiviral response in an IFN-independent manner. Here, we investigated transcription levels of both IL27 subunits in COVID-19 patients. The results show that SARS-CoV-2 infection modulates TLR1/2-MyD88 signaling in PBMCs and monocytes and induces NF-κB activation and expression of NF-κB-target genes that are dependent on a robust pro-inflammatory response, including EBI3; and activates IRF1 signaling which induces IL27p28 mRNA expression. The results suggest that IL27 induces a robust STAT1-dependent pro-inflammatory and antiviral response in an IFN-independent manner in COVID-derived PBMCs and monocytes as a function of a severe clinical course of COVID-19. Similar results were observed in macrophages stimulated with the SARS-CoV-2 spike protein. Thus, IL27 can trigger an antiviral response in the host, suggesting the possibility of novel therapeutics against SARS-CoV-2 infection in humans.
Collapse
Affiliation(s)
- Juan Felipe Valdés-López
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| |
Collapse
|
16
|
Deng YH, Li B, Chen SN, Ren L, Zhang BD, Liu LH, Liu S, Nie P. Molecular characterization of nineteen cytokine receptor family B (CRFB) members, CRFB1, CRFB2, CRFB4-17, with three CRFB9 and two CRFB14 in a cyprinid fish, the blunt snout bream Megalobrama amblycephala. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 145:104725. [PMID: 37146740 DOI: 10.1016/j.dci.2023.104725] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/03/2023] [Accepted: 05/03/2023] [Indexed: 05/07/2023]
Abstract
The class II cytokine receptor family members are receptors of class 2 helical cytokines in mammals, and are named cytokine receptor family B (CRFB) in fish. In zebrafish, sixteen members, including CRFB1, CRFB2 and CRFB4-17 were reported. With the availability of genome sequence, a total of nineteen CRFBs was identified in the blunt snout bream (Megalobrama amblycephala), including CRFB1, CRFB2, CRFB4-17 with the presence of three CRFB9 isoforms, and two CRFB14 isoforms. These CRFB molecules contain well conserved features, such as fibronectin type III (FNIII) domain, transmembrane and intracellular domains as other class II cytokine receptors, and are phylogenetically grouped into thirteen clades with their homologues from other species of fish. The CRFB genes were constitutively expressed in organs/tissues examined in the fish. The finding of more CRFB members in the bream may provide clues to understand possible receptor-ligand interaction and their diversity from an evolutionary point of view.
Collapse
Affiliation(s)
- Yu Hang Deng
- State Key Laboratory of Freshwater Ecology and Biotechnology, and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Bo Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China; School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - Shan Nan Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China
| | - Li Ren
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan Province, China
| | - Bai Dong Zhang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - Lan Hao Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - Shaojun Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan Province, China.
| | - P Nie
- State Key Laboratory of Freshwater Ecology and Biotechnology, and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China; School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong Province, 266237, China; The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, China.
| |
Collapse
|
17
|
McFarlane A, Pohler E, Moraga I. Molecular and cellular factors determining the functional pleiotropy of cytokines. FEBS J 2023; 290:2525-2552. [PMID: 35246947 PMCID: PMC10952290 DOI: 10.1111/febs.16420] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/26/2022] [Accepted: 03/03/2022] [Indexed: 11/30/2022]
Abstract
Cytokines are soluble factors vital for mammalian physiology. Cytokines elicit highly pleiotropic activities, characterized by their ability to induce a wide spectrum of functional responses in a diverse range of cell subsets, which makes their study very challenging. Cytokines activate signalling via receptor dimerization/oligomerization, triggering activation of the JAK (Janus kinase)/STAT (signal transducer and activator of transcription) signalling pathway. Given the strong crosstalk and shared usage of key components of cytokine signalling pathways, a long-standing question in the field pertains to how functional diversity is achieved by cytokines. Here, we discuss how biophysical - for example, ligand-receptor binding affinity and topology - and cellular - for example, receptor, JAK and STAT protein levels, endosomal compartment - parameters contribute to the modulation and diversification of cytokine responses. We review how these parameters ultimately converge into a common mechanism to fine-tune cytokine signalling that involves the control of the number of Tyr residues phosphorylated in the receptor intracellular domain upon cytokine stimulation. This results in different kinetics of STAT activation, and induction of specific gene expression programs, ensuring the generation of functional diversity by cytokines using a limited set of signalling intermediaries. We describe how these first principles of cytokine signalling have been exploited using protein engineering to design cytokine variants with more specific and less toxic responses for immunotherapy.
Collapse
Affiliation(s)
- Alison McFarlane
- Division of Cell Signalling and ImmunologySchool of Life SciencesUniversity of DundeeUK
| | - Elizabeth Pohler
- Division of Cell Signalling and ImmunologySchool of Life SciencesUniversity of DundeeUK
| | - Ignacio Moraga
- Division of Cell Signalling and ImmunologySchool of Life SciencesUniversity of DundeeUK
| |
Collapse
|
18
|
Qin A. An anti-cancer surveillance by the interplay between interferon-beta and retinoblastoma protein RB1. Front Oncol 2023; 13:1173467. [PMID: 37182173 PMCID: PMC10174298 DOI: 10.3389/fonc.2023.1173467] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/05/2023] [Indexed: 05/16/2023] Open
Abstract
Interferon-beta (IFN-β), an extracellular cytokine that initiates signaling pathways for gene regulation, has been demonstrated to function as a tumor suppressor protein through lentiviral gene transduction. In this article, I review the relevant previous works and propose a cell cycle-based, tumor suppressor protein-mediated mechanism of anti-cancer surveillance. IFN-β induces a tumor cell cycle alteration that leads to S phase accumulation, senescence entry, and a loss of tumorigenicity in solid tumor cells. IFN-β does not show a significant cell cycle effect in their normal counterparts. Retinoblastoma protein RB1, another tumor suppressor protein, tightly controls the cell cycle and differentiation of normal cells, preventing them from being significantly impacted by the IFN-β effect. The interplay between IFN-β and RB1 acts as a mechanism of cell cycle-based, tumor suppressor protein-mediated anti-cancer surveillance that can selectively suppress solid tumor or proliferating transformed cells from the loss of control leading to cancer. This mechanism has important implications for the treatment of solid tumors.
Collapse
Affiliation(s)
- Albert Qin
- Medical Research & Clinical Operations, PharmaEssentia Corporation, Taipei, Taiwan
| |
Collapse
|
19
|
Eskandari SK, Allos H, Safadi JM, Sulkaj I, Sanders JSF, Cravedi P, Ghobrial IM, Berger SP, Azzi JR. Type I interferons augment regulatory T cell polarization in concert with ancillary cytokine signals. FRONTIERS IN TRANSPLANTATION 2023; 2:1149334. [PMID: 38993887 PMCID: PMC11235373 DOI: 10.3389/frtra.2023.1149334] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 03/24/2023] [Indexed: 07/13/2024]
Abstract
In the transplant community, research efforts exploring endogenous alternatives to inducing tolerogenic allo-specific immune responses are much needed. In this regard, CD4 + FoxP3+ regulatory T cells (Tregs) are appealing candidates due to their intrinsic natural immunosuppressive qualities. To date, various homeostatic factors that dictate Treg survival and fitness have been elucidated, particularly the non-redundant roles of antigenic CD3ζ/T-cell-receptor, co-stimulatory CD28, and cytokine interleukin (IL-)2 dependent signaling. Many of the additional biological signals that affect Tregs remain to be elucidated, however, especially in the transplant context. Previously, we demonstrated an unexpected link between type I interferons (IFNs) and Tregs in models of multiple myeloma (MM)-where MM plasmacytes escaped immunological surveillance by enhancing type I IFN signaling and precipitating upregulated Treg responses that could be overturned with specific knockdown of type I IFN signaling. Here, we elaborated on these findings by assessing the role of type I IFN signaling (IFN-α and -β) on Treg homeostasis within an alloimmune context. Specifically, we studied the induction of Tregs from naïve CD4 T cells. Using in vitro and in vivo models of murine skin allotransplantation, we found that type I IFN indeed spatiotemporally enhanced the polarization of naïve CD4 T cells into FoxP3+ Tregs. Notably, however, this effect was not independent of, and rather co-dependent on, ancillary cytokine signals including IL-2. These findings provide evidence for the relevance of type I IFN pathway in modulating FoxP3+ Treg responses and, by extension, stipulate an additional means of facilitating Treg fitness via type I IFNs.
Collapse
Affiliation(s)
- Siawosh K. Eskandari
- Transplantation Research Center, Division of Nephrology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Hazim Allos
- Transplantation Research Center, Division of Nephrology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Jenelle M. Safadi
- Transplantation Research Center, Division of Nephrology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ina Sulkaj
- Transplantation Research Center, Division of Nephrology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Graduate Program in Immunology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Jan S. F. Sanders
- Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Paolo Cravedi
- Translational Transplant Research Center, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Irene M. Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Stefan P. Berger
- Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jamil R. Azzi
- Transplantation Research Center, Division of Nephrology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
20
|
Zanin N, Viaris de Lesegno C, Podkalicka J, Meyer T, Gonzalez Troncoso P, Bun P, Danglot L, Chmiest D, Urbé S, Piehler J, Blouin CM, Lamaze C. STAM and Hrs interact sequentially with IFN-α Receptor to control spatiotemporal JAK-STAT endosomal activation. Nat Cell Biol 2023; 25:425-438. [PMID: 36797476 DOI: 10.1038/s41556-022-01085-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/21/2022] [Indexed: 02/18/2023]
Abstract
Activation of the JAK-STAT pathway by type I interferons (IFNs) requires clathrin-dependent endocytosis of the IFN-α and -β receptor (IFNAR), indicating a role for endosomal sorting in this process. The molecular machinery that brings the selective activation of IFN-α/β-induced JAK-STAT signalling on endosomes remains unknown. Here we show that the constitutive association of STAM with IFNAR1 and TYK2 kinase at the plasma membrane prevents TYK2 activation by type I IFNs. IFN-α-stimulated IFNAR endocytosis delivers the STAM-IFNAR complex to early endosomes where it interacts with Hrs, thereby relieving TYK2 inhibition by STAM and triggering signalling of IFNAR at the endosome. In contrast, when stimulated by IFN-β, IFNAR signalling occurs independently of Hrs as IFNAR is sorted to a distinct endosomal subdomain. Our results identify the molecular machinery that controls the spatiotemporal activation of IFNAR by IFN-α and establish the central role of endosomal sorting in the differential regulation of JAK-STAT signalling by IFN-α and IFN-β.
Collapse
Affiliation(s)
- Natacha Zanin
- Membrane Mechanics and Dynamics of Intracellular Signaling Laboratory, Institut Curie-Centre de Recherche, PSL Research University, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Centre National de la Recherche Scientifique (CNRS), Paris, France.,Namur Research Institute for Life Sciences (NARILIS), URBC, University of Namur, Namur, Belgium
| | - Christine Viaris de Lesegno
- Membrane Mechanics and Dynamics of Intracellular Signaling Laboratory, Institut Curie-Centre de Recherche, PSL Research University, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Joanna Podkalicka
- Membrane Mechanics and Dynamics of Intracellular Signaling Laboratory, Institut Curie-Centre de Recherche, PSL Research University, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Centre National de la Recherche Scientifique (CNRS), Paris, France.,Laboratoire Physico-Chimie Curie, Institut Curie, PSL Research University, Sorbonne Université, Paris, France.,Laboratory of Cytobiochemistry, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | - Thomas Meyer
- Department of Biology and Center for Cellular Nanoanalytics, University of Osnabruck, Osnabruck, Germany
| | - Pamela Gonzalez Troncoso
- Membrane Mechanics and Dynamics of Intracellular Signaling Laboratory, Institut Curie-Centre de Recherche, PSL Research University, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Philippe Bun
- Membrane Traffic in Healthy and Diseased Brain, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Université de Paris, Paris, France.,NeurImag Imaging Facility, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Université de Paris, Paris, France
| | - Lydia Danglot
- Membrane Traffic in Healthy and Diseased Brain, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Université de Paris, Paris, France.,NeurImag Imaging Facility, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Université de Paris, Paris, France
| | - Daniela Chmiest
- Membrane Mechanics and Dynamics of Intracellular Signaling Laboratory, Institut Curie-Centre de Recherche, PSL Research University, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Centre National de la Recherche Scientifique (CNRS), Paris, France.,Department of Biochemistry, CIIL Biomedical Research Center, University of Lausanne, Epalinges, Switzerland
| | - Sylvie Urbé
- Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Jacob Piehler
- Department of Biology and Center for Cellular Nanoanalytics, University of Osnabruck, Osnabruck, Germany
| | - Cédric M Blouin
- Membrane Mechanics and Dynamics of Intracellular Signaling Laboratory, Institut Curie-Centre de Recherche, PSL Research University, Paris, France. .,Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France. .,Centre National de la Recherche Scientifique (CNRS), Paris, France.
| | - Christophe Lamaze
- Membrane Mechanics and Dynamics of Intracellular Signaling Laboratory, Institut Curie-Centre de Recherche, PSL Research University, Paris, France. .,Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France. .,Centre National de la Recherche Scientifique (CNRS), Paris, France.
| |
Collapse
|
21
|
Hernández-Sarmiento LJ, Valdés-López JF, Urcuqui-Inchima S. American-Asian- and African lineages of Zika virus induce differential pro-inflammatory and Interleukin 27-dependent antiviral responses in human monocytes. Virus Res 2023; 325:199040. [PMID: 36610657 PMCID: PMC10194209 DOI: 10.1016/j.virusres.2023.199040] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 01/06/2023]
Abstract
Zika virus (ZIKV) is an arbovirus that belongs to the Flaviviridae family and inflammatory responses play a critical role in ZIKV pathogenesis. As a first-line defense, monocytes are key components of innate immunity and host response to viruses. Monocytes are considered the earliest blood cell type to be infected by ZIKV and have been shown to be associated with ZIKV pathogenesis. The first ZIKV epidemic was reported in Africa and Asia although, it is less well known whether African- and Asian- lineages of ZIKV have different impacts on host immune response. We studied the pro-inflammatory and antiviral response of ZIKV-infected monocytes using publicly available RNA-seq analysis (GSE103114). We compared the transcriptomic profiles of human monocytes infected with ZIKV Puerto Rico strain (PRVABC59), American-Asian lineage, and ZIKV Nigeria strain (IBH30656), African lineage. We validated RNA-seq results by ELISA or RT-qPCR, in human monocytes infected with a clinical isolate of ZIKV from Colombia (American-Asian lineage), or with ZIKV from Dakar (African lineage). The transcriptomic analysis showed that ZIKV Puerto Rico strain promotes a higher pro-inflammatory response through TLR2 signaling and NF-kB activation and induces a strong IL27-dependent antiviral activity than ZIKV Nigeria strain. Furthermore, human monocytes are more susceptible to infection with ZIKV from Colombia than ZIKV from Dakar. Likewise, Colombian ZIKV isolate activated IL27 signaling and induced a robust antiviral response in an IFN-independent manner. Moreover, we show that treatment of monocytes with IL27 results in decreased release of ZIKV particles in a dose-dependent manner with an EC50 =2.870 ng/mL for ZIKV from Colombia and EC50 =10.23 ng/mL to ZIKV from Dakar. These findings highlight the differential inflammatory response and antiviral activity of monocytes infected with different lineages of ZIKV and may help better management of ZIKV-infected patients.
Collapse
Affiliation(s)
| | - Juan Felipe Valdés-López
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| |
Collapse
|
22
|
Emerging principles of cytokine pharmacology and therapeutics. Nat Rev Drug Discov 2023; 22:21-37. [PMID: 36131080 DOI: 10.1038/s41573-022-00557-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2022] [Indexed: 01/10/2023]
Abstract
Cytokines are secreted signalling proteins that play essential roles in the initiation, maintenance and resolution of immune responses. Although the unique ability of cytokines to control immune function has garnered clinical interest in the context of cancer, autoimmunity and infectious disease, the use of cytokine-based therapeutics has been limited. This is due, in part, to the ability of cytokines to act on many cell types and impact diverse biological functions, resulting in dose-limiting toxicity or lack of efficacy. Recent studies combining structural biology, protein engineering and receptor pharmacology have unlocked new insights into the mechanisms of cytokine receptor activation, demonstrating that many aspects of cytokine function are highly tunable. Here, we discuss the pharmacological principles underlying these efforts to overcome cytokine pleiotropy and enhance the therapeutic potential of this important class of signalling molecules.
Collapse
|
23
|
Wang Y, Liu M, Guo X, Zhang B, Li H, Liu Y, Han J, Jia L, Li L. Endogenous Retrovirus Elements Are Co-Expressed with IFN Stimulation Genes in the JAK-STAT Pathway. Viruses 2022; 15:60. [PMID: 36680099 PMCID: PMC9861321 DOI: 10.3390/v15010060] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Background: Endogenous retrovirus (ERV) elements can act as proximal regulatory elements in promoting interferon (IFN) responses. Previous relevant studies have mainly focused on IFN-stimulated genes (ISGs). However, the role of ERV elements as cis-regulatory motifs in regulating genes of the JAK-STAT pathway remains poorly understood. In our study, we analyzed the changes in ERV elements and genes under both IFN stimulation and blockade of the signaling pathway. Methods: The effects of interferon on cells under normal conditions and knockout of the receptor were compared based on the THP1_IFNAR1_KO and THP1_IFNAR2_mutant cell lines. The correlation between differentially expressed ERVs (DHERVs) and differentially expressed genes (DEGs) as DEHERV-G pairs was explored with construction of gene regulatory networks related to ERV and induced by proinflammatory cytokines. Results: A total of 430 DEHERV loci and 190 DEGs were identified in 842 DEHERV-G pairs that are common to the three groups. More than 87% of DEHERV-G pairs demonstrated a consistent expression pattern. ISGs such as AIM2, IFIT1, IFIT2, IFIT3, STAT1, and IRF were activated via the JAK-STAT pathway in response to interferon stimulation. Thus, STAT1, STAT2, and IRF1 appear to play core roles in regulatory networks and are closely associated with ERVs. Conclusions: The RNA expression of ISGs and ERV elements is correlated, indicating that ERV elements are closely linked to host innate immune responses.
Collapse
Affiliation(s)
- Yanglan Wang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Mengying Liu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xing Guo
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing 100071, China
- Department of Microbiology, School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Bohan Zhang
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing 100071, China
| | - Hanping Li
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing 100071, China
| | - Yongjian Liu
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing 100071, China
| | - Jingwan Han
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing 100071, China
| | - Lei Jia
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing 100071, China
| | - Lin Li
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing 100071, China
| |
Collapse
|
24
|
Harrer DC, Schenkel C, Bezler V, Kaljanac M, Hartley J, Barden M, Pan H, Holzinger A, Herr W, Abken H. CAR Triggered Release of Type-1 Interferon Limits CAR T-Cell Activities by an Artificial Negative Autocrine Loop. Cells 2022; 11:cells11233839. [PMID: 36497099 PMCID: PMC9737386 DOI: 10.3390/cells11233839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
The advent of chimeric antigen receptor (CAR) T cells expedited the field of cancer immunotherapy enabling durable remissions in patients with refractory hematological malignancies. T cells redirected for universal cytokine-mediated killing (TRUCKs), commonly referred to as "fourth generation" CAR T-cells, are designed to release engineered payloads upon CAR-induced T-cell activation. Building on the TRUCK technology, we aimed to generate CAR T-cells with a CAR-inducible artificial, self-limiting autocrine loop. To this end, we engineered CAR T-cells with CAR triggered secretion of type-1 interferons (IFNs). At baseline, IFNα and IFNβ CAR T-cells showed similar capacities in cytotoxicity and cytokine secretion compared to conventional CAR T-cells. However, under "stress" conditions of repetitive rounds of antigen stimulation using BxPC-3 pancreas carcinoma cells as targets, anti-tumor activity faded in later rounds while being fully active in destructing carcinoma cells during first rounds of stimulation. Mechanistically, the decline in activity was primarily based on type-1 IFN augmented CAR T-cell apoptosis, which was far less the case for CAR T-cells without IFN release. Such autocrine self-limiting loops can be used for applications where transient CAR T-cell activity and persistence upon target recognition is desired to avoid lasting toxicities.
Collapse
Affiliation(s)
- Dennis Christoph Harrer
- Department Hematology and Internal Oncology, University Hospital Regensburg, 93053 Regensburg, Germany
- Leibniz Institute for Immunotherapy, Division Genetic Immunotherapy, and Chair for Genetic Immunotherapy, University Regensburg, 93053 Regensburg, Germany
- Correspondence:
| | - Charlotte Schenkel
- Department Hematology and Internal Oncology, University Hospital Regensburg, 93053 Regensburg, Germany
- Leibniz Institute for Immunotherapy, Division Genetic Immunotherapy, and Chair for Genetic Immunotherapy, University Regensburg, 93053 Regensburg, Germany
| | - Valerie Bezler
- Leibniz Institute for Immunotherapy, Division Genetic Immunotherapy, and Chair for Genetic Immunotherapy, University Regensburg, 93053 Regensburg, Germany
| | - Marcell Kaljanac
- Leibniz Institute for Immunotherapy, Division Genetic Immunotherapy, and Chair for Genetic Immunotherapy, University Regensburg, 93053 Regensburg, Germany
| | - Jordan Hartley
- Leibniz Institute for Immunotherapy, Division Genetic Immunotherapy, and Chair for Genetic Immunotherapy, University Regensburg, 93053 Regensburg, Germany
| | - Markus Barden
- Leibniz Institute for Immunotherapy, Division Genetic Immunotherapy, and Chair for Genetic Immunotherapy, University Regensburg, 93053 Regensburg, Germany
| | - Hong Pan
- Leibniz Institute for Immunotherapy, Division Genetic Immunotherapy, and Chair for Genetic Immunotherapy, University Regensburg, 93053 Regensburg, Germany
| | - Astrid Holzinger
- Leibniz Institute for Immunotherapy, Division Genetic Immunotherapy, and Chair for Genetic Immunotherapy, University Regensburg, 93053 Regensburg, Germany
| | - Wolfgang Herr
- Department Hematology and Internal Oncology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Hinrich Abken
- Leibniz Institute for Immunotherapy, Division Genetic Immunotherapy, and Chair for Genetic Immunotherapy, University Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
25
|
Cytokine pathway variants modulate platelet production: IFNA16 is a thrombocytosis susceptibility locus in humans. Blood Adv 2022; 6:4884-4900. [PMID: 35381074 PMCID: PMC9631663 DOI: 10.1182/bloodadvances.2021005648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 03/09/2022] [Indexed: 02/08/2023] Open
Abstract
Inflammatory stimuli have divergent effects on peripheral platelet counts, although the mechanisms of thrombocytopenic and thrombocytotic responses remain poorly understood. A candidate gene approach targeting 326 polymorphic genes enriched in thrombopoietic and cytokine signaling pathways was applied to identify single nucleotide variants (SNVs) implicated in enhanced platelet responses in cohorts with reactive thrombocytosis (RT) or essential (myeloproliferative neoplasm [MPN]) thrombocytosis (ET). Cytokine profiles incorporating a 15-member subset, pathway topology, and functional interactive networks were distinct between ET and RT, consistent with distinct regulatory pathways of exaggerated thrombopoiesis. Genetic studies using aggregate (ET + RT) or ET-restricted cohorts identified associations with 2 IFNA16 (interferon-α16) SNVs, and the ET associations were validated in a second independent cohort (P = .0002). Odds ratio of the combined ET cohort (n = 105) was 4.92, restricted to the JAK2V617F-negative subset (odds ratio, 5.01). ET substratification analysis by variant IFNA16 exhibited a statistically significant increase in IFN-α16 levels (P = .002) among 16 quantifiable cytokines. Recombinantly expressed variant IFN-α16 encompassing 3 linked non-synonymous SNVs (E65H95P133) retained comparable antiviral and pSTAT signaling profiles as native IFN-α16 (V65D95A133) or IFN-α2, although both native and variant IFN-α16 showed stage-restricted differences (compared with IFN-α2) of IFN-regulated genes in CD34+-stimulated megakaryocytes. These data implicate IFNA16 (IFN-α16 gene product) as a putative susceptibility locus (driver) within the broader disrupted cytokine network evident in MPNs, and they provide a framework for dissecting functional interactive networks regulating stress or MPN thrombopoiesis.
Collapse
|
26
|
Chen J, Guan Y, Guan H, Mu Y, Ding Y, Zou J, Ouyang S, Chen X. Molecular and Structural Basis of Receptor Binding and Signaling of a Fish Type I IFN with Three Disulfide Bonds. THE JOURNAL OF IMMUNOLOGY 2022; 209:806-819. [DOI: 10.4049/jimmunol.2200202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/15/2022] [Indexed: 01/04/2023]
Abstract
Abstract
In mammals, type I IFNs, which commonly contain one or two disulfide bonds, activate the JAK-STAT signaling pathway through binding to the common cell surface receptor formed by IFN-α/β receptor (IFNAR)1 and IFNAR2 subunits. Although type I IFNs are also known to be essential for antiviral defense in teleost fish, very little is known about mechanisms underlying the recognition of fish type I IFNs by associated receptors. In this study, we demonstrate that a type I IFN of large yellow croaker Larimichthys crocea (LcIFNi), belonging to a new subgroup of fish type I IFNs, triggers antiviral response via the conserved JAK-STAT pathway through stable binding with a heterodimeric receptor comprising subunits LcCRFB5 and LcCRFB2. LcIFNi binds to LcCRFB5 with a much higher affinity than to LcCRFB2. Furthermore, we determined the crystal structure of LcIFNi at a 1.39 Å resolution. The high-resolution structure is, to our knowledge, the first reported structure of a type I IFN with three disulfide bonds, all of which were found to be indispensable for folding and stability of LcIFNi. Using structural analysis, mutagenesis, and biochemical assays, we identified key LcIFNi residues involved in receptor interaction and proposed a structural model of LcIFNi bound to the LcCRFB2–LcCRFB5 receptor. The results show that LcIFNi–LcCRFB2 exhibits a similar binding pattern to human IFN-ω–IFNAR2, whereas the binding pattern of LcIFNi–LcCRFB5 is quite different from that of IFN-ω–IFNAR1. Altogether, our findings reveal the structural basis for receptor interaction and signaling of a type I IFN with three disulfide bonds and provide new insights into the mechanisms underlying type I IFN recognition in teleosts.
Collapse
Affiliation(s)
- Jingjie Chen
- *Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yanyun Guan
- *Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Hongxin Guan
- †Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Yinnan Mu
- *Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yang Ding
- *Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Jun Zou
- ‡Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China; and
| | - Songying Ouyang
- †Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Xinhua Chen
- *Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- §Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, China
| |
Collapse
|
27
|
Premraj A, Aleyas AG, Nautiyal B, Rasool TJ. Beta interferons from the extant camelids: Unique among eutherian mammals. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 133:104443. [PMID: 35568245 PMCID: PMC9095258 DOI: 10.1016/j.dci.2022.104443] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/20/2022] [Accepted: 05/08/2022] [Indexed: 06/12/2023]
Abstract
The COVID-19 pandemic is a wake-up call on the zoonotic viral spillover events and the need to be prepared for future outbreaks. Zoonotic RNA viruses like the Middle East respiratory syndrome coronavirus (MERS-CoV) are potential pathogens that could trigger the next pandemic. Dromedary camels are the only known animal source of MERS-CoV zoonotic infections, but little is known about the molecular antiviral response in this species. IFN-β and other type-I interferons provide the first line of defense against invading pathogens in the host immune response. We identified the IFNB gene of the dromedary camel and all extant members of the family Camelidae. Camelid IFN-β is unique with an even number of cysteines in the mature protein compared to other eutherian mammals with an odd number of cysteines. The viral mimetic poly(I:C) strongly induced IFN-β expression in camel kidney cells. Induction of IFN-β expression upon infection with camelpox virus was late and subdued when compared to poly(I:C) treatment. Prokaryotically expressed recombinant dromedary IFN-β induced expression of IFN-responsive genes in camel kidney cells. Further, recombinant IFN-β conferred antiviral resistance to camel kidney cells against the cytopathic effects of the camelpox virus, an endemic zoonotic pathogen. IFN-β from this unique group of mammals will offer insights into antiviral immune mechanisms and aid in the development of specific antivirals against pathogens that have the potential to be the next zoonotic pandemic.
Collapse
Affiliation(s)
- Avinash Premraj
- Camel Biotechnology Center, Presidential Camels & Camel Racing Affairs Centre, Department of the President's Affairs, PO Box 17292, Al Ain, United Arab Emirates
| | - Abi George Aleyas
- Camel Biotechnology Center, Presidential Camels & Camel Racing Affairs Centre, Department of the President's Affairs, PO Box 17292, Al Ain, United Arab Emirates
| | - Binita Nautiyal
- Camel Biotechnology Center, Presidential Camels & Camel Racing Affairs Centre, Department of the President's Affairs, PO Box 17292, Al Ain, United Arab Emirates
| | - Thaha Jamal Rasool
- Camel Biotechnology Center, Presidential Camels & Camel Racing Affairs Centre, Department of the President's Affairs, PO Box 17292, Al Ain, United Arab Emirates.
| |
Collapse
|
28
|
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
|
29
|
Hauptstein N, Pouyan P, Wittwer K, Cinar G, Scherf-Clavel O, Raschig M, Licha K, Lühmann T, Nischang I, Schubert US, Pfaller CK, Haag R, Meinel L. Polymer selection impacts the pharmaceutical profile of site-specifically conjugated Interferon-α2a. J Control Release 2022; 348:881-892. [PMID: 35764249 DOI: 10.1016/j.jconrel.2022.05.060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/12/2022] [Accepted: 05/15/2022] [Indexed: 12/15/2022]
Abstract
Conjugation of poly(ethylene glycol) (PEG) to biologics is a successful strategy to favorably impact the pharmacokinetics and efficacy of the resulting bioconjugate. We compare bioconjugates synthesized by strain-promoted azide-alkyne cycloaddition (SPAAC) using PEG and linear polyglycerol (LPG) of about 20 kDa or 40 kDa, respectively, with an azido functionalized human Interferon-α2a (IFN-α2a) mutant. Site-specific PEGylation and LPGylation resulted in IFN-α2a bioconjugates with improved in vitro potency compared to commercial Pegasys. LPGylated bioconjugates had faster disposition kinetics despite comparable hydrodynamic radii to their PEGylated analogues. Overall exposure of the PEGylated IFN-α2a with a 40 kDa polymer exceeded Pegasys, which, in return, was similar to the 40 kDa LPGylated conjugates. The study points to an expanded polymer design space through which the selected polymer class may result in a different distribution of the studied bioconjugates.
Collapse
Affiliation(s)
- Niklas Hauptstein
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Paria Pouyan
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Kevin Wittwer
- Paul-Ehrlich-Institute, Division of Veterinary Medicine, Paul-Ehrlich-Str. 51-59, 63225 Langen, Germany
| | - Gizem Cinar
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstr. 10, 07743 Jena, Germany; Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Oliver Scherf-Clavel
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Martina Raschig
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Kai Licha
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Tessa Lühmann
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Ivo Nischang
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstr. 10, 07743 Jena, Germany; Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Ulrich S Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstr. 10, 07743 Jena, Germany; Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Christian K Pfaller
- Paul-Ehrlich-Institute, Division of Veterinary Medicine, Paul-Ehrlich-Str. 51-59, 63225 Langen, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Lorenz Meinel
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany; Helmholtz Institute for RNA-Based Infection Research (HIRI), 97080 Würzburg, Germany.
| |
Collapse
|
30
|
Duncan CJA, Skouboe MK, Howarth S, Hollensen AK, Chen R, Børresen ML, Thompson BJ, Stremenova Spegarova J, Hatton CF, Stæger FF, Andersen MK, Whittaker J, Paludan SR, Jørgensen SE, Thomsen MK, Mikkelsen JG, Heilmann C, Buhas D, Øbro NF, Bay JT, Marquart HV, de la Morena MT, Klejka JA, Hirschfeld M, Borgwardt L, Forss I, Masmas T, Poulsen A, Noya F, Rouleau G, Hansen T, Zhou S, Albrechtsen A, Alizadehfar R, Allenspach EJ, Hambleton S, Mogensen TH. Life-threatening viral disease in a novel form of autosomal recessive IFNAR2 deficiency in the Arctic. J Exp Med 2022; 219:e20212427. [PMID: 35442417 PMCID: PMC9026249 DOI: 10.1084/jem.20212427] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 02/28/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022] Open
Abstract
Type I interferons (IFN-I) play a critical role in human antiviral immunity, as demonstrated by the exceptionally rare deleterious variants of IFNAR1 or IFNAR2. We investigated five children from Greenland, Canada, and Alaska presenting with viral diseases, including life-threatening COVID-19 or influenza, in addition to meningoencephalitis and/or hemophagocytic lymphohistiocytosis following live-attenuated viral vaccination. The affected individuals bore the same homozygous IFNAR2 c.157T>C, p.Ser53Pro missense variant. Although absent from reference databases, p.Ser53Pro occurred with a minor allele frequency of 0.034 in their Inuit ancestry. The serine to proline substitution prevented cell surface expression of IFNAR2 protein, small amounts of which persisted intracellularly in an aberrantly glycosylated state. Cells exclusively expressing the p.Ser53Pro variant lacked responses to recombinant IFN-I and displayed heightened vulnerability to multiple viruses in vitro-a phenotype rescued by wild-type IFNAR2 complementation. This novel form of autosomal recessive IFNAR2 deficiency reinforces the essential role of IFN-I in viral immunity. Further studies are warranted to assess the need for population screening.
Collapse
Affiliation(s)
- Christopher J A Duncan
- Clinical and Translational Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Morten K Skouboe
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Sophie Howarth
- Clinical and Translational Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Anne K Hollensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Rui Chen
- Clinical and Translational Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Malene L Børresen
- Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Benjamin J Thompson
- Clinical and Translational Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Jarmila Stremenova Spegarova
- Clinical and Translational Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Catherine F Hatton
- Clinical and Translational Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Frederik F Stæger
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Mette K Andersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - John Whittaker
- Clinical and Translational Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Søren R Paludan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Sofie E Jørgensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | | | | | - Carsten Heilmann
- Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Medical Department, Pediatric Section, Dronning Ingrid Hospital, Nuuk, Greenland
| | - Daniela Buhas
- Division of Genetics, Department of Specialized Medicine, McGill University Health Centre, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Nina F Øbro
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jakob T Bay
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Hanne V Marquart
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - M Teresa de la Morena
- Seattle Children's Hospital, Seattle, WA
- Department of Pediatrics, University of Washington, Seattle, WA
| | | | | | - Line Borgwardt
- Center for Genomic Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Isabel Forss
- Center for Genomic Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Tania Masmas
- Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Anja Poulsen
- Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Francisco Noya
- Division of Allergy & Clinical Immunology, Montreal Children's Hospital, Montreal General Hospital, McGill University, Montreal, Quebec, Canada
| | - Guy Rouleau
- The Neuro, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sirui Zhou
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Anders Albrechtsen
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Reza Alizadehfar
- Division of Allergy & Clinical Immunology, Montreal Children's Hospital, Montreal General Hospital, McGill University, Montreal, Quebec, Canada
| | - Eric J Allenspach
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
- Seattle Children's Hospital, Seattle, WA
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
- Brotman Baty Institute for Precision Medicine, Seattle, WA
| | - Sophie Hambleton
- Clinical and Translational Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Trine H Mogensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
31
|
Lipari E, Saporiti S, Eberini I, Massimo L, Mazzarella E, Anderloni G, Rossi M, D'Amici F, Pergola C, Palinsky W, D'Acunto CW, Centola F. Asn25 Deamidation as an Allosteric Tool to Increase IFNβ-1a Biological Activity. J Interferon Cytokine Res 2022; 42:251-266. [PMID: 35527626 DOI: 10.1089/jir.2021.0209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Interferon beta (IFNβ) is a well-known cytokine, belonging to the type I family, that exerts antiviral, immunomodulatory, and antiproliferative activity. It has been reported that the artificially deamidated form of recombinant IFNβ-1a at Asn25 position shows an increased biological activity. As a deepening of the previous study, the molecular mechanism underlying this biological effect was investigated in this work by combining experimental and computational techniques. Specifically, the binding to IFNAR1 and IFNAR2 receptors and the canonical pathway of artificially deamidated IFNβ-1a molecule were analyzed in comparison to the native form. As a result, a change in receptor affinity of deamidated IFNβ-1a with respect to the native form was observed, and to better explore this molecular interaction, molecular dynamics simulations were carried out. Results confirmed, as previously hypothesized, that the N25D mutation can locally change the interaction network of the mutated residue but also that this effect can be propagated throughout the molecule. In fact, many residues not involved in the interaction with IFNAR1 in the native form participate to the recognition in the deamidated molecule, enhancing the binding to IFNAR1 receptor and consequently an increase of signaling cascade activation. In particular, a higher STAT1 phosphorylation and interferon-stimulated gene expression was observed under deamidated IFNβ-1a cell treatment. In conclusion, this study increases the scientific knowledge of deamidated IFNβ-1a, deciphering its molecular mechanism, and opens new perspectives to novel therapeutic strategies.
Collapse
Affiliation(s)
- Elisa Lipari
- Analytical Development Biotech, Merck Serono S.p.A., Rome, Italy (an Affiliate of Merck KGaA, Darmstadt, Germany)
| | - Simona Saporiti
- Dipartimento di Scienze Farmacologiche e Biomolecolari and Università degli Studi di Milano, Milano, Italy
| | - Ivano Eberini
- Dipartimento di Scienze Farmacologiche e Biomolecolari and Università degli Studi di Milano, Milano, Italy.,Data Science Research Center (DSRC), Università degli Studi di Milano, Milano, Italy
| | - Luigia Massimo
- Analytical Development Biotech, Merck Serono S.p.A., Rome, Italy (an Affiliate of Merck KGaA, Darmstadt, Germany).,Dipartimento di Farmacia, Università degli Studi di Salerno, Fisciano, Italy
| | - Enrico Mazzarella
- Analytical Development Biotech, Merck Serono S.p.A., Rome, Italy (an Affiliate of Merck KGaA, Darmstadt, Germany).,Sezione di Medicina Interna e Malattie Metaboliche, Dipartimento di Medicina Interna e Specialistica, DIBIMIS, Università di Palermo, Palermo, Italy
| | - Giulia Anderloni
- Analytical Development Biotech, Merck Serono S.p.A., Rome, Italy (an Affiliate of Merck KGaA, Darmstadt, Germany).,Sezione di Medicina Interna e Malattie Metaboliche, Dipartimento di Medicina Interna e Specialistica, DIBIMIS, Università di Palermo, Palermo, Italy
| | - Mara Rossi
- Global Analytical Pharmaceutical Science and Innovation, Merck Serono S.p.A., Rome, Italy (an Affiliate of Merck KGaA, Darmstadt, Germany)
| | - Fabio D'Amici
- Analytical Development Biotech, Merck Serono S.p.A., Rome, Italy (an Affiliate of Merck KGaA, Darmstadt, Germany)
| | - Carlo Pergola
- Analytical Development Biotech, Merck Serono S.p.A., Rome, Italy (an Affiliate of Merck KGaA, Darmstadt, Germany)
| | - Wolf Palinsky
- Biotech Development Programme, Merck Biopharma, Aubonne, Switzerland (an Affiliate of Merck KGaA, Darmstadt, Germany)
| | - Cosimo Walter D'Acunto
- Analytical Development Biotech, Merck Serono S.p.A., Rome, Italy (an Affiliate of Merck KGaA, Darmstadt, Germany)
| | - Fabio Centola
- Global Analytical Pharmaceutical Science and Innovation, Merck Serono S.p.A., Rome, Italy (an Affiliate of Merck KGaA, Darmstadt, Germany)
| |
Collapse
|
32
|
Huijser E, Göpfert J, Brkic Z, van Helden-Meeuwsen CG, Jansen S, Mandl T, Olsson P, Schrijver B, Schreurs MWJ, van Daele PLA, Dik WA, Versnel MA. Serum interferon-α2 measured by single-molecule array associates with systemic disease manifestations in Sjögren's syndrome. Rheumatology (Oxford) 2022; 61:2156-2166. [PMID: 34505866 PMCID: PMC9071525 DOI: 10.1093/rheumatology/keab688] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 09/03/2021] [Accepted: 09/03/2021] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES Type I IFN (IFN-I) activation is a prominent feature of primary SS (pSS), SLE and SSc. Ultrasensitive single-molecule array (Simoa) technology has facilitated the measurement of subfemtomolar concentrations of IFNs. Here we aimed to measure IFN-α2 in serum from pSS, SLE and SSc using a Simoa immunoassay and correlate these levels to blood IFN-stimulated gene (ISG) expression and disease activity. METHODS Serum IFN-α2 was measured in patients with pSS (n = 85 and n = 110), SLE (n = 24) and SSc (n = 23) and healthy controls (HCs; n = 68) using an IFN-α Simoa assay on an HD-X analyser. IFN-I pathway activation was additionally determined from serum by an IFN-I reporter assay and paired samples of whole blood ISG expression of IFI44, IFI44L, IFIT1, IFIT3 and MxA by RT-PCR or myxovirus resistance protein 1 (MxA) protein ELISA. RESULTS Serum IFN-α2 levels were elevated in pSS (median 61.3 fg/ml) compared with HCs (median ≤5 fg/ml, P < 0.001) and SSc (median 11.6 fg/ml, P = 0.043), lower compared with SLE (median 313.5 fg/ml, P = 0.068) and positively correlated with blood ISG expression (r = 0.66-0.94, P < 0.001). Comparable to MxA ELISA [area under the curve (AUC) 0.93], IFN-α2 measurement using Simoa identified pSS with high ISG expression (AUC 0.90) with 80-93% specificity and 71-84% sensitivity. Blinded validation in an independent pSS cohort yielded a comparable accuracy. Multiple regression indicated independent associations of autoantibodies, IgG, HCQ treatment, cutaneous disease and a history of extraglandular manifestations with serum IFN-α2 concentrations in pSS. CONCLUSION Simoa serum IFN-α2 reflects blood ISG expression in pSS, SLE and SSc. In light of IFN-targeting treatments, Simoa could potentially be applied for patient stratification or retrospective analysis of historical cohorts.
Collapse
Affiliation(s)
- Erika Huijser
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jens Göpfert
- Department of Applied Biomarkers and Immunoassays, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Zana Brkic
- Department of Internal Medicine, Division of Clinical Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Sanne Jansen
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Thomas Mandl
- Department of Clinical Sciences Malmö, Division of Rheumatology, Lund University, Malmö, Sweden
| | - Peter Olsson
- Department of Clinical Sciences Malmö, Division of Rheumatology, Lund University, Malmö, Sweden
| | - Benjamin Schrijver
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marco W J Schreurs
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Paul L A van Daele
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Clinical Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Willem A Dik
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marjan A Versnel
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
33
|
Yen M, Ren J, Liu Q, Glassman CR, Sheahan TP, Picton LK, Moreira FR, Rustagi A, Jude KM, Zhao X, Blish CA, Baric RS, Su LL, Garcia KC. Facile discovery of surrogate cytokine agonists. Cell 2022; 185:1414-1430.e19. [PMID: 35325595 PMCID: PMC9021867 DOI: 10.1016/j.cell.2022.02.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/11/2022] [Accepted: 02/22/2022] [Indexed: 12/26/2022]
Abstract
Cytokines are powerful immune modulators that initiate signaling through receptor dimerization, but natural cytokines have structural limitations as therapeutics. We present a strategy to discover cytokine surrogate agonists by using modular ligands that exploit induced proximity and receptor dimer geometry as pharmacological metrics amenable to high-throughput screening. Using VHH and scFv to human interleukin-2/15, type-I interferon, and interleukin-10 receptors, we generated combinatorial matrices of single-chain bispecific ligands that exhibited diverse spectrums of functional activities, including potent inhibition of SARS-CoV-2 by surrogate interferons. Crystal structures of IL-2R:VHH complexes revealed that variation in receptor dimer geometries resulted in functionally diverse signaling outputs. This modular platform enabled engineering of surrogate ligands that compelled assembly of an IL-2R/IL-10R heterodimer, which does not naturally exist, that signaled through pSTAT5 on T and natural killer (NK) cells. This "cytokine med-chem" approach, rooted in principles of induced proximity, is generalizable for discovery of diversified agonists for many ligand-receptor systems.
Collapse
Affiliation(s)
- Michelle Yen
- Departments of Molecular and Cellular Physiology, and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Junming Ren
- Departments of Molecular and Cellular Physiology, and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Qingxiang Liu
- Departments of Molecular and Cellular Physiology, and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Caleb R Glassman
- Departments of Molecular and Cellular Physiology, and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Timothy P Sheahan
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lora K Picton
- Departments of Molecular and Cellular Physiology, and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Fernando R Moreira
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Arjun Rustagi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kevin M Jude
- Departments of Molecular and Cellular Physiology, and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xiang Zhao
- Departments of Molecular and Cellular Physiology, and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Catherine A Blish
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Ralph S Baric
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leon L Su
- Departments of Molecular and Cellular Physiology, and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - K Christopher Garcia
- Departments of Molecular and Cellular Physiology, and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
34
|
Payet CA, You A, Fayet OM, Dragin N, Berrih-Aknin S, Le Panse R. Myasthenia Gravis: An Acquired Interferonopathy? Cells 2022; 11:cells11071218. [PMID: 35406782 PMCID: PMC8997999 DOI: 10.3390/cells11071218] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 01/12/2023] Open
Abstract
Myasthenia gravis (MG) is a rare autoimmune disease mediated by antibodies against components of the neuromuscular junction, particularly the acetylcholine receptor (AChR). The thymus plays a primary role in AChR-MG patients. In early-onset AChR-MG and thymoma-associated MG, an interferon type I (IFN-I) signature is clearly detected in the thymus. The origin of this chronic IFN-I expression in the thymus is not yet defined. IFN-I subtypes are normally produced in response to viral infection. However, genetic diseases called interferonopathies are associated with an aberrant chronic production of IFN-I defined as sterile inflammation. Some systemic autoimmune diseases also share common features with interferonopathies. This review aims to analyze the pathogenic role of IFN-I in these diseases as compared to AChR-MG in order to determine if AChR-MG could be an acquired interferonopathy.
Collapse
Affiliation(s)
- Cloé A Payet
- Sorbonne University, INSERM, Institute of Myology, Center of Research in Myology, F-75013 Paris, France
| | - Axel You
- Sorbonne University, INSERM, Institute of Myology, Center of Research in Myology, F-75013 Paris, France
| | - Odessa-Maud Fayet
- Sorbonne University, INSERM, Institute of Myology, Center of Research in Myology, F-75013 Paris, France
| | - Nadine Dragin
- Sorbonne University, INSERM, Institute of Myology, Center of Research in Myology, F-75013 Paris, France
| | - Sonia Berrih-Aknin
- Sorbonne University, INSERM, Institute of Myology, Center of Research in Myology, F-75013 Paris, France
| | - Rozen Le Panse
- Sorbonne University, INSERM, Institute of Myology, Center of Research in Myology, F-75013 Paris, France
| |
Collapse
|
35
|
Goldmann O, Sauerwein T, Molinari G, Rohde M, Förstner KU, Medina E. Cytosolic Sensing of Intracellular Staphylococcus aureus by Mast Cells Elicits a Type I IFN Response That Enhances Cell-Autonomous Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1675-1685. [PMID: 35321877 DOI: 10.4049/jimmunol.2100622] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 01/20/2022] [Indexed: 06/14/2023]
Abstract
Strategically located at mucosal sites, mast cells are instrumental in sensing invading pathogens and modulating the quality of the ensuing immune responses depending on the nature of the infecting microbe. It is believed that mast cells produce type I IFN (IFN-I) in response to viruses, but not to bacterial infections, because of the incapacity of bacterial pathogens to internalize within mast cells, where signaling cascades leading to IFN-I production are generated. However, we have previously reported that, in contrast with other bacterial pathogens, Staphylococcus aureus can internalize into mast cells and therefore could trigger a unique response. In this study, we have investigated the molecular cross-talk between internalized S. aureus and the human mast cells HMC-1 using a dual RNA sequencing approach. We found that a proportion of internalized S. aureus underwent profound transcriptional reprogramming within HMC-1 cells to adapt to the nutrients and stress encountered in the intracellular environment and remained viable. HMC-1 cells, in turn, recognized intracellular S. aureus via cGMP-AMP synthase-STING-TANK-binding kinase 1 signaling pathway, leading to the production of IFN-I. Bacterial internalization and viability were crucial for IFN-I induction because inhibition of S. aureus internalization or infection with heat-killed bacteria completely prevented the production of IFN-I by HMC-1 cells. Feeding back in an autocrine manner in S. aureus-harboring HMC-1 cells and in a paracrine manner in noninfected neighboring HMC-1 cells, IFN-I promoted a cell-autonomous antimicrobial state by inducing the transcription of IFN-I-stimulated genes. This study provides unprecedented evidence of the capacity of mast cells to produce IFN-I in response to a bacterial pathogen.
Collapse
Affiliation(s)
- Oliver Goldmann
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Till Sauerwein
- Institute for Molecular Infection Biology, University of Würzburg, 97080 Würzburg, Germany
- ZB MED-Information Centre for Life Science, 50931 Cologne, Germany
| | - Gabriella Molinari
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; and
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; and
| | - Konrad U Förstner
- Institute for Molecular Infection Biology, University of Würzburg, 97080 Würzburg, Germany
- ZB MED-Information Centre for Life Science, 50931 Cologne, Germany
- TH Köln, University of Applied Sciences, Faculty of Information Science and Communication Studies, 50678 Cologne, Germany
| | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany;
| |
Collapse
|
36
|
Xue W, Ding C, Qian K, Liao Y. The Interplay Between Coronavirus and Type I IFN Response. Front Microbiol 2022; 12:805472. [PMID: 35317429 PMCID: PMC8934427 DOI: 10.3389/fmicb.2021.805472] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 12/24/2021] [Indexed: 12/14/2022] Open
Abstract
In the past few decades, newly evolved coronaviruses have posed a global threat to public health and animal breeding. To control and prevent the coronavirus-related diseases, understanding the interaction of the coronavirus and the host immune system is the top priority. Coronaviruses have evolved multiple mechanisms to evade or antagonize the host immune response to ensure their replication. As the first line and main component of innate immune response, type I IFN response is able to restrict virus in the initial infection stage; it is thus not surprising that the primary aim of the virus is to evade or antagonize the IFN response. Gaining a profound understanding of the interaction between coronaviruses and type I IFN response will shed light on vaccine development and therapeutics. In this review, we provide an update on the current knowledge on strategies employed by coronaviruses to evade type I IFN response.
Collapse
Affiliation(s)
- Wenxiang Xue
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Chan Ding
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Kun Qian
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Ying Liao
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- *Correspondence: Ying Liao,
| |
Collapse
|
37
|
TLR3-driven IFN-β antagonizes STAT5-activating cytokines and suppresses innate type 2 response in the lung. J Allergy Clin Immunol 2022; 149:1044-1059.e5. [PMID: 34428519 PMCID: PMC8859010 DOI: 10.1016/j.jaci.2021.07.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/16/2021] [Accepted: 07/29/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Group 2 innate lymphoid cells (ILC2s) are involved in type 2 immune responses in mucosal organs and are associated with various allergic diseases in humans. Studies are needed to understand the molecules and pathways that control ILC2s. OBJECTIVE The aims of this study were to develop a mouse model that limits the innate type 2 immune response in the lung and to investigate the immunologic mechanisms involved in regulation of lung ILC2s. METHODS Naive BALB/c mice were administered various Toll-like receptor agonists and exposed intranasally to the fungal allergen Alternaria alternata. The mechanisms were investigated using gene knockout mice as well as cultures of lung cells and isolated lung ILC2s. RESULTS Polyinosinic-polycytidylic acid, or poly (I:C), effectively inhibited innate type 2 response to A alternata. Poly (I:C) promoted production of IFNα, -β, and -γ, and its inhibitory effects were dependent on the IFN-α/β receptor pathway. IFN-β was 100 times more potent than IFN-α at inhibiting type 2 cytokine production by lung ILC2s. Signal transducer and activator of transcription 5 (STAT5)-activating cytokines, including IL-2, IL-7, and thymic stromal lymphopoietin, but not IL-33, promoted survival and proliferation of lung ILC2s in vitro, while IFN-β blocked these effects. Expression of the transcription factor GATA3, which is critical for differentiation and maintenance of ILC2s, was inhibited by IFN-β. CONCLUSIONS IFN-β blocks the effects of STAT5-activating cytokines on lung ILC2s and inhibits their survival and effector functions. Administration of IFN-β may provide a new strategy to treat diseases involving ILC2s.
Collapse
|
38
|
Valdés-López JF, Fernandez GJ, Urcuqui-Inchima S. Synergistic Effects of Toll-Like Receptor 1/2 and Toll-Like Receptor 3 Signaling Triggering Interleukin 27 Gene Expression in Chikungunya Virus-Infected Macrophages. Front Cell Dev Biol 2022; 10:812110. [PMID: 35223841 PMCID: PMC8863767 DOI: 10.3389/fcell.2022.812110] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/17/2022] [Indexed: 12/19/2022] Open
Abstract
Chikungunya virus (CHIKV) is the etiological agent of chikungunya fever (CHIKF), a self-limiting disease characterized by myalgia and severe acute or chronic arthralgia. CHIKF is associated with immunopathology and high levels of pro-inflammatory factors. CHIKV is known to have a wide range of tropism in human cell types, including keratinocytes, fibroblasts, endothelial cells, monocytes, and macrophages. Previously, we reported that CHIKV-infected monocytes-derived macrophages (MDMs) express high levels of interleukin 27 (IL27), a heterodimeric cytokine consisting of IL27p28 and EBI3 subunits, that triggers JAK-STAT signaling and promotes pro-inflammatory and antiviral response, in interferon (IFN)-independent manner. Based on the transcriptomic analysis, we now report that induction of IL27-dependent pro-inflammatory and antiviral response in CHIKV-infected MDMs relies on two signaling pathways: an early signal dependent on recognition of CHIKV-PAMPs by TLR1/2-MyD88 to activate NF-κB-complex that induces the expression of EBI3 mRNA; and second signaling dependent on the recognition of intermediates of CHIKV replication (such as dsRNA) by TLR3-TRIF, to activate IRF1 and the induction of IL27p28 mRNA expression. Both signaling pathways were required to produce a functional IL27 protein involved in the induction of ISGs, including antiviral proteins, cytokines, CC- and CXC- chemokines in an IFN-independent manner in MDMs. Furthermore, we reported that activation of TLR4 by LPS, both in human MDMs and murine BMDM, results in the induction of both subunits of IL27 that trigger strong IL27-dependent pro-inflammatory and antiviral response independent of IFNs signaling. Our findings are a significant contribution to the understanding of molecular and cellular mechanisms of CHIKV infection.
Collapse
|
39
|
Hauptstein N, Meinel L, Lühmann T. Bioconjugation strategies and clinical implications of Interferon-bioconjugates. Eur J Pharm Biopharm 2022; 172:157-167. [PMID: 35149191 DOI: 10.1016/j.ejpb.2022.02.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/24/2022] [Accepted: 02/05/2022] [Indexed: 02/08/2023]
Abstract
Interferons (IFN) are immunomodulating, antiviral and antiproliferative cytokines for treatment of multiple indications, including cancer, hepatitis, and autoimmune disease. The first IFNs were discovered in 1957, first approved in 1986, and are nowadays listed in the WHO model list of essential Medicines. Three classes of IFNs are known; IFN-α2a and IFN-β belonging to type-I IFNs, IFN-γ a type-II IFN approved for some hereditary diseases and IFN-λs, which form the newest class of type-III IFNs. IFN-λs were discovered in the last decade with fascinating yet under discovered pharmaceutical potential. This article reviews available IFN drugs, their field and route of application, while also outlining available and future strategies for bioconjugation to further optimize pharmaceutical and clinical performances of all three available IFN classes.
Collapse
Affiliation(s)
- Niklas Hauptstein
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, DE-97074, Würzburg, Germany
| | - Lorenz Meinel
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, DE-97074, Würzburg, Germany; Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), DE-97080 Würzburg, Germany
| | - Tessa Lühmann
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, DE-97074, Würzburg, Germany.
| |
Collapse
|
40
|
Determination of molecular signatures and pathways common to brain tissues of autism spectrum disorder: Insights from comprehensive bioinformatics approach. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.100871] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
41
|
Chen Y, Chen D, Wang Q, Xu Y, Huang X, Haglund F, Su H. Immunological Classification of Pancreatic Carcinomas to Identify Immune Index and Provide a Strategy for Patient Stratification. Front Immunol 2022; 12:719105. [PMID: 35111149 PMCID: PMC8801451 DOI: 10.3389/fimmu.2021.719105] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
Background Cancer immunotherapy has produced significant positive clinical effects in a variety of tumor types. However, pancreatic ductal adenocarcinoma (PDAC) is widely considered to be a "cold" cancer with poor immunogenicity. Our aim is to determine the detailed immune features of PDAC to seek new treatment strategies. Methods The immune cell abundance of PDAC patients was evaluated with the single-sample gene set enrichment analysis (ssGSEA) using 119 immune gene signatures. Based on these data, patients were classified into different immune subtypes (ISs) according to immune gene signatures. We analyzed their response patterns to immunotherapy in the datasets, then established an immune index to reflect the different degrees of immune infiltration through linear discriminant analysis (LDA). Finally, potential prognostic markers associated with the immune index were identified based on weighted correlation network analysis (WGCNA) that was functionally validated in vitro. Results Three ISs were identified in PDAC, of which IS3 had the best prognosis across all three cohorts. The different expressions of immune profiles among the three ISs indicated a distinct responsiveness to immunotherapies in PDAC subtypes. By calculating the immune index, we found that the IS3 represented higher immune infiltration, while IS1 represented lower immune infiltration. Among the investigated signatures, we identified ZNF185, FANCG, and CSTF2 as risk factors associated with immune index that could potentially facilitate diagnosis and could be therapeutic target markers in PDAC patients. Conclusions Our findings identified immunologic subtypes of PDAC with distinct prognostic implications, which allowed us to establish an immune index to represent the immune infiltration in each subtype. These results show the importance of continuing investigation of immunotherapy and will allow clinical workers to personalized treatment more effectively in PDAC patients.
Collapse
Affiliation(s)
- Yi Chen
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Didi Chen
- Department of Radiation Oncology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiang Wang
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Yajing Xu
- Department of Radiation Oncology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaowei Huang
- Department of Radiation Oncology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Felix Haglund
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Huafang Su
- Department of Radiation Oncology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
42
|
Layton DS, Mara K, Dai M, Malaver-Ortega LF, Gough TJ, Bruce K, Jenkins KA, Bean AGD. Interferon Signaling in Chickens Plays a Crucial Role in Inhibiting Influenza Replication in DF1 Cells. Microorganisms 2022; 10:133. [PMID: 35056582 PMCID: PMC8781551 DOI: 10.3390/microorganisms10010133] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 01/17/2023] Open
Abstract
Influenza A viruses (IAV) pose a constant threat to human and poultry health. Of particular interest are the infections caused by highly pathogenic avian influenza (HPAI) viruses, such as H5N1, which cause significant production issues. In response to influenza infection, cells activate immune mechanisms that lead to increased interferon (IFN) production. To investigate how alterations in the interferon signaling pathway affect the cellular response to infection in the chicken, we used CRISPR/Cas9 to generate a chicken cell line that lacks a functional the type I interferon receptor (IFNAR1). We then assessed viral infections with the WSN strain of influenza. Cells lacking a functional IFNAR1 receptor showed reduced expression of the interferon stimulated genes (ISG) such as Protein Kinase R (PKR) and Myxovirus resistance (Mx) and were more susceptible to viral infection with WSN. We further investigated the role or IFNAR1 on low pathogenicity avian influenza (LPAI) strains (H7N9) and a HPAI strain (H5N1). Intriguingly, Ifnar-/- cells appeared more resistant than WT cells when infected with HPAI virus, potentially indicating a different interaction between H5N1 and the IFN signaling pathway. Our findings support that ChIFNAR1 is a key component of the chicken IFN signaling pathway and these data add contributions to the field of host-avian pathogen interaction and innate immunity in chickens.
Collapse
Affiliation(s)
- Daniel S. Layton
- CSIRO Health and Biosecurity, Australian Centre for Disease Preparedness (ACDP), Geelong, VIC 3220, Australia; (K.M.); (M.D.); (T.J.G.); (K.B.); (K.A.J.); (A.G.D.B.)
| | - Kostlend Mara
- CSIRO Health and Biosecurity, Australian Centre for Disease Preparedness (ACDP), Geelong, VIC 3220, Australia; (K.M.); (M.D.); (T.J.G.); (K.B.); (K.A.J.); (A.G.D.B.)
| | - Meiling Dai
- CSIRO Health and Biosecurity, Australian Centre for Disease Preparedness (ACDP), Geelong, VIC 3220, Australia; (K.M.); (M.D.); (T.J.G.); (K.B.); (K.A.J.); (A.G.D.B.)
| | - Luis Fernando Malaver-Ortega
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Clayton Campus, Monash University, Clayton, VIC 3800, Australia;
| | - Tamara J. Gough
- CSIRO Health and Biosecurity, Australian Centre for Disease Preparedness (ACDP), Geelong, VIC 3220, Australia; (K.M.); (M.D.); (T.J.G.); (K.B.); (K.A.J.); (A.G.D.B.)
| | - Kerri Bruce
- CSIRO Health and Biosecurity, Australian Centre for Disease Preparedness (ACDP), Geelong, VIC 3220, Australia; (K.M.); (M.D.); (T.J.G.); (K.B.); (K.A.J.); (A.G.D.B.)
| | - Kristie A. Jenkins
- CSIRO Health and Biosecurity, Australian Centre for Disease Preparedness (ACDP), Geelong, VIC 3220, Australia; (K.M.); (M.D.); (T.J.G.); (K.B.); (K.A.J.); (A.G.D.B.)
| | - Andrew G. D. Bean
- CSIRO Health and Biosecurity, Australian Centre for Disease Preparedness (ACDP), Geelong, VIC 3220, Australia; (K.M.); (M.D.); (T.J.G.); (K.B.); (K.A.J.); (A.G.D.B.)
| |
Collapse
|
43
|
Haji Abdolvahab M, Vafaee R, Arab SS, Behmanesh M. Next-generation version of recombinant human interferon-beta: A molecular dynamic simulation study. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.101036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
44
|
Gu H, Zhang Y, Zeng W, Xia Y. Participation of interferons in psoriatic inflammation. Cytokine Growth Factor Rev 2021; 64:12-20. [PMID: 34953718 DOI: 10.1016/j.cytogfr.2021.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 12/17/2022]
Abstract
Interferons are multifunctional cytokines not expressed in the skin under normal physiological conditions. However, they are overexpressed in serum and skin lesions of patients with psoriasis and play an important role in the pathogenesis of the disease. Interferons act directly on skin resident cells and recruit and modulate inflammatory cells, thereby exacerbating psoriatic inflammation. They upregulate the expression of relevant cytokines and chemokines, facilitate excessive proliferation of keratinocytes, and enhance the formation of poorly differentiated dermal microvessels. In this review, we summarized the pathogenic effect of interferons on psoriasis and also discussed the therapeutic strategies targeting interferons.
Collapse
Affiliation(s)
- Hanjiang Gu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yufei Zhang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Weihui Zeng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
45
|
Mohseni G, Li J, Ariston Gabriel AN, Du L, Wang YS, Wang C. The Function of cGAS-STING Pathway in Treatment of Pancreatic Cancer. Front Immunol 2021; 12:781032. [PMID: 34858438 PMCID: PMC8630697 DOI: 10.3389/fimmu.2021.781032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 10/25/2021] [Indexed: 12/23/2022] Open
Abstract
The activation of stimulator of interferon genes (STING) signalling pathway has been suggested to promote the immune responses against malignancy. STING is activated in response to the detection of cytosolic DNA and can induce type I interferons and link innate immunity with the adaptive immune system. Due to accretive evidence demonstrating that the STING pathway regulates the immune cells of the tumor microenvironment (TME), STING as a cancer biotherapy has attracted considerable attention. Pancreatic cancer, with a highly immunosuppressive TME, remains fatal cancer. STING has been applied to the treatment of pancreatic cancer through distinct strategies. This review reveals the role of STING signalling on pancreatic tumors and other diseases related to the pancreas. We then discuss new advances of STING in either monotherapy or combination methods for pancreatic cancer immunotherapy.
Collapse
Affiliation(s)
- Ghazal Mohseni
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Clinical Laboratory Diagnostics, School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Abakundana Nsenga Ariston Gabriel
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Clinical Laboratory Diagnostics, School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yun-Shan Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
46
|
Shemesh M, Lochte S, Piehler J, Schreiber G. IFNAR1 and IFNAR2 play distinct roles in initiating type I interferon-induced JAK-STAT signaling and activating STATs. Sci Signal 2021; 14:eabe4627. [PMID: 34813358 DOI: 10.1126/scisignal.abe4627] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Maya Shemesh
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Sara Lochte
- Department of Biology and Center of Cellular Nanoanalytics, University of Osnabrück, 49076 Osnabrück, Germany
| | - Jacob Piehler
- Department of Biology and Center of Cellular Nanoanalytics, University of Osnabrück, 49076 Osnabrück, Germany
| | - Gideon Schreiber
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
47
|
Giorgetti SI, Etcheverrigaray M, Terry F, Martin W, De Groot AS, Ceaglio N, Oggero M, Mufarrege EF. Development of highly stable and de-immunized versions of recombinant alpha interferon: Promising candidates for the treatment of chronic and emerging viral diseases. Clin Immunol 2021; 233:108888. [PMID: 34798238 PMCID: PMC8595249 DOI: 10.1016/j.clim.2021.108888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/04/2021] [Accepted: 11/13/2021] [Indexed: 12/23/2022]
Abstract
Human interferon alpha (hIFN-α) administration constitutes the current FDA approved therapy for chronic Hepatitis B and C virus infections. Additionally, hIFN-α treatment efficacy was recently demonstrated in patients with COVID-19. Thus, hIFN-α constitutes a therapeutic alternative for those countries where vaccination is inaccessible and for people who did not respond effectively to vaccination. However, hIFN-α2b exhibits a short plasma half-life resulting in the occurrence of severe side effects. To optimize the cytokine's pharmacokinetic profile, we developed a hyperglycosylated IFN, referred to as GMOP-IFN. Given the significant number of reports showing neutralizing antibodies (NAb) formation after hIFN-α administration, here we applied the DeFT (De-immunization of Functional Therapeutics) approach to develop functional, de-immunized versions of GMOP-IFN. Two GMOP-IFN variants exhibited significantly reduced ex vivo immunogenicity and null antiproliferative activity, while preserving antiviral function. The results obtained in this work indicate that the new de-immunized GMOP-IFN variants constitute promising candidates for antiviral therapy.
Collapse
Affiliation(s)
- Sofía Inés Giorgetti
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Ciudad Universitaria, Ruta Nacional 168 Km 472.4, C.C. 242. (S3000ZAA), Santa Fe, Argentina
| | - Marina Etcheverrigaray
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Ciudad Universitaria, Ruta Nacional 168 Km 472.4, C.C. 242. (S3000ZAA), Santa Fe, Argentina
| | | | | | - Anne Searls De Groot
- EpiVax, Inc., Providence, RI, USA; Institute for Immunology and Informatics, University of Rhode Island, RI, USA
| | - Natalia Ceaglio
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Ciudad Universitaria, Ruta Nacional 168 Km 472.4, C.C. 242. (S3000ZAA), Santa Fe, Argentina
| | - Marcos Oggero
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Ciudad Universitaria, Ruta Nacional 168 Km 472.4, C.C. 242. (S3000ZAA), Santa Fe, Argentina
| | - Eduardo Federico Mufarrege
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Ciudad Universitaria, Ruta Nacional 168 Km 472.4, C.C. 242. (S3000ZAA), Santa Fe, Argentina.
| |
Collapse
|
48
|
Thiecke MJ, Yang EJ, Burren OS, Ray-Jones H, Spivakov M. Prioritisation of Candidate Genes Underpinning COVID-19 Host Genetic Traits Based on High-Resolution 3D Chromosomal Topology. Front Genet 2021; 12:745672. [PMID: 34759959 PMCID: PMC8573080 DOI: 10.3389/fgene.2021.745672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/30/2021] [Indexed: 11/30/2022] Open
Abstract
Genetic variants showing associations with specific biological traits and diseases detected by genome-wide association studies (GWAS) commonly map to non-coding DNA regulatory regions. Many of these regions are located considerable distances away from the genes they regulate and come into their proximity through 3D chromosomal interactions. We previously developed COGS, a statistical pipeline for linking GWAS variants with their putative target genes based on 3D chromosomal interaction data arising from high-resolution assays such as Promoter Capture Hi-C (PCHi-C). Here, we applied COGS to COVID-19 Host Genetic Consortium (HGI) GWAS meta-analysis data on COVID-19 susceptibility and severity using our previously generated PCHi-C results in 17 human primary cell types and SARS-CoV-2-infected lung carcinoma cells. We prioritise 251 genes putatively associated with these traits, including 16 out of 47 genes highlighted by the GWAS meta-analysis authors. The prioritised genes are expressed in a broad array of tissues, including, but not limited to, blood and brain cells, and are enriched for genes involved in the inflammatory response to viral infection. Our prioritised genes and pathways, in conjunction with results from other prioritisation approaches and targeted validation experiments, will aid in the understanding of COVID-19 pathology, paving the way for novel treatments.
Collapse
Affiliation(s)
| | - Emma J Yang
- Functional Gene Control Group, MRC London Institute of Medical Sciences, London, United Kingdom.,Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Oliver S Burren
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Helen Ray-Jones
- Functional Gene Control Group, MRC London Institute of Medical Sciences, London, United Kingdom.,Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Mikhail Spivakov
- Functional Gene Control Group, MRC London Institute of Medical Sciences, London, United Kingdom.,Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London, United Kingdom
| |
Collapse
|
49
|
Kirby D, Parmar B, Fathi S, Marwah S, Nayak CR, Cherepanov V, MacParland S, Feld JJ, Altan-Bonnet G, Zilman A. Determinants of Ligand Specificity and Functional Plasticity in Type I Interferon Signaling. Front Immunol 2021; 12:748423. [PMID: 34691060 PMCID: PMC8529159 DOI: 10.3389/fimmu.2021.748423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/14/2021] [Indexed: 11/13/2022] Open
Abstract
The Type I Interferon family of cytokines all act through the same cell surface receptor and induce phosphorylation of the same subset of response regulators of the STAT family. Despite their shared receptor, different Type I Interferons have different functions during immune response to infection. In particular, they differ in the potency of their induced anti-viral and anti-proliferative responses in target cells. It remains not fully understood how these functional differences can arise in a ligand-specific manner both at the level of STAT phosphorylation and the downstream function. We use a minimal computational model of Type I Interferon signaling, focusing on Interferon-α and Interferon-β. We validate the model with quantitative experimental data to identify the key determinants of specificity and functional plasticity in Type I Interferon signaling. We investigate different mechanisms of signal discrimination, and how multiple system components such as binding affinity, receptor expression levels and their variability, receptor internalization, short-term negative feedback by SOCS1 protein, and differential receptor expression play together to ensure ligand specificity on the level of STAT phosphorylation. Based on these results, we propose phenomenological functional mappings from STAT activation to downstream anti-viral and anti-proliferative activity to investigate differential signal processing steps downstream of STAT phosphorylation. We find that the negative feedback by the protein USP18, which enhances differences in signaling between Interferons via ligand-dependent refractoriness, can give rise to functional plasticity in Interferon-α and Interferon-β signaling, and explore other factors that control functional plasticity. Beyond Type I Interferon signaling, our results have a broad applicability to questions of signaling specificity and functional plasticity in signaling systems with multiple ligands acting through a bottleneck of a small number of shared receptors.
Collapse
Affiliation(s)
- Duncan Kirby
- Department of Physics, University of Toronto, Toronto, ON, Canada
| | - Baljyot Parmar
- Department of Physics, University of Toronto, Toronto, ON, Canada
| | - Sepehr Fathi
- Department of Physics, University of Toronto, Toronto, ON, Canada
| | - Sagar Marwah
- Ajmera Family Transplant Centre, Toronto General Research Institute, Departments of Laboratory Medicine and Pathobiology and Immunology, University of Toronto, Toronto, ON, Canada
| | - Chitra R Nayak
- Department of Physics, University of Toronto, Toronto, ON, Canada.,Department of Physics, Tuskegee University, Tuskegee, AL, United States
| | - Vera Cherepanov
- Sandra Rotman Centre for Global Health, Toronto General Research Institute, University of Toronto, Toronto, ON, Canada
| | - Sonya MacParland
- Ajmera Family Transplant Centre, Toronto General Research Institute, Departments of Laboratory Medicine and Pathobiology and Immunology, University of Toronto, Toronto, ON, Canada
| | - Jordan J Feld
- Toronto Centre for Liver Disease, University Health Network, Toronto, ON, Canada
| | - Grégoire Altan-Bonnet
- Immunodynamics Group, Laboratory of Integrative Cancer Immunology, Center for Cancer Research (CCR), National Cancer Institute (NCI), Bethesda, MD, United States
| | - Anton Zilman
- Department of Physics, University of Toronto, Toronto, ON, Canada.,Institute for Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
50
|
Huang KCY, Chiang SF, Ke TW, Chen TW, Hu CH, Yang PC, Chang HY, Liang JA, Chen WTL, Chao KSC. DNMT1 constrains IFNβ-mediated anti-tumor immunity and PD-L1 expression to reduce the efficacy of radiotherapy and immunotherapy. Oncoimmunology 2021; 10:1989790. [PMID: 38283033 PMCID: PMC10813565 DOI: 10.1080/2162402x.2021.1989790] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/03/2021] [Indexed: 10/20/2022] Open
Abstract
Radiotherapy can boost the therapeutic response to immune checkpoint inhibitors (ICIs) by recruiting T lymphocytes and upregulating PD-L1 expression within the tumor microenvironment (TME). However, in some cases, tumor PD-L1 expression cannot be induced, even in the presence of abundant T lymphocytes, in locally advanced colorectal cancer patients who receive preoperative neoadjuvant concurrent chemoradiotherapy (CCRT). In this study, we found that PD-L1 promoter methylation is negatively correlated with tumor PD-L1 expression and is an independent biomarker for locally advanced colorectal cancer patients. PD-L1 methylation (mCD274) was significantly associated with shorter disease-free survival (cg15837913 loci, p = .0124). By multivariate Cox proportional hazards analyses including influent factors, mCD274 was classified as an independent prognostic factor for poor 5-year DFS [cg15837913, hazard ratio: HR = 4.06, 95% CI = 1.407-11.716, p = .01]. We found that the immunomodulatory agent DNA methyltransferase inhibitor (DNMTi) led to demethylation of the PD-L1 promoter and increased radiotherapy-induced PD-L1 upregulation via interferon β (IFNβ). DNMTi not only induced tumor PD-L1 expression but increased the expression of immune-related genes as well as intratumoral T cell infiltration in vivo. Furthermore, DNMTi strongly enhanced the response to combined treatment with radiotherapy and anti-PD-L1 inhibitors, and prolonged survival in microsatellite stability (MSS) colorectal model. Therefore, DNMTi remodeled the tumor microenvironment to improve the effect of radiotherapy and anti-PD-L1 immunotherapy by directly triggering tumor PD-L1 expression and eliciting stronger immune responses, which may provide potential clinical benefits to colorectal cancer patients in the future.
Collapse
Affiliation(s)
- Kevin Chih-Yang Huang
- Department of Biomedical Imaging and Radiological Science, China Medical University, TaichungTaiwan
- Translation Research Core, China Medical University Hospital, China Medical University, TaichungTaiwan
| | - Shu-Fen Chiang
- Lab of Precision Medicine, Feng-Yuan Hospital, TaichungTaiwan
| | - Tao-Wei Ke
- Department of Colorectal Surgery, China Medical University Hospital, China Medical University, TaichungTaiwan
- School of Chinese Medicine, China Medical University, TaichungTaiwan
| | - Tsung-Wei Chen
- Department of Pathology, Asia University Hospital, Asia University, TaichungTaiwan
- Graduate Institute of Biomedical Science, China Medical University, TaichungTaiwan
| | - Ching-Han Hu
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, TaichungTaiwan
| | - Pei-Chen Yang
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, TaichungTaiwan
| | - Hsin-Yu Chang
- Translation Research Core, China Medical University Hospital, China Medical University, TaichungTaiwan
| | - Ji-An Liang
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Radiotherapy, School of Medicine, China Medical University, TaichungTaiwan
| | - William Tzu-Liang Chen
- Department of Colorectal Surgery, China Medical University Hospital, China Medical University, TaichungTaiwan
- Department of Surgery, School of Medicine, China Medical University, TaichungTaiwan
- Department of Colorectal Surgery, China Medical University HsinChu Hospital, China Medical University, HsinChuTaiwan
| | - K. S. Clifford Chao
- Graduate Institute of Biomedical Science, China Medical University, TaichungTaiwan
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, TaichungTaiwan
- Department of Radiotherapy, School of Medicine, China Medical University, TaichungTaiwan
| |
Collapse
|