1
|
Han J, Wu M, Liu Z. Dysregulation in IFN-γ signaling and response: the barricade to tumor immunotherapy. Front Immunol 2023; 14:1190333. [PMID: 37275859 PMCID: PMC10233742 DOI: 10.3389/fimmu.2023.1190333] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/14/2023] [Indexed: 06/07/2023] Open
Abstract
Interferon-gamma (IFN-γ) has been identified as a crucial factor in determining the responsiveness to immunotherapy. Produced primarily by natural killer (NK) and T cells, IFN-γ promotes activation, maturation, proliferation, cytokine expression, and effector function in immune cells, while simultaneously inducing antigen presentation, growth arrest, and apoptosis in tumor cells. However, tumor cells can hijack the IFN-γ signaling pathway to mount IFN-γ resistance: rather than increasing antigenicity and succumbing to death, tumor cells acquire stemness characteristics and express immunosuppressive molecules to defend against antitumor immunity. In this review, we summarize the potential mechanisms of IFN-γ resistance occurring at two critical stages: disrupted signal transduction along the IFNG/IFNGR/JAK/STAT pathway, or preferential expression of specific interferon-stimulated genes (ISGs). Elucidating the molecular mechanisms through which tumor cells develop IFN-γ resistance help identify promising therapeutic targets to improve immunotherapy, with broad application value in conjugation with targeted, antibody or cellular therapies.
Collapse
Affiliation(s)
- Jiashu Han
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of General Surgery, Peking Union Medical College Hospital (CAMS), Beijing, China
| | - Mengwei Wu
- Department of General Surgery, Peking Union Medical College Hospital (CAMS), Beijing, China
| | - Ziwen Liu
- Department of General Surgery, Peking Union Medical College Hospital (CAMS), Beijing, China
| |
Collapse
|
2
|
Tarasov SA, Gorbunov EA, Don ES, Emelyanova AG, Kovalchuk AL, Yanamala N, Schleker ASS, Klein-Seetharaman J, Groenestein R, Tafani JP, van der Meide P, Epstein OI. Insights into the Mechanism of Action of Highly Diluted Biologics. THE JOURNAL OF IMMUNOLOGY 2020; 205:1345-1354. [PMID: 32727888 DOI: 10.4049/jimmunol.2000098] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 07/04/2020] [Indexed: 12/12/2022]
Abstract
The therapeutic use of Abs in cancer, autoimmunity, transplantation, and other fields is among the major biopharmaceutical advances of the 20th century. Broader use of Ab-based drugs is constrained because of their high production costs and frequent side effects. One promising approach to overcome these limitations is the use of highly diluted Abs, which are produced by gradual reduction of an Ab concentration to an extremely low level. This technology was used to create a group of drugs for the treatment of various diseases, depending on the specificity of the used Abs. Highly diluted Abs to IFN-γ (hd-anti-IFN-γ) have been demonstrated to be efficacious against influenza and other respiratory infections in a variety of preclinical and clinical studies. In the current study, we provide evidence for a possible mechanism of action of hd-anti-IFN-γ. Using high-resolution solution nuclear magnetic resonance spectroscopy, we show that the drug induced conformational changes in the IFN-γ molecule. Chemical shift changes occurred in the amino acids located primarily at the dimer interface and at the C-terminal region of IFN-γ. These molecular changes could be crucial for the function of the protein, as evidenced by an observed hd-anti-IFN-γ-induced increase in the specific binding of IFN-γ to its receptor in U937 cells, enhanced induced production of IFN-γ in human PBMC culture, and increased survival of influenza A-infected mice.
Collapse
Affiliation(s)
- Sergey A Tarasov
- OOO "NPF "Materia Medica Holding," 127473 Moscow, Russian Federation.,The Institute of General Pathology and Pathophysiology, 125315 Moscow, Russian Federation
| | | | - Elena S Don
- OOO "NPF "Materia Medica Holding," 127473 Moscow, Russian Federation.,The Institute of General Pathology and Pathophysiology, 125315 Moscow, Russian Federation
| | - Alexandra G Emelyanova
- OOO "NPF "Materia Medica Holding," 127473 Moscow, Russian Federation.,The Institute of General Pathology and Pathophysiology, 125315 Moscow, Russian Federation
| | | | - Naveena Yanamala
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260
| | - A Sylvia S Schleker
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260
| | - Judith Klein-Seetharaman
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260
| | | | | | | | - Oleg I Epstein
- OOO "NPF "Materia Medica Holding," 127473 Moscow, Russian Federation.,The Institute of General Pathology and Pathophysiology, 125315 Moscow, Russian Federation
| |
Collapse
|
3
|
Noncanonical IFN Signaling, Steroids, and STATs: A Probable Role of V-ATPase. Mediators Inflamm 2019; 2019:4143604. [PMID: 31275057 PMCID: PMC6558600 DOI: 10.1155/2019/4143604] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 04/15/2019] [Indexed: 11/27/2022] Open
Abstract
A small group of only seven transcription factors known as STATs (signal transducer and activator of transcription) are considered to be canonical determinants of specific gene activation for a plethora of ligand/receptor systems. The activation of STATs involves a family of four tyrosine kinases called JAK kinases. JAK1 and JAK2 activate STAT1 in the cytoplasm at the heterodimeric gamma interferon (IFNγ) receptor, while JAK1 and TYK2 activate STAT1 and STAT2 at the type I IFN heterodimeric receptor. The same STATs and JAKs are also involved in signaling by functionally different cytokines, growth factors, and hormones. Related to this, IFNγ-activated STAT1 binds to the IFNγ-activated sequence (GAS) element, but so do other STATs that are not involved in IFNγ signaling. Activated JAKs such as JAK2 and TYK2 are also involved in the epigenetics of nucleosome unwrapping for exposure of DNA to transcription. Furthermore, activated JAKs and STATs appear to function coordinately for specific gene activation. These complex events have not been addressed in canonical STAT signaling. Additionally, the function of noncoding enhancer RNAs, including their role in enhancer/promoter interaction is not addressed in the canonical STAT signaling model. In this perspective, we show that JAK/STAT signaling, involving membrane receptors, is essentially a variation of cytoplasmic nuclear receptor signaling. Focusing on IFN signaling, we showed that ligand, IFN receptor, the JAKs, and the STATs all undergo endocytosis and ATP-dependent nuclear translocation to promoters of genes specifically activated by IFNs. We argue here that the vacuolar ATPase (V-ATPase) proton pump probably plays a key role in endosomal membrane crossing by IFNs for receptor cytoplasmic binding. Signaling of nuclear receptors such as those of estrogen and dihydrotestosterone provides templates for making sense of the specificity of gene activation by closely related cytokines, which has implications for lymphocyte phenotypes.
Collapse
|
4
|
Monette A, Bergeron D, Ben Amor A, Meunier L, Caron C, Mes-Masson AM, Kchir N, Hamzaoui K, Jurisica I, Lapointe R. Immune-enrichment of non-small cell lung cancer baseline biopsies for multiplex profiling define prognostic immune checkpoint combinations for patient stratification. J Immunother Cancer 2019; 7:86. [PMID: 30922393 PMCID: PMC6437930 DOI: 10.1186/s40425-019-0544-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 02/22/2019] [Indexed: 12/21/2022] Open
Abstract
Background Permanence of front-line management of lung cancer by immunotherapies requires predictive companion diagnostics identifying immune-checkpoints at baseline, challenged by the size and heterogeneity of biopsy specimens. Methods An innovative, tumor heterogeneity reducing, immune-enriched tissue microarray was constructed from baseline biopsies, and multiplex immunofluorescence was used to profile 25 immune-checkpoints and immune-antigens. Results Multiple immune-checkpoints were ranked, correlated with antigen presenting and cytotoxic effector lymphocyte activity, and were reduced with advancing disease. Immune-checkpoint combinations on TILs were associated with a marked survival advantage. Conserved combinations validated on more than 11,000 lung, breast, gastric and ovarian cancer patients demonstrate the feasibility of pan-cancer companion diagnostics. Conclusions In this hypothesis-generating study, deepening our understanding of immune-checkpoint biology, comprehensive protein-protein interaction and pathway mapping revealed that redundant immune-checkpoint interactors associate with positive outcomes, providing new avenues for the deciphering of molecular mechanisms behind effects of immunotherapeutic agents targeting immune-checkpoints analyzed. Electronic supplementary material The online version of this article (10.1186/s40425-019-0544-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anne Monette
- Institut du cancer de Montréal, Montréal, Québec, Canada. .,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St-Denis, Tour Viger, Room R10-432, Montréal, Québec, H2X 0A9, Canada. .,Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Canada.
| | - Derek Bergeron
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Canada
| | - Amira Ben Amor
- Medicine Faculty of Tunis, Department of Immunology and Histology, Tunis El Manar University, Tunis, Tunisia
| | - Liliane Meunier
- Institut du cancer de Montréal, Montréal, Québec, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St-Denis, Tour Viger, Room R10-432, Montréal, Québec, H2X 0A9, Canada
| | - Christine Caron
- Institut du cancer de Montréal, Montréal, Québec, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St-Denis, Tour Viger, Room R10-432, Montréal, Québec, H2X 0A9, Canada
| | - Anne-Marie Mes-Masson
- Institut du cancer de Montréal, Montréal, Québec, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St-Denis, Tour Viger, Room R10-432, Montréal, Québec, H2X 0A9, Canada.,Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Canada
| | | | - Kamel Hamzaoui
- Medicine Faculty of Tunis, Department of Immunology and Histology, Tunis El Manar University, Tunis, Tunisia.,Abderrahmen Mami Hospital, Homeostasis and cell immune dysfunction Research Unit, Ariana, Tunisia
| | - Igor Jurisica
- Krembil Research Institute, UHN, 60 Leonard Avenue, Toronto, Ontario, M5T 0S8, Canada.,Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Réjean Lapointe
- Institut du cancer de Montréal, Montréal, Québec, Canada. .,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St-Denis, Tour Viger, Room R10-432, Montréal, Québec, H2X 0A9, Canada. .,Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Canada.
| |
Collapse
|
5
|
Rameshwar P, Moore CA, Shah NN, Smith CP. An Update on the Therapeutic Potential of Stem Cells. Methods Mol Biol 2018; 1842:3-27. [PMID: 30196398 DOI: 10.1007/978-1-4939-8697-2_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The seeming setbacks noted for stem cells underscore the need for experimental studies for safe and efficacious application to patients. Both clinical and experimental researchers have gained valuable knowledge on the characteristics of stem cells, and their behavior in different microenvironment. This introductory chapter focuses on adult mesenchymal stem cells (MSCs) based on the predominance in the clinic. MSCs can be influenced by inflammatory mediators to exert immune suppressive properties, commonly referred to as "licensing." Interestingly, while there are questions if other stem cells can be delivered across allogeneic barrier, there is no question on the ability of MSCs to provide this benefit. This property has been a great advantage since MSCs could be available for immediate application as "off-the-shelf" stem cells for several disorders, tissue repair and gene/drug delivery. Despite the benefit of MSCs, it is imperative that research continues with the various types of stem cells. The method needed to isolate these cells is outlined in this book. In parallel, safety studies are needed; particularly links to oncogenic event. In summary, this introductory chapter discusses several potential areas that need to be addressed for safe and efficient delivery of stem cells, and argue for the incorporation of microenvironmental factors in the studies. The method described in this chapter could be extrapolated to the field of chimeric antigen receptor T-cells (CAR-T). This will require application to stem cell hierarchy of memory T-cells.
Collapse
Affiliation(s)
- Pranela Rameshwar
- Department of Medicine-Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA.
| | - Caitlyn A Moore
- Division of Hematology/Oncology, Department of Medicine, University of Medicine and Dentistry of New Jersey-Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Niloy N Shah
- Division of Hematology/Oncology, Department of Medicine, University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark, NJ, USA
| | - Caroline P Smith
- Division of Hematology/Oncology, Department of Medicine, University of Medicine and Dentistry of New Jersey-Rutgers-New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
6
|
Londino JD, Gulick DL, Lear TB, Suber TL, Weathington NM, Masa LS, Chen BB, Mallampalli RK. Post-translational modification of the interferon-gamma receptor alters its stability and signaling. Biochem J 2017; 474:3543-3557. [PMID: 28883123 PMCID: PMC5967388 DOI: 10.1042/bcj20170548] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/28/2017] [Accepted: 09/05/2017] [Indexed: 12/28/2022]
Abstract
The IFN gamma receptor 1 (IFNGR1) binds IFN-γ and activates gene transcription pathways crucial for controlling bacterial and viral infections. Although decreases in IFNGR1 surface levels have been demonstrated to inhibit IFN-γ signaling, little is known regarding the molecular mechanisms controlling receptor stability. Here, we show in epithelial and monocytic cell lines that IFNGR1 displays K48 polyubiquitination, is proteasomally degraded, and harbors three ubiquitin acceptor sites at K277, K279, and K285. Inhibition of glycogen synthase kinase 3 beta (GSK3β) destabilized IFNGR1 while overexpression of GSK3β increased receptor stability. We identified critical serine and threonine residues juxtaposed to ubiquitin acceptor sites that impacted IFNGR1 stability. In CRISPR-Cas9 IFNGR1 generated knockout cell lines, cellular expression of IFNGR1 plasmids encoding ubiquitin acceptor site mutations demonstrated significantly impaired STAT1 phosphorylation and decreased STAT1-dependent gene induction. Thus, IFNGR1 undergoes rapid site-specific polyubiquitination, a process modulated by GSK3β. Ubiquitination appears to be necessary for efficient IFNGR1-dependent gamma gene induction and represents a relatively uncharacterized regulatory mechanism for this receptor.
Collapse
Affiliation(s)
- James D Londino
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Dexter L Gulick
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Travis B Lear
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Tomeka L Suber
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Nathaniel M Weathington
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Luke S Masa
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Bill B Chen
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Rama K Mallampalli
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, U.S.A.
- Medical Specialty Service Line, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, U.S.A
- Department of Cell Biology and Physiology and Bioengineering, University of Pittsburgh, Pittsburgh, PA, U.S.A
| |
Collapse
|
7
|
Green DS, Young HA, Valencia JC. Current prospects of type II interferon γ signaling and autoimmunity. J Biol Chem 2017; 292:13925-13933. [PMID: 28652404 PMCID: PMC5572907 DOI: 10.1074/jbc.r116.774745] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Interferon γ (IFNγ) is a pleiotropic protein secreted by immune cells. IFNγ signals through the IFNγ receptor, a protein complex that mediates downstream signaling events. Studies into IFNγ signaling have provided insight into the general concepts of receptor signaling, receptor internalization, regulation of distinct signaling pathways, and transcriptional regulation. Although IFNγ is the central mediator of the adaptive immune response to pathogens, it has been shown to be involved in several non-infectious physiological processes. This review will provide an introduction into IFNγ signaling biology and the functional roles of IFNγ in the autoimmune response.
Collapse
Affiliation(s)
- Daniel S Green
- From the Women's Malignancy Branch, Translational Genomics Section, Center for Cancer, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Howard A Young
- Cancer and Inflammation Program, Center for Cancer Research, NCI-Frederick, National Institutes of Health, Frederick, Maryland 21702-1201.
| | - Julio C Valencia
- Cancer and Inflammation Program, Center for Cancer Research, NCI-Frederick, National Institutes of Health, Frederick, Maryland 21702-1201.
| |
Collapse
|
8
|
Noncanonical IFN Signaling: Mechanistic Linkage of Genetic and Epigenetic Events. Mediators Inflamm 2016; 2016:9564814. [PMID: 28077919 PMCID: PMC5203919 DOI: 10.1155/2016/9564814] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 11/08/2016] [Indexed: 11/17/2022] Open
Abstract
The canonical model of cytokine signaling via the JAK/STAT pathway dominates our view of signal transduction but provides no insight into the significance of the simultaneous presence of activated JAKs and STATs in the nucleus of cells treated with cytokines. Such a mechanistic shortcoming challenges the usefulness of the model in its present form. Focusing on the interferon (IFN) cytokines, we have developed a noncanonical model of IFN signaling that naturally connects activated JAKs and STATs at or near response elements of genes that are activated by the IFNs. Specifically, cells treated with IFNγ showed association of activated STAT1α and JAK2 at the GAS element of genes activated by IFNγ. For IFNα treated cells, the association involved activated STAT1α and TYK2 JAK kinase at the ISRE promoter. The power of the noncanonical model is that it provides mechanistic insight into specific gene activation at the level of the associated epigenetics, akin to that of steroid/steroid receptor signaling.
Collapse
|
9
|
Endocytic regulation of cytokine receptor signaling. Cytokine Growth Factor Rev 2016; 32:63-73. [DOI: 10.1016/j.cytogfr.2016.07.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/13/2016] [Indexed: 12/11/2022]
|
10
|
Serban AI, Stanca L, Geicu OI, Dinischiotu A. AGEs-Induced IL-6 Synthesis Precedes RAGE Up-Regulation in HEK 293 Cells: An Alternative Inflammatory Mechanism? Int J Mol Sci 2015; 16:20100-17. [PMID: 26307981 PMCID: PMC4613191 DOI: 10.3390/ijms160920100] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/12/2015] [Accepted: 08/06/2015] [Indexed: 01/21/2023] Open
Abstract
Advanced glycation end products (AGEs) can activate the inflammatory pathways involved in diabetic nephropathy. Understanding these molecular pathways could contribute to therapeutic strategies for diabetes complications. We evaluated the modulation of inflammatory and oxidative markers, as well as the protective mechanisms employed by human embryonic kidney cells (HEK 293) upon exposure to 200 μg/mL bovine serum albumine (BSA) or AGEs–BSA for 12, 24 and 48 h. The mRNA and protein expression levels of AGEs receptor (RAGE) and heat shock proteins (HSPs) 27, 60 and 70, the activity of antioxidant enzymes and the expression levels of eight cytokines were analysed. Cell damage via oxidative mechanisms was evaluated by glutathione and malondialdehyde levels. The data revealed two different time scale responses. First, the up-regulation of interleukin-6 (IL-6), HSP 27 and high catalase activity were detected as early as 12 h after exposure to AGEs–BSA, while the second response, after 24 h, consisted of NF-κB p65, RAGE, HSP 70 and inflammatory cytokine up-regulation, glutathione depletion, malondialdehyde increase and the activation of antioxidant enzymes. IL-6 might be important in the early ignition of inflammatory responses, while the cellular redox imbalance, RAGE activation and NF-κB p65 increased expression further enhance inflammatory signals in HEK 293 cells.
Collapse
Affiliation(s)
- Andreea Iren Serban
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agronomical Sciences and Veterinary Medicine Bucharest, 105 Splaiul Independentei, district 5, Bucharest 050097, Romania.
| | - Loredana Stanca
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agronomical Sciences and Veterinary Medicine Bucharest, 105 Splaiul Independentei, district 5, Bucharest 050097, Romania.
| | - Ovidiu Ionut Geicu
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agronomical Sciences and Veterinary Medicine Bucharest, 105 Splaiul Independentei, district 5, Bucharest 050097, Romania.
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, district 5, Bucharest 050095, Romania.
| | - Anca Dinischiotu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, district 5, Bucharest 050095, Romania.
| |
Collapse
|
11
|
Poosti F, Bansal R, Yazdani S, Prakash J, Post E, Klok P, van den Born J, de Borst MH, van Goor H, Poelstra K, Hillebrands JL. Selective delivery of IFN-γ to renal interstitial myofibroblasts: a novel strategy for the treatment of renal fibrosis. FASEB J 2015; 29:1029-42. [PMID: 25466892 DOI: 10.1096/fj.14-258459] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Renal fibrosis leads to end-stage renal disease demanding renal replacement therapy because no adequate treatment exists. IFN-γ is an antifibrotic cytokine that may attenuate renal fibrosis. Systemically administered IFN-γ causes side effects that may be prevented by specific drug targeting. Interstitial myofibroblasts are the effector cells in renal fibrogenesis. Here, we tested the hypothesis that cell-specific delivery of IFN-γ to platelet-derived growth factor receptor β (PDGFRβ)-expressing myofibroblasts attenuates fibrosis in an obstructive nephropathy [unilateral ureteral obstruction (UUO)] mouse model. PEGylated IFN-γ conjugated to PDGFRβ-recognizing peptide [(PPB)-polyethylene glycol (PEG)-IFN-γ] was tested in vitro and in vivo for antifibrotic properties and compared with free IFN-γ. PDGFRβ expression was >3-fold increased (P < 0.05) in mouse fibrotic UUO kidneys and colocalized with α-smooth muscle actin-positive (SMA(+)) myofibroblasts. In vitro, PPB-PEG-IFN-γ significantly inhibited col1a1, col1a2, and α-SMA mRNA expression in TGF-β-activated NIH3T3 fibroblasts (P < 0.05). In vivo, PPB-PEG-IFN-γ specifically accumulated in PDGFRβ-positive myofibroblasts. PPB-PEG-IFN-γ treatment significantly reduced renal collagen I, fibronectin, and α-SMA mRNA and protein expression. Compared with vehicle treatment, PPB-PEG-IFN-γ preserved tubular morphology, reduced interstitial T-cell infiltration, and attenuated lymphangiogenesis (all P < 0.05) without affecting peritubular capillary density. PPB-PEG-IFN-γ reduced IFN-γ-related side effects as manifested by reduced major histocompatibility complex class II expression in brain tissue (P < 0.05 vs. free IFN-γ). Our findings demonstrate that specific targeting of IFN-γ to PDGFRβ-expressing myofibroblasts attenuates renal fibrosis and reduces systemic adverse effects.
Collapse
Affiliation(s)
- Fariba Poosti
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Ruchi Bansal
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Saleh Yazdani
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Jai Prakash
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Eduard Post
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Pieter Klok
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Jacob van den Born
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Martin H de Borst
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Harry van Goor
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Klaas Poelstra
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Jan-Luuk Hillebrands
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| |
Collapse
|
12
|
Ahmed CM, Johnson HM. Short peptide type I interferon mimetics: therapeutics for experimental allergic encephalomyelitis, melanoma, and viral infections. J Interferon Cytokine Res 2014; 34:802-9. [PMID: 24811478 DOI: 10.1089/jir.2014.0041] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The classical canonical model of interferon (IFN) signaling focuses solely on the activation of STAT transcription factors, which limits the model in terms of specific gene activation, associated epigenetic events, and IFN mimetic development. Accordingly, we have developed a noncanonical model of IFN signaling and report the development of short type I IFN peptide mimetic peptides based on the model. The mimetics, human IFNα1(152-189), human IFNβ(150-187), and ovine IFNτ(156-195) are derived from the C-terminus of the parent IFNs and function intracellularly based on the noncanonical model. Vaccinia virus produces a decoy IFN receptor (B18R) that inhibits type I IFN, but the IFN mimetics bypass B18R for effective antiviral activity. By contrast, both parent IFNs and mimetics inhibited vesicular stomatitis virus. The mimetics also possessed anti-tumor activity against murine melanoma B16 tumor cells in culture and in mice, including synergizing with suppressor of cytokine signaling 1 antagonist. Finally, the mimetics were potent therapeutics against experimental allergic encephalomyelitis, a mouse model of multiple sclerosis. The mimetics lack toxic side effects of the parent IFNs and, thus, are a potent therapeutic replacement of IFNs as therapeutics.
Collapse
Affiliation(s)
- Chulbul M Ahmed
- Department of Microbiology and Cell Science, University of Florida , Gainesville, Florida
| | | |
Collapse
|
13
|
Fu JP, Chen SN, Zou PF, Huang B, Guo Z, Zeng LB, Qin QW, Nie P. IFN-γ in turtle: conservation in sequence and signalling and role in inhibiting iridovirus replication in Chinese soft-shelled turtle Pelodiscus sinensis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 43:87-95. [PMID: 24239708 DOI: 10.1016/j.dci.2013.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 11/03/2013] [Accepted: 11/03/2013] [Indexed: 06/02/2023]
Abstract
The IFN-γ gene was identified in a turtle, the Chinese soft-shelled turtle, Pelodiscus sinensis, with its genome consisting of 4 exons and 3 introns. The deduced amino acid sequence of this gene contains a signal peptide, an IFN-γ family signature motif (130)IQRKAVNELFPT, an NLS motif (155)KRKR and three potential N-glycosylation sites. As revealed by real-time quantitative PCR, the gene was constitutively expressed in all tested organs/tissues, with higher level observed in blood, intestine and thymus. An induced expression of IFN-γ at mRNA level was observed in peripheral blood leucocytes (PBLs) in response to in vitro stimulation of LPS and PolyI:C. The overexpression of IFN-γ in the Chinese soft-shelled turtle artery (STA) cell line resulted in the increase in the expression of transcriptional regulators, such as IRF1, IRF7 and STAT1, and antiviral genes, such as Mx, PKR, implying possibly the existence of a conserved signalling network and role for IFN-γ in the turtle. Furthermore, the infection of soft-shelled turtle iridovirus (STIV) in the cell line transfected with IFN-γ may cause the cell death as demonstrated with the elevated lactate dehydrogenase (LDH) level and cell mortality. However, the mechanism involved in the antiviral activity may require further investigation.
Collapse
Affiliation(s)
- Jian Ping Fu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shan Nan Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province 430072, China
| | - Peng Fei Zou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province 430072, China
| | - Bei Huang
- College of Fisheries, Jimei University, 43 Yindou Road, Xiamen, Fujian Province 361021, China
| | - Zheng Guo
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province 430072, China
| | - Ling Bing Zeng
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, Hubei Province 430223, China
| | - Qi Wei Qin
- Key Laboratory of Tropical Marine Bioresources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou, Guangdong Province 510301, China
| | - Pin Nie
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province 430072, China; College of Fisheries, Jimei University, 43 Yindou Road, Xiamen, Fujian Province 361021, China.
| |
Collapse
|
14
|
Bansal R, Prakash J, De Ruiter M, Poelstra K. Targeted recombinant fusion proteins of IFNγ and mimetic IFNγ with PDGFβR bicyclic peptide inhibits liver fibrogenesis in vivo. PLoS One 2014; 9:e89878. [PMID: 24587093 PMCID: PMC3933682 DOI: 10.1371/journal.pone.0089878] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 01/24/2014] [Indexed: 01/17/2023] Open
Abstract
Hepatic stellate cells (HSCs), following transdifferentiation to myofibroblasts plays a key role in liver fibrosis. Therefore, attempts to attenuate this myofibroblastic phenotype would be a promising therapeutic approach. Interferon gamma (IFNγ) is a potent anti-fibrotic cytokine, but its pleiotropic receptor expression leading to severe adverse effects has limited its clinical application. Since, activated HSC express high-level of platelet derived growth factor beta receptor (PDGFβR), we investigated the potential of PDGFβR-specific targeting of IFNγ and its signaling peptide that lacks IFNγR binding site (mimetic IFNγ or mimIFNγ) in liver fibrosis. We prepared DNA constructs expressing IFNγ, mimIFNγ or BiPPB (PDGFβR-specific bicyclic peptide)-IFNγ, BiPPB-mimIFNγ fusion proteins. Both chimeric proteins alongwith IFNγ and mimIFNγ were produced in E.coli. The expressed proteins were purified and analyzed for PDGFβR-specific binding and in vitro effects. Subsequently, these recombinant proteins were investigated for the liver uptake (pSTAT1α signaling pathway), for anti-fibrotic effects and adverse effects (platelet counts) in CCl4-induced liver fibrogenesis in mice. The purified HSC-targeted IFNγ and mimIFNγ fusion proteins showed PDGFβR-specific binding and significantly reduced TGFβ-induced collagen-I expression in human HSC (LX2 cells), while mouse IFNγ and mimIFNγ did not show any effect. Conversely, mouse IFNγ and BiPPB-IFNγ induced activation and dose-dependent nitric oxide release in mouse macrophages (express IFNγR while lack PDGFβR), which was not observed with mimIFNγ and BiPPB-mimIFNγ, due to the lack of IFNγR binding sites. In vivo, targeted BiPPB-IFNγ and BiPPB-mimIFNγ significantly activated intrahepatic IFNγ-signaling pathway compared to IFNγ and mimIFNγ suggesting increased liver accumulation. Furthermore, the targeted fusion proteins ameliorated liver fibrogenesis in mice by significantly reducing collagen and α-SMA expression and potentiating collagen degradation. IFNγ also induced reduction in fibrogenesis but showed significant decrease in platelet counts, which was restored with targeted proteins. These results suggest that these rationally designed proteins can be further developed as novel anti-fibrotic therapeutics.
Collapse
Affiliation(s)
- Ruchi Bansal
- Department of Controlled Drug Delivery (Targeted Therapeutics), MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
- Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands
- * E-mail:
| | - Jai Prakash
- Department of Controlled Drug Delivery (Targeted Therapeutics), MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
- Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands
| | - Marieke De Ruiter
- Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands
| | - Klaas Poelstra
- Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
15
|
Bansal R, Prakash J, De Ruiter M, Poelstra K. Interferon gamma peptidomimetic targeted to hepatic stellate cells ameliorates acute and chronic liver fibrosis in vivo. J Control Release 2014; 179:18-24. [PMID: 24491909 DOI: 10.1016/j.jconrel.2014.01.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 01/20/2014] [Accepted: 01/23/2014] [Indexed: 01/21/2023]
Abstract
Hepatic stellate cells play a crucial role in the pathogenesis of hepatic fibrosis. Thus, pharmacological inhibition of pro-fibrotic activities of these cells might lead to an effective therapy for this disease. Among the potent anti-fibrotics, interferon gamma (IFNγ), a proinflammatory cytokine, is highly efficacious but it failed in clinical trials due to the poor efficacy and multiple adverse effects attributed to the ubiquitous IFNγ receptor (IFNγR) expression. To resolve these drawbacks, we chemically synthesized a chimeric molecule containing (a) IFNγ signaling peptide (IFNγ peptidomimetic, mimγ) that retains the agonistic activities of IFNγ but lacks an extracellular receptor recognition sequence for IFNγR; coupled via heterobifunctional PEG linker to (b) bicyclic platelet derived growth factor beta receptor (PDGFβR)-binding peptide (BiPPB) to induce internalization into the stellate cells that express PDGFβR. The synthesized targeted IFNγ peptidomimetic (mimγ-BiPPB) was extensively investigated for its anti-fibrotic and adverse effects in acute and chronic CCl4-induced liver fibrosis models in mice. Treatment with mimγ-BiPPB, after the onset of disease, markedly inhibited both early and established hepatic fibrosis as reflected by a reduced intrahepatic α-SMA, desmin and collagen-I mRNA expression and protein levels. While untargeted mimγ and BiPPB had no effect, and native IFNγ only induced a moderate reduction. Additionally, no off-target effects, e.g. systemic inflammation, were found with mimγ-BiPPB, which were substantially observed in mice treated with native IFNγ. The present study highlights the beneficial effects of a novel BiPPB mediated cell-specific targeting of IFNγ peptidomimetic to the disease-inducing cells and therefore represents a highly potential therapeutic approach to treat fibrotic diseases.
Collapse
Affiliation(s)
- Ruchi Bansal
- Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, The Netherlands; Department of Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands.
| | - Jai Prakash
- Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, The Netherlands; Department of Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Marieke De Ruiter
- Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, The Netherlands
| | - Klaas Poelstra
- Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, The Netherlands
| |
Collapse
|
16
|
Ahmed CM, Johnson HM. The role of a non-canonical JAK-STAT pathway in IFN therapy of poxvirus infection and multiple sclerosis: An example of Occam's Broom? JAKSTAT 2013; 2:e26227. [PMID: 24416655 PMCID: PMC3876437 DOI: 10.4161/jkst.26227] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 08/19/2013] [Accepted: 08/20/2013] [Indexed: 11/21/2022] Open
Abstract
Signaling by cytokines such as the interferons (IFNs) involves Janus kinases (JAKs) and signal transducer and activator of transcription (STAT) transcription factors. The beauty of the classical model of JAK-STAT signaling is its simplicity in that JAK-activated STATs in the nucleus are responsible for specific gene activation. The fact that many ligands, growth factors, and hormones use the same STAT transcription factors, but exert different functions at the level of the cell, tissue, and organ would suggest significant shortcomings in the classical model. Our studies have resulted in the development of a non-canonical, more complex model of IFN signaling that bears a striking resemblance to that of steroid hormone (SH)/steroid receptor (SR) signaling. Thus, both types I and II IFN signaling involves nuclear translocation of complexed ligand, receptor, activated JAKs, and activated STATs to the promoters of the genes that are specifically activated by the IFNs, where they are involved in specific gene activation and epigenetic remodeling. Receptor intracellular domains play an important role in binding the C-terminus of the IFNs, which is the basis for our development of IFN mimetics. The IFN mimetics are not recognized by poxvirus decoy receptors, since the decoy receptors compete for extracellular binding and not intracellular binding. Further, the type I IFN mimetics provide therapeutic protection against experimental allergic encephalomyelitis (EAE), a model of multiple sclerosis, without the side effects. Extracellular receptor binding by intact IFN is the primary reason for undesirable side effects of flu-like symptoms, bone-marrow suppression, and weight loss. The non-canonical model of IFN signaling thus provides insight into the specificity of such signaling and a mechanism for development of IFN mimetics. It is our contention that this model applies to other cytokines.
Collapse
Affiliation(s)
- Chulbul M Ahmed
- Department of Microbiology and Cell Science; University of Florida; Gainesville, FL USA
| | - Howard M Johnson
- Department of Microbiology and Cell Science; University of Florida; Gainesville, FL USA
| |
Collapse
|
17
|
Johnson HM, Noon-Song EN, Dabelic R, Ahmed CM. IFN signaling: how a non-canonical model led to the development of IFN mimetics. Front Immunol 2013; 4:202. [PMID: 23898330 PMCID: PMC3722551 DOI: 10.3389/fimmu.2013.00202] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 07/05/2013] [Indexed: 12/16/2022] Open
Abstract
The classical model of cytokine signaling dominates our view of specific gene activation by cytokines such as the interferons (IFNs). The importance of the model extends beyond cytokines and applies to hormones such as growth hormone (GH) and insulin, and growth factors such as epidermal growth factor (EGF) and fibroblast growth factor (FGF). According to this model, ligand activates the cell via interaction with the extracellular domain of the receptor. This results in activation of receptor or receptor-associated tyrosine kinases, primarily of the Janus activated kinase (JAK) family, phosphorylation and dimerization of the signal transducer and activator of transcription (STAT) transcription factors, which dissociate from the receptor cytoplasmic domain and translocate to the nucleus. This view ascribes no further role to the ligand, JAK kinase, or receptor in either specific gene activation or the associated epigenetic events. The presence of dimeric STATs in the nucleus essentially explains it all. Our studies have resulted in the development of a non-canonical, more complex model of IFNγ signaling that is akin to that of steroid hormone (SH)/steroid receptor (SR) signaling. We have shown that ligand, receptor, activated JAKs, and STATs are associated with specific gene activation, where the receptor subunit IFNGR1 functions as a co-transcription factor and the JAKs are involved in associated epigenetic events. We found that the type I IFN system functions similarly. The fact that GH receptor, insulin receptor, EGF receptor, and FGF receptor undergo nuclear translocation upon ligand binding suggests that they may also function similarly. The SH/SR nature of type I and II IFN signaling provides insight into the specificity of signaling by members of cytokine families. The non-canonical model could also provide better understanding to more complex cytokine families such as those of IL-2 and IL-12, whose members often use the same JAKs and STATs, but also have different functions and properties.
Collapse
Affiliation(s)
- Howard M Johnson
- Department of Microbiology and Cell Science, University of Florida , Gainesville, FL , USA
| | | | | | | |
Collapse
|
18
|
Ahmed CM, Noon-Song EN, Kemppainen K, Pascalli MP, Johnson HM. Type I IFN receptor controls activated TYK2 in the nucleus: implications for EAE therapy. J Neuroimmunol 2013; 254:101-9. [PMID: 23110939 PMCID: PMC3534922 DOI: 10.1016/j.jneuroim.2012.10.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 09/25/2012] [Accepted: 10/09/2012] [Indexed: 11/28/2022]
Abstract
Recent studies have suggested that activated wild-type and mutant Janus kinase JAK2 play a role in the epigenetics of histone modification, where it phosphorylates histone H3 on tyrosine 41(H3pY41). We showed that type I IFN signaling involves activated TYK2 in the nucleus. ChIP-PCR demonstrated the presence of receptor subunits IFNAR1 and IFNAR2 along with TYK2, STAT1, and H3pY41 specifically at the promoter of the OAS1 gene in IFN treated cells. A complex of IFNAR1, TYK2, and STAT1α was also shown in the nucleus by immunoprecipitation. IFN treatment was required for TYK2 activation in the nucleus. The presence of IFNAR1, IFNAR2, and activated STAT1 and STAT2, as well as the type I IFN in the nucleus of treated cells was confirmed by the combination of Western blotting and confocal microscopy. Trimethylated histone H3 lysine 9 underwent demethylation and subsequent acetylation specifically in the region of the OAS1 promoter. Resultant N-terminal truncated IFN mimetics functioned intracellularly as antivirals as well as therapeutics against experimental allergic encephalomyelitis without the undesirable side effects that limit the therapeutic efficacy of IFNβ in treatment of multiple sclerosis. The findings indicate that IFN signaling is complex like that of steroid signaling.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Apoptosis/physiology
- B-Lymphocytes/drug effects
- B-Lymphocytes/enzymology
- B-Lymphocytes/ultrastructure
- Cell Line, Transformed
- Cell Nucleus/drug effects
- Cell Nucleus/enzymology
- Chromatin Immunoprecipitation
- Culture Media, Serum-Free/pharmacology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/enzymology
- Enzyme Activation/drug effects
- Enzyme Activation/physiology
- Female
- Green Fluorescent Proteins/metabolism
- HeLa Cells/ultrastructure
- Humans
- Interferon-alpha/chemistry
- Mice
- Mice, Inbred Strains
- Peptides/pharmacology
- Peptides/therapeutic use
- Receptor, Interferon alpha-beta/genetics
- Receptor, Interferon alpha-beta/metabolism
- STAT Transcription Factors/metabolism
- TYK2 Kinase/metabolism
- Time Factors
- Transfection
Collapse
Affiliation(s)
- Chulbul M Ahmed
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, United States.
| | | | | | | | | |
Collapse
|
19
|
Yancoski J, Sadat MA, Aksentijevich N, Bernasconi A, Holland SM, Rosenzweig SD. A novel internalization motif regulates human IFN-γ R1 endocytosis. J Leukoc Biol 2012; 92:301-8. [PMID: 22595141 PMCID: PMC3395421 DOI: 10.1189/jlb.0212057] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 04/19/2012] [Accepted: 04/20/2012] [Indexed: 11/24/2022] Open
Abstract
This study tested the hypothesis that the IFN-γ R1 287-YVSLI-91 intracellular motif regulates its endocytosis. IFN-γ exerts its biological activities by interacting with a specific cell-surface RC composed of two IFN-γ R1 and two IFN-γ R2 chains. Following IFN-γ binding and along with the initiation of signal transduction, the ligand and IFN-γ R1 are internalized. Two major types of consensus-sorting signals are described in receptors, which are rapidly internalized from the plasma membrane to intracellular compartments: tyrosine-based and dileucine-based internalization motifs. Transfection of HEK 293 cells and IFN-γ R1-deficient fibroblasts with WT and site-directed, mutagenesis-generated mutant IFN-γ R1 expression vectors helped us to identify region IFN-γ R1 287-YVSLI-291 as the critical domain required for IFN-γ-induced IFN-γ R1 internalization and Y287 and LI290-291 as part of a common structure essential for receptor endocytosis and function. This new endocytosis motif, YxxLI, shares characteristics of tyrosine-based and dileucine-based internalization motifs and is highly conserved in IFN-γ Rs across species. The IFN-γ R1 270-LI-271 dileucine motif, previously thought to be involved in this receptor endocytosis, showed to be unnecessary for receptor endocytosis.
Collapse
Affiliation(s)
- Judith Yancoski
- Servicio de Inmunología y Reumatología, Hospital Nacional de Pediatría “J. P. Garrahan,” Buenos Aires, Argentina; and
| | - Mohammed A. Sadat
- Infectious Diseases Susceptibility Unit, Laboratory of Host Defenses, and
| | | | - Andrea Bernasconi
- Servicio de Inmunología y Reumatología, Hospital Nacional de Pediatría “J. P. Garrahan,” Buenos Aires, Argentina; and
| | - Steven M. Holland
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
20
|
Steroid-like signalling by interferons: making sense of specific gene activation by cytokines. Biochem J 2012; 443:329-38. [PMID: 22452815 DOI: 10.1042/bj20112187] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Many cytokines, hormones and growth factors use the JAK (Janus kinase)/STAT (signal transducer and activator of transcription) pathway for cell signalling and specific gene activation. In the classical model, ligand is said to interact solely with the receptor extracellular domain, which triggers JAK activation of STATs at the receptor cytoplasmic domain. Activated STATs are then said to carry out nuclear events of specific gene activation. Given the limited number of STATs (seven) and the activation of the same STATs by cytokines with different functions, the mechanism of the specificity of their signalling is not obvious. Focusing on IFNγ (interferon γ), we have shown that ligand, receptor and activated JAKs are involved in nuclear events that are associated with specific gene activation, where the receptor subunit IFNGR1 (IFNγ receptor 1) functions as a transcription/co-transcription factor and the JAKs are involved in key epigenetic events. RTKs (receptor tyrosine kinases) such as EGFR [EGF (epidermal growth factor) receptor] and FGFR [FGF (fibroblast growth factor) receptor] also undergo nuclear translocation in association with their respective ligands. EGFR and FGFR, like IFNGR1, have been shown to function as transcription/co-transcription factors. The RTKs also regulate other kinases that have epigenetic effects. Our IFNγ model, as well as the RTKs EGFR and FGFR, have similarities to that of steroid receptor signalling. These systems consist of ligand-receptor-co-activator complexes at the genes that they activate. The co-activators consist of transcription factors and kinases, of which the latter play an important role in the associated epigenetics. It is our view that signalling by cytokines such as IFNγ is but a variation of specific gene activation by steroid hormones.
Collapse
|
21
|
Abstract
Stem cells are considered as potential therapy for inflammatory disorders, tissue repair, and gene delivery, among others. The heterogeneity of a disease and the underlying disorder of a patient bring up the question on the method by which stem cells should be delivered. This summary discusses potential complex interactions among mediators at sites to tissue insults with stem cells. The chapter selects mesenchymal stem cells (MSCs) as a model, although the discussion is relevant to all stem cells. The review examines how MSCs and their differentiated cells can develop cross communication with soluble factors and cells within the region of tissue damage. Inflammatory cytokines, IL-1, TNFα, and TGFβ are selected to explain how they can affect the responses of MSCs, while predisposing the stem cells to oncogenic event. By understanding the varied functions of MSCs, one will be able to intervene to form a balance in functions, ultimately to achieve safety and efficient application. Cytokines can affect the expression of pluripotent genes such as REST and Oct-4. REST is a critical gene in the decision of a cell to express or repress neural genes. Since cytokines can affect microRNAs, the review incorporates this family of molecules as mediators of cytokine effects. IFNγ, although an inflammatory mediator, is central to the expression of MHC-II on MSCs. Therefore, it is included to discuss its role in the transplantation of stem cells across allogeneic barrier. In summary, this chapter discusses several potential areas that need to be addressed for safe and efficient delivery of stem cells, and argue for the incorporation of microenvironmental factors in the studies.
Collapse
|
22
|
Li F, Li QH, Wang JY, Zhan CY, Xie C, Lu WY. Effects of interferon-gamma liposomes targeted to platelet-derived growth factor receptor-beta on hepatic fibrosis in rats. J Control Release 2011; 159:261-70. [PMID: 22226772 DOI: 10.1016/j.jconrel.2011.12.023] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 10/11/2011] [Accepted: 12/19/2011] [Indexed: 12/13/2022]
Abstract
No drugs have been approved clinically for the therapy of hepatic fibrosis. Though interferon-γ (IFN-γ) is a highly effective anti-fibrotic agent in vitro and in some animal models in vivo, its anti-fibrotic potential in clinical trials has been disappointing, due to unwanted off-target effects and a short half-life period which results in poor efficacy. The aims of this study are to develop a new targeted drug delivery system to selectively deliver IFN-γ to hepatic stellate cells (HSCs) and to investigate whether it will improve the anti-fibrotic effect of IFN-γ and reduce its side effects in fibrotic livers. Sterically stable liposomes (SSLs) were modified by cyclic peptides (pPB) with a specific affinity for platelet-derived growth factor receptor-β (PDGFR-β), and then IFN-γ was encapsulated in the targeted liposomes (pPB-SSL-IFN-γ). In vitro, pPB-SSL was found to be taken up and internalized by cultured activated HSCs. The binding of FITC-labeled pPB-SSL to activated HSCs was in a time-dependent and concentration-dependent manner, which could be inhibited by excess unlabelled pPB-SSL, PDGF-BB, suramin or monensin. The inhibitory effect of pPB-SSL-IFN-γ on the proliferation of activated HSCs was respectively 7.24-fold and 2.95-fold higher than that of free IFN-γ and IFN-γ encapsulated in untargeted SSLs. In healthy rats, the tissue distribution, living-body tracing image analyses and pharmacokinetics study showed that pPB-SSL-IFN-γ accumulated mainly in the livers and had a longer half-life than free IFN-γ (3.98±0.52h vs. 0.21±0.03h). Furthermore, in rats with hepatic fibrosis induced by thioacetamide injection, FITC-labeled pPB-SSL was found to predominantly localize in activated HSCs by immunofluorescent double staining for FITC and albumin or α-smooth muscle actin (α-SMA). The enhanced anti-fibrotic effect of pPB-SSL-IFN-γ treatnment was indicated by significant decreases in the histologic Ishak stage, collagen I-staining positive areas, and α-SMA expression levels in fibrotic livers. In addition, pPB-SSL-IFN-γ treatment improved the leukopenia caused by low- and high-dosage free IFN-γ treatments. In conclusion, IFN-γ encapsulated in pPB-SSL had an extended circulation half-life and was selectively delivered to activated HSCs, which enhanced the anti-fibrotic effect of IFN-γ and reduced its side-effects in rats with hepatic fibrosis. Thus, pPB-SSL-IFN-γ may be an effective agent for the therapy of hepatic fibrosis.
Collapse
Affiliation(s)
- Feng Li
- Department of Gastroenterology, Zhongshan Hospital affiliated to Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
23
|
Hongqin T, Xinyu L, Heng G, Lanfang X, Yongfang W, Shasha S. Triptolide inhibits IFN-γ signaling via the Jak/STAT pathway in HaCaT keratinocytes. Phytother Res 2011; 25:1678-85. [PMID: 21433155 DOI: 10.1002/ptr.3471] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 12/23/2010] [Accepted: 02/10/2011] [Indexed: 01/04/2023]
Abstract
Interferon-gamma (IFN-γ) signaling in keratinocytes plays an important role in IFN-γ-mediated skin inflammation involved in psoriasis. Blocking IFN-γ signal transduction in keratinocytes could be a strategy for controlling inflammatory skin disorders. Tripterygium wilfordii Hook. F. (T. wilfordii) has been used effectively in psoriasis treatment in China. Its therapeutic mechanism on IFN-γ-dependent inflammation has not been elucidated. Triptolide is one of main components of T. wilfordii's antiinflammatory and immune effects. This study aimed to explore the effects of triptolide on an rhIFN-γ-stimulated human keratinocyte cell line (HaCaT) in culture. The expression of IFN-γ receptor α (IFN-γRα), phospho-Janus kinase2 (pJak2), phospho-signal transducer and activator of transcription 1 (pSTAT1) and suppressor of cytokine signaling 1 (SOCS1) was detected by western blotting. The expression of intercellular adhesion molecule-1 (ICAM-1) on the HaCaT cell surface was determined by cell-surface ELISA. The results demonstrated that triptolide inhibited the expression of IFN-γRα (IC₅₀ = 1.37 × 10⁻⁸ M), pJak2 (IC₅₀ = 2.82 × 10⁻⁹ M) and pSTAT1 (IC₅₀ = 1.29 × 10⁻⁹ M) in HaCaT cells. The expression of SOCS1 was up-regulated (ED₅₀ = 3.32 × 10⁻¹¹ M). Triptolide also significantly reduced the expression of ICAM-1 on the HaCaT cell surface (IC₅₀ = 5.82 × 10⁻¹⁰ M). This study suggests that triptolide may contribute to the therapeutic value of T. wilfordii by modulating the IFN-γ signal pathway in IFN-γ-dependent skin inflammatory diseases, including psoriasis.
Collapse
Affiliation(s)
- Tu Hongqin
- Department of Pharmacology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | | | | | | | | | | |
Collapse
|
24
|
Bansal R, Prakash J, de Ruijter M, Beljaars L, Poelstra K. Peptide-modified albumin carrier explored as a novel strategy for a cell-specific delivery of interferon gamma to treat liver fibrosis. Mol Pharm 2011; 8:1899-909. [PMID: 21800888 DOI: 10.1021/mp200263q] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Excessive accumulation of the extracellular matrix proteins primarily produced by activated hepatic stellate cells (HSC) leads to liver fibrosis. To date, no successful therapeutic intervention is available for the treatment of this disease. Platelet derived growth factor beta receptor (PDGFβR) is highly upregulated on disease-inducing activated HSC and thus can be used for delivery of antifibrotic drugs to increase therapeutic efficacy with reduced adverse effects. Interferon gamma (IFNγ) has been recognized as a potent antifibrotic cytokine; however, poor pharmacokinetics and side effects due to frequent administration have limited its clinical use. For HSC-specific delivery, a PDGFβR-specific drug delivery carrier (PPB-HSA) was developed by modifying albumin with PDGFβR-recognizing cyclic peptides. Subsequently, IFNγ was conjugated to PPB-HSA via bifunctional PEG linkers to synthesize PPB-HSA-PEG-IFNγ. In vitro, PPB-HSA-PEG-IFNγ retained complete biological activity similar to unmodified IFNγ and showed PDGFβR-specific binding to human HSC and primary culture-activated rat HSC. In TGFβ-stimulated mouse fibroblasts and human HSC, PPB-HSA-PEG-IFNγ induced significant reduction in crucial fibrotic parameters. In vivo, the conjugate rapidly accumulated into PDGFβR-expressing HSC in fibrotic livers and activated IFNγ-mediated pstat1α signaling pathway. Furthermore, in a CCl(4)-induced acute liver injury model in mice, treatment with HSC-targeted IFNγ strongly ameliorated hepatic fibrogenesis by inducing significant reduction (about 60%; p < 0.01) in collagen I and α-SMA expression as well as enhanced fibrolysis (increased MMP/TIMP ratio; p < 0.05) while free unmodified IFNγ was ineffective. Furthermore, in contrast to free native IFNγ, the conjugate did not induce macrophage infiltration and IL-1β expression in the liver. In conclusion, these data demonstrate the enhanced antifibrotic efficacy and reduced off-target effects of PPB-HSA-PEG-IFNγ conjugate showing the potential of cell-specific targeting of IFNγ for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Ruchi Bansal
- Department of Pharmacokinetics, Toxicology and Targeting, Graduate School for Drug Exploration, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
| | | | | | | | | |
Collapse
|
25
|
Bansal R, Prakash J, Post E, Beljaars L, Schuppan D, Poelstra K. Novel engineered targeted interferon-gamma blocks hepatic fibrogenesis in mice. Hepatology 2011; 54:586-96. [PMID: 21538439 DOI: 10.1002/hep.24395] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 04/19/2011] [Indexed: 01/13/2023]
Abstract
UNLABELLED Liver fibrogenesis is a process tightly controlled by endogenous anti- and pro-fibrogenic factors. Interferon gamma (IFNγ) is a potent antifibrogenic cytokine in vitro and might therefore represent a powerful therapeutic entity. However, its poor pharmacokinetics and adverse effects, due to the presence of IFNγ receptors on nearly all cells, prevented its clinical application so far. We hypothesized that delivery of IFNγ specifically to the disease-inducing cells and concurrently avoiding its binding to nontarget cells might increase therapeutic efficacy and avoid side effects. We conjugated IFNγ to a cyclic peptide recognizing the platelet-derived growth factor beta receptor (PDGFβR) which is strongly up-regulated on activated hepatic stellate cells (HSC), the key effector cells responsible for hepatic fibrogenesis. The IFNγ conjugates were analyzed in vitro for PDGFβR-specific binding and biological effects and in vivo in acute (early) and chronic (progressive and established) carbon-tetrachloride-induced liver fibrosis in mice. The targeted-IFNγ construct showed PDGFβR-specific binding to fibroblasts and HSC and inhibited their activation in vitro. In vivo, the targeted-IFNγ construct attenuated local HSC activation in an acute liver injury model. In the established liver fibrosis model, it not only strongly inhibited fibrogenesis but also induced fibrolysis. In contrast, nontargeted IFNγ was ineffective in both models. Moreover, in contrast to unmodified IFNγ, our engineered targeted-IFNγ did not induce IFNγ-related side effects such as systemic inflammation, hyperthermia, elevated plasma triglyceride levels, and neurotropic effects. CONCLUSION This study presents a novel HSC-targeted engineered-IFNγ, which in contrast to systemic IFNγ, blocked liver fibrogenesis and is devoid of side effects, by specifically acting on the key pathogenic cells within the liver.
Collapse
Affiliation(s)
- Ruchi Bansal
- Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
26
|
Johnson HM, Noon-Song E, Ahmed CM. Controlling Nuclear Jaks and Stats for Specific Gene Activation by Ifn γ and Other Cytokines: A Possible Steroid-like Connection. ACTA ACUST UNITED AC 2011; 2. [PMID: 22924155 DOI: 10.4172/2155-9899.1000112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The mechanism of specific gene activation by cytokines that use JAK/STAT signalling pathway is unknown. There are four different types of JAKs and seven different types of STATs. In the classical model of signaling, ligand interacts solely with the receptor extracellular domain, which triggers JAK activation at the receptor cytoplasmic domain. Activated STATs are then said to carry out nuclear events of specific gene activation, including associated epigenetic changes that cause heterochromatin destabilization. Ligand, receptor, and JAKs play no further role in the classical model. Given the limited number of STATs and the activation of the same STATs by cytokines with different functions, the mechanism of the specificity of their signalling is not obvious. Focusing on gamma interferon (IFNγ), we have shown that ligand, receptor, and activated JAKs are involved in nuclear events that are associated with specific gene activation. In this model, receptor subunit IFNGR1 functions as a transcription/cotranscription factor and the JAKs are involved in key epigenetic events that are required for specific gene activation. The model has implications for gene activation in cancer as well as stem cell differentiation.
Collapse
Affiliation(s)
- Howard M Johnson
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | | | | |
Collapse
|
27
|
Ahmed CMI, Dabelic R, Martin JP, Jager LD, Haider SM, Johnson HM. Enhancement of antiviral immunity by small molecule antagonist of suppressor of cytokine signaling. THE JOURNAL OF IMMUNOLOGY 2010; 185:1103-13. [PMID: 20543109 DOI: 10.4049/jimmunol.0902895] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Suppressors of cytokine signaling (SOCSs) are negative regulators of both innate and adaptive immunity via inhibition of signaling by cytokines such as type I and type II IFNs. We have developed a small peptide antagonist of SOCS-1 that corresponds to the activation loop of JAK2. SOCS-1 inhibits both type I and type II IFN activities by binding to the kinase activation loop via the kinase inhibitory region of the SOCS. The antagonist, pJAK2(1001-1013), inhibited the replication of vaccinia virus and encephalomyocarditis virus in cell culture, suggesting that it possesses broad antiviral activity. In addition, pJAK2(1001-1013) protected mice against lethal vaccinia and encephalomyocarditis virus infection. pJAK2(1001-1013) increased the intracellular level of the constitutive IFN-beta, which may play a role in the antagonist antiviral effect at the cellular level. Ab neutralization suggests that constitutive IFN-beta may act intracellularly, consistent with recent findings on IFN-gamma intracellular signaling. pJAK2(1001-1013) also synergizes with IFNs as per IFN-gamma mimetic to exert a multiplicative antiviral effect at the level of transcription, the cell, and protection of mice against lethal viral infection. pJAK2(1001-1013) binds to the kinase inhibitory region of both SOCS-1 and SOCS-3 and blocks their inhibitory effects on the IFN-gamma activation site promoter. In addition to a direct antiviral effect and synergism with IFN, the SOCS antagonist also exhibits adjuvant effects on humoral and cellular immunity as well as an enhancement of polyinosinic-polycytidylic acid activation of TLR3. The SOCS antagonist thus presents a novel and effective approach to enhancement of host defense against viruses.
Collapse
Affiliation(s)
- Chulbul M I Ahmed
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA.
| | | | | | | | | | | |
Collapse
|
28
|
Luheshi NM, Rothwell NJ, Brough D. Dual functionality of interleukin-1 family cytokines: implications for anti-interleukin-1 therapy. Br J Pharmacol 2010; 157:1318-29. [PMID: 19681864 PMCID: PMC2765320 DOI: 10.1111/j.1476-5381.2009.00331.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Dysregulated inflammation contributes to disease pathogenesis in both the periphery and the brain. Cytokines are coordinators of inflammation and were originally defined as secreted mediators, released from expressing cells to activate plasma membrane receptors on responsive cells. However, a group of cytokines is now recognized as having dual functionality. In addition to their extracellular effects, these cytokines act inside the nuclei of cytokine-expressing or cytokine-responsive cells. Interleukin-1 (IL-1) family cytokines are key pro-inflammatory mediators, and blockade of the IL-1 system in inflammatory diseases is an attractive therapeutic goal. All current therapies target IL-1 extracellular actions. Here we review evidence that suggests IL-1 family members have dual functionality. Several IL-1 family members have been detected inside the nuclei of IL-1-expressing or IL-1-responsive cells, and intranuclear IL-1 is reported to regulate gene transcription and mRNA splicing. However, further work is required to determine the impact of IL-1 intranuclear actions on disease pathogenesis. The intranuclear actions of IL-1 family members represent a new and potentially important area of IL-1 biology and may have implications for the future development of anti-IL-1 therapies.
Collapse
Affiliation(s)
- N M Luheshi
- Faculty of Life Sciences, University of Manchester, Manchester, UK.
| | | | | |
Collapse
|
29
|
Beurel E, Jope RS. Differential regulation of STAT family members by glycogen synthase kinase-3. J Biol Chem 2008; 283:21934-44. [PMID: 18550525 PMCID: PMC2494932 DOI: 10.1074/jbc.m802481200] [Citation(s) in RCA: 137] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Revised: 05/14/2008] [Indexed: 12/22/2022] Open
Abstract
Excessive neuroinflammation contributes to many neurological disorders and is poorly controlled therapeutically. The signal transducer and activator of transcription (STAT) family of transcription factors has a central role in inflammatory reactions, being stimulated by multiple cytokines and interferons and regulating the expression of many proteins involved in inflammation. We found that STAT3 activation is highly dependent on glycogen synthase kinase-3 (GSK3). Inhibitors of GSK3 greatly reduced (>75%) the activating STAT3 tyrosine phosphorylation in mouse primary astrocytes, microglia, and macrophage-derived RAW264.7 cells induced by interferon-gamma (IFNgamma), IFNalpha, interleukin-6, or insulin. GSK3 inhibitors blocked STAT3 DNA binding activity and the expression of STAT3-induced GFAP and Bcl-3. GSK3 dependence was selective for activation of STAT3 and STAT5, whereas STAT1 and STAT6 activation were GSK3-independent. Knockdown of the two GSK3 isoforms showed STAT3 and STAT5 activation were dependent on GSK3beta, but not GSK3alpha. The regulatory mechanism involved GSK3beta binding STAT3 and promoting its association with the IFNgamma receptor-associated intracellular signaling complex responsible for activating STAT3. Furthermore, GSK3beta associated with the IFNgamma receptor and was activated by stimulation with IFNgamma. Thus, inhibitors of GSK3 reduce the activation of STAT3 and STAT5, providing a mechanism to differentially regulate STATs to modulate the inflammatory response.
Collapse
Affiliation(s)
- Eléonore Beurel
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama, Birmingham, Alabama 35294-0017, USA
| | | |
Collapse
|
30
|
Fulcher AJ, Ahmed CMI, Noon-Song EN, Kwan RYQ, Subramaniam PS, Johnson HM, Jans DA. Interferon gamma is recognised by importin alpha/beta: enhanced nuclear localising and transactivation activities of an interferon gamma mimetic. FEBS Lett 2008; 582:1569-74. [PMID: 18405666 DOI: 10.1016/j.febslet.2008.03.054] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2008] [Revised: 03/04/2008] [Accepted: 03/07/2008] [Indexed: 11/30/2022]
Abstract
Interferon (IFN) gamma's ability to localise in the nucleus and function in gene activation has been known for some time, although the role of the conventional nuclear transporting importin molecules is unclear. Here, we demonstrate for the first time the direct recognition of IFNgamma and an IFNgamma mimetic peptide by IMPalpha and the IMPalpha/beta heterodimer, where the IFNgamma mimetic shows higher affinity. Significantly, this correlates well both with in vivo ability to target green fluorescent protein to the nucleus in transfected cells as determined by quantitative confocal laser scanning microscopy, as well as GAS promoter activity of a luciferase reporter. This has important implications for IFNgamma's anti-viral action, and the potential use of the IFNgamma mimetic in antiviral therapies.
Collapse
Affiliation(s)
- Alex J Fulcher
- Nuclear Signaling Laboratory, Department of Biochemistry and Molecular Biology, Monash University, P.O. Box 13D, Victoria, Clayton 3800, Australia
| | | | | | | | | | | | | |
Collapse
|
31
|
Wang T, Holland JW, Carrington A, Zou J, Secombes CJ. Molecular and Functional Characterization of IL-15 in Rainbow TroutOncorhynchus mykiss:A Potent Inducer of IFN-γ Expression in Spleen Leukocytes. THE JOURNAL OF IMMUNOLOGY 2007; 179:1475-88. [PMID: 17641013 DOI: 10.4049/jimmunol.179.3.1475] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-15 is a member of the common gamma-chain family of cytokines that possess a heterogeneous repertoire of activities on various cells of the immune system. We report here the first functional characterization of a fish IL-15 in rainbow trout. The trout IL-15 gene is 6-kb long and contains six exons and five introns that transcribe into a 1.2-kb mRNA containing seven out-of-frame AUG initiation codons and translate into a 193-aa peptide. Potential sites for transcriptional activators and repressors have been identified in the trout IL-15 gene. Like IL-15 from other species, trout IL-15 is closely linked to an INPP4B gene, but there is also a BCL10 gene located between the IL-15 and INPP4B genes. Three alternative splicing variants of the trout IL-15 gene have also been identified and their expression in vivo was studied. Trout IL-15 expression is present in all the tissues and cell lines studied. Recombinant trout IFN-gamma selectively increased IL-15 expression but had little effect on other cytokines such as IL-1 beta and IL-11. Recombinant trout IL-15 preferentially stimulated splenic leukocytes from healthy fish, where it induced a large increase in IFN-gamma expression, with little, if any, effect on IL-1 beta expression. This effect was quite long-lived, and was still apparent 24 h poststimulation. Although the exact cell types being affected have still to be determined, it is clear that once produced IL-15 will have a profound affect on the ability of the fish immune system to activate antimicrobial defenses and genes induced themselves by IFN-gamma.
Collapse
Affiliation(s)
- Tiehui Wang
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | | | | | | | | |
Collapse
|
32
|
Ahmed CM, Martin JP, Johnson HM. IFN mimetic as a therapeutic for lethal vaccinia virus infection: possible effects on innate and adaptive immune responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 178:4576-83. [PMID: 17372016 DOI: 10.4049/jimmunol.178.7.4576] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have developed small peptide mimetics of IFN-gamma that can bypass the poxvirus virulence factor B8R protein, which binds to intact IFN-gamma and prevents its interaction with receptor extracellular domain. Thus, these peptides inhibit vaccinia virus replication in cell culture where intact IFN-gamma is ineffective. We demonstrate here that the mouse IFN-gamma-mimetic peptide, IFN-gamma(95-132), protects C57BL/6 mice against overwhelming lethal vaccinia virus infection. The mimetic peptide was synthesized with an attached lipophilic group for penetration of cell plasma membrane. Injection of mimetic i.p. before and at the time of intranasal (10(6) PFU) or i.p. (10(7) PFU) challenge with virus resulted in complete protection at 200 microg of mimetic and 40-60% protection at 5 microg of mimetic. Initiation of treatment of mice with IFN-gamma mimetic up to 2 days postinfection resulted in complete protection against death, whereas initiation of treatment at 6 days postinfection resulted in 40% protection. Administration of mimetic by the oral route also completely protected mice against the intranasal route of a lethal dose of vaccinia virus challenge. In addition to its direct antiviral effect, the mimetic also possessed adjuvant effects in boosting humoral and cellular immunity to vaccinia virus. The combination of antiviral and adjuvant effects by the IFN mimetic probably plays a role in its potent anti-vaccinia virus properties. These results suggest an effective therapeutic against ongoing, lethal poxvirus infections that taps into innate and adaptive host defenses.
Collapse
Affiliation(s)
- Chulbul M Ahmed
- Department of Microbiology and Cell Science, University of Florida, Museum Road, Gainesville, FL 32611, USA.
| | | | | |
Collapse
|
33
|
Mujtaba MG, Patel CB, Patel RA, Flowers LO, Burkhart MA, Waiboci LW, Martin J, Haider MI, Ahmed CM, Johnson HM. The gamma interferon (IFN-gamma) mimetic peptide IFN-gamma (95-133) prevents encephalomyocarditis virus infection both in tissue culture and in mice. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2006; 13:944-52. [PMID: 16893996 PMCID: PMC1539128 DOI: 10.1128/cvi.00021-06] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2005] [Revised: 04/06/2006] [Accepted: 06/13/2006] [Indexed: 01/12/2023]
Abstract
We have demonstrated previously that the C-terminal gamma interferon (IFN-gamma) mimetic peptide consisting of residues 95 to 133 [IFN-gamma(95-133)], which contains the crucial IFN-gamma nuclear localization sequence (NLS), has antiviral activity in tissue culture. Here we evaluate the efficacy of this peptide and its derivatives first in vitro and then in an animal model of lethal viral infection with the encephalomyocarditis (EMC) virus. Deletion of the NLS region from the IFN-gamma mimetic peptide IFN-gamma(95-133) resulted in loss of antiviral activity. However, the NLS region does not have antiviral activity in itself. Replacing the NLS region of IFN-gamma(95-133) with the NLS region of the simian virus 40 large T antigen retains the antiviral activity in tissue culture. IFN-gamma(95-133) prevented EMC virus-induced lethality in mice in a dose-dependent manner compared to controls. Mice treated with IFN-gamma(95-133) had no or low EMC virus titers in their internal organs, whereas control mice had consistently high viral titers, especially in the heart tissues. Injection of B8R protein, which is encoded by poxviruses as a defense mechanism to neutralize host IFN-gamma, did not inhibit IFN-gamma(95-133) protection against a lethal dose of EMC virus, whereas mice treated with rat IFN-gamma were not protected. The data presented here show that the IFN-gamma mimetic peptide IFN-gamma(95-133) prevents EMC virus infection in vivo and in vitro and may have potential against other lethal viruses, such as the smallpox virus, which encodes the B8R protein.
Collapse
Affiliation(s)
- Mustafa G Mujtaba
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida 32611, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Ahmed CMI, Johnson HM. IFN-gamma and its receptor subunit IFNGR1 are recruited to the IFN-gamma-activated sequence element at the promoter site of IFN-gamma-activated genes: evidence of transactivational activity in IFNGR1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 177:315-21. [PMID: 16785527 DOI: 10.4049/jimmunol.177.1.315] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We have shown previously that IFN-gamma and one of its receptor subunits, IFNGR1, are translocated to the nucleus, together with STAT1alpha as one macromolecular complex, via the classical importin-dependent pathway. In this study, we have identified the nuclear targets of IFN-gamma and IFNGR1. By chromatin immunoprecipitation followed by PCR, IFN-gamma, its receptor subunit IFNGR1, and STAT1alpha were found to be associated with the IFN-gamma-activated sequence (GAS) in the promoter of two of the genes stimulated by IFN-gamma. Immunoprecipitated chromatin also showed the association of the IFN-gamma, IFNGR1, and STAT1alpha on the same DNA sequence. Examination of nuclear extracts from WISH cells treated with IFN-gamma revealed the specific binding of IFN-gamma, IFNGR1, and STAT1alpha to biotinylated GAS nucleotide sequence. Association of IFN-gamma, IFNGR1, and STAT1alpha with the GAS promoter was also demonstrated by EMSA. Transfection with a GAS-luciferase gene together with the IFNGR1 and nonsecreted IFN-gamma resulted in enhanced reporter activity. In addition, IFNGR1 fused to the yeast GAL4 DNA binding domain resulted in enhanced transcription from a GAL4 response element, suggesting the presence of a trans activation domain in IFNGR1. Our observations put IFN-gamma and its receptor subunit, IFNGR1, in direct contact with the promoter region of IFN-gamma-activated genes with associated increased activity, thus suggesting a transcriptional/cotranscriptional role for IFN-gamma/IFNGR1 as well as a possible role in determining the specificity of IFN-gamma action.
Collapse
Affiliation(s)
- Chulbul M I Ahmed
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA.
| | | |
Collapse
|
35
|
Ahmed CMI, Burkhart MA, Subramaniam PS, Mujtaba MG, Johnson HM. Peptide mimetics of gamma interferon possess antiviral properties against vaccinia virus and other viruses in the presence of poxvirus B8R protein. J Virol 2005; 79:5632-9. [PMID: 15827178 PMCID: PMC1082775 DOI: 10.1128/jvi.79.9.5632-5639.2005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
We have developed peptide mimetics of gamma interferon (IFN-gamma) that play a direct role in the activation and nuclear translocation of STAT1alpha transcription factor. These mimetics do not act through recognition by the extracellular domain of IFN-gamma receptor but rather bind to the cytoplasmic domain of the receptor chain 1, IFNGR-1, and thereby initiate the cellular signaling. Thus, we hypothesized that these mimetics would bypass the poxvirus virulence factor B8R protein that binds to intact IFN-gamma and prevents its interaction with the receptor. Human and murine IFN-gamma mimetic peptides were introduced into an adenoviral vector for intracellular expression. Murine IFN-gamma mimetic peptide was also expressed via chemical synthesis with an attached lipophilic group for penetration of cell plasma membrane. In contrast to intact human IFN-gamma, the mimetics did not bind poxvirus B8R protein, a homolog of the IFN-gamma receptor extracellular domain. Expression of B8R protein in WISH cells did not block the antiviral effect of the mimetics against encephalomyocarditis or vesicular stomatitis virus, while the antiviral activity of human IFN-gamma was neutralized. Consistent with the antiviral activity, the upregulation of MHC class I molecules on WISH cells by the IFN-gamma mimetics was not affected by B8R protein, while IFN-gamma-induced upregulation was blocked. Finally, the mimetics, but not IFN-gamma, inhibited vaccinia virus replication in African green monkey kidney BSC-40 cells. The data presented demonstrate that small peptide mimetics of IFN-gamma can avoid the B8R virulence factor for poxviruses and, thus, are potential candidates for antivirals against smallpox virus.
Collapse
Affiliation(s)
- Chulbul M I Ahmed
- University of Florida, Department of Microbiology and Cell Science, Building 981, Room 1052, P.O. Box 110700, Gainesville, FL 32611-0700, USA.
| | | | | | | | | |
Collapse
|
36
|
Shin-Ya M, Hirai H, Satoh E, Kishida T, Asada H, Aoki F, Tsukamoto M, Imanishi J, Mazda O. Intracellular interferon triggers Jak/Stat signaling cascade and induces p53-dependent antiviral protection. Biochem Biophys Res Commun 2005; 329:1139-46. [PMID: 15752772 DOI: 10.1016/j.bbrc.2005.02.088] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2005] [Indexed: 10/25/2022]
Abstract
Intracellular interferons (IFNs) exert biological functions similar to those of extracellular IFNs, but the signal transduction pathway triggered by the intracellular ligands has not been fully revealed. We investigated the signaling cascade by sequence-specific knockdown of signaling molecules by means of the RNA interference. Truncated IFN-beta gene was constructed so that the N-terminal secretory signal sequence was deleted (SD.IFN-beta). Cells transfected with this construct showed phosphorylation and activation of the STAT1 without any detectable secretion of the cytokine. The MHC class I expression was significantly augmented, while the augmentation was suppressed by short interfering RNA duplexes specific for JAK1, TYK2, and IFN-alpha/beta receptor (IFNAR) 1 and 2c chains. The SD.IFN-beta also induced p53 and phosphorylation of p53 at Ser(15). Specific silencing of p53 abrogated the antiviral effect of SD.IFN-beta, suggesting that the tumor suppressor is critically involved in antiviral defense mediated by intracellular IFN.
Collapse
Affiliation(s)
- Masaharu Shin-Ya
- Department of Microbiology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Johnson HM, Subramaniam PS, Olsnes S, Jans DA. Trafficking and signaling pathways of nuclear localizing protein ligands and their receptors. Bioessays 2004; 26:993-1004. [PMID: 15351969 DOI: 10.1002/bies.20086] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Interaction of ligands such as epidermal growth factor and interferon-gamma with the extracellular domains of their plasma membrane receptors results in internalization followed by translocation into the nucleus of the ligand and/or receptor. There has been reluctance, however, to ascribe signaling importance to this, the focus instead being on second messenger pathways, including mobilization of kinases and inducible transcription factors (TFs). The latter, however, fails to explain the fact that so many ligands stimulate the same second messenger cascades/TFs, and yet show distinct gene activation profiles. This is particularly apt in the case of the seven STAT TFs that are held to be the mediators of the distinct cellular functions of over 60 ligands. The current review focuses on five representative nuclear localizing ligands for which there is documentation of translocation into the cytosol and nucleus through well-characterized pathways, in addition to a role in gene activation by ligand/receptor in the nucleus.
Collapse
Affiliation(s)
- Howard M Johnson
- Department of Microbiology and Cell Science, University of Florida, FL, USA.
| | | | | | | |
Collapse
|