1
|
Aliaga-Gaspar P, Brichette-Mieg I, Fernández-Arjona M, Rodríguez-Bada JL, López-Moreno Y, Serrano-Castro P, Fernández-Fernández O, Ciano-Petersen NL, Oliver-Martos B. Recombinant soluble type I interferon receptor exerts antiviral activity by inducing proteins related to autophagy. Biomed Pharmacother 2024; 181:117678. [PMID: 39577364 DOI: 10.1016/j.biopha.2024.117678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/25/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
The soluble type I IFN receptor (sIFNAR2) is produced by alternative splicing and is present in body fluids. Although it can modulate IFN-ß activity, its biological role remains unknown. METHODS An in-silico study was conducted to compare the structure of recombinant human soluble IFNAR2 (r-sIFNAR2) with its native form. The antiviral activity of r-sIFNAR2, produced in BL21-bacteria and CHO cells, was tested using a cytopathic effect assay including appropriate controls. Viability and toxicity were assessed by MTT assays. Proteomic analysis using mass spectrometry was conducted in the A549/EMCV bioassay to elucidate the mechanism of action, and then it was validated by Western blot. RESULTS The BL21-sIFNAR2 had a sequence identity of 83.6 % with the native form, showing variations only in terminal regions. BL21-sIFNAR2 and CHO-sIFNAR2 showed significantly higher percentage of cell viability compared to the viral control, similar to IFN-ß. Cell viability with BL21-sIFNAR2 was comparable to the cell control across all tested concentrations. Proteomic analysis revealed an up regulation of pathways related with autophagy (macroautophagy, autophagy, pexophagy, and mitophagy) with an SQSTM1 overexpression that was then confirmed by Western Blot, especially after virus infection. However, pathways related to interferon signaling, and antiviral mechanisms mediated by IFN-stimulated genes were down-regulated. CONCLUSION r-sIFNAR2 exhibits significant antiviral activity regardless of the expression system used for its production and good safety profile, suggesting its use as a potential antiviral drug. Proteins related to autophagy are involved in the protection from the virus. This study highlights the biological relevance of soluble cytokine receptors as effectors so far overlooked.
Collapse
Affiliation(s)
- Pablo Aliaga-Gaspar
- Neuroimmunology and Neuroinflammation group. Biomedical Research Institute of Málaga-IBIMA Plataforma Bionand Hospital Regional Universitario de Málaga, Málaga, Spain; Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Isabel Brichette-Mieg
- Neuroimmunology and Neuroinflammation group. Biomedical Research Institute of Málaga-IBIMA Plataforma Bionand Hospital Regional Universitario de Málaga, Málaga, Spain
| | - MdM Fernández-Arjona
- Neuroimmunology and Neuroinflammation group. Biomedical Research Institute of Málaga-IBIMA Plataforma Bionand Hospital Regional Universitario de Málaga, Málaga, Spain
| | - José Luis Rodríguez-Bada
- Neuroimmunology and Neuroinflammation group. Biomedical Research Institute of Málaga-IBIMA Plataforma Bionand Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Yolanda López-Moreno
- Neuroimmunology and Neuroinflammation group. Biomedical Research Institute of Málaga-IBIMA Plataforma Bionand Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Pedro Serrano-Castro
- Neuroimmunology and Neuroinflammation group. Biomedical Research Institute of Málaga-IBIMA Plataforma Bionand Hospital Regional Universitario de Málaga, Málaga, Spain; Red Andaluza de Investigación Clínica y Traslacional en Neurología (Neuro-RECA), Málaga, Spain; Department of Medicine and Dermatology, Faculty of Medicine, University of Málaga, Málaga, Spain
| | | | - Nicolás Lundahl Ciano-Petersen
- Neuroimmunology and Neuroinflammation group. Biomedical Research Institute of Málaga-IBIMA Plataforma Bionand Hospital Regional Universitario de Málaga, Málaga, Spain; Red Andaluza de Investigación Clínica y Traslacional en Neurología (Neuro-RECA), Málaga, Spain.
| | - Begoña Oliver-Martos
- Neuroimmunology and Neuroinflammation group. Biomedical Research Institute of Málaga-IBIMA Plataforma Bionand Hospital Regional Universitario de Málaga, Málaga, Spain; Facultad de Medicina, Universidad de Málaga, Málaga, Spain; Department of Cell Biology, Genetics and Physiology, Physiology Area, Faculty of Science University of Malaga, Málaga, Spain.
| |
Collapse
|
2
|
de Weerd NA, Vivian JP, Lim SS, Huang SUS, Hertzog PJ. Structural integrity with functional plasticity: what type I IFN receptor polymorphisms reveal. J Leukoc Biol 2021; 108:909-924. [PMID: 33448473 DOI: 10.1002/jlb.2mr0420-152r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 03/21/2020] [Accepted: 03/26/2020] [Indexed: 12/13/2022] Open
Abstract
The type I IFNs activate an array of signaling pathways, which are initiated after IFNs bind their cognate receptors, IFNα/β receptor (IFNAR)1 and IFNAR2. These signals contribute to many aspects of human health including defense against pathogens, cancer immunosurveillance, and regulation of inflammation. How these cytokines interact with their receptors influences the quality of these signals. As such, the integrity of receptor structure is pivotal to maintaining human health and the response to immune stimuli. This review brings together genome wide association studies and clinical reports describing the association of nonsynonymous IFNAR1 and IFNAR2 polymorphisms with clinical disease, including altered susceptibility to viral and bacterial pathogens, autoimmune diseases, cancer, and adverse reactions to live-attenuated vaccines. We describe the amino acid substitutions or truncations induced by these polymorphisms and, using the knowledge of IFNAR conformational changes, IFNAR-IFN interfaces and overall structure-function relationship of the signaling complexes, we hypothesize the effect of these polymorphisms on receptor structure. That these predicted changes to IFNAR structure are associated with clinical manifestations of human disease, highlights the importance of IFNAR structural integrity to maintaining functional quality of these receptor-mediated responses. Type I IFNs are pivotal to innate immune responses and ultimately, to human health. Understanding the consequences of altered structure on the actions of these clinically significant cell receptors provides important information on the roles of IFNARs in health and disease.
Collapse
Affiliation(s)
- Nicole A de Weerd
- Centre for Innate Immunity and Infectious Diseases, Department of Molecular and Translational Science, Hudson Institute of Medical Research and Monash University, Clayton, Victoria, Australia
| | - Julian P Vivian
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute and Australian Research Council Centre for Excellence for Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia
| | - San S Lim
- Centre for Innate Immunity and Infectious Diseases, Department of Molecular and Translational Science, Hudson Institute of Medical Research and Monash University, Clayton, Victoria, Australia
| | - Stephanie U-Shane Huang
- Centre for Innate Immunity and Infectious Diseases, Department of Molecular and Translational Science, Hudson Institute of Medical Research and Monash University, Clayton, Victoria, Australia
| | - Paul J Hertzog
- Centre for Innate Immunity and Infectious Diseases, Department of Molecular and Translational Science, Hudson Institute of Medical Research and Monash University, Clayton, Victoria, Australia
| |
Collapse
|
3
|
Hurtado-Guerrero I, Hernáez B, Pinto-Medel MJ, Calonge E, Rodriguez-Bada JL, Urbaneja P, Alonso A, Mena-Vázquez N, Aliaga P, Issazadeh-Navikas S, Pavia J, Leyva L, Alcamí J, Alcamí A, Fernández Ó, Oliver-Martos B. Antiviral, Immunomodulatory and Antiproliferative Activities of Recombinant Soluble IFNAR2 without IFN-ß Mediation. J Clin Med 2020; 9:jcm9040959. [PMID: 32244308 PMCID: PMC7230527 DOI: 10.3390/jcm9040959] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/26/2020] [Accepted: 03/29/2020] [Indexed: 12/21/2022] Open
Abstract
Soluble receptors of cytokines are able to modify cytokine activities and therefore the immune system, and some have intrinsic biological activities without mediation from their cytokines. The soluble interferon beta (IFN-ß) receptor is generated through alternative splicing of IFNAR2 and has both agonist and antagonist properties for IFN-ß, but its role is unknown. We previously demonstrated that a recombinant human soluble IFN-ß receptor showed intrinsic therapeutic efficacy in a mouse model of multiple sclerosis. Here we evaluate the potential biological activities of recombinant sIFNAR2 without the mediation of IFN-ß in human cells. Recombinant sIFNAR2 down-regulated the production of IL-17 and IFN-ɣ and reduced the cell proliferation rate. Moreover, it showed a strong antiviral activity, fully protecting the cell monolayer after being infected by the virus. Specific inhibitors completely abrogated the antiviral activity of IFN-ß, but not that of the recombinant sIFNAR2, and there was no activation of the JAK-STAT signaling pathway. Consequently, r-sIFNAR2 exerts immunomodulatory, antiproliferative and antiviral activities without IFN-ß mediation, and could be a promising treatment against viral infections and immune-mediated diseases.
Collapse
Affiliation(s)
- Isaac Hurtado-Guerrero
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
- Neuroinflammation Unit, Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, Copenhagen Biocentre, University of Copenhagen, 2200 Copenhagen, Denmark;
| | - Bruno Hernáez
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain; (B.H.); (A.A.)
| | - María J. Pinto-Medel
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
| | - Esther Calonge
- AIDS Immunopathology Unit, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda 28220 Madrid, Spain; (E.C.); (J.A.)
| | - José L. Rodriguez-Bada
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
| | - Patricia Urbaneja
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
| | - Ana Alonso
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
| | - Natalia Mena-Vázquez
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC de Reumatología, Hospital Regional Universitario de Málaga, 29009 Málaga, Spain
| | - Pablo Aliaga
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Shohreh Issazadeh-Navikas
- Neuroinflammation Unit, Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, Copenhagen Biocentre, University of Copenhagen, 2200 Copenhagen, Denmark;
| | - José Pavia
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- Departamento de Farmacología y Pediatría, Facultad de Medicina. Universidad de Málaga, 29010 Málaga, Spain
| | - Laura Leyva
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
| | - José Alcamí
- AIDS Immunopathology Unit, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda 28220 Madrid, Spain; (E.C.); (J.A.)
- HIV Unit, Infectious Disease Service, Hospital Universitari de Bellvitge, 08907 Barcelona, Spain
| | - Antonio Alcamí
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain; (B.H.); (A.A.)
| | - Óscar Fernández
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- Departamento de Farmacología y Pediatría, Facultad de Medicina. Universidad de Málaga, 29010 Málaga, Spain
| | - Begoña Oliver-Martos
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
- Correspondence: ; Tel.: +34-951-290-223
| |
Collapse
|
4
|
Órpez-Zafra T, Pavía J, Hurtado-Guerrero I, Pinto-Medel MJ, Rodriguez Bada JL, Urbaneja P, Suardíaz M, Villar LM, Comabella M, Montalban X, Alvarez-Cermeño JC, Leyva L, Fernández Ó, Oliver-Martos B. Decreased soluble IFN-β receptor (sIFNAR2) in multiple sclerosis patients: A potential serum diagnostic biomarker. Mult Scler 2016; 23:937-945. [DOI: 10.1177/1352458516667564] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Background: The soluble isoform of the interferon-β (IFN-β) receptor (sIFNAR2) could modulate the activity of both endogenous and systemically administered IFN-β. Previously, we described lower serum sIFNAR2 levels in untreated multiple sclerosis (MS) than in healthy controls (HCs). Objective: To assess sIFNAR2 levels in a new cohort of MS patients and HCs, as well as in patients with clinically isolated syndrome (CIS) and with other inflammatory neurological disorders (OIND) and to assess its ability as a diagnostic biomarker. Methods: The cross-sectional study included 148 MS (84 treatment naive and 64 treated), 87 CIS, 42 OIND, and 96 HCs. Longitudinal study included 94 MS pretreatment and after 1 year of therapy with IFN-β, glatiramer acetate (GA), or natalizumab. sIFNAR2 serum levels were measured by a quantitative ELISA developed and validated in our laboratory. Results: Naive MS and CIS patients showed significantly lower sIFNAR2 levels than HCs and OIND patients. The sensitivity and specificity to discriminate between MS and OIND, for a sIFNAR2 cutoff value of 122.02 ng/mL, were 70.1%, and 79.4%, respectively. sIFNAR2 increased significantly in IFN-β-treated patients during the first year of therapy in contrast to GA- and natalizumab-treated patients who showed non-significant changes. Conclusion: The results suggest that sIFNAR2 could be a potential diagnostic biomarker for MS.
Collapse
Affiliation(s)
- Teresa Órpez-Zafra
- Unidad de Gestión Clínica de Neurociencias, Instituto de Biomedicina de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Jose Pavía
- Departamento de Farmacología y Pediatría, Facultad de Medicina, Instituto de Biomedicina de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
| | - Isaac Hurtado-Guerrero
- Unidad de Gestión Clínica de Neurociencias, Instituto de Biomedicina de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Maria J Pinto-Medel
- Unidad de Gestión Clínica de Neurociencias, Instituto de Biomedicina de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain/Red Española de Esclerosis Múltiple (REEM)
| | - Jose Luis Rodriguez Bada
- Unidad de Gestión Clínica de Neurociencias, Instituto de Biomedicina de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Patricia Urbaneja
- Unidad de Gestión Clínica de Neurociencias, Instituto de Biomedicina de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Margarita Suardíaz
- Unidad de Gestión Clínica de Neurociencias, Instituto de Biomedicina de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain/Red Española de Esclerosis Múltiple (REEM)
| | - Luisa M Villar
- Servicio de Inmunología, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain/Red Española de Esclerosis Múltiple (REEM)
| | - Manuel Comabella
- Department de Neurología-Neuroinmunología, Centre d’Esclerosi Múltiple de Catalunya, Vall d’Hebron Institut de Recerca, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain/Red Española de Esclerosis Múltiple (REEM)
| | - Xavier Montalban
- Department de Neurología-Neuroinmunología, Centre d’Esclerosi Múltiple de Catalunya, Vall d’Hebron Institut de Recerca, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain/Red Española de Esclerosis Múltiple (REEM)
| | - Jose C Alvarez-Cermeño
- Servicio de Neurología, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain/Red Española de Esclerosis Múltiple (REEM)
| | - Laura Leyva
- Unidad de Gestión Clínica de Neurociencias, Instituto de Biomedicina de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain/Red Española de Esclerosis Múltiple (REEM)
| | - Óscar Fernández
- Unidad de Gestión Clínica de Neurociencias, Instituto de Biomedicina de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain/Red Española de Esclerosis Múltiple (REEM)
| | - Begoña Oliver-Martos
- Unidad de Gestión Clínica de Neurociencias, Instituto de Biomedicina de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain/Red Española de Esclerosis Múltiple (REEM)
| |
Collapse
|
5
|
Development and validation of an ELISA for quantification of soluble IFN-β receptor: assessment in multiple sclerosis. Bioanalysis 2015; 7:2869-80. [DOI: 10.4155/bio.15.208] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Aim: The soluble isoform of the IFN-β receptor (sIFNAR2) can bind IFN-β and modulate its activity, although its role in autoimmune diseases remains unknown. Methods: A recombinant human sIFNAR2 protein was cloned, expressed and purified after which we developed and validated an ELISA for its quantification in human serum. Serum sIFNAR2 were assessed in multiple sclerosis (MS) patients and healthy controls. Results: The ELISA has a dynamic range of 3.9–250 ng/ml and a detection limit of 2.44 ng/ml. Serum sIFNAR2 were significantly lower in untreated-MS patients than in healthy controls. Conclusion: The ELISA is suitable for quantification of sIFNAR2 in serum and should facilitate the study of sIFNAR2 in neuroimmunological diseases such as MS.
Collapse
|
6
|
Rubinstein MP, Salem ML, Doedens AL, Moore CJ, Chiuzan C, Rivell GL, Cole DJ, Goldrath AW. G-CSF/anti-G-CSF antibody complexes drive the potent recovery and expansion of CD11b+Gr-1+ myeloid cells without compromising CD8+ T cell immune responses. J Hematol Oncol 2013; 6:75. [PMID: 24279871 PMCID: PMC3850648 DOI: 10.1186/1756-8722-6-75] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 09/02/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Administration of recombinant G-CSF following cytoreductive therapy enhances the recovery of myeloid cells, minimizing the risk of opportunistic infection. Free G-CSF, however, is expensive, exhibits a short half-life, and has poor biological activity in vivo. METHODS We evaluated whether the biological activity of G-CSF could be improved by pre-association with anti-G-CSF mAb prior to injection into mice. RESULTS We find that the efficacy of G-CSF therapy can be enhanced more than 100-fold by pre-association of G-CSF with an anti-G-CSF monoclonal antibody (mAb). Compared with G-CSF alone, administration of G-CSF/anti-G-CSF mAb complexes induced the potent expansion of CD11b+Gr-1+ myeloid cells in mice with or without concomitant cytoreductive treatment including radiation or chemotherapy. Despite driving the dramatic expansion of myeloid cells, in vivo antigen-specific CD8+ T cell immune responses were not compromised. Furthermore, injection of G-CSF/anti-G-CSF mAb complexes heightened protective immunity to bacterial infection. As a measure of clinical value, we also found that antibody complexes improved G-CSF biological activity much more significantly than pegylation. CONCLUSIONS Our findings provide the first evidence that antibody cytokine complexes can effectively expand myeloid cells, and furthermore, that G-CSF/anti-G-CSF mAb complexes may provide an improved method for the administration of recombinant G-CSF.
Collapse
Affiliation(s)
- Mark P Rubinstein
- Department of Biological Sciences, The University of California, San Diego, La Jolla, CA 92093, USA
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, 86 Jonathan Lucas Street, HO506, SC 29403, USA
| | - Mohamed L Salem
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
- Zoology Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Andrew L Doedens
- Department of Biological Sciences, The University of California, San Diego, La Jolla, CA 92093, USA
| | - Caitlin J Moore
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Cody Chiuzan
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Guillermo L Rivell
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
| | - David J Cole
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Ananda W Goldrath
- Department of Biological Sciences, The University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
7
|
Piehler J, Thomas C, Garcia KC, Schreiber G. Structural and dynamic determinants of type I interferon receptor assembly and their functional interpretation. Immunol Rev 2012; 250:317-34. [PMID: 23046138 PMCID: PMC3986811 DOI: 10.1111/imr.12001] [Citation(s) in RCA: 176] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Type I interferons (IFNs) form a network of homologous cytokines that bind to a shared, heterodimeric cell surface receptor and engage signaling pathways that activate innate and adaptive immune responses. The ability of IFNs to mediate differential responses through the same cell surface receptor has been subject of a controversial debate and has important medical implications. During the past decade, a comprehensive insight into the structure, energetics, and dynamics of IFN recognition by its two-receptor subunits, as well as detailed correlations with their functional properties on the level of signal activation, gene expression, and biological responses were obtained. All type I IFNs bind the two-receptor subunits at the same sites and form structurally very similar ternary complexes. Differential IFN activities were found to be determined by different lifetimes and ligand affinities toward the receptor subunits, which dictate assembly and dynamics of the signaling complex in the plasma membrane. We present a simple model, which explains differential IFN activities based on rapid endocytosis of signaling complexes and negative feedback mechanisms interfering with ternary complex assembly. More insight into signaling pathways as well as endosomal signaling and trafficking will be required for a comprehensive understanding, which will eventually lead to therapeutic applications of IFNs with increased efficacy.
Collapse
Affiliation(s)
- Jacob Piehler
- Department of Biology, University of Osnabrück, Osnabrück, Germany
| | - Christoph Thomas
- Departments of Molecular and Cellular Physiology, and Structural Biology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - K. Christopher Garcia
- Departments of Molecular and Cellular Physiology, and Structural Biology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Gideon Schreiber
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
8
|
Witte K, Witte E, Sabat R, Wolk K. IL-28A, IL-28B, and IL-29: promising cytokines with type I interferon-like properties. Cytokine Growth Factor Rev 2010; 21:237-51. [PMID: 20655797 DOI: 10.1016/j.cytogfr.2010.04.002] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
IL-28A, IL-28B and IL-29 (also designated type III interferons) constitute a new subfamily within the IL-10-interferon family. They are produced by virtually any nucleated cell type, particularly dendritic cells, following viral infection or activation with bacterial components, and mediate their effects via the IL-28R1/IL-10R2 receptor complex. Although IL-28/IL-29 are closer to the IL-10-related cytokines in terms of gene structure, protein structure, and receptor usage, they display type I interferon-like anti-viral and cytostatic activities. Unlike type I interferons, the target cell populations of IL-28/IL-29 are restricted and mainly include epithelial cells and hepatocytes. These properties suggest that IL-28/IL-29 are potential therapeutic alternatives to type I interferons in terms of viral infections and tumors. This review describes the current knowledge about these cytokines.
Collapse
Affiliation(s)
- Katrin Witte
- Interdisciplinary Group of Molecular Immunopathology, Dermatology/Medical Immunology, University Hospital Charité, Charitéplatz 1, 10117 Berlin, Germany
| | | | | | | |
Collapse
|
9
|
Schreiber G, Walter MR. Cytokine-receptor interactions as drug targets. Curr Opin Chem Biol 2010; 14:511-9. [PMID: 20619718 DOI: 10.1016/j.cbpa.2010.06.165] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Revised: 05/30/2010] [Accepted: 06/08/2010] [Indexed: 12/24/2022]
Abstract
Cytokines are essential proteins that exert potent control over entire cell populations to fight infections and other pathologies, but can by themselves cause disease. Therefore, cytokine-related drugs act either by stimulating or blocking their activities. Our knowledge of the structures of cytokine-receptor complexes, the biophysical basis of their binding, and their mode of biological activation has substantially increased in recent years. This knowledge has been translated into new drugs and drug candidates. This review summarizes our current understanding of the receptor-mediated activity of cytokines, their relation to health and disease, and the agents in use to activate and block their actions.
Collapse
Affiliation(s)
- Gideon Schreiber
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel.
| | | |
Collapse
|
10
|
Hermesh T, Moltedo B, Moran TM, López CB. Antiviral instruction of bone marrow leukocytes during respiratory viral infections. Cell Host Microbe 2010; 7:343-53. [PMID: 20478536 PMCID: PMC2874206 DOI: 10.1016/j.chom.2010.04.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Revised: 02/04/2010] [Accepted: 03/26/2010] [Indexed: 12/19/2022]
Abstract
Respiratory viral infections trigger a robust inflammatory response in the lung, producing cytokines, chemokines, and growth factors that promote infiltration of effector leukocytes. Whereas the role of chemokines and infiltrating leukocytes in antiviral immunity is well studied, the effect that lung cytokines have on leukocytes in distal hematopoietic and lymphoid tissues and their role in antiviral immunity is unknown. We show that, during infection with influenza or Sendai virus, the lung communicates with the sterile bone marrow, the primary site of hematopoiesis, through type I interferons. While in the bone marrow, leukocytes exposed to type I interferons activate an antiviral transcriptional program and become resistant to infection with different viruses. The protected bone marrow leukocytes are capable of migrating to the infected lung and contribute to virus clearance. These findings show that appropriate instruction of cells during their development in the bone marrow is needed for effective control of infection.
Collapse
Affiliation(s)
- Tamar Hermesh
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | - Bruno Moltedo
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | - Thomas M. Moran
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | - Carolina B. López
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| |
Collapse
|
11
|
Schmeisser H, Gorshkova I, Brown PH, Kontsek P, Schuck P, Zoon KC. Two interferons alpha influence each other during their interaction with the extracellular domain of human type interferon receptor subunit 2. Biochemistry 2007; 46:14638-49. [PMID: 18027911 DOI: 10.1021/bi7012036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The interaction between two human interferons alpha (IFN-alphas) and the extracellular (EC) domain of human type I IFN receptor subunit 2 (IFNAR2) was analyzed. Previous experiments using Daudi cells showed that IFN-alpha21b and some IFN-alpha hybrids (made from IFN-alpha2c and 21b) competed poorly for the IFN-alpha2b binding site. This study examined the causes of the poor competition between these IFN-alphas. IFN-alpha2c and the IFN hybrid CM3 {IFN-alpha21b(1-75)(81-95)/IFN-alpha2c(76-80) (96-166), Y86K} were selected for this study based on their cell binding and biological properties. Competitive binding ELISA, native electrophoresis followed by Western blot, electrospray ionization mass spectrometry (ESI-MS), surface plasmon resonance biosensor (SPR) analysis, as well as neutralization of antiproliferative activities on Daudi cells in the presence of soluble IFNAR2-EC show evidence that each of the described IFN-alpha subtypes affected the binding of the other IFN-alpha to IFNAR2-EC by affecting the stability of the complex, i.e., dissociation of the complex. Moreover, native electrophoresis with different IFNAR2-EC mutants showed that IFN-alpha2c and CM3 utilize different amino acids in the binding domain of IFNAR2-EC. In addition to that, analytical ultracentrifugation (AUC) revealed differences in the oligomeric state of the two studied interferons. Our results demonstrated that two individual IFN-alphas interact differentially with IFNAR2-EC and influence each other during this interaction. This study contributes to the understanding of the mutual interaction between multiple IFN-alpha subtypes during the competition for binding to the receptor.
Collapse
Affiliation(s)
- Hana Schmeisser
- National Institute of Allergy and Infectious Diseases and National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | |
Collapse
|
12
|
Beltrao P, Kiel C, Serrano L. Structures in systems biology. Curr Opin Struct Biol 2007; 17:378-84. [PMID: 17574836 DOI: 10.1016/j.sbi.2007.05.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2007] [Revised: 04/11/2007] [Accepted: 05/29/2007] [Indexed: 11/18/2022]
Abstract
Oil and water do not normally mix, and apparently structural biology and systems biology look like two different universes. It can be argued that structural biology could play a very important role in systems biology. Although at the final stage of understanding a signal transduction pathway, a cell, an organ or a living system, structures could be obviated, we need them to be able to reach that stage. Structures of macromolecules, especially molecular machines, could provide quantitative parameters, help to elucidate functional networks or enable rational designed perturbation experiments for reverse engineering. The role of structural biology in systems biology should be to provide enough understanding so that macromolecules can be translated into dots or even into equations devoid of atoms.
Collapse
Affiliation(s)
- Pedro Beltrao
- European Molecular Biology Laboratory, Meyerhofstrasse 1, Heidelberg D69115, Germany
| | | | | |
Collapse
|
13
|
Wolk K, Witte E, Hoffmann U, Doecke WD, Endesfelder S, Asadullah K, Sterry W, Volk HD, Wittig BM, Sabat R. IL-22 induces lipopolysaccharide-binding protein in hepatocytes: a potential systemic role of IL-22 in Crohn's disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 178:5973-81. [PMID: 17442982 DOI: 10.4049/jimmunol.178.9.5973] [Citation(s) in RCA: 217] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Crohn's disease (CD) is a common, chronic, inflammatory bowel disease characterized by intestinal infiltration of activated immune cells and distortion of the intestinal architecture. In this study, we demonstrate that IL-22, a cytokine that is mainly produced by activated Th1 and Th17 cells, was present in high quantities in the blood of CD patients in contrast to IFN-gamma and IL-17. In a mouse colitis model, IL-22 mRNA expression was elevated predominantly in the inflamed intestine but also in the mesenteric lymph nodes. IL-22BP, the soluble receptor for IL-22, demonstrated an affinity to IL-22 that was at least 4-fold higher than its membrane-bound receptor, and its strong constitutive expression in the intestine and lymph nodes was decreased in the inflamed intestine. To investigate the possible role of systemic IL-22 in CD, we then administered IL-22 to healthy mice and found an up-regulation of LPS-binding protein (LBP) blood levels reaching concentrations known to neutralize LPS. This systemic up-regulation was associated with increased hepatic but not renal or pulmonary LBP mRNA levels. IL-22 also enhanced the secretion of LBP in human primary hepatocytes and HepG2 hepatoma cells in vitro. This increase was mainly transcriptionally regulated and synergistic with that of other LBP inducers. Finally, elevated LBP levels were detected in CD patients and the mouse colitis model. These data suggest that systemic IL-22 may contribute to the prevention of systemic inflammation provoked by LPS present in the blood of CD patients through its induction of hepatic LBP.
Collapse
Affiliation(s)
- Kerstin Wolk
- Interdisciplinary Group of Molecular Immunopathology, Dermatology/Medical Immunology, Campus Charité Mitte, University Hospital Charité, Charitéplatz 1, Berlin, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Rubinstein MP, Kovar M, Purton JF, Cho JH, Boyman O, Surh CD, Sprent J. Converting IL-15 to a superagonist by binding to soluble IL-15R{alpha}. Proc Natl Acad Sci U S A 2006; 103:9166-71. [PMID: 16757567 PMCID: PMC1482584 DOI: 10.1073/pnas.0600240103] [Citation(s) in RCA: 331] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
IL-15 is normally presented in vivo as a cell-associated cytokine bound to IL-15Ralpha. We show here that the biological activity of soluble IL-15 is much improved after interaction with recombinant soluble IL-15Ralpha; after injection, soluble IL-15/IL-15Ralpha complexes rapidly induce strong and selective expansion of memory-phenotype CD8(+) cells and natural killer cells. These findings imply that binding of IL-15Ralpha to IL-15 may create a conformational change that potentiates IL-15 recognition by the betagamma(c) receptor on T cells. The enhancing effect of IL-15Ralpha binding may explain why IL-15 normally functions as a cell-associated cytokine. Significantly, the results with IL-2, a soluble cytokine, are quite different; thus, IL-2 function is markedly inhibited by binding to soluble IL-2Ralpha.
Collapse
Affiliation(s)
- Mark P. Rubinstein
- *Department of Immunology, IMM4, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037
| | - Marek Kovar
- Department of Immunology and Gnotobiology, Institute of Microbiology, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague, Czech Republic
| | - Jared F. Purton
- *Department of Immunology, IMM4, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037
| | - Jae-Ho Cho
- *Department of Immunology, IMM4, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst NSW 2010, Australia; and
| | - Onur Boyman
- *Department of Immunology, IMM4, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037
| | - Charles D. Surh
- *Department of Immunology, IMM4, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037
| | - Jonathan Sprent
- *Department of Immunology, IMM4, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst NSW 2010, Australia; and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
15
|
He W, Ohashi K, Sugimoto C, Onuma M. Theileria orientalis: cloning a cDNA encoding a protein similar to thiol protease with haemoglobin-binding activity. Exp Parasitol 2005; 111:143-53. [PMID: 16139835 DOI: 10.1016/j.exppara.2005.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2005] [Revised: 05/31/2005] [Accepted: 06/07/2005] [Indexed: 11/17/2022]
Abstract
A gene encoding a protein (Tocp1) from Theileria orientalis was isolated from a cDNA library and the deduced amino acid sequence of Tocp1 has 476 amino acids. The primary structure of Tocp1 is similar to eukaryotic thiol proteases (EC 3.4.22.-), but no enzymatic activity was observed with the substitution of essential cysteine at the cysteine active site for glycine. Southern blot analysis showed that multiple genes similar to Tocp1 were present in the parasite genome. Sequence analysis of the genome of the parasite showed that there are at least five different genes similar to Tocp1. Tocp1 transcripts were detected in the T. orientalis piroplasma by Northern blot analysis. Western blot analysis showed that Tocp1 was expressed in the piroplasm of T. orientalis. To address the role of Tocp1 in the life cycle of T. orientalis, Tocp1 was expressed using pET32 expression system. Binding affinity to haemoglobin was demonstrated by enzyme-linked immunosorbent assay.
Collapse
Affiliation(s)
- Weiyong He
- Laboratory of Infectious Diseases, Department of Disease Control, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan.
| | | | | | | |
Collapse
|
16
|
Abstract
The year 2004 represents a milestone for the biosensor research community: in this year, over 1000 articles were published describing experiments performed using commercially available systems. The 1038 papers we found represent an approximately 10% increase over the past year and demonstrate that the implementation of biosensors continues to expand at a healthy pace. We evaluated the data presented in each paper and compiled a 'top 10' list. These 10 articles, which we recommend every biosensor user reads, describe well-performed kinetic, equilibrium and qualitative/screening studies, provide comparisons between binding parameters obtained from different biosensor users, as well as from biosensor- and solution-based interaction analyses, and summarize the cutting-edge applications of the technology. We also re-iterate some of the experimental pitfalls that lead to sub-optimal data and over-interpreted results. We are hopeful that the biosensor community, by applying the hints we outline, will obtain data on a par with that presented in the 10 spotlighted articles. This will ensure that the scientific community at large can be confident in the data we report from optical biosensors.
Collapse
Affiliation(s)
- Rebecca L Rich
- Center for Biomolecular Interaction Analysis, University of Utah, Salt Lake City, UT 84132, USA
| | | |
Collapse
|
17
|
Peleg-Shulman T, Tsubery H, Mironchik M, Fridkin M, Schreiber G, Shechter Y. Reversible PEGylation: A Novel Technology To Release Native Interferon α2 over a Prolonged Time Period. J Med Chem 2004; 47:4897-904. [PMID: 15369394 DOI: 10.1021/jm0497693] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Many peptide and protein drugs have a short circulatory half-life in vivo. The covalent attachment of polyethylene glycol (PEG) chains (PEGylation) can overcome this deficiency, but pegylated peptides and proteins are often inactive. In this study, we present a novel PEG-IFNalpha2 conjugate, PEG(40)-FMS-IFNalpha2, capable of regenerating native interferon alpha2 (IFNalpha2) at a slow rate under physiological conditions. A 2-sulfo-9-fluorenylmethoxycarbonyl (FMS) containing bifunctional reagent, MAL-FMS-NHS, has been synthesized, enabling the linkage of a 40 kDa PEG-SH to IFNalpha2 through a slowly hydrolyzable bond. By use of a BIAcore binding assay, the in vitro rate of regeneration of native interferon was estimated to have a half-life of 65 h. Following subcutaneous administration to rats and monitoring circulating antiviral activity, active IFNalpha2 levels peaked at 50 h, with substantial levels still being detected 200 h after administration. This value contrasts with a half-life of about 1 h measured for unmodified interferon. The concentration of active IFNalpha2 scaled linearly with the quantity injected. Comparing subcutaneous to intravenous administration of PEG(40)-FMS-IFNalpha2, we found that the long circulatory lifetime of IFNalpha2 was affected both by the slow rate of absorption of the PEGylated protein from the subcutaneous volume and by the slow rate of discharge from the PEG in circulation. A numerical simulation of the results was in good agreement with the results observed in vivo. The pharmacokinetic profile of this novel IFNalpha2 conjugate combines a prolonged maintenance in vivo with the regeneration of active-native IFNalpha2, ensuring ready access to peripheral tissues and thus an overall advantage over currently used formulations.
Collapse
Affiliation(s)
- Tal Peleg-Shulman
- Department of Biological Chemistry and Organic Chemistry, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | |
Collapse
|