1
|
Michalska Z, Ostaszewska A, Fularczyk M, Dzierżyńska M, Bielak K, Morytz J, Sieradzan AK, Archacka K, Brzoska E, Rodziewicz-Motowidło S, Ciemerych MA. In Vitro Bioactivity Evaluation of IL-4 and SDF-1 Mimicking Peptides Engineered to Enhance Skeletal Muscle Reconstruction. J Biomed Mater Res A 2025; 113:e37898. [PMID: 40087853 DOI: 10.1002/jbm.a.37898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025]
Abstract
Skeletal muscle regeneration depends on satellite cells, which, in response to injury, activate, proliferate, and reconstruct damaged tissue. However, under certain conditions, such as large injuries or myopathies, this process may not be properly executed, and muscle function may be affected. Thus, pro-regenerative actions, such as the use of various factors or cells, are widely tested as a tool to improve muscle regeneration. In the current study, we designed peptides derived from the IL-4 and SDF-1 proteins, namely IL-4-X, IL-4-Y, SDF-1-X, and SDF-1-Y. We showed that these peptides can bind to appropriate receptors and can adopt proper structure in solution. Importantly, we documented, using in vitro culture, that they do not negatively affect the cells that are present and active in skeletal muscles, such as myoblasts and fibroblasts, bone marrow stromal cells, as well as induced pluripotent stem cells, which can serve as a source of myoblasts. The presence of peptides did not affect cell proliferation compared to untreated cells. In vitro culture and differentiation protocols documented that selected IL-4 and SDF-1 peptides increased cell migration and inhibited undesirable adipogenic differentiation. Thus, we proved that these peptides are safe to use in in vivo studies aimed at improving skeletal muscle regeneration.
Collapse
Affiliation(s)
- Zuzanna Michalska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Anna Ostaszewska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Martyna Fularczyk
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Maria Dzierżyńska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Kacper Bielak
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Justyna Morytz
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Adam K Sieradzan
- Department of Theoretical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Karolina Archacka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Edyta Brzoska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | | | - Maria A Ciemerych
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland
| |
Collapse
|
2
|
Vaz DC, Rodrigues JR, Loureiro-Ferreira N, Müller TD, Sebald W, Redfield C, Brito RMM. Lessons on protein structure from interleukin-4: All disulfides are not created equal. Proteins 2024; 92:219-235. [PMID: 37814578 DOI: 10.1002/prot.26611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 09/09/2023] [Accepted: 09/25/2023] [Indexed: 10/11/2023]
Abstract
Interleukin-4 (IL-4) is a hematopoietic cytokine composed by a four-helix bundle stabilized by an antiparallel beta-sheet and three disulfide bonds: Cys3-Cys127, Cys24-Cys65, and Cys46-Cys99. IL-4 is involved in several immune responses associated to infection, allergy, autoimmunity, and cancer. Besides its physiological relevance, IL-4 is often used as a "model" for protein design and engineering. Hence, to understand the role of each disulfide in the structure and dynamics of IL-4, we carried out several spectroscopic analyses (circular dichroism [CD], fluorescence, nuclear magnetic resonance [NMR]), and molecular dynamics (MD) simulations on wild-type IL-4 and four IL-4 disulfide mutants. All disulfide mutants showed loss of structure, altered interhelical angles, and looser core packings, showing that all disulfides are relevant for maintaining the overall fold and stability of the four-helix bundle motif, even at very low pH. In the absence of the disulfide connecting both protein termini Cys3-Cys127, C3T-IL4 showed a less packed protein core, loss of secondary structure (~9%) and fast motions on the sub-nanosecond time scale (lower S2 order parameters and larger τc correlation time), especially at the two protein termini, loops, beginning of helix A and end of helix D. In the absence of Cys24-Cys65, C24T-IL4 presented shorter alpha-helices (14% loss in helical content), altered interhelical angles, less propensity to form the small anti-parallel beta-sheet and increased dynamics. Simultaneously deprived of two disulfides (Cys3-Cys127 and Cys24-Cys65), IL-4 formed a partially folded "molten globule" with high 8-anilino-1-naphtalenesulphonic acid-binding affinity and considerable loss of secondary structure (~50%decrease), as shown by the far UV-CD, NMR, and MD data.
Collapse
Affiliation(s)
- Daniela C Vaz
- School of Health Sciences, Polytechnic of Leiria, Leiria, Portugal
- Chemistry Department, Faculty of Sciences and Technology, University of Coimbra, Coimbra Chemistry Centre, Institute of Molecular Sciences, Coimbra, Portugal
- Laboratory of Separation and Reaction Engineering-Laboratory of Catalysis and Materials (LSRE-LCM), School of Technology and Management, Polytechnic of Leiria, Leiria, Portugal
- Associate Laboratory in Chemical Engineering (ALiCE), University of Porto, Porto, Portugal
| | - J Rui Rodrigues
- Laboratory of Separation and Reaction Engineering-Laboratory of Catalysis and Materials (LSRE-LCM), School of Technology and Management, Polytechnic of Leiria, Leiria, Portugal
- Associate Laboratory in Chemical Engineering (ALiCE), University of Porto, Porto, Portugal
| | | | - Thomas D Müller
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University of Würzburg, Würzburg, Germany
| | - Walter Sebald
- Department of Physiological Chemistry II, Theodor-Boveri-Institute (Biocentre), University of Würzburg, Würzburg, Germany
| | - Christina Redfield
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Rui M M Brito
- Chemistry Department, Faculty of Sciences and Technology, University of Coimbra, Coimbra Chemistry Centre, Institute of Molecular Sciences, Coimbra, Portugal
| |
Collapse
|
3
|
Thomas S, Fiebig JE, Kuhn EM, Mayer DS, Filbeck S, Schmitz W, Krischke M, Gropp R, Mueller TD. Design of Glycoengineered IL-4 Antagonists Employing Chemical and Biosynthetic Glycosylation. ACS OMEGA 2023; 8:24841-24852. [PMID: 37483220 PMCID: PMC10357448 DOI: 10.1021/acsomega.3c00726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/13/2023] [Indexed: 07/25/2023]
Abstract
Interleukin-4 (IL-4) plays a key role in atopic diseases. It coordinates T-helper cell differentiation to subtype 2, thereby directing defense toward humoral immunity. Together with Interleukin-13, IL-4 further induces immunoglobulin class switch to IgE. Antibodies of this type activate mast cells and basophilic and eosinophilic granulocytes, which release pro-inflammatory mediators accounting for the typical symptoms of atopic diseases. IL-4 and IL-13 are thus major targets for pharmaceutical intervention strategies to treat atopic diseases. Besides neutralizing antibodies against IL-4, IL-13, or its receptors, IL-4 antagonists can present valuable alternatives. Pitrakinra, an Escherichia coli-derived IL-4 antagonist, has been evaluated in clinical trials for asthma treatment in the past; however, deficits such as short serum lifetime and potential immunogenicity among others stopped further development. To overcome such deficits, PEGylation of therapeutically important proteins has been used to increase the lifetime and proteolytic stability. As an alternative, glycoengineering is an emerging strategy used to improve pharmacokinetics of protein therapeutics. In this study, we have established different strategies to attach glycan moieties to defined positions in IL-4. Different chemical attachment strategies employing thiol chemistry were used to attach a glucose molecule at amino acid position 121, thereby converting IL-4 into a highly effective antagonist. To enhance the proteolytic stability of this IL-4 antagonist, additional glycan structures were introduced by glycoengineering utilizing eucaryotic expression. IL-4 antagonists with a combination of chemical and biosynthetic glycoengineering could be useful as therapeutic alternatives to IL-4 neutralizing antibodies already used to treat atopic diseases.
Collapse
Affiliation(s)
- Sarah Thomas
- Department
of Molecular Plant Physiology and Biophysics, Julius-von-Sachs Institute of the University Wuerzburg, Julius-von-Sachs Platz 2, D-97082 Wuerzburg, Germany
| | - Juliane E. Fiebig
- Department
of Molecular Plant Physiology and Biophysics, Julius-von-Sachs Institute of the University Wuerzburg, Julius-von-Sachs Platz 2, D-97082 Wuerzburg, Germany
| | - Eva-Maria Kuhn
- Department
of Molecular Plant Physiology and Biophysics, Julius-von-Sachs Institute of the University Wuerzburg, Julius-von-Sachs Platz 2, D-97082 Wuerzburg, Germany
| | - Dominik S. Mayer
- Department
of Molecular Plant Physiology and Biophysics, Julius-von-Sachs Institute of the University Wuerzburg, Julius-von-Sachs Platz 2, D-97082 Wuerzburg, Germany
| | - Sebastian Filbeck
- Department
of Molecular Plant Physiology and Biophysics, Julius-von-Sachs Institute of the University Wuerzburg, Julius-von-Sachs Platz 2, D-97082 Wuerzburg, Germany
| | - Werner Schmitz
- Department
of Biochemistry and Molecular Biology, Biocenter
of the University Wuerzburg, Am Hubland, D-97074 Wuerzburg, Germany
| | - Markus Krischke
- Department
of Pharmaceutical Biology, Julius-von-Sachs
Institute of the University Wuerzburg, Julius-von-Sachs Platz 2, D-97082 Wuerzburg, Germany
| | - Roswitha Gropp
- Department
of General- Visceral-, Vascular- and Transplantation Surgery, Hospital of the LMU, Nussbaumstr. 20, 80336 Munich, Germany
| | - Thomas D. Mueller
- Department
of Molecular Plant Physiology and Biophysics, Julius-von-Sachs Institute of the University Wuerzburg, Julius-von-Sachs Platz 2, D-97082 Wuerzburg, Germany
| |
Collapse
|
4
|
Walker K, Baravalle R, Holyfield R, Kalms J, Wright H, Seewooruthun C, Muskett FW, Scott-Tucker A, Merritt A, Henry A, Lawson ADG, Hall G, Prosser C, Carr MD. Identification and characterisation of anti-IL-13 inhibitory single domain antibodies provides new insights into receptor selectivity and attractive opportunities for drug discovery. Front Immunol 2023; 14:1216967. [PMID: 37483614 PMCID: PMC10359924 DOI: 10.3389/fimmu.2023.1216967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/19/2023] [Indexed: 07/25/2023] Open
Abstract
Interleukin-13 (IL-13) is a cytokine involved in T-cell immune responses and is a well validated therapeutic target for the treatment of asthma, along with other allergic and inflammatory diseases. IL-13 signals through a ternary signalling complex formed with the receptors IL-13Rα1 and IL-4Rα. This complex is assembled by IL-13 initially binding IL-13Rα1, followed by association of the binary IL-13:IL-13Rα1 complex with IL-4Rα. The receptors are shared with IL-4, but IL-4 initially binds IL-4Rα. Here we report the identification and characterisation of a diverse panel of single-domain antibodies (VHHs) that bind to IL-13 (KD 40 nM-5.5 μM) and inhibit downstream IL-13 signalling (IC50 0.2-53.8 μM). NMR mapping showed that the VHHs recognise a number of epitopes on IL-13, including previously unknown allosteric sites. Further NMR investigation of VHH204 bound to IL-13 revealed a novel allosteric mechanism of inhibition, with the antibody stabilising IL-13 in a conformation incompatible with receptor binding. This also led to the identification of a conformational equilibrium for free IL-13, providing insights into differing receptor signalling complex assembly seen for IL-13 compared to IL-4, with formation of the IL-13:IL-13Rα1 complex required to stabilise IL-13 in a conformation with high affinity for IL-4Rα. These findings highlight new opportunities for therapeutic targeting of IL-13 and we report a successful 19F fragment screen of the IL-13:VHH204 complex, including binding sites identified for several hits. To our knowledge, these 19F containing fragments represent the first small-molecules shown to bind to IL-13 and could provide starting points for a small-molecule drug discovery programme.
Collapse
Affiliation(s)
- Kayleigh Walker
- Leicester Institute of Structural and Chemical Biology, and Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - Roberta Baravalle
- Leicester Institute of Structural and Chemical Biology, and Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - Rachel Holyfield
- Leicester Institute of Structural and Chemical Biology, and Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - Jacqueline Kalms
- Leicester Institute of Structural and Chemical Biology, and Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
- UCB Biopharma, UCB Pharma, Slough, United Kingdom
| | - Helena Wright
- Leicester Institute of Structural and Chemical Biology, and Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - Chitra Seewooruthun
- Leicester Institute of Structural and Chemical Biology, and Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - Frederick W. Muskett
- Leicester Institute of Structural and Chemical Biology, and Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | | | - Andy Merritt
- LifeArc, Centre for Therapeutics Discovery, Stevenage Bioscience Catalyst, Stevenage, United Kingdom
| | | | | | - Gareth Hall
- Leicester Institute of Structural and Chemical Biology, and Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - Christine Prosser
- Leicester Institute of Structural and Chemical Biology, and Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
- UCB Biopharma, UCB Pharma, Slough, United Kingdom
| | - Mark D. Carr
- Leicester Institute of Structural and Chemical Biology, and Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
5
|
Ding Y, Wang Y, Zhang Y, Dang B, Hu S, Zhao C, Huang Y, Zheng G, Ma T, Zhang T. Alpha-linolenic acid improves nasal mucosa epithelial barrier function in allergic rhinitis by arresting CD4 + T cell differentiation via IL-4Rα-JAK2-STAT3 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154825. [PMID: 37178572 DOI: 10.1016/j.phymed.2023.154825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 04/07/2023] [Accepted: 04/16/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND Allergic rhinitis (AR) defined as inflammation and tissue remodeling of the nasal mucosa in atopic individuals after allergen exposure. Alpha-linolenic acid [cis-9, cis-12, cis-15-octadecatrienoic acid (18:3)] (ALA) as dietary supplementation can reduce inflammation and allergic symptoms. OBJECTIVE To evaluate the potential therapeutic effect and mechanism of ALA in AR mouse model. METHODS Ovalbumin sensitized AR mouse model were challenged with oral ALA administration. Nasal symptoms, tissue pathology, immune cell infiltration and goblet cell hyperplasia were investigated. Levels of IgE, TNF-β, IFN-γ, IL-2, IL-4, IL-5, IL-12, IL-13 and IL-25 were determined by ELISA in serum and nasal fluid. Quantitative RT-PCR and immunofluorescence were performed for occludin and zonula occludens-1 expression. CD3+CD4+ T-cells from peripheral blood and splenic lymphocytes were isolated and Th1/Th2 ratio were determined. Mouse naive CD4+ T cell were isolated and Th1/Th2 ratio, IL-4Rα expression, and IL5/IL13 secretion were determined. IL-4Rα-JAK2-STAT3 pathway change in AR mice were performed by western blot. RESULTS Ovalbumin induced AR, nasal symptoms, pathological performance, IgE, and cytokine production. ALA treated mice showed reduced nasal symptoms, nasal inflammation, nasal septum thickening, goblet cell hyperplasia, and eosinophil infiltration. In serum and nasal fluid of ovalbumin challenged mice, ALA decreased IgE, IL-4 levels, and the increase of Th2-cells. ALA prevented the disruption of the epithelial cell barrier in ovalbumin-challenged AR mice. Simultaneously, ALA prevents IL-4 induced barrier disruption. ALA treatment of AR by affecting the differentiation stage of CD4+T cells and block IL-4Rα-JAK2-STAT3 pathway. CONCLUSION This study suggests that ALA has the potential therapeutic effect to ovalbumin-induced AR. ALA can affect the differentiation stage of CD4+T cells and improve epithelial barrier functions through IL-4Rα-JAK2-STAT3 pathways. CLINICAL IMPLICATION ALA might be considered as drug candidate for improving epithelial barrier function through Th1/Th2 ratio recovery in AR.
Collapse
Affiliation(s)
- Yuanyuan Ding
- College of Pharmacy, Xi'an Jiaotong University, Yanta West Road, Xi'an 710061, China
| | - Yuejin Wang
- College of Pharmacy, Xi'an Jiaotong University, Yanta West Road, Xi'an 710061, China
| | - Yonghui Zhang
- College of Pharmacy, Xi'an Jiaotong University, Yanta West Road, Xi'an 710061, China
| | - Baowen Dang
- College of Pharmacy, Xi'an Jiaotong University, Yanta West Road, Xi'an 710061, China
| | - Shiting Hu
- College of Pharmacy, Xi'an Jiaotong University, Yanta West Road, Xi'an 710061, China
| | - Chenrui Zhao
- College of Pharmacy, Xi'an Jiaotong University, Yanta West Road, Xi'an 710061, China
| | - Yihan Huang
- College of Pharmacy, Xi'an Jiaotong University, Yanta West Road, Xi'an 710061, China
| | - Guodong Zheng
- College of Pharmacy, Xi'an Jiaotong University, Yanta West Road, Xi'an 710061, China
| | - Tianyou Ma
- School of Public Health, Xi'an Jiaotong University Health Science Center, Yanta West Road, Xi'an, Shaanxi 710061, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi 710061, China.
| | - Tao Zhang
- College of Pharmacy, Xi'an Jiaotong University, Yanta West Road, Xi'an 710061, China.
| |
Collapse
|
6
|
Gärtner Y, Bitar L, Zipp F, Vogelaar CF. Interleukin-4 as a therapeutic target. Pharmacol Ther 2023; 242:108348. [PMID: 36657567 DOI: 10.1016/j.pharmthera.2023.108348] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/06/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023]
Abstract
Interleukin-4 (IL-4) is a pleiotropic cytokine mainly known for its role in type 2 immunity. Therapies antagonizing or blocking IL-4 activity have been developed to counteract diseases such as atopic dermatitis and asthma. In contrast, other disorders experimentally benefit from IL-4-related effects and IL-4 recently demonstrated beneficial activity in experimental stroke, spinal cord injury and the animal model of multiple sclerosis. To exploit IL-4-related activity for therapeutic concepts, current experimental efforts include modifying the pathway without inducing type 2 immune response and targeting of the cytokine to specific tissues. Here, we review different activities of IL-4 as well as therapeutic strategies.
Collapse
Affiliation(s)
- Yvonne Gärtner
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lynn Bitar
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Christina Francisca Vogelaar
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
7
|
Shankar A, McAlees JW, Lewkowich IP. Modulation of IL-4/IL-13 cytokine signaling in the context of allergic disease. J Allergy Clin Immunol 2022; 150:266-276. [PMID: 35934680 PMCID: PMC9371363 DOI: 10.1016/j.jaci.2022.06.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 11/29/2022]
Abstract
Aberrant activation of CD4 TH2 cells and excessive production of TH2 cytokines such as IL-4 and IL-13 have been implicated in the pathogenesis of allergic diseases. Generally, IL-4 and IL-13 utilize Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathways for induction of inflammatory gene expression and the effector functions associated with disease pathology in many allergic diseases. However, it is increasingly clear that JAK/STAT pathways activated by IL-4/IL-13 can themselves be modulated in the presence of other intracellular signaling programs, thereby changing the overall tone and/or magnitude of IL-4/IL-13 signaling. Apart from direct activation of the canonic JAK/STAT pathways, IL-4 and IL-13 also induce proinflammatory gene expression and effector functions through activation of additional signaling cascades. These alternative signaling cascades contribute to several specific aspects of IL-4/IL-13-associated cellular and molecular responses. A more complete understanding of IL-4/IL-13 signaling pathways, including the precise conditions under which noncanonic signaling pathways are activated, and the impact of these pathways on cellular- and host-level responses, will better allow us to design agents that target specific pathologic outcomes or tailor therapies for the treatment of uncommon disease endotypes.
Collapse
|
8
|
Mastouri M, Baachaoui S, Mosbah A, Raouafi N. In silico screening for oligopeptides useful as capture and reporting probes for interleukin-6 biosensing. RSC Adv 2022; 12:13003-13013. [PMID: 35497015 PMCID: PMC9049833 DOI: 10.1039/d2ra01496c] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/24/2022] [Indexed: 11/21/2022] Open
Abstract
IL-6 is an important interleukin associated with inflammation and several diseases such as cancer. Evaluation of its levels in human blood sera is a critical step for an accurate diagnosis of the diseases. Our goal is to design peptides that can selectively bind in different poses with good affinities to IL-6. For this purpose, we started from the crystal structures of different IL-6/protein complexes available in the Protein Data Bank (PDB) to select short peptides in the interaction zones, in which we intentionally introduced point mutations to increase their stability and affinity. To examine their usefulness as capture and reporting probes for the IL-6 biosensing, the five peptides and their interaction with IL-6 were studied in saline aqueous solution. Molecular docking, MD, and MM-PBSA were used to investigate the affinity and stability of these complexes. The conformational changes, the distance between the mass centers, the gyration radii, and the numbers of hydrogen bonds were analyzed to select the most suitable candidates. Three peptides, namely CTE17, CAY15 and CSE25, have the highest affinities presenting significant numbers of residues that have contact frequencies greater than 50% of simulation run time and are the most promising candidates. CTE17 and CSE25 showed they can form a stable sandwich with the target protein. For sake of comparison, we examined the previously known peptides (FND20, INL19 and CEK17) having affinity to IL-6 and the affinity of the lead i.e. CSE25 to two other interleukin family members (IL-4 and to IL-10).
Collapse
Affiliation(s)
- Mohamed Mastouri
- Sensors and Biosensors Group, Laboratory of Analytical Chemistry & Electrochemistry (LR99ES15), Faculty of Science, University of Tunis El Manar 2092 Tunis El Manar Tunisia
| | - Sabrine Baachaoui
- Sensors and Biosensors Group, Laboratory of Analytical Chemistry & Electrochemistry (LR99ES15), Faculty of Science, University of Tunis El Manar 2092 Tunis El Manar Tunisia
| | - Amor Mosbah
- BVBGR Laboratory (LR11ES31), ISBST, Biotechnopole Sidi Thabet, University of Manouba Ariana 2020 Tunisia
| | - Noureddine Raouafi
- Sensors and Biosensors Group, Laboratory of Analytical Chemistry & Electrochemistry (LR99ES15), Faculty of Science, University of Tunis El Manar 2092 Tunis El Manar Tunisia
| |
Collapse
|
9
|
Foerster J, Molęda A. Virus-Like Particle-Mediated Vaccination against Interleukin-13 May Harbour General Anti-Allergic Potential beyond Atopic Dermatitis. Viruses 2020; 12:v12040438. [PMID: 32294982 PMCID: PMC7232523 DOI: 10.3390/v12040438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 03/31/2020] [Accepted: 04/09/2020] [Indexed: 12/23/2022] Open
Abstract
Virus-like particle (VLP)-based anti-infective prophylactic vaccination has been established in clinical use. Although validated in proof-of-concept clinical trials in humans, no VLP-based therapeutic vaccination against self-proteins to modulate chronic disease has yet been licensed. The present review summarises recent scientific advances, identifying interleukin-13 as an excellent candidate to validate the concept of anti-cytokine vaccination. Based on numerous clinical studies, long-term elimination of IL-13 is not expected to trigger target-related serious adverse effects and is likely to be safer than combined targeting of IL-4/IL-13. Furthermore, recently published results from large-scale trials confirm that elimination of IL-13 is highly effective in atopic dermatitis, an exceedingly common condition, as well as eosinophilic esophagitis. The distinctly different mode of action of a polyclonal vaccine response is discussed in detail, suggesting that anti-IL-13 vaccination has the potential of outperforming monoclonal antibody-based approaches. Finally, recent data have identified a subset of follicular T helper cells dependent on IL-13 which selectively trigger massive IgE accumulation in response to anaphylactoid allergens. Thus, prophylactic IL-13 vaccination may have broad application in a number of allergic conditions.
Collapse
MESH Headings
- Animals
- Anti-Allergic Agents/administration & dosage
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/immunology
- Antibodies, Neutralizing/immunology
- Cytokines/metabolism
- Dermatitis, Atopic/immunology
- Dermatitis, Atopic/metabolism
- Dermatitis, Atopic/prevention & control
- Humans
- Interleukin-13/antagonists & inhibitors
- Interleukin-13/immunology
- Ligands
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
- Vaccination
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/therapeutic use
Collapse
|
10
|
Sequeida A, Castillo A, Cordero N, Wong V, Montero R, Vergara C, Valenzuela B, Vargas D, Valdés N, Morales J, Tello M, Sandino AM, Maisey K, Imarai M. The Atlantic salmon interleukin 4/13 receptor family: Structure, tissue distribution and modulation of gene expression. FISH & SHELLFISH IMMUNOLOGY 2020; 98:773-787. [PMID: 31734286 DOI: 10.1016/j.fsi.2019.11.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 06/10/2023]
Abstract
Interleukin (IL)-4 and IL-13 play a central role in T helper 2 immune response in mammals. The cell signalling is mediated by the type I heterodimeric receptor containing the IL-4Rα and γC chains, and the type II receptors formed by IL-4Rα and IL-13Rα1. In salmonid species, three paralogues of the IL-4 and IL-13 cytokines have been reported, il-4/13a, il-4/13b1 and il-4/13b2. In regard to receptors, two paralogues of each IL-4/13 receptor chains have been identified in rainbow trout while five genes named γc1, il-4rα, il-13rα1a, il-13rα1b, and il-13rα2 have identified in Atlantic salmon. Since Atlantic salmon is an important farmed fish species, the aim of this work was to get new insights into distribution, structure and expression regulation of the IL-4/13 receptors in salmon. By using qRT-PCR, it was shown that all γc1, il-4rα, il-13rα1a, il-13rα1b, and il-13rα2 receptor chains were expressed in lymphoid and non-lymphoid tissues of healthy salmon, nonetheless γC expression was higher in lymphoid than non-lymphoid tissues. The in silico structural analysis and homology modelling of the predicted receptor proteins showed that domains and most motifs present in the superior vertebrate chains are conserved in salmon suggesting a conserved role for these receptor chains. Only IL-13Rα1B is a receptor chain with a unique structure that seem not to be present in higher vertebrates but in fish species. In order to determine the regulatory role of IL-4/13 on the expression of receptor chains, Atlantic salmon il-4/13A gene was synthetized and cloned in pET15b. The recombinant IL-4/13A was produced in E. coli and the activity of the purified cytokine was confirmed in vitro. The regulatory role of IL-4/13A on the expression of their potential receptors was tested in salmon receiving the recombinant cytokine and effects were compared with those of the control group. The results showed that IL-4/13A induced the expression of its own gene and GATA-3, in the head kidney of fish but not in the spleen, while IL-10 increased in both lymphoid organs indicating a regulatory role of this cytokine on the induction of Th2 responses in salmon. IFN-γ and MHC class II were also later induced in head kidney. In regard to the expression of the receptor chains, IL-4/13A upregulated the expression of γC, IL-13Rα1A and IL-13Rα2A in the spleen but not in the head kidney of salmon, indicating a role on the modulation of cell signalling for the Th2 response. Furthermore, Piscirickettsia salmonis infection of Atlantic salmon occurred with an increase of γC and IL-13Rα1A suggesting a potential role of the IL-4/13 system in bacterial immunity or pathogenesis. This study contributes to a better understanding of the IL-4/13A system in salmon, which as a key axis for Th2 response may be involved not only in pathogen elimination but also in adaptive immune repair that seems critical tolerance to infectious diseases.
Collapse
Affiliation(s)
- Alvaro Sequeida
- Laboratory of Immunology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile.
| | - Andrés Castillo
- Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Natalia Cordero
- Laboratory of Immunology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile; Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Valentina Wong
- Laboratory of Immunology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile.
| | - Ruth Montero
- Laboratory of Comparative Immunology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile; Laboratory for Comparative Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Claudio Vergara
- Laboratory of Immunology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile; Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Beatriz Valenzuela
- Laboratory of Immunology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile; Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Deborah Vargas
- Laboratory of Virology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile; Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Natalia Valdés
- Laboratory of Metagenomics, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile.
| | - Jonathan Morales
- Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Mario Tello
- Laboratory of Metagenomics, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile; Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Ana María Sandino
- Laboratory of Virology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile; Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Kevin Maisey
- Laboratory of Comparative Immunology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile.
| | - Mónica Imarai
- Laboratory of Immunology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile; Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| |
Collapse
|
11
|
Characterization of cryptic allosteric site at IL-4Rα: New paradigm towards IL-4/IL-4R inhibition. Int J Biol Macromol 2018; 123:239-245. [PMID: 30391589 DOI: 10.1016/j.ijbiomac.2018.10.204] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/27/2018] [Accepted: 10/29/2018] [Indexed: 12/26/2022]
Abstract
Interleukin-4(IL-4), an anti-inflammatory cytokine, plays significant role in pathogenesis of various diseases such as asthma, tumors, and HIV infections. These responses are mediated by expression of IL-4R (receptor) on various hematopoietic and non-hematopoietic cells surfaces. To date, the X-ray crystal structure of unbound (i.e. free) IL-4R is not reported which hampers active research on the molecular interaction mechanism between IL-4 and IL-4R. To investigate the missing gaps about stable binding mode of IL-4 and drug-ability of IL-4R active site, modelling and molecular dynamics (MD) simulation of IL-4/IL-4R complex was performed. Drug-ability of the target protein changed after modelling the loop region near C-terminal of IL-4R protein. This led to the identification of a novel druggable site other than the reported interfacial site. Our analysis showed that the modelled residues Ser111 and Ser164-Lys167 are part of newly discovered allosteric site, which underwent major fluctuation after association with its ligand protein (IL-4). The results indicated possible role of this cryptic allosteric site in IL-4/IL-4R signaling pathway that might help us to block IL-4/IL-4R association to prevent various allergic and malignant diseases.
Collapse
|
12
|
Macalino SJY, Basith S, Clavio NAB, Chang H, Kang S, Choi S. Evolution of In Silico Strategies for Protein-Protein Interaction Drug Discovery. Molecules 2018; 23:E1963. [PMID: 30082644 PMCID: PMC6222862 DOI: 10.3390/molecules23081963] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/03/2018] [Accepted: 08/04/2018] [Indexed: 12/14/2022] Open
Abstract
The advent of advanced molecular modeling software, big data analytics, and high-speed processing units has led to the exponential evolution of modern drug discovery and better insights into complex biological processes and disease networks. This has progressively steered current research interests to understanding protein-protein interaction (PPI) systems that are related to a number of relevant diseases, such as cancer, neurological illnesses, metabolic disorders, etc. However, targeting PPIs are challenging due to their "undruggable" binding interfaces. In this review, we focus on the current obstacles that impede PPI drug discovery, and how recent discoveries and advances in in silico approaches can alleviate these barriers to expedite the search for potential leads, as shown in several exemplary studies. We will also discuss about currently available information on PPI compounds and systems, along with their usefulness in molecular modeling. Finally, we conclude by presenting the limits of in silico application in drug discovery and offer a perspective in the field of computer-aided PPI drug discovery.
Collapse
Affiliation(s)
- Stephani Joy Y Macalino
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea.
| | - Shaherin Basith
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea.
| | - Nina Abigail B Clavio
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea.
| | - Hyerim Chang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea.
| | - Soosung Kang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea.
| | - Sun Choi
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea.
| |
Collapse
|
13
|
Sequeida A, Maisey K, Imarai M. Interleukin 4/13 receptors: An overview of genes, expression and functional role in teleost fish. Cytokine Growth Factor Rev 2017; 38:66-72. [PMID: 28988781 DOI: 10.1016/j.cytogfr.2017.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Revised: 09/26/2017] [Accepted: 09/26/2017] [Indexed: 12/11/2022]
Abstract
In superior vertebrates, Interleukin 4 (IL-4) and Interleukin 13 (IL-13) play key and diverse roles to support immune responses acting on cell surface receptors. When stimulated, receptors activate intracellular signalling cascades switching cell phenotypes according to stimuli. In teleost fish, Interleukin 4/13 (IL-4/13) is the ancestral family cytokine related to both IL-4 and IL-13. Every private and common receptor subunit for IL-4/13 have in fish at least two paralogues and, as in mammals, soluble forms are also part of the receptor system. Reports for findings of fish IL-4/13 receptors have covered comparative analysis, transcriptomic profiles and to a lesser extent, functional analysis regarding ligand-receptor interactions and their biological effects. This review addresses available information from fish IL-4/13 receptors and discusses overall implications on teleost immunity, summarized gene induction strategies and pathogen-induced gene modulation, which may be useful tools to enhance immune response. Additionally, we present novel coding sequences for Atlantic salmon (Salmo salar) common gamma chain receptor (γC), Interleukin 13 receptor alpha 1A chain (IL-13Rα1A) and Interleukin 13 receptor alpha 1B chain (IL-13Rα1B).
Collapse
Affiliation(s)
- A Sequeida
- Laboratory of Immunology, Center for Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363 Santiago, Chile
| | - K Maisey
- Laboratory of Immunology, Center for Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363 Santiago, Chile; Laboratory of Comparative Immunology, Center for Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile,Av. Bernardo O'Higgins, 3363 Santiago, Chile
| | - M Imarai
- Laboratory of Immunology, Center for Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363 Santiago, Chile.
| |
Collapse
|
14
|
Kabashima K, Nomura T. Revisiting murine models for atopic dermatitis and psoriasis with multipolar cytokine axes. Curr Opin Immunol 2017; 48:99-107. [PMID: 28915378 DOI: 10.1016/j.coi.2017.08.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 07/18/2017] [Accepted: 08/16/2017] [Indexed: 12/18/2022]
Abstract
Atopic dermatitis (AD) and psoriasis are one of the common skin diseases. Animal models are a powerful tool to analyze these diseases, which are complicated by multiple cytokine pathways. However, many discrepancies between the human diseases and murine models have been noticed. Therefore, investigators should be aware of the differences between the murine AD models and human AD when translating murine data to human skin diseases. This review highlights the differences between the inflammatory profiles between murine models and human diseases focusing on AD and psoriasis.
Collapse
Affiliation(s)
- Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara, Sakyoku, Kyoto 606-8507, Japan; Singapore Immunology Network (SIgN) and Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore.
| | - Takashi Nomura
- Department of Dermatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara, Sakyoku, Kyoto 606-8507, Japan
| |
Collapse
|
15
|
Ligand-induced type II interleukin-4 receptor dimers are sustained by rapid re-association within plasma membrane microcompartments. Nat Commun 2017; 8:15976. [PMID: 28706306 PMCID: PMC5519985 DOI: 10.1038/ncomms15976] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 05/16/2017] [Indexed: 12/14/2022] Open
Abstract
The spatiotemporal organization of cytokine receptors in the plasma membrane is still debated with models ranging from ligand-independent receptor pre-dimerization to ligand-induced receptor dimerization occurring only after receptor uptake into endosomes. Here, we explore the molecular and cellular determinants governing the assembly of the type II interleukin-4 receptor, taking advantage of various agonists binding the receptor subunits with different affinities and rate constants. Quantitative kinetic studies using artificial membranes confirm that receptor dimerization is governed by the two-dimensional ligand–receptor interactions and identify a critical role of the transmembrane domain in receptor dimerization. Single molecule localization microscopy at physiological cell surface expression levels, however, reveals efficient ligand-induced receptor dimerization by all ligands, largely independent of receptor binding affinities, in line with the similar STAT6 activation potencies observed for all IL-4 variants. Detailed spatiotemporal analyses suggest that kinetic trapping of receptor dimers in actin-dependent microcompartments sustains robust receptor dimerization and signalling. The contribution of ligands for cytokine receptor dimerization is still not fully understood. Here, the authors show the efficient ligand-induced dimerization of type II interleukin-4 receptor at the plasma membrane and the kinetic trapping of signalling complexes by actin-dependent membrane microdomains.
Collapse
|
16
|
Smith LJ, van Gunsteren WF, Hansen N. Using Complementary NMR Data Sets To Detect Inconsistencies and Model Flaws in the Structure Determination of Human Interleukin-4. J Phys Chem B 2017. [PMID: 28640620 DOI: 10.1021/acs.jpcb.7b03647] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The derivation of protein structure from values of observable quantities measured in NMR experiments is a rather nontrivial task due to (i) the limited number of data compared to degrees of freedom of a protein, (ii) the uncertainty inherent to the function connecting an observable quantity to molecular structure, (iii) the finite quality of biomolecular models and force fields used in structure refinement, and (iv) the conformational freedom of a protein in aqueous solution, which requires extensive conformational sampling and appropriate conformational averaging when calculating or restraining to sets of NMR data. The protein interleukin-4 (IL-4) has been taken as a test case using NOE distances, S2 order parameters, and 3J-couplings as test data and the former two types of data as restraints. It is shown that, by combining sets of different, complementary NMR data as restraints in MD simulations, inconsistencies in the data or flaws in the model and procedures used to derive protein structure from NMR data can be detected. This leads to an improved structural interpretation of such data particularly in more mobile loop regions.
Collapse
Affiliation(s)
- Lorna J Smith
- Department of Chemistry, University of Oxford, Inorganic Chemistry Laboratory , South Parks Road, Oxford OX1 3QR, United Kingdom
| | - Wilfred F van Gunsteren
- Laboratory of Physical Chemistry, Swiss Federal Institute of Technology, ETH , CH-8093 Zürich, Switzerland
| | - Niels Hansen
- Institute of Thermodynamics and Thermal Process Engineering, University of Stuttgart , D-70569 Stuttgart, Germany
| |
Collapse
|
17
|
Lühmann T, Spieler V, Werner V, Ludwig MG, Fiebig J, Mueller TD, Meinel L. Interleukin-4-Clicked Surfaces Drive M2 Macrophage Polarization. Chembiochem 2016; 17:2123-2128. [DOI: 10.1002/cbic.201600480] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Indexed: 01/27/2023]
Affiliation(s)
- Tessa Lühmann
- Institute for Pharmacy and Food Chemistry; University of Würzburg; Am Hubland 97074 Würzburg Germany
| | - Valerie Spieler
- Institute for Pharmacy and Food Chemistry; University of Würzburg; Am Hubland 97074 Würzburg Germany
| | - Vera Werner
- Institute for Pharmacy and Food Chemistry; University of Würzburg; Am Hubland 97074 Würzburg Germany
| | | | - Juliane Fiebig
- Lehrstuhl für Botanik I Molekulare Pflanzenphysik und Biophysik; University of Würzburg; Julius-von-Sachs-Platz 2 97082 Würzburg Germany
| | - Thomas D. Mueller
- Lehrstuhl für Botanik I Molekulare Pflanzenphysik und Biophysik; University of Würzburg; Julius-von-Sachs-Platz 2 97082 Würzburg Germany
| | - Lorenz Meinel
- Institute for Pharmacy and Food Chemistry; University of Würzburg; Am Hubland 97074 Würzburg Germany
| |
Collapse
|
18
|
Interleukin-4 receptor signaling and its binding mechanism: A therapeutic insight from inhibitors tool box. Cytokine Growth Factor Rev 2016; 32:3-15. [PMID: 27165851 DOI: 10.1016/j.cytogfr.2016.04.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/23/2016] [Accepted: 04/15/2016] [Indexed: 01/23/2023]
Abstract
Studies on Interlukin-4 (IL-4) disclosed great deal of information about its various physiological and pathological roles. All these roles depend upon its interaction and signaling through either type-I (IL-4Rα/common γ-chain) or type-II (IL-4Rα/IL-13Rα) receptors. Another cytokine, IL-13, shares some of the functions of IL-4, because both cytokines use a common receptor subunit, IL-4Rα. Here in this review, we discuss the structural details of IL-4 and IL-4Rα subunit and the structural similarities between IL-4 and IL-13. We also describe detailed chemistry of type-I and type-II receptor complexes and their signaling pathways. Furthermore, we elaborate the strength of type-II hetero dimer signals in response to IL-4 and IL-13. These cytokines are prime players in pathogenesis of allergic asthma, allergic hypersensitivity, different cancers, and HIV infection. Recent advances in the structural and binding chemistry of these cytokines various types of inhibitors were designed to block the interaction of IL-4 and IL-13 with their receptor, including several IL-4 mutant analogs and IL-4 antagonistic antibodies. Moreover, different targeted immunotoxins, which is a fusion of cytokine protein with a toxin or suicidal gene, are the new class of inhibitors to prevent cancer progression. In addition few small molecular inhibitors such as flavonoids have also been developed which are capable of binding with high affinity to IL-4Rα and, therefore, can be very effective in blocking IL-4-mediated responses.
Collapse
|
19
|
Broughton SE, Nero TL, Dhagat U, Kan WL, Hercus TR, Tvorogov D, Lopez AF, Parker MW. The βc receptor family – Structural insights and their functional implications. Cytokine 2015; 74:247-58. [DOI: 10.1016/j.cyto.2015.02.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 02/06/2015] [Indexed: 11/25/2022]
|
20
|
Obmolova G, Teplyakov A, Malia TJ, Keough E, Luo J, Sweet R, Jacobs SA, Yi F, Hippensteel R, O'Neil KT, Gilliland GL. Induced conformational change in human IL-4 upon binding of a signal-neutralizing DARPin. Proteins 2015; 83:1191-7. [PMID: 25900776 PMCID: PMC5029753 DOI: 10.1002/prot.24815] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 04/08/2015] [Accepted: 04/11/2015] [Indexed: 11/12/2022]
Abstract
The crystal structure of DARPin 44C12V5 that neutralizes IL-4 signaling has been determined alone and bound to human IL-4. A significant conformational change occurs in the IL-4 upon DARPin binding. The DARPin binds to the face of IL-4 formed by the A and C α-helices. The structure of the DARPin remains virtually unchanged. The conformational changes in IL-4 include a reorientation of the C-helix Trp91 side chain and repositioning of CD-loop residue Leu96. Both side chains move by >9 Å, becoming buried in the central hydrophobic region of the IL-4:DARPin interface. This hydrophobic region is surrounded by a ring of hydrophilic interactions comprised of hydrogen bonds and salt bridges and represents a classical "hotspot." The structures also reveal how the DARPin neutralizes IL-4 signaling. Comparing the IL-4:DARPin complex structure with the structures of IL-4 bound to its receptors (Hage et al., Cell 1999; 97, 271-281; La Porte et al., Cell 2008, 132, 259-272), it is found that the DARPin binds to the same IL-4 face that interacts with the junction of the D1 and D2 domains of the IL-4Rα receptors. Signaling is blocked since IL-4 cannot bind to this receptor, which it must do first before initiating a productive receptor complex with either the IL-13α1 or the γc receptor.
Collapse
Affiliation(s)
- Galina Obmolova
- Janssen Research & Development, LLC, Biotechnology Center of ExcellenceSpring HousePennsylvania19477
| | - Alexey Teplyakov
- Janssen Research & Development, LLC, Biotechnology Center of ExcellenceSpring HousePennsylvania19477
| | - Thomas J. Malia
- Janssen Research & Development, LLC, Biotechnology Center of ExcellenceSpring HousePennsylvania19477
| | - Edward Keough
- Janssen Research & Development, LLC, Biotechnology Center of ExcellenceSpring HousePennsylvania19477
| | - Jinquan Luo
- Janssen Research & Development, LLC, Biotechnology Center of ExcellenceSpring HousePennsylvania19477
| | - Raymond Sweet
- Janssen Research & Development, LLC, Biotechnology Center of ExcellenceSpring HousePennsylvania19477
- Present address: Raymond Sweet's current address is 700 Oak Springs Rd.Bryn MawrPA19010
| | - Steven A. Jacobs
- Janssen Research & Development, LLC, Biotechnology Center of ExcellenceSpring HousePennsylvania19477
| | - Fang Yi
- Janssen Research & Development, LLC, Biotechnology Center of ExcellenceSpring HousePennsylvania19477
| | - Randi Hippensteel
- Janssen Research & Development, LLC, Biotechnology Center of ExcellenceSpring HousePennsylvania19477
- Present address: Randi Hippensteel's current address is Eurofins Lancaster Laboratories2425 New Holland PikeLancasterPA17601
| | - Karyn T. O'Neil
- Janssen Research & Development, LLC, Biotechnology Center of ExcellenceSpring HousePennsylvania19477
| | - Gary L. Gilliland
- Janssen Research & Development, LLC, Biotechnology Center of ExcellenceSpring HousePennsylvania19477
| |
Collapse
|
21
|
Zarzycka B, Nicolaes GAF, Lutgens E. Targeting the adaptive immune system: new strategies in the treatment of atherosclerosis. Expert Rev Clin Pharmacol 2015; 8:297-313. [PMID: 25843158 DOI: 10.1586/17512433.2015.1025052] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Atherosclerosis is a lipid-driven chronic inflammatory disease of the arterial wall. Current treatment of atherosclerosis is focused on limiting its risk factors, such as hyperlipidemia or hypertension. However, treatments that target the inflammatory nature of atherosclerosis are still under development. Discovery of novel targets involved in the inflammation of the arterial wall creates opportunities to design new therapeutics that successfully modulate atherosclerosis. Here, we review drug targets that have proven to play pivotal roles in the adaptive immune system in atherosclerosis, and we discuss their potential as novel therapeutics.
Collapse
Affiliation(s)
- Barbara Zarzycka
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands
| | | | | |
Collapse
|
22
|
Zhao HQ, Li WM, Lu ZQ, Sheng ZY, Yao YM. The growing spectrum of anti-inflammatory interleukins and their potential roles in the development of sepsis. J Interferon Cytokine Res 2014; 35:242-51. [PMID: 25517926 DOI: 10.1089/jir.2014.0119] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Sepsis, recognized as a deadly immunological disorder, is one of the major causes of death in intensive care units globally. Traditionally, sepsis was characterized by an excessive systemic proinflammatory response to invasive microbial pathogens. However, failures of highly sophisticated trials directed toward the uncontrolled inflammatory reaction have led to an appeal by experts for reevaluation of the present approach toward sepsis. With accumulated evidence, a principal role for immunosuppression in severe sepsis has been evaluated. Different pathways of negative regulation in the pathophysiological process of sepsis have been investigated. Significant among these regulatory elements are the anti-inflammatory cytokines. In the past few years, several interleukins (ILs) have been identified and characterized, among which IL-35 and IL-37 represent newly identified ones in the spectrum of anti-inflammatory cytokines. In this study, we focus on regulatory cytokines of the IL family (including the old members: IL-4, IL-10, and IL-13, and newly discovered ones: IL-35 and IL-37) to address current knowledge regarding their structural and functional characteristics as well as their roles in the development of sepsis. Although the exact roles for these cytokines are pending further elucidation, the current advances in our understanding of mechanisms that regulate the immune responses during severe sepsis may lead to the identification of new diagnostic or treatment targets.
Collapse
Affiliation(s)
- Hong-qiang Zhao
- 1 Medical School of Chinese People's Liberation Army, the Chinese PLA General Hospital , Beijing, People's Republic of China
| | | | | | | | | |
Collapse
|
23
|
Spangler JB, Moraga I, Mendoza JL, Garcia KC. Insights into cytokine-receptor interactions from cytokine engineering. Annu Rev Immunol 2014; 33:139-67. [PMID: 25493332 DOI: 10.1146/annurev-immunol-032713-120211] [Citation(s) in RCA: 202] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cytokines exert a vast array of immunoregulatory actions critical to human biology and disease. However, the desired immunotherapeutic effects of native cytokines are often mitigated by toxicity or lack of efficacy, either of which results from cytokine receptor pleiotropy and/or undesired activation of off-target cells. As our understanding of the structural principles of cytokine-receptor interactions has advanced, mechanism-based manipulation of cytokine signaling through protein engineering has become an increasingly feasible and powerful approach. Modified cytokines, both agonists and antagonists, have been engineered with narrowed target cell specificities, and they have also yielded important mechanistic insights into cytokine biology and signaling. Here we review the theory and practice of cytokine engineering and rationalize the mechanisms of several engineered cytokines in the context of structure. We discuss specific examples of how structure-based cytokine engineering has opened new opportunities for cytokines as drugs, with a focus on the immunotherapeutic cytokines interferon, interleukin-2, and interleukin-4.
Collapse
Affiliation(s)
- Jamie B Spangler
- Howard Hughes Medical Institute, Department of Molecular and Cellular Physiology, Department of Structural Biology, Stanford University School of Medicine, Stanford, California 94305; , , ,
| | | | | | | |
Collapse
|
24
|
Malecek K, Grigoryan A, Zhong S, Gu WJ, Johnson LA, Rosenberg SA, Cardozo T, Krogsgaard M. Specific increase in potency via structure-based design of a TCR. THE JOURNAL OF IMMUNOLOGY 2014; 193:2587-99. [PMID: 25070852 DOI: 10.4049/jimmunol.1302344] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Adoptive immunotherapy with Ag-specific T lymphocytes is a powerful strategy for cancer treatment. However, most tumor Ags are nonreactive "self" proteins, which presents an immunotherapy design challenge. Recent studies have shown that tumor-specific TCRs can be transduced into normal PBLs, which persist after transfer in ∼30% of patients and effectively destroy tumor cells in vivo. Although encouraging, the limited clinical responses underscore the need for enrichment of T cells with desirable antitumor capabilities prior to patient transfer. In this study, we used structure-based design to predict point mutations of a TCR (DMF5) that enhance its binding affinity for an agonist tumor Ag-MHC (peptide-MHC [pMHC]), Mart-1 (27L)-HLA-A2, which elicits full T cell activation to trigger immune responses. We analyzed the effects of selected TCR point mutations on T cell activation potency and analyzed cross-reactivity with related Ags. Our results showed that the mutated TCRs had improved T cell activation potency while retaining a high degree of specificity. Such affinity-optimized TCRs have demonstrated to be very specific for Mart-1 (27L), the epitope for which they were structurally designed. Although of somewhat limited clinical relevance, these studies open the possibility for future structural-based studies that could potentially be used in adoptive immunotherapy to treat melanoma while avoiding adverse autoimmunity-derived effects.
Collapse
Affiliation(s)
- Karolina Malecek
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016; Program in Structural Biology, New York University School of Medicine, New York, NY 10016
| | - Arsen Grigoryan
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016
| | - Shi Zhong
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| | - Wei Jun Gu
- Department of Chemistry, New York University, New York, NY 10012
| | - Laura A Johnson
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Steven A Rosenberg
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Timothy Cardozo
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016; Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016
| | - Michelle Krogsgaard
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016; Program in Structural Biology, New York University School of Medicine, New York, NY 10016; Interdisciplinary Melanoma Cooperative Group, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
25
|
Li W, Holsinger RMD, Kruse CA, Flügel A, Graeber MB. The potential for genetically altered microglia to influence glioma treatment. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2014; 12:750-62. [PMID: 24047526 DOI: 10.2174/18715273113126660171] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 12/06/2012] [Accepted: 12/06/2012] [Indexed: 01/06/2023]
Abstract
Diffuse and unstoppable infiltration of brain and spinal cord tissue by neoplastic glial cells is the single most important therapeutic problem posed by the common glioma group of tumors: astrocytoma, oligoastrocytoma, oligodendroglioma, their malignant variants and glioblastoma. These neoplasms account for more than two thirds of all malignant central nervous system tumors. However, most glioma research focuses on an examination of the tumor cells rather than on host-specific, tumor micro-environmental cells and factors. This can explain why existing diffuse glioma therapies fail and why these tumors have remained incurable. Thus, there is a great need for innovation. We describe a novel strategy for the development of a more effective treatment of diffuse glioma. Our approach centers on gaining control over the behavior of the microglia, the defense cells of the CNS, which are manipulated by malignant glioma and support its growth. Armoring microglia against the influences from glioma is one of our research goals. We further discuss how microglia precursors may be genetically enhanced to track down infiltrating glioma cells.
Collapse
Affiliation(s)
- W Li
- Brain and Mind Research Institute, The University of Sydney, Camperdown, NSW, Australia.
| | | | | | | | | |
Collapse
|
26
|
Ahmed N, Dhanapala P, Sadli N, Barrow CJ, Suphioglu C. Mimtags: the use of phage display technology to produce novel protein-specific probes. J Immunol Methods 2014; 405:121-9. [PMID: 24530568 DOI: 10.1016/j.jim.2014.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 02/04/2014] [Accepted: 02/04/2014] [Indexed: 11/16/2022]
Abstract
In recent times the use of protein-specific probes in the field of proteomics has undergone evolutionary changes leading to the discovery of new probing techniques. Protein-specific probes serve two main purposes: epitope mapping and detection assays. One such technique is the use of phage display in the random selection of peptide mimotopes (mimtags) that can tag epitopes of proteins, replacing the use of monoclonal antibodies in detection systems. In this study, phage display technology was used to screen a random peptide library with a biologically active purified human interleukin-4 receptor (IL-4R) and interleukin-13 (IL-13) to identify mimtag candidates that interacted with these proteins. Once identified, the mimtags were commercially synthesised, biotinylated and used for in vitro immunoassays. We have used phage display to identify M13 phage clones that demonstrated specific binding to IL-4R and IL-13 cytokine. A consensus in binding sequences was observed and phage clones characterised had identical peptide sequence motifs. Only one was synthesised for use in further immunoassays, demonstrating significant binding to either IL-4R or IL-13. We have successfully shown the use of phage display to identify and characterise mimtags that specifically bind to their target epitope. Thus, this new method of probing proteins can be used in the future as a novel tool for immunoassay and detection technique, which is cheaper and more rapidly produced and therefore a better alternative to the use of monoclonal antibodies.
Collapse
Affiliation(s)
- Nayyar Ahmed
- NeuroAllergy Research Laboratory (NARL), Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, Victoria 3216, Australia; School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, Victoria 3216 Australia
| | - Pathum Dhanapala
- NeuroAllergy Research Laboratory (NARL), Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, Victoria 3216, Australia; School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, Victoria 3216 Australia
| | - Nadia Sadli
- NeuroAllergy Research Laboratory (NARL), Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, Victoria 3216, Australia; School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, Victoria 3216 Australia
| | - Colin J Barrow
- School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, Victoria 3216 Australia
| | - Cenk Suphioglu
- NeuroAllergy Research Laboratory (NARL), Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, Victoria 3216, Australia; School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, Victoria 3216 Australia.
| |
Collapse
|
27
|
Ma B, Nussinov R. Druggable orthosteric and allosteric hot spots to target protein-protein interactions. Curr Pharm Des 2014; 20:1293-301. [PMID: 23713780 PMCID: PMC6361532 DOI: 10.2174/13816128113199990073] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 05/21/2013] [Indexed: 11/22/2022]
Abstract
Drug designing targeting protein-protein interactions is challenging. Because structural elucidation and computational analysis have revealed the importance of hot spot residues in stabilizing these interactions, there have been on-going efforts to develop drugs which bind the hot spots and out-compete the native protein partners. The question arises as to what are the key 'druggable' properties of hot spots in protein-protein interactions and whether these mimic the general hot spot definition. Identification of orthosteric (at the protein- protein interaction site) and allosteric (elsewhere) druggable hot spots is expected to help in discovering compounds that can more effectively modulate protein-protein interactions. For example, are there any other significant features beyond their location in pockets in the interface? The interactions of protein-protein hot spots are coupled with conformational dynamics of protein complexes. Currently increasing efforts focus on the allosteric drug discovery. Allosteric drugs bind away from the native binding site and can modulate the native interactions. We propose that identification of allosteric hot spots could similarly help in more effective allosteric drug discovery. While detection of allosteric hot spots is challenging, targeting drugs to these residues has the potential of greatly increasing the hot spot and protein druggability.
Collapse
Affiliation(s)
| | - Ruth Nussinov
- Basic Science Program, Leidos Biomedical Research, Inc. Cancer and Inflammation Program, NCIFrederick, Frederick, MD 21702.
| |
Collapse
|
28
|
Abstract
Protein action in nature is largely controlled by the level of expression and by post-translational modifications. Post-translational modifications result in a proteome that is at least two orders of magnitude more diverse than the genome. There are three basic types of post-translational modifications: covalent modification of an amino acid side chain, hydrolytic cleavage or isomerization of a peptide bond, and reductive cleavage of a disulfide bond. This review addresses the modification of disulfide bonds. Protein disulfide bonds perform either a structural or a functional role, and there are two types of functional disulfide: the catalytic and allosteric bonds. The allosteric disulfide bonds control the function of the mature protein in which they reside by triggering a change when they are cleaved. The change can be in ligand binding, substrate hydrolysis, proteolysis, or oligomer formation. The allosteric disulfides are cleaved by oxidoreductases or by thiol/disulfide exchange, and the configurations of the disulfides and the secondary structures that they link share some recurring features. How these bonds are being identified using bioinformatics and experimental screens and what the future holds for this field of research are also discussed.
Collapse
Affiliation(s)
- Kristina M Cook
- Lowy Cancer Research Centre and Prince of Wales Clinical School, University of New South Wales, Sydney NSW2052, Australia
| | | |
Collapse
|
29
|
Junttila IS, Creusot RJ, Moraga I, Bates DL, Wong MT, Alonso MN, Suhoski MM, Lupardus P, Meier-Schellersheim M, Engleman EG, Utz PJ, Fathman CG, Paul WE, Garcia KC. Redirecting cell-type specific cytokine responses with engineered interleukin-4 superkines. Nat Chem Biol 2012; 8:990-8. [PMID: 23103943 PMCID: PMC3508151 DOI: 10.1038/nchembio.1096] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 09/20/2012] [Indexed: 01/28/2023]
Abstract
Cytokines dimerize their receptors, with the binding of the 'second chain' triggering signaling. In the interleukin (IL)-4 and IL-13 system, different cell types express varying numbers of alternative second receptor chains (γc or IL-13Rα1), forming functionally distinct type I or type II complexes. We manipulated the affinity and specificity of second chain recruitment by human IL-4. A type I receptor-selective IL-4 'superkine' with 3,700-fold higher affinity for γc was three- to ten-fold more potent than wild-type IL-4. Conversely, a variant with high affinity for IL-13Rα1 more potently activated cells expressing the type II receptor and induced differentiation of dendritic cells from monocytes, implicating the type II receptor in this process. Superkines showed signaling advantages on cells with lower second chain numbers. Comparative transcriptional analysis reveals that the superkines induce largely redundant gene expression profiles. Variable second chain numbers can be exploited to redirect cytokines toward distinct cell subsets and elicit new actions, potentially improving the selectivity of cytokine therapy.
Collapse
Affiliation(s)
- Ilkka S. Junttila
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
- School of Medicine, University of Tampere, 33014, Tampere, Finland
- Fimlab Laboratories, Pirkanmaa Hospital District, 33521, Tampere, Finland
| | - Remi J. Creusot
- Department of Medicine, Division of Immunology & Rheumatology, Stanford University School of Medicine, Stanford, CA, 94305
| | - Ignacio Moraga
- Howard Hughes Medical Institute, Departments of Molecular and Cellular Physiology, Structural Biology, and Program in Immunology, Stanford University School of Medicine, Stanford, CA, 94305
| | - Darren L. Bates
- Howard Hughes Medical Institute, Departments of Molecular and Cellular Physiology, Structural Biology, and Program in Immunology, Stanford University School of Medicine, Stanford, CA, 94305
| | - Michael T. Wong
- Department of Medicine, Division of Immunology & Rheumatology, Stanford University School of Medicine, Stanford, CA, 94305
| | - Michael N. Alonso
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305
| | - Megan M. Suhoski
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305
| | - Patrick Lupardus
- Howard Hughes Medical Institute, Departments of Molecular and Cellular Physiology, Structural Biology, and Program in Immunology, Stanford University School of Medicine, Stanford, CA, 94305
| | - Martin Meier-Schellersheim
- Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Edgar G. Engleman
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305
| | - Paul J. Utz
- Department of Medicine, Division of Immunology & Rheumatology, Stanford University School of Medicine, Stanford, CA, 94305
| | - C. Garrison Fathman
- Department of Medicine, Division of Immunology & Rheumatology, Stanford University School of Medicine, Stanford, CA, 94305
| | - William E. Paul
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - K. Christopher Garcia
- Howard Hughes Medical Institute, Departments of Molecular and Cellular Physiology, Structural Biology, and Program in Immunology, Stanford University School of Medicine, Stanford, CA, 94305
| |
Collapse
|
30
|
Duppatla V, Gjorgjevikj M, Schmitz W, Kottmair M, Mueller TD, Sebald W. Enzymatic deglutathionylation to generate interleukin-4 cysteine muteins with free thiol. Bioconjug Chem 2012; 23:1396-405. [PMID: 22681442 DOI: 10.1021/bc2004389] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Interleukin-4 (IL-4) is a prototypical regulator protein of the immune system that is crucial for the pathogenesis and maintenance of asthma and other atopic diseases. It, together with IL-13, uses the IL-4 receptor α chain (IL-4Rα) to signal into immune and other cells. An IL-4 mutein acting as a dual IL-4/IL-13 receptor antagonist is in clinical development. Here, it is described how IL-4 muteins containing a single engineered cysteine with a free thiol can be prepared and used for site-specific chemical modification. The muteins were initially expressed in E. coli, refolded, and purified, but not in a fully reduced nonconjugated form. Attempts to reduce the cysteine chemically failed because the native disulfide bonds of IL-4 were also reduced under similar conditions. Therefore, an enzymatic procedure was developed to reduce glutathionylated IL-4 cysteine muteins employing glutaredoxin and reduced glutathione. Cysteine muteins engineered at four different positions around the IL-4Rα binding site were enzymatically reduced at different rates. All muteins were prepared with free thiol in reasonable yield and were modified by N-ethylmaleimide (NEM) or maleimido-PEG. The effect on IL-4Rα binding of cysteine substitution and of the site-specific modification by glutathione, N-ethylmaleimide (NEM), or a branched 2.36 kDa poly(ethylene glycol) (PEG) will be discussed.
Collapse
Affiliation(s)
- Viswanadham Duppatla
- Lehrstuhl für Physiologische Chemie II, Theodor-Boveri-Institut für Biowissenschaften (Biozentrum) der Universität Würzburg, Würzburg, Germany.
| | | | | | | | | | | |
Collapse
|
31
|
Promiscuity and specificity in BMP receptor activation. FEBS Lett 2012; 586:1846-59. [PMID: 22710174 DOI: 10.1016/j.febslet.2012.02.043] [Citation(s) in RCA: 231] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 02/28/2012] [Accepted: 02/29/2012] [Indexed: 02/03/2023]
Abstract
Bone Morphogenetic Proteins (BMPs), together with Transforming Growth Factor (TGF)-β and Activins/Inhibins constitute the TGF-β superfamily of ligands. This superfamily is formed by more than 30 structurally related secreted proteins. Since TGF-β members act as morphogens, either a strict relation between a particular ligand to a distinct cellular receptor and/or temporospatial expression patterns of ligands and receptors is expected. Instead, only a limited number of receptors exist implicating promiscuous interactions of ligands and receptors. Furthermore, in complex tissues a multitude of different ligands can be found, which signal via overlapping subsets of receptors. This raises the intriguing question how concerted interactions of different ligands and receptors generate highly specific cellular signals, which are required during development and tissue homeostasis.
Collapse
|
32
|
Abstract
Interleukin-4 (IL4) is a pleiotropic cytokine involved in host protection from gastrointestinal nematodes. Here, we review the structure, function, and evolutionary history of IL4. Cumulative evidence indicates that over 100 million years of eutherian mammalian evolution, IL4 has experienced multiple episodes of positive selection. We argue that IL4 may have evolved in conflict with pathogen-derived antagonists, and therefore diversified to escape antagonism while being constrained to maintain binding to its cellular receptors. Selective pressure driving IL4 diversification may have arisen from ancient episodes of conflict with parasitic worm-derived IL4 antagonists. Descendants of such antagonists may still equip the armamentarium of contemporary gastrointestinal nematodes.
Collapse
Affiliation(s)
- Meenu R Pillai
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | |
Collapse
|
33
|
Yoon DJ, Liu CT, Quinlan DS, Nafisi PM, Kamei DT. Intracellular trafficking considerations in the development of natural ligand-drug molecular conjugates for cancer. Ann Biomed Eng 2011; 39:1235-51. [PMID: 21350890 PMCID: PMC3069328 DOI: 10.1007/s10439-011-0280-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 02/12/2011] [Indexed: 12/01/2022]
Abstract
Overexpressed receptors, characteristic of many cancers, have been targeted by various researchers to achieve a more specific treatment for cancer. A common approach is to use the natural ligand for the overexpressed receptor as a cancer-targeting agent which can deliver a chemically or genetically conjugated toxic molecule. However, it has been found that the therapeutic efficacy of such ligand-drug molecular conjugates can be limited, since they naturally follow the intracellular trafficking pathways of the endogenous ligands. Therefore, a thorough understanding of the intracellular trafficking properties of these ligands can lead to novel design criteria for engineering ligands to be more effective drug carriers. This review presents a few commonly used ligand/receptor systems where intracellular trafficking considerations can potentially improve the therapeutic efficacy of the ligand-drug molecular conjugates.
Collapse
Affiliation(s)
- Dennis J. Yoon
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Christina T. Liu
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Devin S. Quinlan
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Parsa M. Nafisi
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Daniel T. Kamei
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
| |
Collapse
|
34
|
Pierce BG, Haidar JN, Yu Y, Weng Z. Combinations of affinity-enhancing mutations in a T cell receptor reveal highly nonadditive effects within and between complementarity determining regions and chains. Biochemistry 2010; 49:7050-9. [PMID: 20681514 DOI: 10.1021/bi901969a] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Understanding the energetic and structural response to multiple mutations in a protein-protein interface is a key aspect of rational protein design. Here we investigate the cooperativity of combinations of point mutations of a T cell receptor (TCR) that binds in vivo to HLA-A2 MHC and a viral peptide. The mutations were obtained from two sources: a structure-based design study on the TCR alpha chain (nine mutations) and an in vitro selection study on the TCR beta chain (four mutations). In addition to combining the highest-affinity variants from each chain, we tested other combinations of mutations within and among the chains, for a total of 23 TCR mutants that we measured for binding kinetics to the peptide and major histocompatibility complex. A wide range of binding affinities was observed, from 2- to 1000-fold binding improvement versus that of the wild type, with significant nonadditive effects observed within and between TCR chains. This included an amino acid-dependent cooperative interaction between CDR1 and CDR3 residues that are separated by more than 9 A in the wild-type complex. When analyzing the kinetics of the mutations, we found that the association rates were primarily responsible for the cooperativity, while the dissociation rates were responsible for the anticooperativity (less-than-additive energetics). On the basis of structural modeling of anticooperative mutants, we determined that side chain clash between proximal mutants likely led to nonadditive binding energies. These results highlight the complex nature of TCR association and binding and will be informative in future design efforts that combine multiple mutant residues.
Collapse
Affiliation(s)
- Brian G Pierce
- Bioinformatics Program, Boston University, Boston, Massachusetts 02215, USA
| | | | | | | |
Collapse
|
35
|
A selection fit mechanism in BMP receptor IA as a possible source for BMP ligand-receptor promiscuity. PLoS One 2010; 5. [PMID: 20927405 PMCID: PMC2946932 DOI: 10.1371/journal.pone.0013049] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Accepted: 09/02/2010] [Indexed: 11/29/2022] Open
Abstract
Background Members of the TGF-β superfamily are characterized by a highly promiscuous ligand-receptor interaction as is readily apparent from the numeral discrepancy of only seven type I and five type II receptors available for more than 40 ligands. Structural and functional studies have been used to address the question of how specific signals can be deduced from a limited number of receptor combinations and to unravel the molecular mechanisms underlying the protein-protein recognition that allow such limited specificity. Principal Findings In this study we have investigated how an antigen binding antibody fragment (Fab) raised against the extracellular domain of the BMP receptor type IA (BMPR-IA) recognizes the receptor's BMP-2 binding epitope and thereby neutralizes BMP-2 receptor activation. The crystal structure of the complex of the BMPR-IA ectodomain bound to the Fab AbD1556 revealed that the contact surface of BMPR-IA overlaps extensively with the contact surface for BMP-2 interaction. Although the structural epitopes of BMPR-IA to both binding partners coincides, the structures of BMPR-IA in the two complexes differ significantly. In contrast to the structural differences, alanine-scanning mutagenesis of BMPR-IA showed that the functional determinants for binding to the antibody and BMP-2 are almost identical. Conclusions Comparing the structures of BMPR-IA bound to BMP-2 or bound to the Fab AbD1556 with the structure of unbound BMPR-IA shows that binding of BMPR-IA to its interaction partners follows a selection fit mechanism, possibly indicating that the ligand promiscuity of BMPR-IA is inherently encoded by structural adaptability. The functional and structural analysis of the BMPR-IA binding antibody AbD1556 mimicking the BMP-2 binding epitope may thus pave the way for the design of low-molecular weight synthetic receptor binders/inhibitors.
Collapse
|
36
|
Schreiber G, Haran G, Zhou HX. Fundamental aspects of protein-protein association kinetics. Chem Rev 2010; 109:839-60. [PMID: 19196002 DOI: 10.1021/cr800373w] [Citation(s) in RCA: 576] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- G Schreiber
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, 76100, Israel.
| | | | | |
Collapse
|
37
|
Koyanagi M, Kerns JA, Chung L, Zhang Y, Brown S, Moldoveanu T, Malik HS, Bix M. Diversifying selection and functional analysis of interleukin-4 suggests antagonism-driven evolution at receptor-binding interfaces. BMC Evol Biol 2010; 10:223. [PMID: 20649995 PMCID: PMC3017759 DOI: 10.1186/1471-2148-10-223] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 07/22/2010] [Indexed: 12/30/2022] Open
Abstract
Background Interleukin-4 (IL4) is a secreted immunoregulatory cytokine critically involved in host protection from parasitic helminths [1]. Reasoning that helminths may have evolved mechanisms to antagonize IL4 to maximize their dispersal, we explored mammalian IL4 evolution. Results This analysis revealed evidence of diversifying selection at 15 residues, clustered in epitopes responsible for IL4 binding to its Type I and Type II receptors. Such a striking signature of selective pressure suggested either recurrent episodes of pathogen antagonism or ligand/receptor co-evolution. To test the latter possibility, we performed detailed functional analysis of IL4 allotypes expressed by Mus musculus musculus and Mus musculus castaneus, which happen to differ at 5 residues (including three at positively selected sites) in and adjacent to the site 1 epitope that binds the IL4Rα subunit shared by the Type I and Type II IL4 receptors. We show that this intra-species variation affects the ability of IL4 neither to bind IL4 receptor alpha (IL4Rα) nor to signal biological responses through its Type I receptor. Conclusions Our results -- reminiscent of clustered positively selected sites revealing functionally important residues at host-virus interaction interfaces -- are consistent with IL4 having evolved to avoid recurrent pathogen antagonism, while maintaining the capacity to bind and signal through its cognate receptor. This work exposes what may be a general feature of evolutionary conflicts fought by pathogen antagonists at host protein-protein interaction interfaces involved in immune signaling: the emergence of receptor-binding ligand epitopes capable of buffering amino acid variation.
Collapse
Affiliation(s)
- Madoka Koyanagi
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Sebald W, Nickel J, Zhang JL, Mueller TD. Molecular basis of cytokine signalling - theme and variations. FEBS J 2009; 277:106-18. [DOI: 10.1111/j.1742-4658.2009.07480.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
39
|
Carbonell P, Nussinov R, del Sol A. Energetic determinants of protein binding specificity: insights into protein interaction networks. Proteomics 2009; 9:1744-53. [PMID: 19253304 PMCID: PMC7299235 DOI: 10.1002/pmic.200800425] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Indexed: 01/26/2023]
Abstract
One of the challenges of the postgenomic era is to provide a more realistic representation of cellular processes by combining a systems biology description of functional networks with information on their interacting components. Here we carried out a systematic large-scale computational study on a structural protein-protein interaction network dataset in order to dissect thermodynamic characteristics of binding determining the interplay between protein affinity and specificity. As expected, interactions involving specific binding sites display higher affinities than those of promiscuous binding sites. Next, in order to investigate a possible role of modular distribution of hot spots in binding specificity, we divided binding sites into modules previously shown to be energetically independent. In general, hot spots that interact with different partners are located in different modules. We further observed that common hot spots tend to interact with partners exhibiting common binding motifs, whereas different hot spots tend to interact with partners with different motifs. Thus, energetic properties of binding sites provide insights into the way proteins modulate interactions with different partners. Knowledge of those factors playing a role in protein specificity is important for understanding how proteins acquire additional partners during evolution. It should also be useful in drug design.
Collapse
Affiliation(s)
- Pablo Carbonell
- Bioinformatics Research Unit, Research and Development Division, Fujirebio Inc., Komiya-cho, Hachioji-shi, Tokyo 192-0031, Japan
| | - Ruth Nussinov
- Basic Research Program, SAIC-Frederick, Inc., Center for Cancer Research Nanobiology Program, NCI-Frederick, Frederick, MD 21702
- Sackler Inst. of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Antonio del Sol
- Bioinformatics Research Unit, Research and Development Division, Fujirebio Inc., Komiya-cho, Hachioji-shi, Tokyo 192-0031, Japan
| |
Collapse
|
40
|
del Sol A, Carbonell P. The modular organization of domain structures: insights into protein-protein binding. PLoS Comput Biol 2008; 3:e239. [PMID: 18069884 PMCID: PMC2134966 DOI: 10.1371/journal.pcbi.0030239] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2007] [Accepted: 10/17/2007] [Indexed: 12/21/2022] Open
Abstract
Domains are the building blocks of proteins and play a crucial role in protein–protein interactions. Here, we propose a new approach for the analysis and prediction of domain–domain interfaces. Our method, which relies on the representation of domains as residue-interacting networks, finds an optimal decomposition of domain structures into modules. The resulting modules comprise highly cooperative residues, which exhibit few connections with other modules. We found that non-overlapping binding sites in a domain, involved in different domain–domain interactions, are generally contained in different modules. This observation indicates that our modular decomposition is able to separate protein domains into regions with specialized functions. Our results show that modules with high modularity values identify binding site regions, demonstrating the predictive character of modularity. Furthermore, the combination of modularity with other characteristics, such as sequence conservation or surface patches, was found to improve our predictions. In an attempt to give a physical interpretation to the modular architecture of domains, we analyzed in detail six examples of protein domains with available experimental binding data. The modular configuration of the TEM1-β-lactamase binding site illustrates the energetic independence of hotspots located in different modules and the cooperativity of those sited within the same modules. The energetic and structural cooperativity between intramodular residues is also clearly shown in the example of the chymotrypsin inhibitor, where non–binding site residues have a synergistic effect on binding. Interestingly, the binding site of the T cell receptor β chain variable domain 2.1 is contained in one module, which includes structurally distant hot regions displaying positive cooperativity. These findings support the idea that modules possess certain functional and energetic independence. A modular organization of binding sites confers robustness and flexibility to the performance of the functional activity, and facilitates the evolution of protein interactions. Proteins are built by domains, which mediate protein–protein interactions involved in different biological activities. A challenging problem in computational biology is the understanding of the domain–domain interaction mechanism. Here, we propose a new approach for the analysis and prediction of domain–domain binding sites. Our computational approach, which relies on the modular division of 3-D domain structures, identifies modular regions involved in binding and can complement previously introduced predictive methods. Further results illustrate that binding sites display a modular configuration. A detailed analysis of protein domains with available experimental binding data revealed that modules are energetically independent from each other, whereas residues within modules contribute cooperatively to the binding energy. The modular composition of binding surfaces may generate high binding affinity and specificity, and facilitate the appearance of new domain binding partners. This advantageous organization of protein structures has been conserved by evolution and may be used to design an effective drug strategy.
Collapse
Affiliation(s)
- Antonio del Sol
- Bioinformatics Research Unit, Research and Development Division, Fujirebio, Tokyo, Japan.
| | | |
Collapse
|
41
|
LaPorte SL, Juo ZS, Vaclavikova J, Colf LA, Qi X, Heller NM, Keegan AD, Garcia KC. Molecular and structural basis of cytokine receptor pleiotropy in the interleukin-4/13 system. Cell 2008; 132:259-72. [PMID: 18243101 PMCID: PMC2265076 DOI: 10.1016/j.cell.2007.12.030] [Citation(s) in RCA: 421] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2007] [Revised: 11/20/2007] [Accepted: 12/15/2007] [Indexed: 11/19/2022]
Abstract
Interleukin-4 and Interleukin-13 are cytokines critical to the development of T cell-mediated humoral immune responses, which are associated with allergy and asthma, and exert their actions through three different combinations of shared receptors. Here we present the crystal structures of the complete set of type I (IL-4R alpha/gamma(c)/IL-4) and type II (IL-4R alpha/IL-13R alpha1/IL-4, IL-4R alpha/IL-13R alpha1/IL-13) ternary signaling complexes. The type I complex reveals a structural basis for gamma(c)'s ability to recognize six different gamma(c)-cytokines. The two type II complexes utilize an unusual top-mounted Ig-like domain on IL-13R alpha1 for a novel mode of cytokine engagement that contributes to a reversal in the IL-4 versus IL-13 ternary complex assembly sequences, which are mediated through substantially different recognition chemistries. We also show that the type II receptor heterodimer signals with different potencies in response to IL-4 versus IL-13 and suggest that the extracellular cytokine-receptor interactions are modulating intracellular membrane-proximal signaling events.
Collapse
MESH Headings
- Amino Acid Sequence
- Binding Sites
- Cell Line
- Cell Line, Tumor
- Crystallography, X-Ray
- Dimerization
- Dose-Response Relationship, Drug
- Histidine/metabolism
- Humans
- Hydrogen Bonding
- Hydrophobic and Hydrophilic Interactions
- Interleukin-13/genetics
- Interleukin-13/isolation & purification
- Interleukin-13/metabolism
- Interleukin-13/pharmacology
- Interleukin-4/genetics
- Interleukin-4/isolation & purification
- Interleukin-4/metabolism
- Interleukin-4/pharmacology
- Kinetics
- Ligands
- Models, Molecular
- Molecular Mimicry
- Molecular Sequence Data
- Phosphorylation
- Protein Binding
- Protein Structure, Secondary
- Protein Structure, Tertiary
- Receptors, Cytokine/chemistry
- Receptors, Cytokine/metabolism
- Receptors, Interleukin-13/chemistry
- Receptors, Interleukin-13/metabolism
- Receptors, Interleukin-4/chemistry
- Receptors, Interleukin-4/metabolism
- Recombinant Proteins/metabolism
- STAT3 Transcription Factor/metabolism
- STAT6 Transcription Factor/metabolism
- Sequence Homology, Amino Acid
- Signal Transduction
- Thermodynamics
- Tyrosine/metabolism
- X-Ray Diffraction
Collapse
Affiliation(s)
- Sherry L. LaPorte
- Howard Hughes Medical Institute, Departments of Molecular and Cellular Physiology, and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Z. Sean Juo
- Howard Hughes Medical Institute, Departments of Molecular and Cellular Physiology, and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Jana Vaclavikova
- Howard Hughes Medical Institute, Departments of Molecular and Cellular Physiology, and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Leremy A. Colf
- Howard Hughes Medical Institute, Departments of Molecular and Cellular Physiology, and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Xiulan Qi
- Center for Vascular and Inflammatory Diseases and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - Nicola M. Heller
- Center for Vascular and Inflammatory Diseases and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - Achsah D. Keegan
- Center for Vascular and Inflammatory Diseases and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - K. Christopher Garcia
- Howard Hughes Medical Institute, Departments of Molecular and Cellular Physiology, and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
42
|
Yang X, Lee J, Brooks J, Wilhelm J, Myszka D, Kasaian MT, Goldman S, Wolf S, Fitz LJ. Binding characterization of the interleukin-13 signaling complex and development of a ternary time-resolved fluorescence resonance energy transfer assay. Anal Biochem 2008; 376:206-12. [PMID: 18328249 DOI: 10.1016/j.ab.2008.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2007] [Revised: 02/06/2008] [Accepted: 02/07/2008] [Indexed: 12/01/2022]
Abstract
Interleukin-13 (IL-13) is a critical mediator of pulmonary pathology associated with asthma. Drugs that block the biological function of IL-13 may be an effective treatment for asthma. IL-13 signals by forming a ternary complex with IL-13Ralpha1 and IL-4R. Genetic variants of IL-13 and of its receptor components have been linked to asthma. One in particular, IL-13R110Q, is associated with increased IgE levels and asthma. We characterized the interactions of the binary complexes composed of IL-13 or IL-13R110Q with IL-13Ralpha1 and the ternary complexes composed of IL-13 or IL-13R110Q and IL-13Ralpha1 with IL-4R using surface plasmon resonance and time-resolved fluorescence resonance energy transfer (TR-FRET). By both biophysical methods, we found no differences between IL-13 and IL-13R110Q binding in either the binary or the ternary complex. IL-4R bound to the IL-13/IL-13Ralpha1 complex with slow on and off rates, resulting in a relatively weak affinity of about 100nM. We developed a TR-FRET assay targeting the interaction between the IL-4R and the binary complex. Two antibodies with known binding epitopes to IL-13 that block binding to either IL-13Ralpha1 or IL-4R inhibited the TR-FRET signal formed by the ternary complex. This assay will be useful to identify and characterize inhibitory molecules of IL-13 function.
Collapse
Affiliation(s)
- Xiaoke Yang
- Wyeth Inflammation, 200 Cambridge Park Drive, Cambridge, MA 02140, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Knackmuss S, Krause S, Engel K, Reusch U, Virchow JC, Mueller T, Kraich M, Little M, Luttmann W, Friedrich K. Specific inhibition of interleukin-13 activity by a recombinant human single-chain immunoglobulin domain directed against the IL-13 receptor alpha1 chain. Biol Chem 2007; 388:325-30. [PMID: 17338640 DOI: 10.1515/bc.2007.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Interleukin-13 (IL-13) is a T-cell-derived pleiotropic cytokine of particular medical importance because of its critical role in the development of allergic asthma. The effects of IL-13 on its target cells are mediated through a dimeric transmembrane receptor (IL-13R), which shares the IL-4Ralpha subunit with the IL-4R system, but contains as a specific component the IL-13Ralpha1 chain. We have generated a set of single-chain Fv fragments with specific binding capacity to the extracellular domain of the human IL-13Ralpha1 receptor. Bacteriophage clones displaying receptor-binding antibody domains were selected from both naive and synthetic libraries by repetitive panning on recombinant and cell surface-expressed recombinant IL-13Ralpha1. Their specific reactivity with native human IL-13Ralpha1 expressed on the surface of transfected cells was demonstrated by flow cytometry. One binder that specifically interfered with cell activation by IL-13 was extensively characterized. This scFv inhibited IL-13-driven gene transcription and cell proliferation in test cell lines, as well as IL-13-induced activation of primary human monocytes in a dose-dependent manner, with an IC(50) below 300 nM. This novel reagent thus constitutes a valuable tool for the further elucidation of IL-13 function in disease and offers potential therapeutic perspectives.
Collapse
Affiliation(s)
- Stefan Knackmuss
- Affimed Therapeutics AG, Im Neuenheimer Feld 582, D-69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Reichmann D, Rahat O, Cohen M, Neuvirth H, Schreiber G. The molecular architecture of protein-protein binding sites. Curr Opin Struct Biol 2007; 17:67-76. [PMID: 17239579 DOI: 10.1016/j.sbi.2007.01.004] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Revised: 12/13/2006] [Accepted: 01/10/2007] [Indexed: 11/16/2022]
Abstract
The formation of specific protein interactions plays a crucial role in most, if not all, biological processes, including signal transduction, cell regulation, the immune response and others. Recent advances in our understanding of the molecular architecture of protein-protein binding sites, which facilitates such diversity in binding affinity and specificity, are enabling us to address key questions. What is the amino acid composition of binding sites? What are interface hotspots? How are binding sites organized? What are the differences between tight and weak interacting complexes? How does water contribute to binding? Can the knowledge gained be translated into protein design? And does a universal code for binding exist, or is it the architecture and chemistry of the interface that enable diverse but specific binding solutions?
Collapse
Affiliation(s)
- Dana Reichmann
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | |
Collapse
|
45
|
Rich RL, Myszka DG. Survey of the year 2006 commercial optical biosensor literature. J Mol Recognit 2007; 20:300-66. [DOI: 10.1002/jmr.862] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|