1
|
Eirich P, Nesterov P, Shityakov S, Skorb EV, Sander B, Broscheit J, Dandekar T, Jones NG, Engstler M. The release of host-derived antibodies bound to the variant surface glycoprotein (VSG) of Trypanosoma brucei cannot be explained by pH-dependent conformational changes of the VSG dimer. OPEN RESEARCH EUROPE 2024; 4:87. [PMID: 38903703 PMCID: PMC11187536 DOI: 10.12688/openreseurope.16783.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 01/26/2024] [Indexed: 06/22/2024]
Abstract
Background Trypanosoma brucei is a protozoan parasite that evades the mammalian host's adaptive immune response by antigenic variation of the highly immunogenic variant surface glycoprotein (VSG). VSGs form a dense surface coat that is constantly recycled through the endosomal system. Bound antibodies are separated in the endosome from the VSG and destroyed in the lysosome. For VSGs it has been hypothesized that pH-dependent structural changes of the VSG could occur in the more acidic environment of the endosome and hence, facilitate the separation of the antibody from the VSG. Methods We used size exclusion chromatography, where molecules are separated according to their hydrodynamic radius to see if the VSG is present as a homodimer at both pH values. To gain information about the structural integrity of the protein we used circular dichroism spectroscopy by exposing the VSG in solution to a mixture of right- and left-circularly polarized light and analysing the absorbed UV spectra. Evaluation of protein stability and molecular dynamics simulations at different pH values was performed using different computational methods. Results We show, for an A2-type VSG, that the dimer size is only slightly larger at pH 5.2 than at pH 7.4. Moreover, the dimer was marginally more stable at lower pH due to the higher affinity (ΔG = 353.37 kcal/mol) between the monomers. Due to the larger size, the predicted epitopes were more exposed to the solvent at low pH. Moderate conformational changes (ΔRMSD = 0.35 nm) in VSG were detected between the dimers at pH 5.2 and pH 7.4 in molecular dynamics simulations, and no significant differences in the protein secondary structure were observed by circular dichroism spectroscopy. Conclusions Thus, the dissociation of anti-VSG-antibodies in endosomes cannot be explained by changes in pH.
Collapse
Affiliation(s)
- Patrick Eirich
- Department of Cell & Developmental Biology, Biocentre, University of Würzburg, Würzburg, Bavaria, 97074, Germany
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Würzburg University Hospital, University of Würzburg, Würzburg, Bavaria, 97080, Germany
| | - Pavel Nesterov
- Infochemistry Scientific Center, Laboratory of Chemoinformatics, ITMO University, Saint Petersburg, Saint Petersburg, 191002, Russian Federation
| | - Sergey Shityakov
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Würzburg University Hospital, University of Würzburg, Würzburg, Bavaria, 97080, Germany
- Infochemistry Scientific Center, Laboratory of Chemoinformatics, ITMO University, Saint Petersburg, Saint Petersburg, 191002, Russian Federation
- Department of Bioinformatics, Biocentre, University of Würzburg, Würzburg, Bavaria, 97074, Germany
| | - Ekaterina V. Skorb
- Infochemistry Scientific Center, Laboratory of Chemoinformatics, ITMO University, Saint Petersburg, Saint Petersburg, 191002, Russian Federation
| | - Bodo Sander
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Bavaria, 97080, Germany
| | - Jens Broscheit
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Würzburg University Hospital, University of Würzburg, Würzburg, Bavaria, 97080, Germany
| | - Thomas Dandekar
- Department of Bioinformatics, Biocentre, University of Würzburg, Würzburg, Bavaria, 97074, Germany
| | - Nicola G. Jones
- Department of Cell & Developmental Biology, Biocentre, University of Würzburg, Würzburg, Bavaria, 97074, Germany
| | - Markus Engstler
- Department of Cell & Developmental Biology, Biocentre, University of Würzburg, Würzburg, Bavaria, 97074, Germany
| |
Collapse
|
2
|
Borio A, Holgado A, Passegger C, Strobl H, Beyaert R, Heine H, Zamyatina A. Exploring Species-Specificity in TLR4/MD-2 Inhibition with Amphiphilic Lipid A Mimicking Glycolipids. Molecules 2023; 28:5948. [PMID: 37630200 PMCID: PMC10459247 DOI: 10.3390/molecules28165948] [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: 07/02/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
The Toll-like receptor 4 (TLR4)/myeloid differentiation factor 2 (MD-2) complex is a key receptor of the innate immune system and a major driver of inflammation that is responsible for the multifaceted defense response to Gram-negative infections. However, dysfunction in the tightly regulated mechanisms of TLR4-mediated signaling leads to the uncontrolled upregulation of local and systemic inflammation, often resulting in acute or chronic disease. Therefore, the TLR4/MD-2 receptor complex is an attractive target for the design and development of anti-inflammatory therapies which aim to control the unrestrained activation of TLR4-mediated signaling. Complex structure-activity relationships and species-specificity behind ligand recognition by the TLR4/MD-2 complex complicate the development of MD-2-specific TLR4 antagonists. The restriction of the conformational flexibility of the disaccharide polar head group is one of the key structural features of the newly developed lipid A-mimicking glycophospholipids, which are potential inhibitors of TLR4-mediated inflammation. Since phosphorylation has a crucial influence on MD-2-ligand interaction, glycolipids with variable numbers and positioning of phosphate groups were synthesized and evaluated for their ability to inhibit TLR4-mediated pro-inflammatory signaling in human and murine immune cells. A bis-phosphorylated glycolipid was found to have nanomolar antagonist activity on human TLR4 while acting as a partial agonist on murine TLR4. The glycolipid inhibited mTLR4/MD-2-mediated cytokine release, acting as an antagonist in the presence of lipopolysaccharide (LPS), but at the same time induced low-level cytokine production.
Collapse
Affiliation(s)
- Alessio Borio
- Institute of Organic Chemistry, Department of Chemistry, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Aurora Holgado
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Department for Biomedical Molecular Biology, Ghent University, Technologiepark 71, B-9052 Ghent, Belgium
| | - Christina Passegger
- Division of Immunology and Pathophysiology, Medical University Graz, Heinrichstraße 31, 8010 Graz, Austria
| | - Herbert Strobl
- Division of Immunology and Pathophysiology, Medical University Graz, Heinrichstraße 31, 8010 Graz, Austria
| | - Rudi Beyaert
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Department for Biomedical Molecular Biology, Ghent University, Technologiepark 71, B-9052 Ghent, Belgium
| | - Holger Heine
- Research Group Innate Immunity, Priority Area Chronic Lung Diseases, Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Parkallee 22, 23845 Borstel, Germany
| | - Alla Zamyatina
- Institute of Organic Chemistry, Department of Chemistry, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| |
Collapse
|
3
|
García MM, Molina-Álvarez M, Rodríguez-Rivera C, Paniagua N, Quesada E, Uranga JA, Rodríguez-Franco MI, Pascual D, Goicoechea C. Antinociceptive and modulatory effect of pathoplastic changes in spinal glia of a TLR4/CD14 blocking molecule in two models of pain in rat. Biomed Pharmacother 2022; 150:112986. [PMID: 35462333 DOI: 10.1016/j.biopha.2022.112986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/02/2022] [Accepted: 04/14/2022] [Indexed: 11/02/2022] Open
Abstract
The role of spinal glia in the development and maintenance of chronic pain has become over the last years a subject of increasing interest. In this regard, toll-like receptor 4 (TLR4) signaling has been proposed as a major trigger mechanism. Hence, in this study we explored the implications of TLR4 inhibition in the periphery and primarily in the CNS, focusing on the impact this inhibition renders in pain development and glia activation in the dorsal horn in two models of pain. Making use of a synthetic cluster of differentiation 14 (CD14)/TLR4 antagonist, the effect of TLR4 blockade on tactile allodynia and heat hyperalgesia was evaluated in osteoarthritic and postoperative rat models. An in vitro parallel artificial membrane permeation assay was performed to determine the proneness of the drug to permeate the blood-brain barrier prior to systemic and central administration. Findings suggest a dominant role of peripheral TLR4 in the model of incisional pain, whilst both peripheral and central TLR4 seem to be responsible for osteoarthritic pain. That is, central and peripheral TLR4 may be differently involved in the etiopathology of diverse types of pain what potentially seems a promising approach in the management of pain.
Collapse
Affiliation(s)
- Miguel M García
- Area of Pharmacology and Nutrition and Bromatology, Department of Basic Health Sciences, School of Health Sciences, Universidad Rey Juan Carlos, High Performance Experimental Pharmacology Research Group, Universidad Rey Juan Carlos (PHARMAKOM), Madrid, Spain; Unidad Asociada I+D+i Instituto de Química Médica (IQM-CSIC)-URJC, Madrid, Spain
| | - Miguel Molina-Álvarez
- Area of Pharmacology and Nutrition and Bromatology, Department of Basic Health Sciences, School of Health Sciences, Universidad Rey Juan Carlos, High Performance Experimental Pharmacology Research Group, Universidad Rey Juan Carlos (PHARMAKOM), Madrid, Spain; Unidad Asociada I+D+i Instituto de Química Médica (IQM-CSIC)-URJC, Madrid, Spain
| | - Carmen Rodríguez-Rivera
- Area of Pharmacology and Nutrition and Bromatology, Department of Basic Health Sciences, School of Health Sciences, Universidad Rey Juan Carlos, High Performance Experimental Pharmacology Research Group, Universidad Rey Juan Carlos (PHARMAKOM), Madrid, Spain; Unidad Asociada I+D+i Instituto de Química Médica (IQM-CSIC)-URJC, Madrid, Spain
| | - Nancy Paniagua
- Area of Pharmacology and Nutrition and Bromatology, Department of Basic Health Sciences, School of Health Sciences, Universidad Rey Juan Carlos, High Performance Experimental Pharmacology Research Group, Universidad Rey Juan Carlos (PHARMAKOM), Madrid, Spain; Unidad Asociada I+D+i Instituto de Química Médica (IQM-CSIC)-URJC, Madrid, Spain
| | - Ernesto Quesada
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain; Unidad Asociada I+D+i Instituto de Química Médica (IQM-CSIC)-URJC, Madrid, Spain
| | - José Antonio Uranga
- Area of Histology, Department of Basic Health Sciences, School of Health Sciences, Universidad Rey Juan Carlos, High Performance Research Group in Physiopathology and Pharmacology of the Digestive System, Universidad Rey Juan Carlos (NEUGUT), Madrid, Spain
| | | | - David Pascual
- Area of Pharmacology and Nutrition and Bromatology, Department of Basic Health Sciences, School of Health Sciences, Universidad Rey Juan Carlos, High Performance Experimental Pharmacology Research Group, Universidad Rey Juan Carlos (PHARMAKOM), Madrid, Spain; Unidad Asociada I+D+i Instituto de Química Médica (IQM-CSIC)-URJC, Madrid, Spain.
| | - Carlos Goicoechea
- Area of Pharmacology and Nutrition and Bromatology, Department of Basic Health Sciences, School of Health Sciences, Universidad Rey Juan Carlos, High Performance Experimental Pharmacology Research Group, Universidad Rey Juan Carlos (PHARMAKOM), Madrid, Spain; Unidad Asociada I+D+i Instituto de Química Médica (IQM-CSIC)-URJC, Madrid, Spain
| |
Collapse
|
4
|
Rawat K, Pal A, Banerjee S, Pal A, Mandal SC, Batabyal S. Ovine CD14- an Immune Response Gene Has a Role Against Gastrointestinal Nematode Haemonchus contortus-A Novel Report. Front Immunol 2021; 12:664877. [PMID: 34335567 PMCID: PMC8324245 DOI: 10.3389/fimmu.2021.664877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
CD14 (also known as the monocyte differentiation antigen) is an important immune response gene known to be primarily responsible for innate immunity against bacterial pathogens, and as a pattern recognition receptor (PRR), binds with LPS (endotoxin), lipoproteins, and lipotechoic acid of bacteria. So far very limited work has been conducted in parasitic immunology. In the current study, we reported the role of CD14 in parasitic immunology in livestock species (sheep) for the first time. Ovine CD14 is characterized as a horse-shoe shaped bent solenoid with a hydrophobic amino-terminal pocket for CD14 along with domains. High mutation frequency was observed, out of total 41 mutations identified, 23 mutations were observed to be thermodynamically unstable and 11 mutations were deleterious in nature, causing major functional alteration of important domains of CD14, an indication of variations in individual susceptibility for sheep against Haemonchus contortus infestations. In silico studies with molecular docking reveal a role of immune response against Haemonchus contortus in sheep, which is later confirmed with experimental evidence through differential mRNA expression analysis for sheep, which revealed better expression of CD14 in Haemonchus contortus infected sheep compared to that of non-infected sheep. We confirmed the above findings with supportive evidence through haematological and biochemical analyses. Phylogenetic analysis was conducted to assess the evolutionary relationship with respect to humans and it was observed that sheep may well be used as model organisms due to better genetic closeness compared to that of mice.
Collapse
Affiliation(s)
- Kavita Rawat
- Department of BioChemistry, West Bengal University of Animal and Fishery Sciences, Kolkata, India
| | - Aruna Pal
- Department of LFC, West Bengal University of Animal and Fishery Sciences, Kolkata, India
| | - Samiddha Banerjee
- Department of Animal Science, Visva Bharati University, Bolpur, India
| | - Abantika Pal
- Department of Computer Science, Indian Institute of Technology, Kharagpur, India
| | - Subhas Chandra Mandal
- Department of Parasitology, West Bengal University of Animal and Fishery Sciences, Kolkata, India
| | - Subhasis Batabyal
- Department of BioChemistry, West Bengal University of Animal and Fishery Sciences, Kolkata, India
| |
Collapse
|
5
|
Heine H, Adanitsch F, Peternelj TT, Haegman M, Kasper C, Ittig S, Beyaert R, Jerala R, Zamyatina A. Tailored Modulation of Cellular Pro-inflammatory Responses With Disaccharide Lipid A Mimetics. Front Immunol 2021; 12:631797. [PMID: 33815382 PMCID: PMC8012497 DOI: 10.3389/fimmu.2021.631797] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/17/2021] [Indexed: 01/08/2023] Open
Abstract
Pro-inflammatory signaling mediated by Toll-like receptor 4 (TLR4)/myeloid differentiation-2 (MD-2) complex plays a crucial role in the instantaneous protection against infectious challenge and largely contributes to recovery from Gram-negative infection. Activation of TLR4 also boosts the adaptive immunity which is implemented in the development of vaccine adjuvants by application of minimally toxic TLR4 activating ligands. The modulation of pro-inflammatory responses via the TLR4 signaling pathway was found beneficial for management of acute and chronic inflammatory disorders including asthma, allergy, arthritis, Alzheimer disease pathology, sepsis, and cancer. The TLR4/MD-2 complex can recognize the terminal motif of Gram-negative bacterial lipopolysaccharide (LPS)—a glycophospholipid lipid A. Although immense progress in understanding the molecular basis of LPS-induced TLR4-mediated signaling has been achieved, gradual, and predictable TLR4 activation by structurally defined ligands has not yet been attained. We report on controllable modulation of cellular pro-inflammatory responses by application of novel synthetic glycolipids—disaccharide-based lipid A mimetics (DLAMs) having picomolar affinity for TLR4/MD-2. Using crystal structure inspired design we have developed endotoxin mimetics where the inherently flexible β(1 → 6)-linked diglucosamine backbone of lipid A is replaced by a conformationally restricted α,α-(1↔1)-linked disaccharide scaffold. The tertiary structure of the disaccharide skeleton of DLAMs mirrors the 3-dimensional shape of TLR4/MD-2 bound E. coli lipid A. Due to exceptional conformational rigidity of the sugar scaffold, the specific 3D organization of DLAM must be preserved upon interaction with proteins. These structural factors along with specific acylation and phosphorylation pattern can ensure picomolar affinity for TLR4 and permit efficient dimerization of TLR4/MD-2/DLAM complexes. Since the binding pose of lipid A in the binding pocket of MD-2 (±180°) is crucial for the expression of biological activity, the chemical structure of DLAMs was designed to permit a predefined binding orientation in the binding groove of MD-2, which ensured tailored and species-independent (human and mice) TLR4 activation. Manipulating phosphorylation and acylation pattern at the sugar moiety facing the secondary dimerization interface allowed for adjustable modulation of the TLR4-mediated signaling. Tailored modulation of cellular pro-inflammatory responses by distinct modifications of the molecular structure of DLAMs was attained in primary human and mouse immune cells, lung epithelial cells and TLR4 transfected HEK293 cells.
Collapse
Affiliation(s)
- Holger Heine
- Research Group Innate Immunity, Research Center Borstel - Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Florian Adanitsch
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Tina Tinkara Peternelj
- Department of Biotechnology, National Institute of Chemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Mira Haegman
- Unit of Molecular Signal Transduction in Inflammation, Department of Biomedical Molecular Biology, Ghent University, Center for Inflammation Research, VIB, Ghent, Belgium
| | | | - Simon Ittig
- Biozentrum University of Basel, Basel, Switzerland
| | - Rudi Beyaert
- Unit of Molecular Signal Transduction in Inflammation, Department of Biomedical Molecular Biology, Ghent University, Center for Inflammation Research, VIB, Ghent, Belgium
| | - Roman Jerala
- Department of Biotechnology, National Institute of Chemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Alla Zamyatina
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| |
Collapse
|
6
|
Garcia MM, Goicoechea C, Molina-Álvarez M, Pascual D. Toll-like receptor 4: A promising crossroads in the diagnosis and treatment of several pathologies. Eur J Pharmacol 2020; 874:172975. [PMID: 32017939 DOI: 10.1016/j.ejphar.2020.172975] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 12/20/2019] [Accepted: 01/29/2020] [Indexed: 12/26/2022]
Abstract
Toll-like receptor 4 (TLR4) is expressed in a wide variety of cells and is the central component of the mammalian innate immune system. Since its discovery in 1997, TLR4 has been assigned an ever-increasing number of functions that extend from pathogen recognition to tissue damage identification and promotion of the intrinsic "damage repair response" in pain, intestinal, respiratory and vascular disorders. Precisely, the finding of conserved sequence homology among species along with the molecular and functional characterisation of the TLR4 gene enabled researchers to envisage a common operating system in the activation of innate immunity and the initiation of plastic changes at the onset of chronic pain. Malfunctioning in other conditions was conceived in parallel. In this respect, "pivot" proteins and pathway redundancy are not just evolutionary leftovers but essential for normal functioning or cell survival. Indeed, at present, TLR4 single nucleotide polymorphisms (SNP) and their association with certain dysfunctions and diseases are being confirmed in different pools of patients. However, despite its ability to trigger pathogen infection or alternatively tissue injury communications to immune system, TLR4 targeting might not be considered a panacea. This review article represents a compilation of what we know about TLR4 from clinics and basic research on the 20th anniversary of its discovery. Understanding how to fine-tune the interaction between TLR4 and its specific ligands may lead in the next decades to the development of promising new treatments, reducing polypharmacy and probably having an impact on drug use in numerous pathologies.
Collapse
Affiliation(s)
- Miguel M Garcia
- Area of Pharmacology, Nutrition and Bromatology, Department of Basic Health Sciences, Universidad Rey Juan Carlos, Avda, Atenas S/n, 28922, Alcorcón, Spain
| | - Carlos Goicoechea
- Area of Pharmacology, Nutrition and Bromatology, Department of Basic Health Sciences, Universidad Rey Juan Carlos, Avda, Atenas S/n, 28922, Alcorcón, Spain
| | - Miguel Molina-Álvarez
- Area of Pharmacology, Nutrition and Bromatology, Department of Basic Health Sciences, Universidad Rey Juan Carlos, Avda, Atenas S/n, 28922, Alcorcón, Spain
| | - David Pascual
- Area of Pharmacology, Nutrition and Bromatology, Department of Basic Health Sciences, Universidad Rey Juan Carlos, Avda, Atenas S/n, 28922, Alcorcón, Spain.
| |
Collapse
|
7
|
Borio A, Holgado A, Garate JA, Beyaert R, Heine H, Zamyatina A. Disaccharide-Based Anionic Amphiphiles as Potent Inhibitors of Lipopolysaccharide-Induced Inflammation. ChemMedChem 2018; 13:2317-2331. [PMID: 30276970 DOI: 10.1002/cmdc.201800505] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/18/2018] [Indexed: 01/08/2023]
Abstract
Despite significant advances made in the last decade in the understanding of molecular mechanisms of sepsis and in the development of clinically relevant therapies, sepsis remains the leading cause of mortality in intensive care units with increasing incidence worldwide. Toll-like receptor 4 (TLR4)-a transmembrane pattern-recognition receptor responsible for propagating the immediate immune response to Gram-negative bacterial infection-plays a central role in the pathogenesis of sepsis and chronic inflammation-related disorders. TLR4 is complexed with the lipopolysaccharide (LPS)-sensing protein myeloid differentiation-2 (MD-2) which represents a preferred target for establishing new anti-inflammatory treatment strategies. Herein we report the development, facile synthesis, and biological evaluation of novel disaccharide-based TLR4⋅MD-2 antagonists with potent anti-endotoxic activity at micromolar concentrations. A series of synthetic anionic glycolipids entailing amide-linked β-ketoacyl lipid residues was prepared in a straightforward manner by using a single orthogonally protected nonreducing diglucosamine scaffold. Suppression of the LPS-induced release of interleukin-6 and tumor necrosis factor was monitored and confirmed in human immune cells (MNC and THP1) and mouse macrophages. Structure-activity relationship studies and molecular dynamics simulations revealed the structural basis for the high-affinity interaction between anionic glycolipids and MD-2, and highlighted two compounds as leads for the development of potential anti-inflammatory therapeutics.
Collapse
Affiliation(s)
- Alessio Borio
- Department of Chemistry, University of Natural Resources and Life Sciences, Muthgasse 18, 1190, Vienna, Austria
| | - Aurora Holgado
- Department for Biomedical Molecular Biology, Unit of Molecular Signal Transduction in Inflammation, Ghent University, Center for Inflammation Research, VIB, Technologiepark 927, 9052, Ghent, Belgium
| | - Jose Antonio Garate
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile
| | - Rudi Beyaert
- Department for Biomedical Molecular Biology, Unit of Molecular Signal Transduction in Inflammation, Ghent University, Center for Inflammation Research, VIB, Technologiepark 927, 9052, Ghent, Belgium
| | - Holger Heine
- Research Group Innate Immunity, Research Center Borstel-Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Disease (DZL), Parkallee 22, 23845, Borstel, Germany
| | - Alla Zamyatina
- Department of Chemistry, University of Natural Resources and Life Sciences, Muthgasse 18, 1190, Vienna, Austria
| |
Collapse
|
8
|
Yang D, Han Z, Alam MM, Oppenheim JJ. High-mobility group nucleosome binding domain 1 (HMGN1) functions as a Th1-polarizing alarmin. Semin Immunol 2018; 38:49-53. [PMID: 29503123 DOI: 10.1016/j.smim.2018.02.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 02/26/2018] [Indexed: 12/16/2022]
Abstract
High-mobility group (HMG) nucleosome binding domain 1 (HMGN1), which previously was thought to function only as a nucleosome-binding protein that regulates chromatin structure, histone modifications, and gene expression, was recently discovered to be an alarmin that contributes extracellularly to the generation of innate and adaptive immune responses. HMGN1 promotes DC recruitment through interacting with a Gαi protein-coupled receptor (GiPCR) and activates DCs predominantly through triggering TLR4. HMGN1 preferentially promotes Th1-type immunity, which makes it relevant for the fields of vaccinology, autoimmunity, and oncoimmunology. Here, we discuss the alarmin properties of HMGN1 and update recent advances on its roles in immunity and potential applications for immunotherapy of tumors.
Collapse
Affiliation(s)
- De Yang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, National Institute of Health, USA.
| | - Zhen Han
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, National Institute of Health, USA
| | - Md Masud Alam
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, National Institute of Health, USA
| | - Joost J Oppenheim
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, National Institute of Health, USA.
| |
Collapse
|
9
|
Abstract
In humans and other mammals, recognition of endotoxins—abundant surface lipopolysaccharides (LPS) of Gram-negative bacteria—provides a potent stimulus for induction of inflammation and mobilization of host defenses. The structurally unique lipid A region of LPS is the principal determinant of this pro-inflammatory activity. This region of LPS is normally buried within the bacterial outer membrane and aggregates of purified LPS, making even more remarkable its picomolar potency and the ability of discrete variations in lipid A structure to markedly alter the pro-inflammatory activity of LPS. Two recognition systems—MD-2/TLR4 and “LPS-sensing” cytosolic caspases—together confer LPS responsiveness at the host cell surface, within endosomes, and at sites physically accessible to the cytosol. Understanding how the lipid A of LPS is delivered and recognized at these diverse sites is crucial to understanding how the magnitude and character of the inflammatory responses are regulated.
Collapse
Affiliation(s)
- Jerrold Weiss
- Inflammation Program and Departments of Internal Medicine and Microbiology, University of Iowa, Iowa City, Iowa, USA.,Veterans Affairs Medical Center, Iowa City, Iowa, USA
| | - Jason Barker
- Inflammation Program and Departments of Internal Medicine and Microbiology, University of Iowa, Iowa City, Iowa, USA.,Veterans Affairs Medical Center, Iowa City, Iowa, USA
| |
Collapse
|
10
|
Facchini FA, Coelho H, Sestito SE, Delgado S, Minotti A, Andreu D, Jiménez-Barbero J, Peri F. Co-administration of Antimicrobial Peptides Enhances Toll-like Receptor 4 Antagonist Activity of a Synthetic Glycolipid. ChemMedChem 2018; 13:280-287. [PMID: 29265636 PMCID: PMC5900894 DOI: 10.1002/cmdc.201700694] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/02/2017] [Indexed: 12/21/2022]
Abstract
This study examines the effect of co‐administration of antimicrobial peptides and the synthetic glycolipid FP7, which is active in inhibiting inflammatory cytokine production caused by TLR4 activation and signaling. The co‐administration of two lipopolysaccharide (LPS)‐neutralizing peptides (a cecropin A–melittin hybrid peptide and a human cathelicidin) enhances by an order of magnitude the potency of FP7 in blocking the TLR4 signal. Interestingly, this is not an additional effect of LPS neutralization by peptides, because it also occurs if cells are stimulated by the plant lectin phytohemagglutinin, a non‐LPS TLR4 agonist. Our data suggest a dual mechanism of action for the peptides, not exclusively based on LPS binding and neutralization, but also on a direct effect on the LPS‐binding proteins of the TLR4 receptor complex. NMR experiments in solution show that peptide addition changes the aggregation state of FP7, promoting the formation of larger micelles. These results suggest a relationship between the aggregation state of lipid A‐like ligands and the type and intensity of the TLR4 response.
Collapse
Affiliation(s)
- Fabio A Facchini
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milano, Italy
| | - Helena Coelho
- Molecular Recognition & Host-Pathogen Interactions Programme, CIC bioGUNE, Bizkaia Technology Park, Building 801A, 48170, Derio, Spain.,Department of Organic Chemistry II, Faculty of Science & Technology, University of the Basque Country, 48940, Leioa, Bizkaia, Spain.,UCIBIO, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516, Caparica, Portugal
| | - Stefania E Sestito
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milano, Italy
| | - Sandra Delgado
- Molecular Recognition & Host-Pathogen Interactions Programme, CIC bioGUNE, Bizkaia Technology Park, Building 801A, 48170, Derio, Spain
| | - Alberto Minotti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milano, Italy
| | - David Andreu
- Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona Biomedical Research Park, Dr. Aiguader 88, 08003, Barcelona, Spain
| | - Jesús Jiménez-Barbero
- Molecular Recognition & Host-Pathogen Interactions Programme, CIC bioGUNE, Bizkaia Technology Park, Building 801A, 48170, Derio, Spain.,Department of Organic Chemistry II, Faculty of Science & Technology, University of the Basque Country, 48940, Leioa, Bizkaia, Spain.,Ikerbasque, Basque Foundation for Science, Maria Diaz de Haro 13, 48009, Bilbao, Spain
| | - Francesco Peri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milano, Italy
| |
Collapse
|
11
|
Wacker MA, Teghanemt A, Weiss JP, Barker JH. High-affinity caspase-4 binding to LPS presented as high molecular mass aggregates or in outer membrane vesicles. Innate Immun 2017; 23:336-344. [PMID: 28409545 DOI: 10.1177/1753425917695446] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Caspases of the non-canonical inflammasome (caspases -4, -5, and -11) directly bind endotoxin (LOS/LPS) and can be activated in the absence of any co-factors. Models of LPS-induced caspase activation have postulated that 1:1 binding of endotoxin monomers to caspase trigger caspase oligomerization and activation, analogous to that established for endotoxin-induced activation of MD-2/TLR4. However, using metabolically radiolabeled LOS and LPS, we now show high affinity and selective binding of caspase-4 to high molecular mass aggregates of purified endotoxin and to endotoxin-rich outer membrane vesicles without formation of 1:1 endotoxin:caspase complexes. Thus, our findings demonstrate markedly different endotoxin recognition properties of caspase-4 from that of MD-2/TLR4 and strongly suggest that activation of caspase-4 (and presumably caspase-5 and caspase-11) are mediated by interactions with activating endotoxin-rich membrane interfaces rather than by endotoxin monomers.
Collapse
Affiliation(s)
- Mark A Wacker
- 1 Department of Biology, Central Michigan University, Mt. Pleasant, MI, USA
| | - Athmane Teghanemt
- 2 Inflammation Program, University of Iowa, and Iowa City VA Health Care System, Iowa City, IA, USA.,3 Department of Internal Medicine, University of Iowa, and Iowa City VA Health Care System, Iowa City, IA, USA
| | - Jerrold P Weiss
- 2 Inflammation Program, University of Iowa, and Iowa City VA Health Care System, Iowa City, IA, USA.,3 Department of Internal Medicine, University of Iowa, and Iowa City VA Health Care System, Iowa City, IA, USA.,4 Department of Microbiology, University of Iowa, and Iowa City VA Health Care System, Iowa City, IA, USA
| | - Jason H Barker
- 2 Inflammation Program, University of Iowa, and Iowa City VA Health Care System, Iowa City, IA, USA.,3 Department of Internal Medicine, University of Iowa, and Iowa City VA Health Care System, Iowa City, IA, USA.,4 Department of Microbiology, University of Iowa, and Iowa City VA Health Care System, Iowa City, IA, USA
| |
Collapse
|
12
|
Mattis DM, Chervin AS, Ranoa DR, Kelley SL, Tapping RI, Kranz DM. Studies of the TLR4-associated protein MD-2 using yeast-display and mutational analyses. Mol Immunol 2015; 68:203-12. [PMID: 26320630 DOI: 10.1016/j.molimm.2015.08.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 08/06/2015] [Accepted: 08/10/2015] [Indexed: 11/19/2022]
Abstract
Bacterial lipopolysaccharide (LPS) activates the innate immune system by forming a complex with myeloid differentiation factor 2 (MD-2) and Toll-like receptor 4 (TLR4), which is present on antigen presenting cells. MD-2 plays an essential role in this activation of the innate immune system as a member of the ternary complex, TLR4:MD-2:LPS. With the goal of further understanding the molecular details of the interaction of MD-2 with LPS and TLR4, and possibly toward engineering dominant negative regulators of the MD-2 protein, here we subjected MD-2 to a mutational analysis using yeast display. The approach included generation of site-directed alanine mutants, and ligand-driven selections of MD-2 mutant libraries. Our findings showed that: (1) proline mutations in the F119-K132 loop that binds LPS were strongly selected for enhanced yeast surface stability, (2) there was a preference for positive-charged side chains (R/K) at residue 120 for LPS binding, and negative-charged side chains (D/E) for TLR4 binding, (3) aromatic residues were strongly preferred at F119 and F121 for LPS binding, and (4) an MD-2 mutant (T84N/D101A/S118A/S120D/K122P) exhibited increased binding to TLR4 but decreased binding to LPS. These studies revealed the impact of specific residues and regions of MD-2 on the binding of LPS and TLR4, and they provide a framework for further directed evolution of the MD-2 protein.
Collapse
Affiliation(s)
- Daiva M Mattis
- Department of Biochemistry, University of Illinois, Urbana, IL 61801, USA
| | - Adam S Chervin
- Department of Biochemistry, University of Illinois, Urbana, IL 61801, USA
| | - Diana R Ranoa
- Department of Microbiology, University of Illinois, Urbana, IL 61801, USA
| | - Stacy L Kelley
- Department of Microbiology, University of Illinois, Urbana, IL 61801, USA
| | - Richard I Tapping
- Department of Microbiology, University of Illinois, Urbana, IL 61801, USA
| | - David M Kranz
- Department of Biochemistry, University of Illinois, Urbana, IL 61801, USA.
| |
Collapse
|
13
|
Gioannini TL, Teghanemt A, Zhang D, Esparza G, Yu L, Weiss J. Purified monomeric ligand.MD-2 complexes reveal molecular and structural requirements for activation and antagonism of TLR4 by Gram-negative bacterial endotoxins. Immunol Res 2015; 59:3-11. [PMID: 24895101 DOI: 10.1007/s12026-014-8543-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A major focus of work in our laboratory concerns the molecular mechanisms and structural bases of Gram-negative bacterial endotoxin recognition by host (e.g., human) endotoxin-recognition proteins that mediate and/or regulate activation of Toll-like receptor (TLR) 4. Here, we review studies of wild-type and variant monomeric endotoxin.MD-2 complexes first produced and characterized in our laboratories. These purified complexes have provided unique experimental reagents, revealing both quantitative and qualitative determinants of TLR4 activation and antagonism. This review is dedicated to the memory of Dr. Theresa L. Gioannini (1949-2014) who played a central role in many of the studies and discoveries that are reviewed.
Collapse
Affiliation(s)
- Theresa L Gioannini
- The Inflammation Program, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 2501 Crosspark Rd, Coralville, IA, 52241, USA
| | | | | | | | | | | |
Collapse
|
14
|
Yang H, Wang H, Ju Z, Ragab AA, Lundbäck P, Long W, Valdes-Ferrer SI, He M, Pribis JP, Li J, Lu B, Gero D, Szabo C, Antoine DJ, Harris HE, Golenbock DT, Meng J, Roth J, Chavan SS, Andersson U, Billiar TR, Tracey KJ, Al-Abed Y. MD-2 is required for disulfide HMGB1-dependent TLR4 signaling. ACTA ACUST UNITED AC 2015; 212:5-14. [PMID: 25559892 PMCID: PMC4291531 DOI: 10.1084/jem.20141318] [Citation(s) in RCA: 267] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Yang et al. show that a disulfide isoform of HMGB1, with a role in TLR4 signaling, physically interacts with and binds MD-2. MD-2 deficiency in macrophage cell lines or in primary mouse macrophages stimulated with HMGB1 implicates MD-2 in TLR4 signaling. They also identify an HGMB1 peptide inhibitor, P5779, which when administered in vivo can protect mice from acetaminophen-induced hepatoxicity, ischemia/reperfusion injury, and sepsis. Innate immune receptors for pathogen- and damage-associated molecular patterns (PAMPs and DAMPs) orchestrate inflammatory responses to infection and injury. Secreted by activated immune cells or passively released by damaged cells, HMGB1 is subjected to redox modification that distinctly influences its extracellular functions. Previously, it was unknown how the TLR4 signalosome distinguished between HMGB1 isoforms. Here we demonstrate that the extracellular TLR4 adaptor, myeloid differentiation factor 2 (MD-2), binds specifically to the cytokine-inducing disulfide isoform of HMGB1, to the exclusion of other isoforms. Using MD-2–deficient mice, as well as MD-2 silencing in macrophages, we show a requirement for HMGB1-dependent TLR4 signaling. By screening HMGB1 peptide libraries, we identified a tetramer (FSSE, designated P5779) as a specific MD-2 antagonist preventing MD-2–HMGB1 interaction and TLR4 signaling. P5779 does not interfere with lipopolysaccharide-induced cytokine/chemokine production, thus preserving PAMP-mediated TLR4–MD-2 responses. Furthermore, P5779 can protect mice against hepatic ischemia/reperfusion injury, chemical toxicity, and sepsis. These findings reveal a novel mechanism by which innate systems selectively recognize specific HMGB1 isoforms. The results may direct toward strategies aimed at attenuating DAMP-mediated inflammation while preserving antimicrobial immune responsiveness.
Collapse
Affiliation(s)
- Huan Yang
- Department of Biomedical Science and Department of Medicinal Chemistry, The Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Haichao Wang
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, NY 11030
| | - Zhongliang Ju
- Department of Biomedical Science and Department of Medicinal Chemistry, The Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Ahmed A Ragab
- Department of Biomedical Science and Department of Medicinal Chemistry, The Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Peter Lundbäck
- Department of Medicine and Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, SE-171 77 Stockholm, Sweden Department of Medicine and Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, SE-171 77 Stockholm, Sweden
| | - Wei Long
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, NY 11030
| | - Sergio I Valdes-Ferrer
- Department of Biomedical Science and Department of Medicinal Chemistry, The Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Mingzhu He
- Department of Biomedical Science and Department of Medicinal Chemistry, The Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - John P Pribis
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Jianhua Li
- Department of Biomedical Science and Department of Medicinal Chemistry, The Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Ben Lu
- Department of Biomedical Science and Department of Medicinal Chemistry, The Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Domokos Gero
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX 77555
| | - Csaba Szabo
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX 77555
| | - Daniel J Antoine
- Medical Research Council Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool L69 3BX, England, UK
| | - Helena E Harris
- Department of Medicine and Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, SE-171 77 Stockholm, Sweden Department of Medicine and Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, SE-171 77 Stockholm, Sweden
| | - Doug T Golenbock
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Jianmin Meng
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Jesse Roth
- Department of Biomedical Science and Department of Medicinal Chemistry, The Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Sangeeta S Chavan
- Department of Biomedical Science and Department of Medicinal Chemistry, The Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Ulf Andersson
- Department of Medicine and Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, SE-171 77 Stockholm, Sweden Department of Medicine and Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, SE-171 77 Stockholm, Sweden
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Kevin J Tracey
- Department of Biomedical Science and Department of Medicinal Chemistry, The Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Yousef Al-Abed
- Department of Biomedical Science and Department of Medicinal Chemistry, The Feinstein Institute for Medical Research, Manhasset, NY 11030
| |
Collapse
|
15
|
Kagan JC, Barton GM. Emerging principles governing signal transduction by pattern-recognition receptors. Cold Spring Harb Perspect Biol 2014; 7:a016253. [PMID: 25395297 PMCID: PMC4355268 DOI: 10.1101/cshperspect.a016253] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The problem of recognizing and disposing of non-self-organisms, whether for nutrients or defense, predates the evolution of multicellularity. Accordingly, the function of the innate immune system is often intimately associated with fundamental aspects of cell biology. Here, we review our current understanding of the links between cell biology and pattern-recognition receptors of the innate immune system. We highlight the importance of receptor localization for the detection of microbes and for the initiation of antimicrobial signaling pathways. We discuss examples that illustrate how pattern-recognition receptors influence, and are influenced by, the general membrane trafficking machinery of mammalian cells. In the future, cell biological analysis likely will rival pure genetic analysis as a tool to uncover fundamental principles that govern host-microbe interactions.
Collapse
Affiliation(s)
- Jonathan C Kagan
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, Massachusetts 02115
| | - Gregory M Barton
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, California 94720-3200
| |
Collapse
|
16
|
Guinan EC, Palmer CD, Mancuso CJ, Brennan L, Stoler-Barak L, Kalish LA, Suter EE, Gallington LC, Huhtelin DP, Mansilla M, Schumann RR, Murray JC, Weiss J, Levy O. Identification of single nucleotide polymorphisms in hematopoietic cell transplant patients affecting early recognition of, and response to, endotoxin. Innate Immun 2013; 20:697-711. [PMID: 24107515 DOI: 10.1177/1753425913505122] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Hematopoietic cell transplant (HCT) is a life-saving therapy for many malignant and non-malignant bone marrow diseases. Associated morbidities are often due to transplant-related toxicities and infections, exacerbated by regimen-induced immune suppression and systemic incursion of bacterial products. Patients undergoing myeloablative conditioning for HCT become endotoxemic and display blood/plasma changes consistent with lipopolysaccharide (LPS)-induced systemic innate immune activation. Herein, we addressed whether patients scheduled for HCT display differences in recognition/response to LPS ex vivo traceable to specific single nucleotide polymorphisms (SNPs). Two SNPs of LPS binding protein (LBP) were associated with changes in plasma LBP levels, with one LBP SNP also associating with differences in efficiency of extraction and transfer of endotoxin to myeloid differentiation factor-2 (MD-2), a step needed for activation of TLR4. None of the examined SNPs of CD14, bactericidal/permeability-increasing protein (BPI), TLR4 or MD-2 were associated with corresponding protein plasma levels or endotoxin delivery to MD-2, but CD14 and BPI SNPs significantly associated with differences in LPS-induced TNF-α release ex vivo and infection frequency, respectively. These findings suggest that specific LBP, CD14 and BPI SNPs might be contributory assessments in studies where clinical outcome may be affected by host response to endotoxin and bacterial infection.
Collapse
Affiliation(s)
- Eva C Guinan
- Boston Children's Hospital, Boston, MA, USA Harvard Medical School, Boston, MA, USA Dana-Farber Cancer Institute, Boston, MA, USA
| | - Christine D Palmer
- Boston Children's Hospital, Boston, MA, USA Harvard Medical School, Boston, MA, USA Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Boston, MA, USA
| | | | | | | | - Leslie A Kalish
- Boston Children's Hospital, Boston, MA, USA Harvard Medical School, Boston, MA, USA
| | | | | | - David P Huhtelin
- University of Iowa and Veterans' Administration Medical Center, Coralville, Iowa City, IA, USA
| | - Maria Mansilla
- Department of Pediatrics, University of Iowa, IA, Iowa City, USA
| | - Ralf R Schumann
- Institute for Microbiology, Charité-University Medical Center, Berlin, Germany
| | - Jeffrey C Murray
- Department of Pediatrics, University of Iowa, IA, Iowa City, USA
| | - Jerrold Weiss
- University of Iowa and Veterans' Administration Medical Center, Coralville, Iowa City, IA, USA
| | - Ofer Levy
- Boston Children's Hospital, Boston, MA, USA Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Tsukamoto H, Ihara H, Ito R, Ukai I, Suzuki N, Kimoto M, Tomioka Y, Ikeda Y. MD-2-dependent human Toll-like receptor 4 monoclonal antibodies detect extracellular association of Toll-like receptor 4 with extrinsic soluble MD-2 on the cell surface. Biochem Biophys Res Commun 2013; 440:31-6. [PMID: 24021278 DOI: 10.1016/j.bbrc.2013.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 09/02/2013] [Indexed: 11/18/2022]
Abstract
MD-2 is essential for lipopolysaccharide (LPS) recognition of Toll-like receptor 4 (TLR4) but not for cell surface expression. The TLR4/MD-2 complex is formed intracellularly through co-expression. Extracellular complex formation remains a matter for debate because of the aggregative nature of secreted MD-2 in the absence of TLR4 co-expression. We demonstrated extracellular complex formation using three independent monoclonal antibodies (mAbs), all of which are specific for complexed TLR4 but unreactive with free TLR4 and MD-2. These mAbs bound to TLR4-expressing Ba/F3 cells only when co-cultured with MD-2-secreting Chinese hamster ovary cells or incubated with conditioned medium from these cells. All three mAbs bound the extracellularly formed complex indistinguishably from the intracellularly formed complex in titration studies. In addition, we demonstrated that two mAbs lost their affinity for TLR4/MD-2 on LPS stimulation, suggesting that these mAbs bound to conformation-sensitive epitopes. This was also found when the extracellularly formed complex was stimulated with LPS. Additionally, we showed that cell surface TLR4 and extrinsically secreted MD-2 are capable of forming the functional complex extracellularly, indicating an additional or alternative pathway for the complex formation.
Collapse
Affiliation(s)
- Hiroki Tsukamoto
- Laboratory of Oncology, Pharmacy Practice and Sciences, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Sendai 980-8578, Japan; Division of Molecular Cell Biology, Department of Biomolecular Sciences, Saga University Faculty of Medicine, 5-1-1 Nabeshima, Saga 849-8501, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Li W, Mochizuki S, Sakurai K. Structural Transition of Lipopolysaccharide and Reduction in the Biological Activity by Amphiphilic Lipid with Cationic Amino Acid. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2013. [DOI: 10.1246/bcsj.20120353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Wenjing Li
- Department of Chemistry and Biochemistry, The University of Kitakyushu
| | | | - Kazuo Sakurai
- Department of Chemistry and Biochemistry, The University of Kitakyushu
- CREST, Japan Science and Technology Agency
| |
Collapse
|
19
|
Teghanemt A, Weiss JP, Gioannini TL. Radioiodination of an endotoxin·MD-2 complex generates a novel sensitive, high-affinity ligand for TLR4. Innate Immun 2013; 19:545-60. [PMID: 23439691 DOI: 10.1177/1753425913475688] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
A purified complex of metabolically labeled [(3)H]lipooligosaccharide (LOS) and recombinant human myeloid differentiation factor 2 (MD-2), [(3)H]LOS·MD-2, has been used to demonstrate pM affinity binding interactions with soluble TLR4 ectodomain (TLR4ecd). For measurement of the binding parameters of membrane-bound TLR4, we took advantage of the stability of endotoxin·MD-2 and tyrosine(s) present on the surface of MD-2 to radioiodinate LOS·MD-2. Radioiodinated LOS·MD-2 generated a reagent with an estimated 1:1 molar ratio of [(125)I] to sMD-2 with 20-fold higher specific radioactivity and TLR4-activating properties comparable to metabolically-labeled LOS·MD-2. LOS·MD-2[(125)I] and [(3)H]LOS·MD-2 have similar affinities for soluble (FLAG) TLR4ecd and for membrane-bound TLR4 in HEK293T/TLR4 cells. In a similar dose-dependent manner, sMD-2 and LOS·MD-2 inhibit LOS·MD-2[(125)I] binding to TLR4 indicating the pM affinity binding of LOS·MD-2[(125)I] is agonist-independent. LOS·MD-2[(125)I] allowed measurement of low levels of cell-surface human or murine TLR4 expressed by stable cell lines (2000-3000 sites/cell) and quantitatively measures low levels of 'MD-2-free' TLR4 (est. 250 molecules/cell) in cells co-expressing TLR4 and MD-2. Occupation of 50-100 TLR4/cell by LOS·MD-2 is sufficient to trigger measurable TLR4-dependent cell activation. LOS·MD-2[(125)I] provides a powerful reagent to measure quantitatively functional human and murine cell-surface TLR4, including in cells where surface TLR4 is potentially functionally significant but not detectable by other methods.
Collapse
Affiliation(s)
- Athmane Teghanemt
- 1Inflammation Program, Department of Internal Medicine, Roy A. and Lucille J. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | | | | |
Collapse
|
20
|
Ranoa DRE, Kelley SL, Tapping RI. Human lipopolysaccharide-binding protein (LBP) and CD14 independently deliver triacylated lipoproteins to Toll-like receptor 1 (TLR1) and TLR2 and enhance formation of the ternary signaling complex. J Biol Chem 2013; 288:9729-9741. [PMID: 23430250 DOI: 10.1074/jbc.m113.453266] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Bacterial lipoproteins are the most potent microbial agonists for the Toll-like receptor 2 (TLR2) subfamily, and this pattern recognition event induces cellular activation, leading to host immune responses. Triacylated bacterial lipoproteins coordinately bind TLR1 and TLR2, resulting in a stable ternary complex that drives intracellular signaling. The sensitivity of TLR-expressing cells to lipoproteins is greatly enhanced by two lipid-binding serum proteins known as lipopolysaccharide-binding protein (LBP) and soluble CD14 (sCD14); however, the physical mechanism that underlies this increased sensitivity is not known. To address this, we measured the ability of LBP and sCD14 to drive ternary complex formation between soluble extracellular domains of TLR1 and TLR2 and a synthetic triacylated lipopeptide agonist. Importantly, addition of substoichiometric amounts of either LBP or sCD14 significantly enhanced formation of a TLR1·TLR2 lipopeptide ternary complex as measured by size exclusion chromatography. However, neither LBP nor sCD14 was physically associated with the final ternary complex. Similar results were obtained using outer surface protein A (OspA), a naturally occurring triacylated lipoprotein agonist from Borrelia burgdorferi. Activation studies revealed that either LBP or sCD14 sensitized TLR-expressing cells to nanogram levels of either the synthetic lipopeptide or OspA lipoprotein agonist. Together, our results show that either LBP or sCD14 can drive ternary complex formation and TLR activation by acting as mobile carriers of triacylated lipopeptides or lipoproteins.
Collapse
Affiliation(s)
- Diana Rose E Ranoa
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Stacy L Kelley
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Richard I Tapping
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; Department of College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801.
| |
Collapse
|
21
|
Kelley SL, Lukk T, Nair SK, Tapping RI. The crystal structure of human soluble CD14 reveals a bent solenoid with a hydrophobic amino-terminal pocket. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:1304-11. [PMID: 23264655 PMCID: PMC3552104 DOI: 10.4049/jimmunol.1202446] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Human monocyte differentiation Ag CD14 is a pattern recognition receptor that enhances innate immune responses to infection by sensitizing host cells to bacterial LPS (endotoxin), lipoproteins, lipoteichoic acid, and other acylated microbial products. CD14 physically delivers these lipidated microbial products to various TLR signaling complexes that subsequently induce intracellular proinflammatory signaling cascades upon ligand binding. The ensuing cellular responses are usually protective to the host but can also result in host fatality through sepsis. In this work, we have determined the x-ray crystal structure of human CD14. The structure reveals a bent solenoid typical of leucine-rich repeat proteins with an amino-terminal pocket that presumably binds acylated ligands including LPS. Comparison of human and mouse CD14 structures shows great similarity in overall protein fold. However, compared with mouse CD14, human CD14 contains an expanded pocket and alternative rim residues that are likely to be important for LPS binding and cell activation. The x-ray crystal structure of human CD14 presented in this article may foster additional ligand-bound structural studies, virtual docking studies, and drug design efforts to mitigate LPS-induced sepsis and other inflammatory diseases.
Collapse
Affiliation(s)
- Stacy L. Kelley
- Department of Biochemistry, University of Illinois at Urbana-Champaign
| | - Tiit Lukk
- Department of Biochemistry, University of Illinois at Urbana-Champaign
| | - Satish K. Nair
- Department of Biochemistry, University of Illinois at Urbana-Champaign
| | - Richard I. Tapping
- Department of Microbiology, University of Illinois at Urbana-Champaign
- College of Medicine, University of Illinois at Urbana-Champaign
| |
Collapse
|
22
|
Feng C, Stamatos NM, Dragan AI, Medvedev A, Whitford M, Zhang L, Song C, Rallabhandi P, Cole L, Nhu QM, Vogel SN, Geddes CD, Cross AS. Sialyl residues modulate LPS-mediated signaling through the Toll-like receptor 4 complex. PLoS One 2012; 7:e32359. [PMID: 22496731 PMCID: PMC3322133 DOI: 10.1371/journal.pone.0032359] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 01/26/2012] [Indexed: 11/19/2022] Open
Abstract
We previously reported that neuraminidase (NA) pretreatment of human PBMCs markedly increased their cytokine response to lipopolysaccharide (LPS). To study the mechanisms by which this occurs, we transfected HEK293T cells with plasmids encoding TLR4, CD14, and MD2 (three components of the LPS receptor complex), as well as a NFκB luciferase reporting system. Both TLR4 and MD2 encoded by the plasmids are α-2,6 sialylated. HEK293T cells transfected with TLR4/MD2/CD14 responded robustly to the addition of LPS; however, omission of the MD2 plasmid abrogated this response. Addition of culture supernatants from MD2 (sMD2)-transfected HEK293T cells, but not recombinant, non-glycosylated MD2 reconstituted this response. NA treatment of sMD2 enhanced the LPS response as did NA treatment of the TLR4/CD14-transfected cell supplemented with untreated sMD2, but optimal LPS-initiated responses were observed with NA-treated TLR4/CD14-transfected cells supplemented with NA-treated sMD2. We hypothesized that removal of negatively charged sialyl residues from glycans on the TLR4 complex would hasten the dimerization of TLR4 monomers required for signaling. Co-transfection of HEK293T cells with separate plasmids encoding either YFP- or FLAG-tagged TLR4, followed by treatment with NA and stimulation with LPS, led to an earlier and more robust time-dependent dimerization of TLR4 monomers on co-immunoprecipitation, compared to untreated cells. These findings were confirmed by fluorescence resonance energy transfer (FRET) analysis. Overexpression of human Neu1 increased LPS-initiated TLR4-mediated NFκB activation and a NA inhibitor suppressed its activation. We conclude that (1) sialyl residues on TLR4 modulate LPS responsiveness, perhaps by facilitating clustering of the homodimers, and that (2) sialic acid, and perhaps other glycosyl species, regulate MD2 activity required for LPS-mediated signaling. We speculate that endogenous sialidase activity mobilized during cell activation may play a role in this regulation.
Collapse
Affiliation(s)
- Chiguang Feng
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Nicholas M. Stamatos
- Institute of Human Virology and Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Anatoliy I. Dragan
- Institute of Fluorescence, University of Maryland, Baltimore County, Baltimore, Maryland, United States of America
| | - Andrei Medvedev
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Melissa Whitford
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Lei Zhang
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Chang Song
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Prasad Rallabhandi
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Leah Cole
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Quan M. Nhu
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Stefanie N. Vogel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Chris D. Geddes
- Institute of Fluorescence, University of Maryland, Baltimore County, Baltimore, Maryland, United States of America
| | - Alan S. Cross
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
23
|
Yu L, Phillips RL, Zhang D, Teghanemt A, Weiss JP, Gioannini TL. NMR studies of hexaacylated endotoxin bound to wild-type and F126A mutant MD-2 and MD-2·TLR4 ectodomain complexes. J Biol Chem 2012; 287:16346-55. [PMID: 22433852 DOI: 10.1074/jbc.m112.343467] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Host response to invasion by many gram-negative bacteria depends upon activation of Toll-like receptor 4 (TLR4) by endotoxin presented as a monomer bound to myeloid differentiation factor 2 (MD-2). Metabolic labeling of hexaacylated endotoxin (LOS) from Neisseria meningitidis with [(13)C]acetate allowed the use of NMR to examine structural properties of the fatty acyl chains of LOS present in TLR4-agonistic and -antagonistic binary and ternary complexes with, respectively, wild-type or mutant (F126A) MD-2 ± TLR4 ectodomain. Chemical shift perturbation indicates that Phe(126) affects the environment and/or position of each of the bound fatty acyl chains both in the binary LOS·MD-2 complex and in the ternary LOS·MD-2·TLR4 ectodomain complex. In both wild-type and mutant LOS·MD-2 complexes, one of the six fatty acyl chains of LOS is more susceptible to paramagnetic attenuation, suggesting protrusion of that fatty acyl chain from the hydrophobic pocket of MD-2, independent of association with TLR4. These findings indicate that re-orientation of the aromatic side chain of Phe(126) is induced by binding of hexaacylated E, preceding interaction with TLR4. This re-arrangement of Phe(126) may act as a "hydrophobic switch," driving agonist-dependent contacts needed for TLR4 dimerization and activation.
Collapse
Affiliation(s)
- Liping Yu
- NMR Core Facility, Roy A. and Lucille J. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52241, USA
| | | | | | | | | | | |
Collapse
|
24
|
Deep sequencing-based expression transcriptional profiling changes during Brucella infection. Microb Pathog 2012; 52:267-77. [PMID: 22342430 DOI: 10.1016/j.micpath.2012.02.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Revised: 01/27/2012] [Accepted: 02/02/2012] [Indexed: 01/18/2023]
Abstract
Brucellosis is a worldwide zoonotic infectious disease that has significant economic effects on animal production and human health. The host macrophage -Brucella interaction is critical to the establishment of infections. Thus, the kinetic transcriptional profile of gene expression in macrophages infected with the Brucella melitensis strain 16M was investigated in the current study using a technology based on deep sequencing. The total RNA was extracted from macrophages 0, 4, and 24 h post-infection. Data analysis showed that in the gene ontology term, the expression of genes in the endoplasmic reticulum, lysosomes, as well as those involved in programmed cell death and apoptosis significantly changed during the first 24 h post-infection. Pathway enrichment analysis indicated that the genes in the apoptosis pathway, NOD-like receptor signaling pathway, Fc gamma R-mediated phagocytosis, lysosome pathway, p53 signaling pathway, and protein processing in the endoplasmic reticulum significantly changed during the first 24 h post-infection. The B-cell receptor and toll-like receptor signaling pathways were also significantly changed 24 h post-infection compared with those 4 h post-infection. The results of the current study can contribute to an improved understanding of the manner by which host cell responses may be manipulated to prevent Brucella infection.
Collapse
|
25
|
Yang D, Postnikov YV, Li Y, Tewary P, de la Rosa G, Wei F, Klinman D, Gioannini T, Weiss JP, Furusawa T, Bustin M, Oppenheim JJ. High-mobility group nucleosome-binding protein 1 acts as an alarmin and is critical for lipopolysaccharide-induced immune responses. J Exp Med 2012; 209:157-71. [PMID: 22184635 PMCID: PMC3260868 DOI: 10.1084/jem.20101354] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 11/15/2011] [Indexed: 01/08/2023] Open
Abstract
Alarmins are endogenous mediators capable of promoting the recruitment and activation of antigen-presenting cells (APCs), including dendritic cells (DCs), that can potentially alert host defense against danger signals. However, the relevance of alarmins to the induction of adaptive immune responses remains to be demonstrated. In this study, we report the identification of HMGN1 (high-mobility group nucleosome-binding protein 1) as a novel alarmin and demonstrate that it contributes to the induction of antigen-specific immune responses. HMGN1 induced DC maturation via TLR4 (Toll-like receptor 4), recruitment of APCs at sites of injection, and activation of NF-κB and multiple mitogen-activated protein kinases in DCs. HMGN1 promoted antigen-specific immune response upon co-administration with antigens, and Hmgn1(-/-) mice developed greatly reduced antigen-specific antibody and T cell responses when immunized with antigens in the presence of lipopolysaccharide (LPS). The impaired ability of Hmgn1(-/-) mice to mount antigen-specific immune responses was accompanied by both deficient DC recruitment at sites of immunization and reduced production of inflammatory cytokines. Bone marrow chimera experiments revealed that HMGN1 derived from nonleukocytes was critical for the induction of antigen-specific antibody and T cell responses. Thus, extracellular HMGN1 acts as a novel alarmin critical for LPS-induced development of innate and adaptive immune responses.
Collapse
Affiliation(s)
- De Yang
- Basic Research Program, Scientific Application and International Corporation–Frederick, Inc.; and Laboratory of Molecular Immunoregulation and Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research; National Cancer Institute at Frederick, Frederick, MD 21702
| | - Yuri V. Postnikov
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Yana Li
- Basic Research Program, Scientific Application and International Corporation–Frederick, Inc.; and Laboratory of Molecular Immunoregulation and Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research; National Cancer Institute at Frederick, Frederick, MD 21702
| | - Poonam Tewary
- Basic Research Program, Scientific Application and International Corporation–Frederick, Inc.; and Laboratory of Molecular Immunoregulation and Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research; National Cancer Institute at Frederick, Frederick, MD 21702
| | - Gonzalo de la Rosa
- Basic Research Program, Scientific Application and International Corporation–Frederick, Inc.; and Laboratory of Molecular Immunoregulation and Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research; National Cancer Institute at Frederick, Frederick, MD 21702
| | - Feng Wei
- Basic Research Program, Scientific Application and International Corporation–Frederick, Inc.; and Laboratory of Molecular Immunoregulation and Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research; National Cancer Institute at Frederick, Frederick, MD 21702
| | - Dennis Klinman
- Basic Research Program, Scientific Application and International Corporation–Frederick, Inc.; and Laboratory of Molecular Immunoregulation and Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research; National Cancer Institute at Frederick, Frederick, MD 21702
| | - Theresa Gioannini
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Jerrold P. Weiss
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Takashi Furusawa
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Michael Bustin
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Joost J. Oppenheim
- Basic Research Program, Scientific Application and International Corporation–Frederick, Inc.; and Laboratory of Molecular Immunoregulation and Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research; National Cancer Institute at Frederick, Frederick, MD 21702
| |
Collapse
|
26
|
Gray P, Michelsen KS, Sirois CM, Lowe E, Shimada K, Crother TR, Chen S, Brikos C, Bulut Y, Latz E, Underhill D, Arditi M. Identification of a novel human MD-2 splice variant that negatively regulates Lipopolysaccharide-induced TLR4 signaling. THE JOURNAL OF IMMUNOLOGY 2010; 184:6359-66. [PMID: 20435923 DOI: 10.4049/jimmunol.0903543] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Myeloid differentiation factor 2 (MD-2) is a secreted gp that assembles with TLR4 to form a functional signaling receptor for bacterial LPS. In this study, we have identified a novel alternatively spliced isoform of human MD-2, termed MD-2 short (MD-2s), which lacks the region encoded by exon 2 of the MD-2 gene. Similar to MD-2, MD-2s is glycosylated and secreted. MD-2s also interacted with LPS and TLR4, but failed to mediate LPS-induced NF-kappaB activation and IL-8 production. We show that MD-2s is upregulated upon IFN-gamma, IL-6, and TLR4 stimulation and negatively regulates LPS-mediated TLR4 signaling. Furthermore, MD-2s competitively inhibited binding of MD-2 to TLR4. Our study pinpoints a mechanism that may be used to regulate TLR4 activation at the onset of signaling and identifies MD-2s as a potential therapeutic candidate to treat human diseases characterized by an overly exuberant or chronic immune response to LPS.
Collapse
Affiliation(s)
- Pearl Gray
- Division of Pediatric Infectious Diseases and Immunology, Burns and Allen Research Institute, Cedars-Sinai Medical Center, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90048, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Sera from patients with Crohn’s disease break bacterial lipopolysaccharide tolerance of human intestinal epithelial cells via MD-2 activity. Innate Immun 2010; 16:381-90. [DOI: 10.1177/1753425909357076] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Myeloid differentiation (MD)-2 is linked to the cell surface as a Toll-like receptor (TLR) 4-bound protein though may also function as a soluble receptor to enable the lipopolysaccharide (LPS)-driven response. We recently demonstrated the importance of MD-2 either as a cell-associated or as a soluble receptor in the control of intestinal epithelial cell response toward LPS. High levels of circulating MD-2 were recently proposed as a risk factor for infectious/ inflammatory diseases as septic shock. We hypothesized that MD-2 might be present in sera from patients with inflammatory bowel disease and have pathogenic consequences. We analysed MD-2 activity in sera from patients with inflammatory bowel disease or from healthy subjects. We measured MD-2 activity as the capacity to mediate LPS-driven stimulation of intestinal epithelial cells (HT29). We found that sera from patients with inflammatory bowel disease, particularly Crohn’s disease, endowed HT29 cells with a markedly higher LPS-dependent stimulating capacity as compared to sera from healthy subjects. The effect of sera was specific for LPS activation and was reduced in the presence of anti-MD-2, and anti-TLR4 antibodies. We conclude that sera from patients with inflammatory bowel disease might contain increased MD-2. This might result in higher local availability of the protein leading to a loss of tolerance toward gut microbiota.
Collapse
|
28
|
Prohinar P, Rallabhandi P, Weiss JP, Gioannini TL. Expression of functional D299G.T399I polymorphic variant of TLR4 depends more on coexpression of MD-2 than does wild-type TLR4. THE JOURNAL OF IMMUNOLOGY 2010; 184:4362-7. [PMID: 20212095 DOI: 10.4049/jimmunol.0903142] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Two missense variants (D299G and T399I) of TLR4 are cosegregated in individuals of European descent and, in a number of test systems, result in reduced responsiveness to endotoxin. How these changes within the ectodomain (ecd) of TLR4 affect TLR4 function is unclear. For both wild-type and D299G.T399I TLR4, we used endotoxinCD14 and endotoxinMD-2 complexes of high specific radioactivity to measure: 1) interaction of recombinant MD-2TLR4 with endotoxinCD14 and TLR4 with endotoxinMD-2; 2) expression of functional MD-2TLR4 and TLR4; and 3) MD-2TLR4 and TLR4-dependent cellular endotoxin responsiveness. Both wild-type and D299G.T399I TLR4(ecd) demonstrated high affinity (K(d) approximately 200 pM) interaction of endotoxinCD14 with MD-2TLR4(ecd) and endotoxinMD-2 with TLR4(ecd). However, levels of functional TLR4 were reduced up to 2-fold when D299G.T399I TLR4 was coexpressed with MD-2 and >10-fold when expressed without MD-2, paralleling differences in cellular endotoxin responsiveness. The dramatic effect of the D299G.T399I haplotype on expression of functional TLR4 without MD-2 suggests that cells expressing TLR4 without MD-2 are most affected by these polymorphisms.
Collapse
Affiliation(s)
- Polonca Prohinar
- Department of Internal Medicine, Roy A and Lucille J Carver College of Medicine, University of Iowa, Iowa City, IA 52240, USA
| | | | | | | |
Collapse
|
29
|
Piazza M, Yu L, Teghanemt A, Gioannini T, Weiss J, Peri F. Evidence of a specific interaction between new synthetic antisepsis agents and CD14. Biochemistry 2010; 48:12337-44. [PMID: 19928913 DOI: 10.1021/bi901601b] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Synthetic molecules derived from natural sugars with a positively charged amino group or ammonium salt and two lipophilic chains have been shown to inhibit TLR4 activation in vitro and in vivo. To characterize the mechanism of action of this class of molecules, we investigated possible interactions with the extracellular components that bind and shuttle endotoxin [lipopolysaccharide (LPS)] to TLR4, namely, LBP, CD14, and MD-2. Molecules that inhibited TLR4 activation inhibited LBP.CD14-dependent transfer of endotoxin monomers derived from aggregates of tritiated lipooligosaccharide ([(3)H]LOS) from Neisseria meninigitidis to MD-2.TLR4, resulting in a reduced level of formation of a ([(3)H]LOS.MD-2.TLR4(ECD))(2) (M(r) approximately 190000) complex. This effect was due to inhibition of the transfer of [(3)H]LOS from aggregates in solution to sCD14 with little or no effect on [(3)H]LOS shuttling from [(3)H]LOS.sCD14 to MD-2. These compounds also inhibited transfer of the [(3)H]LOS monomer from full-length CD14 to a truncated, polyhistidine-tagged CD14. Dose-dependent inhibition of the transfer of [(3)H]LOS between the two forms of CD14 was observed with each of three different synthetic compounds that inhibited TLR4 activation but not by another structurally related analogue that lacked TLR4 antagonistic activity. Saturation transfer difference (STD) NMR data showed direct binding to CD14 by the synthetic TLR4 antagonist mediated principally through the lipid chains of the synthetic compound. Taken together, our findings strongly suggest that these compounds inhibit TLR4 activation by endotoxin by competitively occupying CD14 and thereby reducing the level of delivery of activating endotoxin to MD-2.TLR4.
Collapse
Affiliation(s)
- Matteo Piazza
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
The stimulation of TLR4 by LPS activates two distinct signaling pathways leading to the expression of diverse inflammatory genes. Intensive studies over the past decade have revealed the components involved in these signaling pathways, however, more recently the focus has shifted somewhat towards the components that regulate these pathways. Several regulatory mechanisms, including localisation of components, splice variants and inhibitory molecules will be discussed in this review.
Collapse
Affiliation(s)
- Anne F McGettrick
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland.
| | | |
Collapse
|
31
|
Vasl J, Oblak A, Gioannini TL, Weiss JP, Jerala R. Novel roles of lysines 122, 125, and 58 in functional differences between human and murine MD-2. THE JOURNAL OF IMMUNOLOGY 2009; 183:5138-45. [PMID: 19783674 DOI: 10.4049/jimmunol.0901544] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The MD-2/TLR4 complex provides a highly robust mechanism for recognition and response of mammalian innate immunity to Gram-negative bacterial endotoxins. Despite overall close structural and functional similarity, human (h) and murine (m) MD-2 show several species-related differences, including the ability of hMD-2, but not mMD-2, to bind endotoxin (E) in the absence of TLR4. Wild-type mMD-2 can support TLR4-dependent cell activation by E only when mMD-2 and mTLR4 are coexpressed in the same cell. However, replacement of Glu122, Leu125, and/or Asn58 of mMD-2 with the corresponding residues (lysines) of hMD-2 was sufficient to yield soluble extracellular MD-2 that reacted with monomeric E . sCD14 complex to form extracellular monomeric E . MD-2 that activated cells expressing TLR4 without MD-2. Moreover, in contrast to wild-type mMD-2, double and triple mMD-2 mutants also supported E-triggered signaling in combination with human TLR4. Conversely, a K125L mutant of hMD-2 reacted with E . CD14 and activated TLR4 only when coexpressed with TLR4, and not when secreted without TLR4. These findings reveal novel roles of lysines 122, 125, and 58 in human MD-2 that contribute to the functional differences between human and murine MD-2 and, potentially, to differences in the sensitivity of humans and mice to endotoxin.
Collapse
Affiliation(s)
- Jozica Vasl
- Department of Biotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | | | | | | | | |
Collapse
|
32
|
Vamadevan AS, Fukata M, Arnold ET, Thomas LS, Hsu D, Abreu MT. Regulation of Toll-like receptor 4-associated MD-2 in intestinal epithelial cells: a comprehensive analysis. Innate Immun 2009; 16:93-103. [PMID: 19710105 DOI: 10.1177/1753425909339231] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The intestinal epithelium maintains a state of controlled inflammation despite continuous contact with Gram-negative commensal bacteria and lipopolysaccharide (LPS) on its luminal surface. Recognition of LPS by the Toll-like receptor (TLR) 4/MD-2 complex results in pro-inflammatory gene expression and cytokine secretion in intestinal epithelial cells (IECs). We have shown that IECs express low levels of MD-2 and TLR4 and are poorly responsive to LPS. In this study, we did a comprehensive analysis to understand the immune-mediated and epigenetic mechanisms by which IECs down-regulate MD-2 expression. Expression of MD-2 and TLR4 mRNA was examined in human inflammatory bowel disease and intestinal epithelial cell lines (T84, HT-29, Caco-2). Nuclear factor-kappaB transcriptional activation was used as a measure of LPS responsiveness. Intestinal epithelial cells in patients with inflammatory bowel disease exhibited increased expression of MD-2 and TLR4 mRNA. Lipopolysaccharide responsiveness in IECs was polarized to the basolateral membrane. Bisulfite sequencing of the MD-2 promoter demonstrated methylation of CpG dinucleotides. Inhibition of methylation by 5-azacytidine and histone de-actylation by trichostatin A, two forms of epigenetic silencing, resulted in increased mRNA expression of MD-2 in IECs. These results demonstrate various molecular mechanisms by which IECs down-regulate MD-2 and, thereby, protect against dysregulated inflammation to commensal bacteria in the intestinal lumen.
Collapse
Affiliation(s)
- Arunan S Vamadevan
- Division of Gastroenterology, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida 33101, USA
| | | | | | | | | | | |
Collapse
|
33
|
Innate immune sensing and activation of cell surface Toll-like receptors. Semin Immunol 2009; 21:175-84. [DOI: 10.1016/j.smim.2009.05.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2009] [Accepted: 05/06/2009] [Indexed: 12/30/2022]
|
34
|
Tissières P, Araud T, Ochoda A, Drifte G, Dunn-Siegrist I, Pugin J. Cooperation between PU.1 and CAAT/enhancer-binding protein beta is necessary to induce the expression of the MD-2 gene. J Biol Chem 2009; 284:26261-72. [PMID: 19632992 DOI: 10.1074/jbc.m109.042580] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Myeloid differentiation factor 2 (MD-2) binds Gram-negative bacterial lipopolysaccharide with high affinity and is essential for Toll-like receptor 4-dependent signal transduction. MD-2 has recently been recognized as a type II acute phase protein. Plasma concentrations of the soluble form of MD-2 increase markedly during the course of severe infections. Its production is regulated in hepatocytes and myeloid cells by interleukin-6 (IL-6) but not IL-1beta. In the present work we show that two transcription factors (TF), PU.1 and CAAT/enhancer-binding protein beta (C/EBPbeta), participate in the activation of the human MD-2 gene in hepatocytic cells after stimulation with IL-6. PU.1 TF and proximal PU.1 binding sites in the MD-2 promoter were shown to be critical for the basal activity of the promoter as well as for IL-6-induced soluble MD-2 production. Deletions of proximal portions of the MD-2 promoter containing PU.1 and/or NF-IL-6 consensus binding sites as well as site-directed mutagenesis of these binding sites abrogated IL-6-dependent MD-2 gene activation. We show that the cooperation between C/EBPbeta and PU.1 is critical for the transcriptional activation of the MD-2 gene by IL-6. PU.1 was essentially known as a TF involved in the differentiation of myeloid precursor cells and the expression of surface receptors of the innate immunity. Herein, we show that it also participates in the regulation of an acute phase protein, MD-2, in nonmyeloid cells cooperatively with C/EBPbeta, a classical IL-6-inducible TF.
Collapse
Affiliation(s)
- Pierre Tissières
- Intensive Care, University Hospitals of Geneva, Geneva 14, Switzerland
| | | | | | | | | | | |
Collapse
|
35
|
Piazza M, Rossini C, Della Fiorentina S, Pozzi C, Comelli F, Bettoni I, Fusi P, Costa B, Peri F. Glycolipids and benzylammonium lipids as novel antisepsis agents: synthesis and biological characterization. J Med Chem 2009; 52:1209-13. [PMID: 19161283 DOI: 10.1021/jm801333m] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
New glycolipids and a benzylammonium lipid were rationally designed by varying the chemical structure of a D-glucose-derived hit compound active as lipid A antagonist. We report the synthesis of these compounds, their in vitro activity as lipid A antagonists on HEK cells, and the capacity to inhibit LPS-induced septic shock in vivo. The lack of toxicity and the good in vivo activity suggest the use of some compounds of the panel as hits for antisepsis drug development.
Collapse
Affiliation(s)
- Matteo Piazza
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milano, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Sandanger Ø, Ryan L, Bohnhorst J, Iversen AC, Husebye H, Halaas Ø, Landrø L, Aukrust P, Frøland SS, Elson G, Visintin A, Øktedalen O, Damås JK, Sundan A, Golenbock D, Espevik T. IL-10 enhances MD-2 and CD14 expression in monocytes and the proteins are increased and correlated in HIV-infected patients. THE JOURNAL OF IMMUNOLOGY 2009; 182:588-95. [PMID: 19109192 DOI: 10.4049/jimmunol.182.1.588] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Soluble proteins that bind LPS, like myeloid differentiation-2 (MD-2) and CD14, have essential roles in regulating LPS signaling through TLR4. During a gram-negative bacterial infection, the host may control the response by adjusting the levels of soluble MD-2 and CD14. To address the surface expression of MD-2 on human leukocytes, we developed a mAb, IIC1, that recognized MD-2 both free and when bound to TLR4. MD-2 was found on the surface of freshly isolated monocytes, on a subpopulation of CD19(+) B-cells and on CD15(+) neutrophils. LPS transiently reduced the MD-2 levels on monocytes, which is most likely due to endocytosis of the LPS receptor complex since MD-2 colocalized with TLR4 in early endosomes after LPS stimulation. In the absence of LPS, MD-2 partly colocalized with TLR4 in Golgi trans and medial compartments. Cultivating monocytes for 18-20 h resulted in loss of MD-2 expression on the surface, which was reversed either by LPS or IL-10. Furthermore, addition of IL-10, but not LPS, resulted in a considerable increase in mRNA for both MD-2 and CD14. Using ELISA, we demonstrated that IL-10 had a profound dose- and time-related effect on the release of soluble MD-2 and soluble CD14 from monocytes. In HIV-infected patients, the amounts of MD-2, CD14, and IL-10 increased significantly in the patient group with AIDS. Of interest, we found that IL-10, CD14, and MD-2 levels were positively correlated, suggesting that IL-10 may be a driving force for increased release of MD-2 and CD14 during systemic inflammation.
Collapse
Affiliation(s)
- Øystein Sandanger
- Norwegian University of Science and Technology, Institute of Cancer Research and Molecular Medicine, Trondheim, Norway
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|