1
|
Sant'Anna MRV, Pereira-Filho AA, Mendes-Sousa AF, Silva NCS, Gontijo NF, Pereira MH, Koerich LB, D'Avila Pessoa GC, Andersen J, Araujo RN. Inhibition of vertebrate complement system by hematophagous arthropods: inhibitory molecules, mechanisms, physiological roles, and applications. INSECT SCIENCE 2024; 31:1334-1352. [PMID: 38246860 DOI: 10.1111/1744-7917.13317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/28/2023] [Accepted: 12/07/2023] [Indexed: 01/23/2024]
Abstract
In arthropods, hematophagy has arisen several times throughout evolution. This specialized feeding behavior offered a highly nutritious diet obtained during blood feeds. On the other hand, blood-sucking arthropods must overcome problems brought on by blood intake and digestion. Host blood complement acts on the bite site and is still active after ingestion, so complement activation is a potential threat to the host's skin feeding environment and to the arthropod gut enterocytes. During evolution, blood-sucking arthropods have selected, either in their saliva or gut, anticomplement molecules that inactivate host blood complement. This review presents an overview of the complement system and discusses the arthropod's salivary and gut anticomplement molecules studied to date, exploring their mechanism of action and other aspects related to the arthropod-host-pathogen interface. The possible therapeutic applications of arthropod's anticomplement molecules are also discussed.
Collapse
Affiliation(s)
- Mauricio Roberto Vianna Sant'Anna
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil
| | - Adalberto Alves Pereira-Filho
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Naylene Carvalho Sales Silva
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Nelder Figueiredo Gontijo
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil
| | - Marcos Horácio Pereira
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil
| | - Leonardo Barbosa Koerich
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil
| | - Grasielle Caldas D'Avila Pessoa
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil
| | - John Andersen
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Ricardo Nascimento Araujo
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
2
|
Shangguan W, Li X, Wang Y, Huang Z, Dong Y, Feng M, Feng J. Design and Biological Evaluation of the Long-Acting C5-Inhibited Ornithodoros moubata Complement Inhibitor (OmCI) Modified with Fatty Acid. Bioconjug Chem 2024; 35:653-664. [PMID: 38593046 DOI: 10.1021/acs.bioconjchem.4c00126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Disorder of complement response is a significant pathogenic factor causing some autoimmune and inflammation diseases. The Ornithodoros moubata Complement Inhibitor (OmCI), a small 17 kDa natural protein, was initially extracted from soft tick salivary glands. The protein was found binding to complement C5 specifically, inhibiting the activation of the complement pathway, which is a successful therapeutic basis of complement-mediated diseases. However, a short half-life due to rapid renal clearance is a common limitation of small proteins for clinical application. In this study, we extended the half-life of OmCI by modifying it with fatty acid, which was a method used to improve the pharmacokinetics of native peptides and proteins. Five OmCI mutants were initially designed, and single-site cysteine mutation was introduced to each of them. After purification, four OmCI mutants were obtained that showed similar in vitro biological activities. Three mutants of them were subsequently coupled with different fatty acids by nucleophilic substitution. In total, 15 modified derivatives were screened and tested for anticomplement activity in vitro. The results showed that coupling with fatty acid would not significantly affect their complement-inhibitory activity (CH50 and AH50). OmCIT90C-CM02 and OmCIT90C-CM05 were validated as the applicable OmCI bioconjugates for further pharmacokinetic assessments, and both showed improved plasma half-life in mice compared with unmodified OmCI (15.86, 17.96 vs 2.57 h). In summary, our data demonstrated that OmCI conjugated with fatty acid could be developed as the potential long-acting C5 complement inhibitor in the clinic.
Collapse
Affiliation(s)
- Wenwen Shangguan
- School of Pharmacy, Fudan University, 201203 Shanghai, China
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Xiaowan Li
- School of Pharmacy, Fudan University, 201203 Shanghai, China
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Yandan Wang
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, 310014 Hangzhou, China
| | - Zongqing Huang
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
- Shanghai Duomirui Biotechnology Co Ltd, 201203 Shanghai, China
| | - Yuanzhen Dong
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
- Shanghai Duomirui Biotechnology Co Ltd, 201203 Shanghai, China
| | - Meiqing Feng
- School of Pharmacy, Fudan University, 201203 Shanghai, China
| | - Jun Feng
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| |
Collapse
|
3
|
Yang Z, Nicholson SE, Cancio TS, Cancio LC, Li Y. Complement as a vital nexus of the pathobiological connectome for acute respiratory distress syndrome: An emerging therapeutic target. Front Immunol 2023; 14:1100461. [PMID: 37006238 PMCID: PMC10064147 DOI: 10.3389/fimmu.2023.1100461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/27/2023] [Indexed: 03/19/2023] Open
Abstract
The hallmark of acute respiratory distress syndrome (ARDS) pathobiology is unchecked inflammation-driven diffuse alveolar damage and alveolar-capillary barrier dysfunction. Currently, therapeutic interventions for ARDS remain largely limited to pulmonary-supportive strategies, and there is an unmet demand for pharmacologic therapies targeting the underlying pathology of ARDS in patients suffering from the illness. The complement cascade (ComC) plays an integral role in the regulation of both innate and adaptive immune responses. ComC activation can prime an overzealous cytokine storm and tissue/organ damage. The ARDS and acute lung injury (ALI) have an established relationship with early maladaptive ComC activation. In this review, we have collected evidence from the current studies linking ALI/ARDS with ComC dysregulation, focusing on elucidating the new emerging roles of the extracellular (canonical) and intracellular (non-canonical or complosome), ComC (complementome) in ALI/ARDS pathobiology, and highlighting complementome as a vital nexus of the pathobiological connectome for ALI/ARDS via its crosstalking with other systems of the immunome, DAMPome, PAMPome, coagulome, metabolome, and microbiome. We have also discussed the diagnostic/therapeutic potential and future direction of ALI/ARDS care with the ultimate goal of better defining mechanistic subtypes (endotypes and theratypes) through new methodologies in order to facilitate a more precise and effective complement-targeted therapy for treating these comorbidities. This information leads to support for a therapeutic anti-inflammatory strategy by targeting the ComC, where the arsenal of clinical-stage complement-specific drugs is available, especially for patients with ALI/ARDS due to COVID-19.
Collapse
Affiliation(s)
- Zhangsheng Yang
- Combat Casualty Care Research Team (CRT) 3, United States (US) Army Institute of Surgical Research, Joint Base San Antonio (JBSA)-Fort Sam Houston, TX, United States
| | - Susannah E. Nicholson
- Division of Trauma Research, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Tomas S. Cancio
- Combat Casualty Care Research Team (CRT) 3, United States (US) Army Institute of Surgical Research, Joint Base San Antonio (JBSA)-Fort Sam Houston, TX, United States
| | - Leopoldo C. Cancio
- United States (US) Army Burn Center, United States (US) Army Institute of Surgical Research, Joint Base San Antonio (JBSA)-Fort Sam Houston, TX, United States
| | - Yansong Li
- Division of Trauma Research, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- The Geneva Foundation, Immunological Damage Control Resuscitation Program, Tacoma, WA, United States
- *Correspondence: Yansong Li,
| |
Collapse
|
4
|
Stennett A, Friston K, Harris CL, Wollman AJM, Bronowska AK, Madden KS. The case for complement component 5 as a target in neurodegenerative disease. Expert Opin Ther Targets 2023; 27:97-109. [PMID: 36786123 DOI: 10.1080/14728222.2023.2177532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
INTRODUCTION Complement-based drug discovery is undergoing a renaissance, empowered by new advances in structural biology, complement biology and drug development. Certain components of the complement pathway, particularly C1q and C3, have been extensively studied in the context of neurodegenerative disease, and established as key therapeutic targets. C5 also has huge therapeutic potential in this arena, with its druggability clearly demonstrated by the success of C5-inhibitor eculizumab. AREAS COVERED We will discuss the evidence supporting C5 as a target in neurodegenerative disease, along with the current progress in developing different classes of C5 inhibitors and the gaps in knowledge that will help progress in the field. EXPERT OPINION Validation of C5 as a therapeutic target for neurodegenerative disease would represent a major step forward for complement therapeutics research and has the potential to furnish disease-modifying drugs for millions of patients suffering worldwide. Key hurdles that need to be overcome for this to be achieved are understanding how C5a and C5b should be targeted to bring therapeutic benefit and demonstrating the ability to target C5 without creating vulnerability to infection in patients. This requires greater biological elucidation of its precise role in disease pathogenesis, supported by better chemical/biological tools.
Collapse
Affiliation(s)
- Amelia Stennett
- School of Natural and Environmental Sciences, Newcastle University, NE1 7RU, Newcastle-Upon-Tyne, UK
| | - Kallie Friston
- School of Natural and Environmental Sciences, Newcastle University, NE1 7RU, Newcastle-Upon-Tyne, UK
| | - Claire L Harris
- Faculty of Medical Sciences, Newcastle University, NE2 4HH, Newcastle-Upon-Tyne, UK
| | - Adam J M Wollman
- Faculty of Medical Sciences, Newcastle University, NE2 4HH, Newcastle-Upon-Tyne, UK
| | - Agnieszka K Bronowska
- School of Natural and Environmental Sciences, Newcastle University, NE1 7RU, Newcastle-Upon-Tyne, UK
| | - Katrina S Madden
- School of Natural and Environmental Sciences, Newcastle University, NE1 7RU, Newcastle-Upon-Tyne, UK.,Faculty of Medical Sciences, Newcastle University, NE2 4HH, Newcastle-Upon-Tyne, UK
| |
Collapse
|
5
|
Sadik CD, Rashid H, Hammers CM, Diercks GFH, Weidinger A, Beissert S, Schauer F, Fettiplace J, Thaçi D, Ngai Y, Nunn MA, Zillikens D, Horváth B. Evaluation of Nomacopan for Treatment of Bullous Pemphigoid: A Phase 2a Nonrandomized Controlled Trial. JAMA Dermatol 2022; 158:641-649. [PMID: 35507334 PMCID: PMC9069343 DOI: 10.1001/jamadermatol.2022.1156] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Importance Bullous pemphigoid is a difficult-to-treat autoimmune blistering skin disease that predominantly affects older adults and is associated with an increased mortality rate. Objective To examine the safety and therapeutic potential of nomacopan, an inhibitor of leukotriene B4 and complement C5, in patients with bullous pemphigoid. Design, Setting, and Participants This multicenter, single-group, phase 2a nonrandomized controlled trial was conducted in the dermatology departments of universities in the Netherlands and Germany. Participants were enrolled between September 2018 and April 2020. Older adult patients (aged ≥55 years) with mild to moderate, new-onset or relapsing bullous pemphigoid were recruited into the study. Interventions Patients received nomacopan, 90 mg, subcutaneously on day 1 and 30 mg subcutaneously daily until day 42. Main Outcomes and Measures The primary end point was the proportion of patients with grade 3 to 5 (severe) adverse events associated or possibly associated with nomacopan. Secondary end points included mean absolute and percentage changes in the Bullous Pemphigoid Disease Area Index (BPDAI) activity score, the BPDAI pruritus score, and the patient-reported outcome measures Dermatology Life Quality Index (DLQI) and Treatment of Autoimmune Bullous Disease Quality of Life (TABQOL). Results A total of 9 patients (median [range] age, 75 [55-85] years) with bullous pemphigoid were included in the trial, of whom 5 were women (55.6%). No serious adverse events associated with nomacopan were found. The mean (90% CI) BPDAI activity score decreased from 32.0 (8.7) points on day 1 to 19.6 (9.0) points on day 42. Seven of 9 patients (77.8%) responded to nomacopan with a reduction in the BPDAI activity score of at least 8 points between days 1 and 42; in 3 responders, the reduction was 80% or greater. On day 42, the mean (90% CI) BPDAI pruritus score had decreased by 6.8 (4.6) points from 17.6 (4.0) points on day 1. The mean (90% CI) DLQI score decreased from 11.3 (4.2) points at baseline to 6.4 (3.8) points by day 42, and the mean (90% CI) TABQOL score decreased from 14.6 (5.4) points at baseline to 10.3 (5.0) points on day 42. Conclusions and Relevance Results of this nonrandomized controlled trial suggest that nomacopan can be well tolerated in older patients with bullous pemphigoid and may have therapeutic benefits for suppressing acute flares of this disease. A larger, placebo-controlled randomized clinical trial is warranted to confirm this safety profile and to establish nomacopan as a new therapeutic option for bullous pemphigoid. Trial Registration ClinicalTrials.gov Identifier: NCT04035733.
Collapse
Affiliation(s)
- Christian D Sadik
- Department of Dermatology, Allergy, and Venereology, University of Lübeck, Lübeck, Germany.,Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Hanan Rashid
- Department of Dermatology, Center of Blistering Diseases, European Reference Network-Skin, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Christoph M Hammers
- Department of Dermatology, Allergy, and Venereology, University of Lübeck, Lübeck, Germany.,Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Gilles F H Diercks
- Department of Dermatology, Center of Blistering Diseases, European Reference Network-Skin, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anke Weidinger
- Department of Dermatology and Allergy, University of Kiel, Kiel, Germany
| | - Stefan Beissert
- Department of Dermatology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Franziska Schauer
- Department of Dermatology, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | | | - Diamant Thaçi
- Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany.,Institute and Comprehensive Center for Inflammation Medicine, University of Lübeck, Lübeck, Germany
| | | | | | - Detlef Zillikens
- Department of Dermatology, Allergy, and Venereology, University of Lübeck, Lübeck, Germany.,Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Barbara Horváth
- Department of Dermatology, Center of Blistering Diseases, European Reference Network-Skin, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
6
|
Ali A, Zeb I, Alouffi A, Zahid H, Almutairi MM, Ayed Alshammari F, Alrouji M, Termignoni C, Vaz IDS, Tanaka T. Host Immune Responses to Salivary Components - A Critical Facet of Tick-Host Interactions. Front Cell Infect Microbiol 2022; 12:809052. [PMID: 35372098 PMCID: PMC8966233 DOI: 10.3389/fcimb.2022.809052] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/04/2022] [Indexed: 12/15/2022] Open
Abstract
Tick sialome is comprised of a rich cocktail of bioactive molecules that function as a tool to disarm host immunity, assist blood-feeding, and play a vibrant role in pathogen transmission. The adaptation of the tick's blood-feeding behavior has lead to the evolution of bioactive molecules in its saliva to assist them to overwhelm hosts' defense mechanisms. During a blood meal, a tick secretes different salivary molecules including vasodilators, platelet aggregation inhibitors, anticoagulants, anti-inflammatory proteins, and inhibitors of complement activation; the salivary repertoire changes to meet various needs such as tick attachment, feeding, and modulation or impairment of the local dynamic and vigorous host responses. For instance, the tick's salivary immunomodulatory and cement proteins facilitate the tick's attachment to the host to enhance prolonged blood-feeding and to modulate the host's innate and adaptive immune responses. Recent advances implemented in the field of "omics" have substantially assisted our understanding of host immune modulation and immune inhibition against the molecular dynamics of tick salivary molecules in a crosstalk between the tick-host interface. A deep understanding of the tick salivary molecules, their substantial roles in multifactorial immunological cascades, variations in secretion, and host immune responses against these molecules is necessary to control these parasites. In this article, we reviewed updated knowledge about the molecular mechanisms underlying host responses to diverse elements in tick saliva throughout tick invasion, as well as host defense strategies. In conclusion, understanding the mechanisms involved in the complex interactions between the tick salivary components and host responses is essential to decipher the host defense mechanisms against the tick evasion strategies at tick-host interface which is promising in the development of effective anti-tick vaccines and drug therapeutics.
Collapse
Affiliation(s)
- Abid Ali
- Department of Zoology, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Ismail Zeb
- Department of Zoology, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Abdulaziz Alouffi
- King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Hafsa Zahid
- Department of Zoology, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Mashal M. Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Fahdah Ayed Alshammari
- College of Sciences and Literature Microbiology, Nothern Border University, Rafha, Saudi Arabia
| | - Mohammed Alrouji
- College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia
| | - Carlos Termignoni
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Itabajara da Silva Vaz
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Tetsuya Tanaka
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
7
|
Eskandarpour M, Zhang X, Micera A, Zaher S, Larkin FDP, Nunn M, Bonini S, Weston‐Davies W, Calder VL. Allergic eye disease: Blocking LTB4/C5 in vivo suppressed disease and Th2 & Th9 cells. Allergy 2022; 77:660-664. [PMID: 34614221 PMCID: PMC9292376 DOI: 10.1111/all.15128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/27/2021] [Accepted: 10/03/2021] [Indexed: 11/29/2022]
Affiliation(s)
| | - Xiaozhe Zhang
- Ocular Immunology Group UCL Institute of Ophthalmology London UK
| | - Alessandra Micera
- UOSD Research Laboratories IRCCS‐fondazione Bietti University Campus Bio‐Medico Rome Italy
| | - Sarah Zaher
- NIHR Moorfields Clinical Research Facility Moorfields Eye Hospital London UK
- NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology London UK
| | - Frank D. P. Larkin
- NIHR Moorfields Clinical Research Facility Moorfields Eye Hospital London UK
- NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology London UK
| | | | - Stefano Bonini
- Ophthalmology Operative Complex Unit University Campus Bio‐Medico Rome Italy
| | | | - Virginia L. Calder
- Ocular Immunology Group UCL Institute of Ophthalmology London UK
- NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology London UK
| |
Collapse
|
8
|
Braunger K, Ahn J, Jore MM, Johnson S, Tang TTL, Pedersen DV, Andersen GR, Lea SM. Structure and function of a family of tick-derived complement inhibitors targeting properdin. Nat Commun 2022; 13:317. [PMID: 35031611 PMCID: PMC8760278 DOI: 10.1038/s41467-021-27920-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022] Open
Abstract
Activation of the serum-resident complement system begins a cascade that leads to activation of membrane-resident complement receptors on immune cells, thus coordinating serum and cellular immune responses. Whilst many molecules act to control inappropriate activation, Properdin is the only known positive regulator of the human complement system. By stabilising the alternative pathway C3 convertase it promotes complement self-amplification and persistent activation boosting the magnitude of the serum complement response by all triggers. In this work, we identify a family of tick-derived alternative pathway complement inhibitors, hereafter termed CirpA. Functional and structural characterisation reveals that members of the CirpA family directly bind to properdin, inhibiting its ability to promote complement activation, and leading to potent inhibition of the complement response in a species specific manner. We provide a full functional and structural characterisation of a properdin inhibitor, opening avenues for future therapeutic approaches.
Collapse
Affiliation(s)
- Katharina Braunger
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, Oxford, UK
| | - Jiyoon Ahn
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, Oxford, UK
| | - Matthijs M Jore
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, Oxford, UK
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Steven Johnson
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, Oxford, UK.
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute, 21702, Frederick, MD, USA.
| | - Terence T L Tang
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, Oxford, UK
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Dennis V Pedersen
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000, Aarhus, Denmark
| | - Gregers R Andersen
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000, Aarhus, Denmark
| | - Susan M Lea
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, Oxford, UK.
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute, 21702, Frederick, MD, USA.
- Central Oxford Structural Molecular Imaging Centre, University of Oxford, OX1 3RE, Oxford, UK.
| |
Collapse
|
9
|
Horikami D, Fujii W, Aritake K, Murata T. L-PGDS Attenuates Acute Lung Injury by Prostaglandin D 2 in Both Dependent and Independent Ways. THE JOURNAL OF IMMUNOLOGY 2021; 207:2545-2550. [PMID: 34615734 DOI: 10.4049/jimmunol.2100394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 09/06/2021] [Indexed: 11/19/2022]
Abstract
Lipocalin-type PG D synthase (L-PGDS) has two roles: it can be a PGD synthase, or it can be a carrier protein of hydrophobic small molecules. In this study, we investigated the dual roles of L-PGDS in acute lung injury by using L-PGDS-deficient and point-mutated mice, which lack PGD2 producibility but maintain lipocalin ability. Hydrochloride (HCl) administration (0.1 M intratracheally for 6 h) caused hemorrhage and dysfunction in the wild-type (WT) mouse lung. These symptoms were accompanied by an increase in PGD2 production. Both deficiency and point mutation of L-PGDS aggravated the HCl-induced hemorrhage and dysfunction. Although both the gene modifications decreased PGD2 production, only L-PGDS-deficient mice, but not point mutation mice, lacked protein expressions of L-PGDS in the lungs. In the WT mice, HCl administration caused pulmonary edema, indexed as an increase in lung water content and protein leakage in bronchoalveolar lavage fluid. L-PGDS deficiency and point mutation similarly aggravated edema formation. HCl administration also stimulated mucin production and bronchoalveolar lavage fluid leukocyte infiltration in the WT mouse lungs. Of interest, L-PGDS deficiency, but not point mutation, exacerbated these manifestations. Consistently, only L-PGDS deficiency increased the mRNA expression of IL-33, which stimulates mucin production in the inflamed lung. These results show that L-PGDS attenuated HCl-induced acute lung injury progresses in two different ways: L-PGDS produced PGD2, which inhibited pulmonary edema formation, whereas its lipocalin ability decreased mucin formation and inflammatory cell infiltration in the inflamed lung.
Collapse
Affiliation(s)
- Daiki Horikami
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Tokyo, Japan
| | - Wataru Fujii
- Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Tokyo, Japan; and
| | - Kosuke Aritake
- Laboratory of Chemical Pharmacology, Daiichi University of Pharmacy, Fukuoka, Japan
| | - Takahisa Murata
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Tokyo, Japan;
| |
Collapse
|
10
|
Denisov SS, Dijkgraaf I. Immunomodulatory Proteins in Tick Saliva From a Structural Perspective. Front Cell Infect Microbiol 2021; 11:769574. [PMID: 34722347 PMCID: PMC8548845 DOI: 10.3389/fcimb.2021.769574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/28/2021] [Indexed: 12/25/2022] Open
Abstract
To feed successfully, ticks must bypass or suppress the host’s defense mechanisms, particularly the immune system. To accomplish this, ticks secrete specialized immunomodulatory proteins into their saliva, just like many other blood-sucking parasites. However, the strategy of ticks is rather unique compared to their counterparts. Ticks’ tendency for gene duplication has led to a diverse arsenal of dozens of closely related proteins from several classes to modulate the immune system’s response. Among these are chemokine-binding proteins, complement pathways inhibitors, ion channels modulators, and numerous poorly characterized proteins whose functions are yet to be uncovered. Studying tick immunomodulatory proteins would not only help to elucidate tick-host relationships but would also provide a rich pool of potential candidates for the development of immunomodulatory intervention drugs and potentially new vaccines. In the present review, we will attempt to summarize novel findings on the salivary immunomodulatory proteins of ticks, focusing on biomolecular targets, structure-activity relationships, and the perspective of their development into therapeutics.
Collapse
Affiliation(s)
- Stepan S Denisov
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, Netherlands
| | - Ingrid Dijkgraaf
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, Netherlands
| |
Collapse
|
11
|
Eskandarpour M, Nunn MA, Weston-Davies W, Calder VL. Immune-Mediated Retinal Vasculitis in Posterior Uveitis and Experimental Models: The Leukotriene (LT)B4-VEGF Axis. Cells 2021; 10:cells10020396. [PMID: 33671954 PMCID: PMC7919050 DOI: 10.3390/cells10020396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/09/2021] [Accepted: 02/12/2021] [Indexed: 02/07/2023] Open
Abstract
Retinal vascular diseases have distinct, complex and multifactorial pathogeneses yet share several key pathophysiological aspects including inflammation, vascular permeability and neovascularisation. In non-infectious posterior uveitis (NIU), retinal vasculitis involves vessel leakage leading to retinal enlargement, exudation, and macular oedema. Neovascularisation is not a common feature in NIU, however, detection of the major angiogenic factor—vascular endothelial growth factor A (VEGF-A)—in intraocular fluids in animal models of uveitis may be an indication for a role for this cytokine in a highly inflammatory condition. Suppression of VEGF-A by directly targeting the leukotriene B4 (LTB4) receptor (BLT1) pathway indicates a connection between leukotrienes (LTs), which have prominent roles in initiating and propagating inflammatory responses, and VEGF-A in retinal inflammatory diseases. Further research is needed to understand how LTs interact with intraocular cytokines in retinal inflammatory diseases to guide the development of novel therapeutic approaches targeting both inflammatory mediator pathways.
Collapse
Affiliation(s)
- Malihe Eskandarpour
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK;
- Correspondence:
| | - Miles A. Nunn
- Akari Therapeutics Plc, London EC1V 9EL, UK; (M.A.N.); (W.W.-D.)
| | | | - Virginia L. Calder
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK;
| |
Collapse
|
12
|
Krueger S, Curtis JE, Scott DR, Grishaev A, Glenn G, Smith G, Ellingsworth L, Borisov O, Maynard EL. Structural Characterization and Modeling of a Respiratory Syncytial Virus Fusion Glycoprotein Nanoparticle Vaccine in Solution. Mol Pharm 2021; 18:359-376. [PMID: 33322901 PMCID: PMC10467610 DOI: 10.1021/acs.molpharmaceut.0c00986] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The respiratory syncytial virus (RSV) fusion (F) protein/polysorbate 80 (PS80) nanoparticle vaccine is the most clinically advanced vaccine for maternal immunization and protection of newborns against RSV infection. It is composed of a near-full-length RSV F glycoprotein, with an intact membrane domain, formulated into a stable nanoparticle with PS80 detergent. To understand the structural basis for the efficacy of the vaccine, a comprehensive study of its structure and hydrodynamic properties in solution was performed. Small-angle neutron scattering experiments indicate that the nanoparticle contains an average of 350 PS80 molecules, which form a cylindrical micellar core structure and five RSV F trimers that are arranged around the long axis of the PS80 core. All-atom models of full-length RSV F trimers were built from crystal structures of the soluble ectodomain and arranged around the long axis of the PS80 core, allowing for the generation of an ensemble of conformations that agree with small-angle neutron and X-ray scattering data as well as transmission electron microscopy (TEM) images. Furthermore, the hydrodynamic size of the RSV F nanoparticle was found to be modulated by the molar ratio of PS80 to protein, suggesting a mechanism for nanoparticle assembly involving addition of RSV F trimers to and growth along the long axis of the PS80 core. This study provides structural details of antigen presentation and conformation in the RSV F nanoparticle vaccine, helping to explain the induction of broad immunity and observed clinical efficacy. Small-angle scattering methods provide a general strategy to visualize surface glycoproteins from other pathogens and to structurally characterize nanoparticle vaccines.
Collapse
Affiliation(s)
- Susan Krueger
- NIST Center for Neutron Research, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, Maryland 20899, United States
| | - Joseph E Curtis
- NIST Center for Neutron Research, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, Maryland 20899, United States
| | - Daniel R Scott
- Novavax, Inc., 21 Firstfield Road, Gaithersburg, Maryland 20878, United States
| | - Alexander Grishaev
- Institute for Bioscience and Biotechnology Research, National Institute of Standards and Technology, 9600 Gudelsky Drive, Rockville, Maryland 20850, United States
| | - Greg Glenn
- Novavax, Inc., 21 Firstfield Road, Gaithersburg, Maryland 20878, United States
| | - Gale Smith
- Novavax, Inc., 21 Firstfield Road, Gaithersburg, Maryland 20878, United States
| | - Larry Ellingsworth
- Novavax, Inc., 21 Firstfield Road, Gaithersburg, Maryland 20878, United States
| | - Oleg Borisov
- Novavax, Inc., 21 Firstfield Road, Gaithersburg, Maryland 20878, United States
| | | |
Collapse
|
13
|
Leukotriene B 4 and Its Receptor in Experimental Autoimmune Uveitis and in Human Retinal Tissues: Clinical Severity and LTB 4 Dependence of Retinal Th17 Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:320-334. [PMID: 33159884 DOI: 10.1016/j.ajpath.2020.10.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 12/15/2022]
Abstract
Nomacopan, a drug originally derived from tick saliva, has dual functions of sequestering leukotriene B4 (LTB4) and inhibiting complement component 5 (C5) activation. Nomacopan has been shown to provide therapeutic benefit in experimental autoimmune uveitis (EAU). Longer acting forms of nomacopan were more efficacious in mouse EAU models, and the long-acting variant that inhibited only LTB4 was at least as effective as the long-acting variant that inhibited both C5 and LTB4, preventing structural damage to the retina and a significantly reducing effector T helper 17 cells and inflammatory macrophages. Increased levels of LTB4 and C5a (produced upon C5 activation) were detected during disease progression. Activated retinal lymphocytes were shown to express LTB4 receptors (R) in vitro and in inflamed draining lymph nodes. Levels of LTB4R-expressing active/inflammatory retinal macrophages were also increased. Within the draining lymph node CD4+ T-cell population, 30% expressed LTB4R+ following activation in vitro, whereas retinal infiltrating cells expressed LTB4R and C5aR. Validation of expression of those receptors in human uveitis and healthy tissues suggests that infiltrating cells could be targeted by inhibitors of the LTB4-LTB4 receptor 1 (BLT1) pathway as a novel therapeutic approach. This study provides novel data on intraocular LTB4 and C5a in EAU, their associated receptor expression by retinal infiltrating cells in mouse and human tissues, and in attenuating EAU via the dual inhibitor nomacopan.
Collapse
|
14
|
A soft tick Ornithodoros moubata salivary protein OmCI is a potent inhibitor to prevent avian complement activation. Ticks Tick Borne Dis 2019; 11:101354. [PMID: 31866440 DOI: 10.1016/j.ttbdis.2019.101354] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 11/15/2019] [Accepted: 12/02/2019] [Indexed: 01/15/2023]
Abstract
Complement is a key first line innate host defense system in the blood of vertebrates. Upon activation, this powerful defense mechanism can elicit inflammatory responses, lyse non-self-cells, or mark them for opsonophagocytic removal. Blood-feeding arthropods thus require the ability to block host complement activation in the bloodmeal to prevent undesired cell or tissue damage during feeding. The soft tick Ornithodoros moubata produces a complement inhibitory protein, OmCI. This protein binds to a mammalian complement protein C5 and blocks further activation of complement cascades, which results in the prevention of complement-mediated bacterial killing through membrane attack complex. Interestingly, the amino acids involved in OmCI binding are highly conserved among mammalian and avian C5, but the ability of this protein to inhibit the complement from birds remains unclear. Here we demonstrated that OmCI is capable of preventing quail complement-mediated erythrocyte lysis, inhibiting the capability of this animal's complement to eliminate a serum-sensitive Lyme disease bacterial strain. We also found that the ability of OmCI to inhibit quail complement-mediated killing of Lyme disease bacteria can be extended to different domestic and wild birds. Our results illustrate the utility of OmCI to block bird complement. These results provide the foundation for further use of this protein as a tool to study the molecular basis of avian complement and pathogen evasion to such a defense mechanism.
Collapse
|
15
|
Sezin T, Murthy S, Attah C, Seutter M, Holtsche MM, Hammers CM, Schmidt E, Meshrkey F, Mousavi S, Zillikens D, Nunn MA, Sadik CD. Dual inhibition of complement factor 5 and leukotriene B4 synergistically suppresses murine pemphigoid disease. JCI Insight 2019; 4:128239. [PMID: 31391346 DOI: 10.1172/jci.insight.128239] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/09/2019] [Indexed: 11/17/2022] Open
Abstract
The treatment of most autoimmune diseases still relies on systemic immunosuppression and is associated with severe side effects. The development of drugs that more specifically abrogate pathogenic pathways is therefore most desirable. In nature, such specificity is exemplified, e.g., by the soft tick-derived biotherapeutic Coversin, which locally suppresses immune responses by inhibiting complement factor 5 (C5) and leukotriene B4 (LTB4). C5a, a proteolytic fragment of C5, and LTB4 are critical drivers of skin inflammation in pemphigoid diseases (PDs), a group of autoimmune blistering skin diseases. Here, we demonstrate that both Coversin and its mutated form L-Coversin, which inhibits LTB4 only, dose dependently attenuate disease in a model of bullous pemphigoid-like epidermolysis bullosa acquisita (BP-like EBA). Coversin, however, reduces disease more effectively than L-Coversin, indicating that inhibition of C5 and LTB4 synergize in their suppressing effects in this model. Further supporting the therapeutic potential of Coversin in humans, we found that C5a and LTB4 are both present in the blister fluid of patients with BP in quantities inducing the recruitment of granulocytes and that the number of cells expressing their receptors, C5aR1 and BLT1, respectively, is increased in perilesional skin. Collectively, our results highlight Coversin and possibly L-Coversin as potential therapeutics for PDs.
Collapse
Affiliation(s)
- Tanya Sezin
- Department of Dermatology, Allergy, and Venereology
| | | | | | | | | | | | - Enno Schmidt
- Lübeck Institute for Experimental Dermatology, and.,Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | | | | | - Detlef Zillikens
- Department of Dermatology, Allergy, and Venereology.,Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | | | - Christian D Sadik
- Department of Dermatology, Allergy, and Venereology.,Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| |
Collapse
|
16
|
Mans BJ. Chemical Equilibrium at the Tick-Host Feeding Interface:A Critical Examination of Biological Relevance in Hematophagous Behavior. Front Physiol 2019; 10:530. [PMID: 31118903 PMCID: PMC6504839 DOI: 10.3389/fphys.2019.00530] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 04/15/2019] [Indexed: 12/14/2022] Open
Abstract
Ticks secrete hundreds to thousands of proteins into the feeding site, that presumably all play important functions in the modulation of host defense mechanisms. The current review considers the assumption that tick proteins have functional relevance during feeding. The feeding site may be described as a closed system and could be treated as an ideal equilibrium system, thereby allowing modeling of tick-host interactions in an equilibrium state. In this equilibrium state, the concentration of host and tick proteins and their affinities will determine functional relevance at the tick-host interface. Using this approach, many characterized tick proteins may have functional relevant concentrations and affinities at the feeding site. Conversely, the feeding site is not an ideal closed system, but is dynamic and changing, leading to possible overestimation of tick protein concentration at the feeding site and consequently an overestimation of functional relevance. Ticks have evolved different possible strategies to deal with this dynamic environment and overcome the barrier that equilibrium kinetics poses to tick feeding. Even so, cognisance of the limitations that equilibrium binding place on deductions of functional relevance should serve as an important incentive to determine both the concentration and affinity of tick proteins proposed to be functional at the feeding site.
Collapse
Affiliation(s)
- Ben J. Mans
- Epidemiology, Parasites and Vectors, Agricultural Research Council-Onderstepoort Veterinary Research, Pretoria, South Africa
- Department of Veterinary Tropical Diseases, University of Pretoria, Pretoria, South Africa
- Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
| |
Collapse
|
17
|
Michaudel C, Mackowiak C, Maillet I, Fauconnier L, Akdis CA, Sokolowska M, Dreher A, Tan HTT, Quesniaux VF, Ryffel B, Togbe D. Ozone exposure induces respiratory barrier biphasic injury and inflammation controlled by IL-33. J Allergy Clin Immunol 2018; 142:942-958. [DOI: 10.1016/j.jaci.2017.11.044] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 11/08/2017] [Accepted: 11/14/2017] [Indexed: 12/31/2022]
|
18
|
Macpherson A, Liu X, Dedi N, Kennedy J, Carrington B, Durrant O, Heywood S, van den Elsen J, Lawson ADG. The rational design of affinity-attenuated OmCI for the purification of complement C5. J Biol Chem 2018; 293:14112-14121. [PMID: 30030376 PMCID: PMC6130949 DOI: 10.1074/jbc.ra118.004043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/03/2018] [Indexed: 12/04/2022] Open
Abstract
Complement component C5 is the target of the mAb eculizumab and is the focus of a sustained drug discovery effort to prevent complement-induced inflammation in a range of autoimmune diseases. The immune evasion protein OmCI binds to and potently inactivates C5; this tight-binding interaction can be exploited to affinity-purify C5 protein from serum, offering a vastly simplified protocol compared with existing methods. However, breaking the high-affinity interaction requires conditions that risk denaturing or activating C5. We performed structure-guided in silico mutagenesis to identify prospective OmCI residues that contribute significantly to the binding affinity. We tested our predictions in vitro, using site-directed mutagenesis, and characterized mutants using a range of biophysical techniques, as well as functional assays. Our biophysical analyses suggest that the C5–OmCI interaction is complex with potential for multiple binding modes. We present single mutations that lower the affinity of OmCI for C5 and combinations of mutations that significantly decrease or entirely abrogate formation of the complex. The affinity-attenuated forms of OmCI are suitable for affinity purification and allow elution under mild conditions that are nondenaturing or activating to C5. We present the rational design, biophysical characterization, and experimental validation of affinity-reduced forms of OmCI as tool reagents to enable the affinity purification of C5.
Collapse
Affiliation(s)
- Alex Macpherson
- From the UCB-Celltech, Slough SL1 3WE, United Kingdom and .,the Department of Biology and Biochemistry, University of Bath, Bath BA2 7AX, United Kingdom
| | - Xiaofeng Liu
- From the UCB-Celltech, Slough SL1 3WE, United Kingdom and
| | - Neesha Dedi
- From the UCB-Celltech, Slough SL1 3WE, United Kingdom and
| | | | | | - Oliver Durrant
- From the UCB-Celltech, Slough SL1 3WE, United Kingdom and
| | - Sam Heywood
- From the UCB-Celltech, Slough SL1 3WE, United Kingdom and
| | - Jean van den Elsen
- the Department of Biology and Biochemistry, University of Bath, Bath BA2 7AX, United Kingdom
| | | |
Collapse
|
19
|
Sadik CD, Miyabe Y, Sezin T, Luster AD. The critical role of C5a as an initiator of neutrophil-mediated autoimmune inflammation of the joint and skin. Semin Immunol 2018; 37:21-29. [PMID: 29602515 DOI: 10.1016/j.smim.2018.03.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/18/2018] [Accepted: 03/20/2018] [Indexed: 01/15/2023]
Abstract
The deposition of IgG autoantibodies in peripheral tissues and the subsequent activation of the complement system, which leads to the accumulation of the anaphylatoxin C5a in these tissues, is a common hallmark of diverse autoimmune diseases, including rheumatoid arthritis (RA) and pemphigoid diseases (PDs). C5a is a potent chemoattractant for granulocytes and mice deficient in its precursor C5 or its receptor C5aR1 are resistant to granulocyte recruitment and, consequently, to tissue inflammation in several models of autoimmune diseases. However, the mechanism whereby C5a/C5aR regulates granulocyte recruitment in these diseases has remained elusive. Mechanistic studies over the past five years into the role of C5a/C5aR1 in the K/BxN serum arthritis mouse model have provided novel insights into the mechanisms C5a/C5aR1 engages to initiate granulocyte recruitment into the joint. It is now established that the critical actions of C5a/C5aR1 do not proceed in the joint itself, but on the luminal endothelial surface of the joint vasculature, where C5a/C5aR1 mediate the arrest of neutrophils on the endothelium by activating β2 integrin. Then, C5a/C5aR1 induces the release of leukotriene B4 (LTB4) from the arrested neutrophils. The latter, subsequently, initiates by autocrine/paracrine actions via its receptor BLT1 the egress of neutrophils from the blood vessel lumen into the interstitial. Compelling evidence suggests that this C5a/C5aR1-LTB4/BLT1 axis driving granulocyte recruitment in arthritis may represent a more generalizable biological principle critically regulating effector cell recruitment in other IgG autoantibody-induced diseases, such as in pemphigoid diseases. Thus, dual inhibition of C5a and LTB4, as implemented in nature by the lipocalin coversin in the soft-tick Ornithodoros moubata, may constitute a most effective therapeutic principle for the treatment of IgG autoantibody-driven diseases.
Collapse
Affiliation(s)
- Christian D Sadik
- Department of Dermatology, Allergy, and Venereology University of Lübeck, 23538, Lübeck, Germany.
| | - Yoshishige Miyabe
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tanya Sezin
- Department of Dermatology, Allergy, and Venereology University of Lübeck, 23538, Lübeck, Germany
| | - Andrew D Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
20
|
Roversi P, Johnson S, Preston SG, Nunn MA, Paesen GC, Austyn JM, Nuttall PA, Lea SM. Structural basis of cholesterol binding by a novel clade of dendritic cell modulators from ticks. Sci Rep 2017; 7:16057. [PMID: 29167574 PMCID: PMC5700055 DOI: 10.1038/s41598-017-16413-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/13/2017] [Indexed: 12/13/2022] Open
Abstract
Two crystal structures of Japanin, an 18 kDa immune-modulatory lipocalin from the Brown Ear Tick (Rhipicephalus appendiculatus), have been determined at 2.2 and 2.4 Å resolution. In both crystal forms the protein is in complex with cholesterol, which sits in a closed pocket at the centre of the lipocalin barrel. Both crystal forms are dimers, which are also observed in solution. Molecular modelling suggests that previously-described members of a tick protein family bearing high sequence homology to Japanin are also likely to bind cholesterol or cholesterol derivatives.
Collapse
Affiliation(s)
- Pietro Roversi
- Biochemistry Department, University of Oxford, Oxford, OX1 3QU, England, United Kingdom. .,Leicester Institute of Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester, LE1 7RH, England, United Kingdom.
| | - Steven Johnson
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, England, United Kingdom
| | - Stephen G Preston
- Department of Zoology, University of Oxford, Oxford, OX1 3PS, England, United Kingdom
| | - Miles A Nunn
- Akari Therapeutics, Plc, 75/76 Wimpole Street, London, W1G 9RT, England, United Kingdom
| | - Guido C Paesen
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, United Kingdom
| | - Jonathan M Austyn
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, England, United Kingdom
| | - Patricia A Nuttall
- Department of Zoology, University of Oxford, Oxford, OX1 3PS, England, United Kingdom
| | - Susan M Lea
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, England, United Kingdom.
| |
Collapse
|
21
|
Pischke SE, Gustavsen A, Orrem HL, Egge KH, Courivaud F, Fontenelle H, Despont A, Bongoni AK, Rieben R, Tønnessen TI, Nunn MA, Scott H, Skulstad H, Barratt-Due A, Mollnes TE. Complement factor 5 blockade reduces porcine myocardial infarction size and improves immediate cardiac function. Basic Res Cardiol 2017; 112:20. [PMID: 28258298 PMCID: PMC5336537 DOI: 10.1007/s00395-017-0610-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 02/28/2017] [Indexed: 12/31/2022]
Abstract
Inhibition of complement factor 5 (C5) reduced myocardial infarction in animal studies, while no benefit was found in clinical studies. Due to lack of cross-reactivity of clinically used C5 antibodies, different inhibitors were used in animal and clinical studies. Coversin (Ornithodoros moubata complement inhibitor, OmCI) blocks C5 cleavage and binds leukotriene B4 in humans and pigs. We hypothesized that inhibition of C5 before reperfusion will decrease infarct size and improve ventricular function in a porcine model of myocardial infarction. In pigs (Sus scrofa), the left anterior descending coronary artery was occluded (40 min) and reperfused (240 min). Coversin or placebo was infused 20 min after occlusion and throughout reperfusion in 16 blindly randomized pigs. Coversin significantly reduced myocardial infarction in the area at risk by 39% (p = 0.03, triphenyl tetrazolium chloride staining) and by 19% (p = 0.02) using magnetic resonance imaging. The methods correlated significantly (R = 0.92, p < 0.01). Tissue Doppler echocardiography showed increased systolic displacement (31%, p < 0.01) and increased systolic velocity (29%, p = 0.01) in coversin treated pigs. Interleukin-1β in myocardial microdialysis fluid was significantly reduced (31%, p < 0.05) and tissue E-selectin expression was significantly reduced (p = 0.01) in the non-infarcted area at risk by coversin treatment. Coversin ablated plasma C5 activation throughout the reperfusion period and decreased myocardial C5b-9 deposition, while neither plasma nor myocardial LTB4 were significantly reduced. Coversin substantially reduced the size of infarction, improved ventricular function, and attenuated interleukin-1β and E-selectin in this porcine model by inhibiting C5. We conclude that inhibition of C5 in myocardial infarction should be reconsidered.
Collapse
Affiliation(s)
- Soeren E Pischke
- Department of Immunology, Oslo University Hospital, Rikshospitalet, P.b. 4950 Nydalen, 0424, Oslo, Norway.
- K.G. Jebsen IRC, University of Oslo, Oslo, Norway.
- Intervention Centre, Oslo University Hospital, Oslo, Norway.
- Division of Emergencies and Critical Care, Department of Anaesthesiology, Oslo University Hospital, Oslo, Norway.
| | - A Gustavsen
- Department of Immunology, Oslo University Hospital, Rikshospitalet, P.b. 4950 Nydalen, 0424, Oslo, Norway
- K.G. Jebsen IRC, University of Oslo, Oslo, Norway
| | - H L Orrem
- Department of Immunology, Oslo University Hospital, Rikshospitalet, P.b. 4950 Nydalen, 0424, Oslo, Norway
- K.G. Jebsen IRC, University of Oslo, Oslo, Norway
- Division of Emergencies and Critical Care, Department of Anaesthesiology, Oslo University Hospital, Oslo, Norway
| | - K H Egge
- Department of Immunology, Oslo University Hospital, Rikshospitalet, P.b. 4950 Nydalen, 0424, Oslo, Norway
- K.G. Jebsen IRC, University of Oslo, Oslo, Norway
| | - F Courivaud
- Intervention Centre, Oslo University Hospital, Oslo, Norway
| | - H Fontenelle
- Intervention Centre, Oslo University Hospital, Oslo, Norway
| | - A Despont
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - A K Bongoni
- Immunology Research Centre, St. Vincent's Hospital, Melbourne, VIC, Australia
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - R Rieben
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - T I Tønnessen
- Division of Emergencies and Critical Care, Department of Anaesthesiology, Oslo University Hospital, Oslo, Norway
| | - M A Nunn
- Akari Therapeutics Plc, London, UK
| | - H Scott
- Department of Pathology, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - H Skulstad
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, University of Oslo, Oslo, Norway
| | - A Barratt-Due
- Department of Immunology, Oslo University Hospital, Rikshospitalet, P.b. 4950 Nydalen, 0424, Oslo, Norway
- K.G. Jebsen IRC, University of Oslo, Oslo, Norway
- Division of Emergencies and Critical Care, Department of Anaesthesiology, Oslo University Hospital, Oslo, Norway
| | - T E Mollnes
- Department of Immunology, Oslo University Hospital, Rikshospitalet, P.b. 4950 Nydalen, 0424, Oslo, Norway
- K.G. Jebsen IRC, University of Oslo, Oslo, Norway
- Research Laboratory, Nordland Hospital, Bodø, Norway
- Faculty of Health Sciences, K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
22
|
Kuhn N, Schmidt CQ, Schlapschy M, Skerra A. PASylated Coversin, a C5-Specific Complement Inhibitor with Extended Pharmacokinetics, Shows Enhanced Anti-Hemolytic Activity in Vitro. Bioconjug Chem 2016; 27:2359-2371. [PMID: 27598771 DOI: 10.1021/acs.bioconjchem.6b00369] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The Ornithodoros moubata Complement Inhibitor (OmCI) binds complement component 5 (C5) with high affinity and, thus, selectively prevents proteolytic activation of the terminal lytic complement pathway. A recombinant version of OmCI (also known as Coversin and rEV576) has proven efficacious in several animal models of complement-mediated diseases and successfully completed a phase Ia clinical trial. Coversin is a small 17 kDa lipocalin protein which has a very short plasma half-life if not bound to C5; therefore, the drug requires frequent dosing. We have improved the pharmacokinetics of Coversin by N-terminal translational conjugation with a 600 residue polypeptide composed of Pro, Ala, and Ser (PAS) residues. To this end, PAS-Coversin as well as the unmodified Coversin were functionally expressed in the cytoplasm of E. coli and purified to homogeneity. Both versions showed identical affinity to human C5, as determined by surface plasmon resonance measurements, and revealed similar complement inhibitory activity, as measured in ELISAs with human serum. In line with the PEG-like biophysical properties, PASylation dramatically prolonged the plasma half-life of uncomplexed Coversin by a factor ≥50 in mice. In a clinically relevant in vitro model of the complement-mediated disease paroxysmal nocturnal hemoglobinuria (PNH) both versions of Coversin effectively reduced erythrocyte lysis. Unexpectedly, while the IC50 values were comparable, PAS-Coversin reached a substantially lower plateau of residual lysis at saturating inhibitor concentrations. Taken together, our data demonstrate two clinically relevant improvements of PASylated Coversin: markedly increased plasma half-life and considerably reduced background hemolysis of erythrocytes with PNH-induced phenotype.
Collapse
Affiliation(s)
- Nadine Kuhn
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, Technische Universität München , Emil-Erlenmeyer-Forum 5, 85354 Freising (Weihenstephan), Germany
| | - Christoph Q Schmidt
- Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University , Helmholtzstrasse 20, 89081 Ulm, Germany
| | - Martin Schlapschy
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, Technische Universität München , Emil-Erlenmeyer-Forum 5, 85354 Freising (Weihenstephan), Germany.,XL-protein GmbH , Lise-Meitner-Strasse 30, 85354 Freising, Germany
| | - Arne Skerra
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, Technische Universität München , Emil-Erlenmeyer-Forum 5, 85354 Freising (Weihenstephan), Germany.,XL-protein GmbH , Lise-Meitner-Strasse 30, 85354 Freising, Germany
| |
Collapse
|
23
|
Manzano-Román R, Díaz-Martín V, Oleaga A, Obolo-Mvoulouga P, Pérez-Sánchez R. TSGP4 from Ornithodoros moubata: molecular cloning, phylogenetic analysis and vaccine efficacy of a new member of the lipocalin clade of cysteinyl leukotriene scavengers. Vet Parasitol 2016; 227:130-7. [PMID: 27523949 DOI: 10.1016/j.vetpar.2016.08.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/01/2016] [Accepted: 08/03/2016] [Indexed: 01/05/2023]
Abstract
Recently obtained evidence indicated that an orthologue of the O. savignyi TSGP4 salivary lipocalin was present in the saliva of O. moubata. TSGP4 is known to act as a cysteinyl leukotrienes scavenger helping in the prevention of inflammation and oedema at the tick bite site. Since this function seems to be crucial for successful tick feeding, the novel O. moubata TSGP4 turned into a potential vaccine target. The purposes of the current work were: (i) to clone and characterize the O. moubata TSGP4 and, (ii) to produce it as recombinant to evaluate its protective efficacy as vaccine antigen. The results of these experiments indicated that the O. moubata TSGP4 shows high sequence and structural identity with the O. savignyi orthologue suggesting identical function in the physiology of the tick-host relationship. The mature native TSGP4 is not immunogenic when it is inoculated to host with tick saliva during feeding, but host vaccination with the recombinant protein TSGP4 in Freund's adjuvants induced strong humoral immune responses that recognized both the recombinant and native TSGP4 and protected the host with a 14.1% efficacy. So, the O. moubata TSGP4 can be considered a silent salivary antigen; however, in the light of the current results, its inclusion in the current repertory of protective antigens to be targeted by anti-tick vaccines could be controversial.
Collapse
Affiliation(s)
- R Manzano-Román
- Parasitología Animal, Instituto de Recursos Naturales y Agrobiología de Salamanca (IRNASA, CSIC), Cordel de Merinas, 40-52, 37008 Salamanca, Spain.
| | - V Díaz-Martín
- Parasitología Animal, Instituto de Recursos Naturales y Agrobiología de Salamanca (IRNASA, CSIC), Cordel de Merinas, 40-52, 37008 Salamanca, Spain.
| | - A Oleaga
- Parasitología Animal, Instituto de Recursos Naturales y Agrobiología de Salamanca (IRNASA, CSIC), Cordel de Merinas, 40-52, 37008 Salamanca, Spain.
| | - P Obolo-Mvoulouga
- Parasitología Animal, Instituto de Recursos Naturales y Agrobiología de Salamanca (IRNASA, CSIC), Cordel de Merinas, 40-52, 37008 Salamanca, Spain.
| | - R Pérez-Sánchez
- Parasitología Animal, Instituto de Recursos Naturales y Agrobiología de Salamanca (IRNASA, CSIC), Cordel de Merinas, 40-52, 37008 Salamanca, Spain.
| |
Collapse
|
24
|
Jablonka W, Pham V, Nardone G, Gittis A, Silva-Cardoso L, Atella GC, Ribeiro JM, Andersen JF. Structure and Ligand-Binding Mechanism of a Cysteinyl Leukotriene-Binding Protein from a Blood-Feeding Disease Vector. ACS Chem Biol 2016; 11:1934-44. [PMID: 27124118 DOI: 10.1021/acschembio.6b00032] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Blood-feeding disease vectors mitigate the negative effects of hemostasis and inflammation through the binding of small-molecule agonists of these processes by salivary proteins. In this study, a lipocalin protein family member (LTBP1) from the saliva of Rhodnius prolixus, a vector of the pathogen Trypanosoma cruzi, is shown to sequester cysteinyl leukotrienes during feeding to inhibit immediate inflammatory responses. Calorimetric binding experiments showed that LTBP1 binds leukotrienes C4 (LTC4), D4 (LTD4), and E4 (LTE4) but not biogenic amines, adenosine diphosphate, or other eicosanoid compounds. Crystal structures of ligand-free LTBP1 and its complexes with LTC4 and LTD4 reveal a conformational change during binding that brings Tyr114 into close contact with the ligand. LTC4 is cleaved in the complex, leaving free glutathione and a C20 fatty acid. Chromatographic analysis of bound ligands showed only intact LTC4, suggesting that cleavage could be radiation-mediated.
Collapse
Affiliation(s)
- Willy Jablonka
- Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland 20852, United States
| | - Van Pham
- Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland 20852, United States
| | - Glenn Nardone
- Research Technologies Branch, NIAID, National Institutes of Health, Rockville, Maryland 20852, United States
| | - Apostolos Gittis
- Research Technologies Branch, NIAID, National Institutes of Health, Rockville, Maryland 20852, United States
| | - Lívia Silva-Cardoso
- Instituto de Bioquímica Médica
Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Georgia C. Atella
- Instituto de Bioquímica Médica
Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - José M.C. Ribeiro
- Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland 20852, United States
| | - John F. Andersen
- Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland 20852, United States
| |
Collapse
|
25
|
Desoubeaux G, Reichert JM, Sleeman M, Reckamp KL, Ryffel B, Adamczewski JP, Sweeney TD, Vanbever R, Diot P, Owen CA, Page C, Lerondel S, Le Pape A, Heuze-Vourc'h N. Therapeutic monoclonal antibodies for respiratory diseases: Current challenges and perspectives, March 31 - April 1, 2016, Tours, France. MAbs 2016; 8:999-1009. [PMID: 27266390 PMCID: PMC4968091 DOI: 10.1080/19420862.2016.1196521] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Monoclonal antibody (mAb) therapeutics have tremendous potential to benefit patients with lung diseases, for which there remains substantial unmet medical need. To capture the current state of mAb research and development in the area of respiratory diseases, the Research Center of Respiratory Diseases (CEPR-INSERM U1100), the Laboratory of Excellence “MAbImprove,” the GDR 3260 “Antibodies and therapeutic targeting,” and the Grant Research program ARD2020 “Biotherapeutics” invited speakers from industry, academic and government organizations to present their recent research results at the Therapeutic Monoclonal Antibodies for Respiratory Diseases: Current challenges and perspectives congress held March 31 – April 1, 2016 in Tours, France.
Collapse
Affiliation(s)
- Guillaume Desoubeaux
- a Université François-Rabelais , Tours , France.,b INSERM, Center d'Etude des Pathologies Respiratoires , Tours , France.,c Centre Hospitalo-Universitaire de Tours , Tours , France
| | - Janice M Reichert
- d The Antibody Society , Framingham , MA , USA.,e Reichert Biotechnology Consulting LLC , Framingham MA , USA
| | | | - Karen L Reckamp
- g City of Hope, Comprehensive Cancer Center , Duarte , CA , USA
| | - Bernhard Ryffel
- h Université d'Orléans , Orléans , France.,i University of Cape Town, Institute of Infectious Disease and Molecular Medicine (IDM) , Cape Town , South Africa
| | | | | | - Rita Vanbever
- l Université Catholique de Louvain, Louvain Drug Research Institute , Brussels , Belgium
| | - Patrice Diot
- a Université François-Rabelais , Tours , France.,b INSERM, Center d'Etude des Pathologies Respiratoires , Tours , France.,c Centre Hospitalo-Universitaire de Tours , Tours , France
| | - Caroline A Owen
- m Harvard Medical School, Brigham and Women's Hospital , Boston , MA , USA.,n Lovelace Respiratory Research Institute , Albuquerque , NM , USA
| | - Clive Page
- o King's College, Sackler Institute of Pulmonary Pharmacology , London , UK
| | | | - Alain Le Pape
- a Université François-Rabelais , Tours , France.,b INSERM, Center d'Etude des Pathologies Respiratoires , Tours , France.,p PHENOMIN-TAAM CNRS, CIPA , Orléans , France
| | - Nathalie Heuze-Vourc'h
- a Université François-Rabelais , Tours , France.,b INSERM, Center d'Etude des Pathologies Respiratoires , Tours , France
| |
Collapse
|
26
|
Mans BJ, de Castro MH, Pienaar R, de Klerk D, Gaven P, Genu S, Latif AA. Ancestral reconstruction of tick lineages. Ticks Tick Borne Dis 2016; 7:509-35. [DOI: 10.1016/j.ttbdis.2016.02.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/26/2016] [Accepted: 02/02/2016] [Indexed: 01/15/2023]
|
27
|
Jore MM, Johnson S, Sheppard D, Barber NM, Li YI, Nunn MA, Elmlund H, Lea SM. Structural basis for therapeutic inhibition of complement C5. Nat Struct Mol Biol 2016; 23:378-86. [PMID: 27018802 PMCID: PMC5771465 DOI: 10.1038/nsmb.3196] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/02/2016] [Indexed: 01/03/2023]
Abstract
Activation of complement C5 generates the potent anaphylatoxin C5a and leads to pathogen lysis, inflammation and cell damage. The therapeutic potential of C5 inhibition has been demonstrated by eculizumab, one of the world's most expensive drugs. However, the mechanism of C5 activation by C5 convertases remains elusive, thus limiting development of therapeutics. Here we identify and characterize a new protein family of tick-derived C5 inhibitors. Structures of C5 in complex with the new inhibitors, the phase I and phase II inhibitor OmCI, or an eculizumab Fab reveal three distinct binding sites on C5 that all prevent activation of C5. The positions of the inhibitor-binding sites and the ability of all three C5-inhibitor complexes to competitively inhibit the C5 convertase conflict with earlier steric-inhibition models, thus suggesting that a priming event is needed for activation.
Collapse
Affiliation(s)
- Matthijs M Jore
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Steven Johnson
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Devon Sheppard
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Natalie M Barber
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Yang I Li
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Miles A Nunn
- Centre for Ecology and Hydrology, Wallingford, UK
| | - Hans Elmlund
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, Victoria, Australia
| | - Susan M Lea
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
28
|
Skjeflo EW, Sagatun C, Dybwik K, Aam S, Urving SH, Nunn MA, Fure H, Lau C, Brekke OL, Huber-Lang M, Espevik T, Barratt-Due A, Nielsen EW, Mollnes TE. Combined inhibition of complement and CD14 improved outcome in porcine polymicrobial sepsis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2015; 19:415. [PMID: 26612199 PMCID: PMC4662001 DOI: 10.1186/s13054-015-1129-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 11/04/2015] [Indexed: 01/16/2023]
Abstract
Introduction Sepsis is an exaggerated and dysfunctional immune response to infection. Activation of innate immunity recognition systems including complement and the Toll-like receptor family initiate this disproportionate inflammatory response. The aim of this study was to explore the effect of combined inhibition of the complement component C5 and the Toll-like receptor co-factor CD14 on survival, hemodynamic parameters and systemic inflammation including complement activation in a clinically relevant porcine model of polymicrobial sepsis. Methods Norwegian landrace piglets (4 ± 0.5 kg) were blindly randomized to a treatment group (n = 12) receiving the C5 inhibitor coversin (OmCI) and anti-CD14 or to a positive control group (n = 12) receiving saline. Under anesthesia, sepsis was induced by a 2 cm cecal incision and the piglets were monitored in standard intensive care for 8 hours. Three sham piglets had a laparotomy without cecal incision or treatment. Complement activation was measured as sC5b-9 using enzyme immunoassay. Cytokines were measured with multiplex technology. Results Combined C5 and CD14 inhibition significantly improved survival (p = 0.03). Nine piglets survived in the treatment group and four in the control group. The treatment group had significantly lower pulmonary artery pressure (p = 0.04) and ratio of pulmonary artery pressure to systemic artery pressure (p < 0.001). Plasma sC5b-9 levels were significantly lower in the treatment group (p < 0.001) and correlated significantly with mortality (p = 0.006). IL-8 and IL-10 were significantly (p < 0.05) lower in the treatment group. Conclusions Combined inhibition of C5 and CD14 significantly improved survival, hemodynamic parameters and inflammation in a blinded, randomized trial of porcine polymicrobial sepsis.
Collapse
Affiliation(s)
- Espen W Skjeflo
- Research Laboratory, Nordland Hospital, Prinsens Gate 164, 8092, Bodø, Norway. .,Faculty of Health Sciences, K. G. Jebsen TREC, University of Tromsø, 9037, Tromsø, Norway.
| | - Caroline Sagatun
- Department of Surgery, Nordland Hospital, Prinsens Gate 164, 8092, Bodø, Norway.
| | - Knut Dybwik
- Department of Anestesiology, Nordland Hospital, Prinsens Gate 164, 8092, Bodø, Norway. .,Faculty of Professional Studies, University of Nordland, Universitetsaleen 11, 8049, Bodø, Norway.
| | - Sturla Aam
- Faculty of Medicine, Ludwig Maximillian University, Professor Huber Platz 2, 80539, Munich, Germany.
| | - Sven H Urving
- Department of Anestesiology, Nordland Hospital, Prinsens Gate 164, 8092, Bodø, Norway.
| | - Miles A Nunn
- Volution Immuno Pharmaceuticals Limited, 5 Argosy Court, Whitley Business Park, Coventry, CV3 4GA, UK.
| | - Hilde Fure
- Research Laboratory, Nordland Hospital, Prinsens Gate 164, 8092, Bodø, Norway.
| | - Corinna Lau
- Research Laboratory, Nordland Hospital, Prinsens Gate 164, 8092, Bodø, Norway.
| | - Ole-Lars Brekke
- Research Laboratory, Nordland Hospital, Prinsens Gate 164, 8092, Bodø, Norway. .,Faculty of Health Sciences, K. G. Jebsen TREC, University of Tromsø, 9037, Tromsø, Norway.
| | - Markus Huber-Lang
- Department of Traumatology, Center of Surgery, University of Ulm, Albert Einstein Allee 23, 89081, Ulm, Germany.
| | - Terje Espevik
- Centre of Molecular Inflammation Research, and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Høgskoleringen 1, 7491, Trondheim, Norway.
| | - Andreas Barratt-Due
- Department of Immunology, Oslo University Hospital and K.G. Jebsen IRC, University of Oslo, PB 4960 Nydalen, 0424, Oslo, Norway. .,Division of Emergencies and Critical Care, Rikshospitalet, Oslo University Hospital Oslo, Sognsvannsveien 20, 0372, Oslo, Norway.
| | - Erik W Nielsen
- Faculty of Health Sciences, K. G. Jebsen TREC, University of Tromsø, 9037, Tromsø, Norway. .,Department of Anestesiology, Nordland Hospital, Prinsens Gate 164, 8092, Bodø, Norway. .,Faculty of Professional Studies, University of Nordland, Universitetsaleen 11, 8049, Bodø, Norway. .,Department of Immunology, Oslo University Hospital and K.G. Jebsen IRC, University of Oslo, PB 4960 Nydalen, 0424, Oslo, Norway.
| | - Tom E Mollnes
- Research Laboratory, Nordland Hospital, Prinsens Gate 164, 8092, Bodø, Norway. .,Faculty of Health Sciences, K. G. Jebsen TREC, University of Tromsø, 9037, Tromsø, Norway. .,Centre of Molecular Inflammation Research, and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Høgskoleringen 1, 7491, Trondheim, Norway. .,Department of Immunology, Oslo University Hospital and K.G. Jebsen IRC, University of Oslo, PB 4960 Nydalen, 0424, Oslo, Norway.
| |
Collapse
|
29
|
O’Dwyer CA, O’Brien ME, Wormald MR, White MM, Banville N, Hurley K, McCarthy C, McElvaney NG, Reeves EP. The BLT1 Inhibitory Function of α-1 Antitrypsin Augmentation Therapy Disrupts Leukotriene B4Neutrophil Signaling. THE JOURNAL OF IMMUNOLOGY 2015; 195:3628-41. [DOI: 10.4049/jimmunol.1500038] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 08/12/2015] [Indexed: 12/15/2022]
|
30
|
Tambourgi DV, van den Berg CW. Animal venoms/toxins and the complement system. Mol Immunol 2014; 61:153-62. [PMID: 24998802 DOI: 10.1016/j.molimm.2014.06.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 06/10/2014] [Accepted: 06/10/2014] [Indexed: 12/22/2022]
Abstract
Nature is a wealthy source of agents that have been shown to be beneficial to human health, but nature is also a rich source of potential dangerous health damaging compounds. This review will summarise and discuss the agents from the animal kingdom that have been shown to interact with the human complement (C) system. Most of these agents are toxins found in animal venoms and animal secretions. In addition to the mechanism of action of these toxins, their contribution to the field of complement, their role in human pathology and the potential benefit to the venomous animal itself will be discussed. Potential therapeutic applications will also be discussed.
Collapse
Affiliation(s)
| | - Carmen W van den Berg
- Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Cardiff, UK.
| |
Collapse
|
31
|
Abstract
INTRODUCTION As an ecological adaptation venoms have evolved independently in several species of Metazoa. As haematophagous arthropods ticks are mainly considered as ectoparasites due to directly feeding on the skin of animal hosts. Ticks are of major importance since they serve as vectors for several diseases affecting humans and livestock animals. Ticks are rarely considered as venomous animals despite that tick saliva contains several protein families present in venomous taxa and that many Ixodida genera can induce paralysis and other types of toxicoses. Tick saliva was previously proposed as a special kind of venom since tick venom is used for blood feeding that counteracts host defense mechanisms. As a result, the present study provides evidence to reconsider the venomous properties of tick saliva. RESULTS Based on our extensive literature mining and in silico research, we demonstrate that ticks share several similarities with other venomous taxa. Many tick salivary protein families and their previously described functions are homologous to proteins found in scorpion, spider, snake, platypus and bee venoms. This infers that there is a structural and functional convergence between several molecular components in tick saliva and the venoms from other recognized venomous taxa. We also highlight the fact that the immune response against tick saliva and venoms (from recognized venomous taxa) are both dominated by an allergic immunity background. Furthermore, by comparing the major molecular components of human saliva, as an example of a non-venomous animal, with that of ticks we find evidence that ticks resemble more venomous than non-venomous animals. Finally, we introduce our considerations regarding the evolution of venoms in Arachnida. CONCLUSIONS Taking into account the composition of tick saliva, the venomous functions that ticks have while interacting with their hosts, and the distinguishable differences between human (non-venomous) and tick salivary proteins, we consider that ticks should be referred to as venomous ectoparasites.
Collapse
Affiliation(s)
- Alejandro Cabezas-Cruz
- Center for Infection and Immunity of Lille (CIIL), INSERM U1019 – CNRS UMR 8204, Université Lille Nord de France, Institut Pasteur de Lille, Lille, France
- SaBio. Instituto de Investigación de Recursos Cinegéticos, IREC-CSIC-UCLM-JCCM, Ciudad Real 13005, Spain
| | - James J Valdés
- Institute of Parasitology, Biology Centre of the Academy of Sciences of the Czech Republic, České Budějovice, 37005, Czech Republic
| |
Collapse
|
32
|
Barratt-Due A, Thorgersen EB, Egge K, Pischke S, Sokolov A, Hellerud BC, Lindstad JK, Pharo A, Bongoni AK, Rieben R, Nunn M, Scott H, Mollnes TE. Combined inhibition of complement C5 and CD14 markedly attenuates inflammation, thrombogenicity, and hemodynamic changes in porcine sepsis. THE JOURNAL OF IMMUNOLOGY 2013; 191:819-27. [PMID: 23761634 DOI: 10.4049/jimmunol.1201909] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Complement and the TLR family constitute two important branches of innate immunity. We previously showed attenuating effects on inflammation and thromogenicity by inhibiting the TLR coreceptor CD14 in porcine sepsis. In the present study, we explored the effect of the C5 and leukotriene B4 inhibitor Ornithodoros moubata complement inhibitor (OmCI; also known as coversin) alone and combined with anti-CD14 on the early inflammatory, hemostatic, and hemodynamic responses in porcine Escherichia coli-induced sepsis. Pigs were randomly allocated to negative controls (n = 6), positive controls (n = 8), intervention with OmCI (n = 8), or with OmCI and anti-CD14 (n = 8). OmCI ablated C5 activation and formation of the terminal complement complex and significantly decreased leukotriene B4 levels in septic pigs. Granulocyte tissue factor expression, formation of thrombin-antithrombin complexes (p < 0.001), and formation of TNF-α and IL-6 (p < 0.05) were efficiently inhibited by OmCI alone and abolished or strongly attenuated by the combination of OmCI and anti-CD14 (p < 0.001 for all). Additionally, the combined therapy attenuated the formation of plasminogen activator inhibitor-1 (p < 0.05), IL-1β, and IL-8, increased the formation of IL-10, and abolished the expression of wCD11R3 (CD11b) and the fall in neutrophil cell count (p < 0.001 for all). Finally, OmCI combined with anti-CD14 delayed increases in heart rate by 60 min (p < 0.05) and mean pulmonary artery pressure by 30 min (p < 0.01). Ex vivo studies confirmed the additional effect of combining anti-CD14 with OmCI. In conclusion, upstream inhibition of the key innate immunity molecules, C5 and CD14, is a potential broad-acting treatment regimen in sepsis as it efficiently attenuated inflammation and thrombogenicity and delayed hemodynamic changes.
Collapse
Affiliation(s)
- Andreas Barratt-Due
- Department of Immunology, Oslo University Hospital, National Hospital, University of Oslo, 0424 Oslo, Norway.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|