Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Pathol ; 191(2): 320-334, 2021 02.
Article in English | MEDLINE | ID: mdl-33159884

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.


Subject(s)
Leukotriene B4/metabolism , Receptors, Leukotriene B4/metabolism , Retina/metabolism , Uveitis/immunology , Uveitis/metabolism , Animals , Biological Products/pharmacology , Complement C5a/antagonists & inhibitors , Complement C5a/metabolism , Disease Models, Animal , Female , Humans , Leukotriene B4/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Receptors, Leukotriene B4/antagonists & inhibitors , Retina/immunology , Th17 Cells/immunology
2.
Nat Immunol ; 9(7): 753-60, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18536718

ABSTRACT

To provide insight into the structural and functional properties of human complement component 5 (C5), we determined its crystal structure at a resolution of 3.1 A. The core of C5 adopted a structure resembling that of C3, with the domain arrangement at the position corresponding to the C3 thioester being very well conserved. However, in contrast to C3, the convertase cleavage site in C5 was ordered and the C345C domain flexibly attached to the core of C5. Binding of the tick C5 inhibitor OmCI to C5 resulted in stabilization of the global conformation of C5 but did not block the convertase cleavage site. The structure of C5 may render possible a structure-based approach for the design of new selective complement inhibitors.


Subject(s)
Complement C5/chemistry , Complement C5/metabolism , Insect Proteins/metabolism , Protein Structure, Quaternary , Animals , Arthropod Proteins , Binding Sites , Carrier Proteins , Complement C3 , Crystallography, X-Ray , Humans , Insect Proteins/chemistry , Surface Plasmon Resonance
4.
J Immunol ; 192(11): 5324-31, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24790148

ABSTRACT

Sepsis and septic shock, caused by an excessive systemic host-inflammatory response, are associated with high morbidity and mortality. The complement system and TLRs provide important pattern recognition receptors initiating the cytokine storm by extensive cross-talk. We hypothesized that double blockade of complement C5 and the TLR coreceptor CD14 could improve survival of experimental polymicrobial sepsis. Mice undergoing cecal ligation and puncture (CLP)-induced sepsis were treated with neutralizing anti-CD14 Ab biG 53, complement C5 inhibitor coversin (Ornithodoros moubata C inhibitor), or a combination thereof. The inflammatory study (24-h observation) revealed statistically significant increases in 22 of 24 measured plasma biomarkers in the untreated CLP group, comprising 14 pro- and anti-inflammatory cytokines and 8 chemokines, growth factors, and granulocyte activation markers. Single CD14 or C5 blockade significantly inhibited 20 and 19 of the 22 biomarkers, respectively. Combined CD14 and C5 inhibition significantly reduced all 22 biomarkers (mean reduction 85%; range 54-95%) compared with the untreated CLP group. Double blockade was more potent than single treatment and was required to significantly inhibit IL-6 and CXCL1. Combined inhibition significantly reduced morbidity (motility and eyelid movement) and mortality measured over 10 d. In the positive control CLP group, median survival was 36 h (range 24-48 h). Combined treatment increased median survival to 96 h (range 24-240 h) (p = 0.001), whereas survival in the single-treatment groups was not significantly increased (median and range for anti-CD14 and anti-C5 treatment were 36 h [24-48 h] and 48 h [24-96 h]). Combined with standard intervention therapy, specific blockade of CD14 and C5 might represent a promising new therapeutic strategy for treatment of polymicrobial sepsis.


Subject(s)
Antibodies, Neutralizing/pharmacology , Complement C5/antagonists & inhibitors , Complement C5/immunology , Cytokines/immunology , Lipopolysaccharide Receptors/immunology , Sepsis/immunology , Animals , Antibodies, Neutralizing/immunology , Male , Mice , Sepsis/drug therapy , Sepsis/microbiology , Time Factors
6.
PLoS Pathog ; 9(6): e1003450, 2013.
Article in English | MEDLINE | ID: mdl-23825947

ABSTRACT

Hard ticks subvert the immune responses of their vertebrate hosts in order to feed for much longer periods than other blood-feeding ectoparasites; this may be one reason why they transmit perhaps the greatest diversity of pathogens of any arthropod vector. Tick-induced immunomodulation is mediated by salivary components, some of which neutralise elements of innate immunity or inhibit the development of adaptive immunity. As dendritic cells (DC) trigger and help to regulate adaptive immunity, they are an ideal target for immunomodulation. However, previously described immunoactive components of tick saliva are either highly promiscuous in their cellular and molecular targets or have limited effects on DC. Here we address the question of whether the largest and globally most important group of ticks (the ixodid metastriates) produce salivary molecules that specifically modulate DC activity. We used chromatography to isolate a salivary gland protein (Japanin) from Rhipicephalus appendiculatus ticks. Japanin was cloned, and recombinant protein was produced in a baculoviral expression system. We found that Japanin specifically reprogrammes DC responses to a wide variety of stimuli in vitro, radically altering their expression of co-stimulatory and co-inhibitory transmembrane molecules (measured by flow cytometry) and their secretion of pro-inflammatory, anti-inflammatory and T cell polarising cytokines (assessed by Luminex multiplex assays); it also inhibits the differentiation of DC from monocytes. Sequence alignments and enzymatic deglycosylation revealed Japanin to be a 17.7 kDa, N-glycosylated lipocalin. Using molecular cloning and database searches, we have identified a group of homologous proteins in R. appendiculatus and related species, three of which we have expressed and shown to possess DC-modulatory activity. All data were obtained using DC generated from at least four human blood donors, with rigorous statistical analysis. Our results suggest a previously unknown mechanism for parasite-induced subversion of adaptive immunity, one which may also facilitate pathogen transmission.


Subject(s)
Arthropod Proteins/immunology , Dendritic Cells/immunology , Immunologic Factors/immunology , Monocytes/immunology , Rhipicephalus/immunology , Salivary Proteins and Peptides/immunology , Adaptive Immunity/drug effects , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Animals , Arthropod Proteins/genetics , Arthropod Proteins/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Immunologic Factors/genetics , Immunologic Factors/pharmacology , Monocytes/pathology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Rhipicephalus/genetics , Salivary Proteins and Peptides/genetics , Salivary Proteins and Peptides/pharmacology
7.
J Immunol ; 191(2): 819-27, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23761634

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-1ß, 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.


Subject(s)
Arthropod Proteins/pharmacology , Carrier Proteins/pharmacology , Complement C5/antagonists & inhibitors , Leukotriene B4/antagonists & inhibitors , Lipopolysaccharide Receptors/immunology , Sepsis/immunology , Animals , Antithrombin III/biosynthesis , Arterial Pressure/drug effects , Arterial Pressure/immunology , CD11b Antigen/biosynthesis , Escherichia coli/immunology , Escherichia coli Infections/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Heart Rate/drug effects , Heart Rate/immunology , Hemodynamics/drug effects , Immunity, Innate , Inflammation/drug therapy , Inflammation/immunology , Interleukin-10/biosynthesis , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , Leukocyte Count , Lipopolysaccharide Receptors/metabolism , Neutrophils/cytology , Peptide Hydrolases/biosynthesis , Plasminogen Activator Inhibitor 1/biosynthesis , Sus scrofa , Tumor Necrosis Factor-alpha/biosynthesis
8.
Crit Care ; 19: 415, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26612199

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.


Subject(s)
Complement C5/antagonists & inhibitors , Lipopolysaccharide Receptors/metabolism , Sepsis/drug therapy , Toll-Like Receptors/immunology , Animals , Inflammation/blood , Inflammation/mortality , Sepsis/metabolism , Sepsis/microbiology , Sepsis/mortality , Swine , Toll-Like Receptors/metabolism
9.
J Biol Chem ; 288(26): 18789-802, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23625922

ABSTRACT

Molecules that simultaneously inhibit independent or co-dependent proinflammatory pathways may have advantages over conventional monotherapeutics. OmCI is a bifunctional protein derived from blood-feeding ticks that specifically prevents complement (C)-mediated C5 activation and also sequesters leukotriene B4 (LTB4) within an internal binding pocket. Here, we examined the effect of LTB4 binding on OmCI structure and function and investigated the relative importance of C-mediated C5 activation and LTB4 in a mouse model of immune complex-induced acute lung injury (IC-ALI). We describe two crystal structures of bacterially expressed OmCI: one binding a C16 fatty acid and the other binding LTB4 (C20). We show that the C5 and LTB4 binding activities of the molecule are independent of each other and that OmCI is a potent inhibitor of experimental IC-ALI, equally dependent on both C5 inhibition and LTB4 binding for full activity. The data highlight the importance of LTB4 in IC-ALI and activation of C5 by the complement pathway C5 convertase rather than by non-C proteases. The findings suggest that dual inhibition of C5 and LTB4 may be useful for treatment of human immune complex-dependent diseases.


Subject(s)
Acute Lung Injury/metabolism , Antigen-Antibody Complex/pharmacology , Arthropod Proteins/pharmacology , Carrier Proteins/pharmacology , Lipocalins/pharmacology , Acute Lung Injury/immunology , Acute Lung Injury/therapy , Animals , Arthropod Proteins/metabolism , Carrier Proteins/metabolism , Chromatography, Gas , Complement C5/metabolism , Eicosanoids/metabolism , Fatty Acids/metabolism , Immunoenzyme Techniques , Leukotriene B4/metabolism , Lipocalins/metabolism , Male , Mice , Mice, Inbred C57BL , Recombinant Proteins/metabolism , Sheep , Surface Plasmon Resonance , Thrombin/metabolism
10.
Parasit Vectors ; 17(1): 201, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711091

ABSTRACT

PURPOSE: The rising burden of mosquito-borne diseases in Europe extends beyond urban areas, encompassing rural and semi-urban regions near managed and natural wetlands evidenced by recent outbreaks of Usutu and West Nile viruses. While wetland management policies focus on biodiversity and ecosystem services, few studies explore the impact on mosquito vectors. METHODS: Our research addresses this gap, examining juvenile mosquito and aquatic predator communities in 67 ditch sites within a South England coastal marsh subjected to different wetland management tiers. Using joint distribution models, we analyse how mosquito communities respond to abiotic and biotic factors influenced by wetland management. RESULTS: Of the 12 mosquito species identified, Culiseta annulata (Usutu virus vector) and Culex pipiens (Usutu and West Nile virus vector) constitute 47% of 6825 larval mosquitoes. Abundant predators include Coleoptera (water beetles) adults, Corixidae (water boatmen) and Zygoptera (Damselfy) larvae. Models reveal that tier 3 management sites (higher winter water levels, lower agricultural intensity) associated with shade and less floating vegetation are preferred by specific mosquito species. All mosquito species except Anopheles maculipennis s.l., are negatively impacted by potential predators. Culiseta annulata shows positive associations with shaded and turbid water, contrary to preferences of Corixidae predators. CONCLUSIONS: Tier 3 areas managed for biodiversity, characterised by higher seasonal water levels and reduced livestock grazing intensity, provide favourable habitats for key mosquito species that are known vectors of arboviruses, such as Usutu and West Nile. Our findings emphasise the impact of biodiversity-focused wetland management, altering mosquito breeding site vegetation to enhance vector suitability. Further exploration of these trade-offs is crucial for comprehending the broader implications of wetland management.


Subject(s)
Biodiversity , Culicidae , Mosquito Vectors , Wetlands , Animals , Mosquito Vectors/physiology , Mosquito Vectors/virology , Culicidae/classification , Culicidae/physiology , Culicidae/virology , Ecosystem , Larva/physiology , Seasons , United Kingdom , Culex/physiology , Culex/virology , Culex/classification , England
11.
J Immunol ; 187(9): 4913-9, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21964028

ABSTRACT

Experimental evidence suggests that C inhibition and more particularly combined inhibition of C and the TLR coreceptor CD14 may be of therapeutic benefit in sepsis and other inflammatory conditions. A barrier to the testing and further development of many inhibitors is that their activity is species specific. Pig is a relevant species for experimental models of human disease, and this study undertakes a comprehensive comparison of the inhibitory efficacy of the C5 inhibitor Ornithodoros moubata C inhibitor (OmCI) in human and porcine whole blood ex vivo models of Escherichia coli-induced sepsis. The effect of OmCI on complement activity in pigs undergoing E. coli sepsis was also examined. Porcine and human serum, and whole blood anticoagulated with lepirudin, was incubated with E. coli and the effect of OmCI investigated. The ex vivo results were virtually identical in pig and human. OmCI completely ablated the activity of all three C pathways at 0.64 µM. E. coli-induced C activation and expression of CD11b (wCD11R3 in the pig), was abolished ex vivo at 0.32 µM OmCI. Combining anti-CD14 and OmCI reduced the formation of IL-8 and TNF-α more potently than the single inhibitors. OmCI also efficiently bound E. coli-induced leukotriene B(4) in pig and human plasma. In support of our ex vivo findings, in vivo the activity of all C pathways was inhibited at 0.6 mg OmCI/kg pig. In conclusion, OmCI efficiently inhibited pig and human C activation, has accompanying anti-inflammatory effects and is a promising candidate inhibitor for further in vivo studies of sepsis.


Subject(s)
Complement C5a/antagonists & inhibitors , Complement Inactivator Proteins/physiology , Ornithodoros/immunology , Animals , Complement C5a/metabolism , Complement Inactivator Proteins/therapeutic use , Complement Pathway, Alternative/immunology , Complement Pathway, Classical/immunology , Disease Models, Animal , Escherichia coli Infections/immunology , Escherichia coli Infections/prevention & control , Female , Humans , Male , Pilot Projects , Salivary Proteins and Peptides/physiology , Salivary Proteins and Peptides/therapeutic use , Sepsis/immunology , Sepsis/prevention & control , Swine
12.
Br J Pharmacol ; 180(4): 422-440, 2023 02.
Article in English | MEDLINE | ID: mdl-36251578

ABSTRACT

BACKGROUND AND PURPOSE: Traumatic haemorrhage (TH) is the leading cause of potentially preventable deaths that occur during the prehospital phase of care. No effective pharmacological therapeutics are available for critical TH patients yet. Here, we identify terminal complement activation (TCA) as a therapeutic target in combat casualties and evaluate the efficacy of a TCA inhibitor (nomacopan) on organ damage and survival in vivo. EXPERIMENTAL APPROACH: Complement activation products and cytokines were analysed in plasma from 54 combat casualties. The correlations between activated complement pathway(s) and the clinical outcomes in trauma patients were assessed. Nomacopan was administered to rats subjected to lethal TH (blast injury and haemorrhagic shock). Effects of nomacopan on TH were determined using survival rate, organ damage, physiological parameters, and laboratory profiles. KEY RESULTS: Early TCA was associated with systemic inflammatory responses and clinical outcomes in this trauma cohort. Lethal TH in the untreated rats induced early TCA that correlated with the severity of tissue damage and mortality. The addition of nomacopan to a damage-control resuscitation (DCR) protocol significantly inhibited TCA, decreased local and systemic inflammatory responses, improved haemodynamics and metabolism, attenuated tissue and organ damage, and increased survival. CONCLUSION AND IMPLICATIONS: Previous findings of our and other groups revealed that early TCA represents a rational therapeutic target for trauma patients. Nomacopan as a pro-survival and organ-protective drug, could emerge as a promising adjunct to DCR that may significantly reduce the morbidity and mortality in severe TH patients while awaiting transport to critical care facilities.


Subject(s)
Complement C5 , Shock, Hemorrhagic , Rats , Animals , Complement C5/pharmacology , Shock, Hemorrhagic/drug therapy , Complement Activation , Immunologic Factors/pharmacology , Phenotype
13.
Front Pharmacol ; 13: 846683, 2022.
Article in English | MEDLINE | ID: mdl-35350753

ABSTRACT

Votucalis is a biologically active protein in tick (R. appendiculatus) saliva, which specifically binds histamine with high affinity and, therefore, has the potential to inhibit the host's immunological responses at the feeding site. We hypothesized that scavenging of peripherally released endogenous histamine by Votucalis results in both anti-itch and anti-nociceptive effects. To test this hypothesis, adult male mice were subjected to histaminergic itch, as well as peripheral nerve injury that resulted in neuropathic pain. Thus, we selected models where peripherally released histamine was shown to be a key regulator. In these models, the animals received systemic (intraperitoneal, i.p.) or peripheral transdermal (subcutaneous, s.c. or intraplantar, i.pl.) administrations of Votucalis and itch behavior, as well as mechanical and thermal hypersensitivity, were evaluated. Selective histamine receptor antagonists were used to determine the involvement of histamine receptors in the effects produced by Votucalis. We also used the spontaneous object recognition test to confirm the centrally sparing properties of Votucalis. Our main finding shows that in histamine-dependent itch and neuropathic pain models peripheral (s.c. or i.pl.) administration of Votucalis displayed a longer duration of action for a lower dose range, when compared with Votucalis systemic (i.p.) effects. Stronger anti-itch effect was observed after co-administration of Votucalis (s.c.) and antagonists that inhibited peripheral histamine H1 and H2 receptors as well as central histamine H4 receptors indicating the importance of these histamine receptors in itch. In neuropathic mice, Votucalis produced a potent and complete anti-nociceptive effect on mechanical hypersensitivity, while thermal (heat) hypersensitivity was largely unaffected. Overall, our findings further emphasize the key role for histamine in the regulation of histaminergic itch and chronic neuropathic pain. Given the effectiveness of Votucalis after peripheral transdermal administration, with a lack of central effects, we provide here the first evidence that scavenging of peripherally released histamine by Votucalis may represent a novel therapeutically effective and safe long-term strategy for the management of these refractory health conditions.

14.
JAMA Dermatol ; 158(6): 641-649, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35507334

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.


Subject(s)
Autoimmune Diseases , Pemphigoid, Bullous , Aged , Female , Germany , Humans , Male , Neoplasm Recurrence, Local , Pemphigoid, Bullous/drug therapy , Pruritus , Quality of Life
15.
Biochem J ; 429(1): 103-12, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20545626

ABSTRACT

The saliva of blood-feeding parasites is a rich source of peptidase inhibitors that help to overcome the host's defence during host-parasite interactions. Using proteomic analysis, the cystatin OmC2 was demonstrated in the saliva of the soft tick Ornithodoros moubata, an important disease vector transmitting African swine fever virus and the spirochaete Borrelia duttoni. A structural, biochemical and biological characterization of this peptidase inhibitor was undertaken in the present study. Recombinant OmC2 was screened against a panel of physiologically relevant peptidases and was found to be an effective broad-specificity inhibitor of cysteine cathepsins, including endopeptidases (cathepsins L and S) and exopeptidases (cathepsins B, C and H). The crystal structure of OmC2 was determined at a resolution of 2.45 A (1 A=0.1 nm) and was used to describe the structure-inhibitory activity relationship. The biological impact of OmC2 was demonstrated both in vitro and in vivo. OmC2 affected the function of antigen-presenting mouse dendritic cells by reducing the production of the pro-inflammatory cytokines tumour necrosis factor alpha and interleukin-12, and proliferation of antigen-specific CD4+ T-cells. This suggests that OmC2 may suppress the host's adaptive immune response. Immunization of mice with OmC2 significantly suppressed the survival of O. moubata in infestation experiments. We conclude that OmC2 is a promising target for the development of a novel anti-tick vaccine to control O. moubata populations and combat the spread of associated diseases.


Subject(s)
Immunologic Factors/chemistry , Immunologic Factors/physiology , Salivary Cystatins/chemistry , Salivary Cystatins/physiology , Amino Acid Sequence , Animals , Crystallization , Crystallography, X-Ray , Female , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Ornithodoros/chemistry , Ornithodoros/immunology
16.
Cells ; 10(2)2021 02 15.
Article in English | MEDLINE | ID: mdl-33671954

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.


Subject(s)
Inflammation/drug therapy , Receptors, Leukotriene B4/metabolism , Retinal Vasculitis/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Disease Models, Animal , Humans , Receptors, Leukotriene B4/immunology , Retinal Vasculitis/immunology , Uveitis/drug therapy , Uveitis/immunology
17.
Emerg Infect Dis ; 15(9): 1467-70, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19788816

ABSTRACT

When Candida dubliniensis isolates obtained from seabird excrement and from humans in Ireland were compared by using multilocus sequence typing, 13 of 14 avian isolates were genetically distinct from human isolates. The remaining avian isolate was indistinguishable from a human isolate, suggesting that transmission may occur between humans and birds.


Subject(s)
Bird Diseases , Candida , Candidiasis/microbiology , Charadriiformes/microbiology , Zoonoses , Animals , Bird Diseases/microbiology , Bird Diseases/transmission , Candida/classification , Candida/genetics , Candida/isolation & purification , Candidiasis/veterinary , Feces/microbiology , Fungal Proteins/chemistry , Fungal Proteins/genetics , Humans , Ireland , Mycological Typing Techniques , Point Mutation , Sequence Analysis, DNA , Zoonoses/microbiology , Zoonoses/transmission
18.
JCI Insight ; 4(15)2019 08 08.
Article in English | MEDLINE | ID: mdl-31391346

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.


Subject(s)
Biological Products/pharmacology , Complement C5/antagonists & inhibitors , Complement Inactivating Agents/pharmacology , Leukotriene B4/antagonists & inhibitors , Pemphigoid, Bullous/drug therapy , Animals , Biological Products/therapeutic use , Cells, Cultured , Chemotaxis/drug effects , Chemotaxis/immunology , Collagen Type VII/administration & dosage , Collagen Type VII/immunology , Complement C5/immunology , Complement C5/metabolism , Complement Inactivating Agents/therapeutic use , Disease Models, Animal , Granulocytes/immunology , Healthy Volunteers , Humans , Leukotriene B4/immunology , Leukotriene B4/metabolism , Male , Mice , Neutrophils , Pemphigoid, Bullous/immunology , Pemphigoid, Bullous/pathology , Primary Cell Culture , Rabbits , Skin/immunology , Skin/pathology
19.
J Mol Biol ; 369(3): 784-93, 2007 Jun 08.
Article in English | MEDLINE | ID: mdl-17445829

ABSTRACT

The complement (C) system is a potent innate immune defence system against parasites. We have recently characterised and expressed OmCI, a 16 kDa protein derived from the soft tick Ornithodoros moubata that specifically binds C5, thereby preventing C activation. The structure of recombinant OmCI determined at 1.9 A resolution confirms a lipocalin fold and reveals that the protein binds a fatty acid derivative that we have identified by mass spectrometry as ricinoleic acid. We propose that OmCI could sequester one of the fatty acid-derived inflammatory modulators from the host plasma, thereby interfering with the host inflammatory response to the tick bite. Mapping of sequence differences between OmCI and other tick lipocalins with different functions, combined with biochemical investigations of OmCI activity, supports the hypothesis that OmCI acts by preventing interaction with the C5 convertase, rather than by blocking the C5a cleavage site.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Complement C5/chemistry , Complement Inactivator Proteins/chemistry , Complement System Proteins/chemistry , Insect Proteins/chemistry , Amino Acid Sequence , Animals , Arthropod Proteins , Gas Chromatography-Mass Spectrometry , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Ricinoleic Acids/chemistry , Ticks , X-Ray Diffraction
20.
Environ Health Perspect ; 116(11): 1563-7, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19057712

ABSTRACT

On 3 October 2007, 40 participants with diverse expertise attended the workshop Tamiflu and the Environment: Implications of Use under Pandemic Conditions to assess the potential human health impact and environmental hazards associated with use of Tamiflu during an influenza pandemic. Based on the identification and risk-ranking of knowledge gaps, the consensus was that oseltamivir ethylester-phosphate (OE-P) and oseltamivir carboxylate (OC) were unlikely to pose an ecotoxicologic hazard to freshwater organisms. OC in river water might hasten the generation of OC-resistance in wildfowl, but this possibility seems less likely than the potential disruption that could be posed by OC and other pharmaceuticals to the operation of sewage treatment plants. The work-group members agreed on the following research priorities: a) available data on the ecotoxicology of OE-P and OC should be published; b) risk should be assessed for OC-contaminated river water generating OC-resistant viruses in wildfowl; c) sewage treatment plant functioning due to microbial inhibition by neuraminidase inhibitors and other antimicrobials used during a pandemic should be investigated; and d) realistic worst-case exposure scenarios should be developed. Additional modeling would be useful to identify localized areas within river catchments that might be prone to high pharmaceutical concentrations in sewage treatment plant effluent. Ongoing seasonal use of Tamiflu in Japan offers opportunities for researchers to assess how much OC enters and persists in the aquatic environment.


Subject(s)
Influenza, Human/drug therapy , Oseltamivir/therapeutic use , Humans , Risk Assessment
SELECTION OF CITATIONS
SEARCH DETAIL