Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Front Trop Dis ; 42024 Jan 31.
Article in English | MEDLINE | ID: mdl-38500783

ABSTRACT

The parasitic worm-derived immunomodulator, ES-62 rescues defective levels of IL-10-producing regulatory B cells (Bregs) and suppresses chronic Th1/Th17-driven inflammation to protect against joint destruction in the mouse collagen-induced arthritis (CIA) model of rheumatoid arthritis. Such autoimmune arthritis is also associated with dysbiosis of the gut microbiota and disruption of intestinal barrier integrity. We recently further exploited the CIA model to show that ES-62's prevention of joint destruction is associated with protection of intestinal barrier integrity and normalization of the gut microbiota, thereby suppressing the gut pathology that precedes the onset of autoimmunity and joint damage in CIA-mice. As the status of the gut microbiota impacts on immune responses by influencing haematopoiesis, we have therefore investigated whether ES-62 harnesses the homeostatic mechanisms regulating this gut-bone marrow (BM) axis to resolve the chronic inflammation promoting autoimmunity and joint destruction in CIA. Reflecting this, ES-62 was found to counteract the BM myeloid/lymphoid bias typically associated with chronic inflammation and infection. This was achieved primarily by ES-62 acting to maintain the levels of lymphoid lineages (B220+ and CD3+ cells) observed in naïve, healthy mice but lost from the BM of CIA-mice. Moreover, ES-62's ability to prevent bone-destroying osteoclastogenesis was found to be associated with its suppression of CIA-induced upregulation of osteoclast progenitors (OCPs) in the BM. Critically, and supporting ES-62's targeting of the gut-BM axis, this rewiring of inflammatory haematopoiesis was lost in mice with a depleted microbiome. Underlining the importance of ES-62's actions in restoring steady-state haematopoiesis, the BM levels of B and T lymphoid cells were shown to be inversely correlated, whilst the levels of OCPs positively correlated, with the severity of joint damage in CIA-mice.

2.
Biochem Soc Trans ; 52(1): 55-63, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38334208

ABSTRACT

Parasitic worms are pathogens of major medical and veterinary importance. They have evolved highly effective and sophisticated strategies of immune system manipulation, typically involving actively excreted/secreted (E-S) products. These molecules dampen and regulate the host immune responses that would otherwise result in parasite expulsion, thereby enabling the worms to survive in the host for many years, and they can also help prevent the potentially serious tissue damage that the worms can induce. Reflecting these E-S product-associated anti-inflammatory activities, there is also increasing evidence that parasitic worms and their products may serendipitously protect against allergic and autoimmune conditions and in addition, comorbidities of ageing that are associated with inflammatory responses, like type 2 diabetes and obesity. Research in this area has to date generally focused on identifying the cellular and effector targets of immunomodulation induced by the worm E-S products. However, increasing evidence that they can induce stably imprinted phenotypes of haematopoietic and stromal cells which promote their long-lasting survival has recently ignited interest in the ability of the molecules to epigenetically rewire cells to 'resolve and repair' phenotypes. Here, we review and discuss these new data in the context of their potential for exploitation in identifying novel gene signatures for the development of advanced and safe therapeutics for chronic inflammatory diseases.


Subject(s)
Autoimmune Diseases , Diabetes Mellitus, Type 2 , Helminths , Animals , Immune System , Epigenesis, Genetic
3.
Front Immunol ; 14: 1285069, 2023.
Article in English | MEDLINE | ID: mdl-38077318

ABSTRACT

Mice develop pathology in the lungs as they age and this may be accelerated by a high calorie diet (HCD). ES-62 is a protein secreted by the parasitic worm Acanthocheilonema viteae that is immunomodulatory by virtue of covalently attached phosphorylcholine (PC) moieties. In this study, we show that weekly treatment of C57BL/6J mice with ES-62 protected against pathology in the lungs in male but not female mice fed a HCD from 10 weeks of age as shown by reductions in cellular infiltration and airway remodelling, particularly up to 160 days of age. ES-62 also reduced gene expression of the cytokines IL-4 and IL-17 and in addition the TLR/IL-1R adaptor MyD88, in the lungs of male mice although HCD-induced increases in these inflammatory markers were not detected until between 340 and 500 days of age. A combination of two drug-like ES-62 PC-based small molecule analogues (SMAs), produced broadly similar protective effects in the lungs of male mice with respect to both lung pathology and inflammatory markers, in addition to a decrease in HCD-induced IL-5 expression. Overall, our data show that ES-62 and its SMAs offer protection against HCD-accelerated pathological changes in the lungs during ageing. Given the targeting of Th2 cytokines and IL-17, we discuss this protection in the context of ES-62's previously described amelioration of airway hyper-responsiveness in mouse models of asthma.


Subject(s)
Acanthocheilonema , Interleukin-17 , Male , Animals , Mice , Interleukin-17/metabolism , Mice, Inbred C57BL , Cytokines/metabolism , Obesity , Aging , Lung/metabolism
4.
Trends Parasitol ; 39(9): 739-748, 2023 09.
Article in English | MEDLINE | ID: mdl-37487870

ABSTRACT

Despite the introduction of novel treatment strategies, management of rheumatic disorders remains associated with substantial unmet clinical need. Of interest therefore, it has recently become apparent that there is a global inverse relationship between the incidence of such conditions and parasitic helminth infection, with striking examples involving rheumatoid arthritis (RA)/systemic lupus erythematosus (SLE) patients and filarial nematode worm infection in studies in India. Such findings reflect that helminths are master manipulators of the immune system, particularly in being able to modulate proinflammatory responses. The aim of this article is thus to consider findings to date on this exciting and intriguing research area to form an opinion on whether parasitic worms may be exploited to generate novel therapies for rheumatic diseases.


Subject(s)
Arthritis, Rheumatoid , Helminths , Lupus Erythematosus, Systemic , Rheumatic Diseases , Animals , Arthritis, Rheumatoid/drug therapy , India
5.
Front Immunol ; 13: 953053, 2022.
Article in English | MEDLINE | ID: mdl-36105811

ABSTRACT

Despite significant increases in human lifespan over the last century, adoption of high calorie diets (HCD) has driven global increases in type-2 diabetes, obesity and cardiovascular disease, disorders precluding corresponding improvements in healthspan. Reflecting that such conditions are associated with chronic systemic inflammation, evidence is emerging that infection with parasitic helminths might protect against obesity-accelerated ageing, by virtue of their evolution of survival-promoting anti-inflammatory molecules. Indeed, ES-62, an anti-inflammatory secreted product of the filarial nematode Acanthocheilonema viteae, improves the healthspan of both male and female C57BL/6J mice undergoing obesity-accelerated ageing and also extends median lifespan in male animals, by positively impacting on inflammatory, adipose metabolic and gut microbiome parameters of ageing. We therefore explored whether ES-62 affects the osteoimmunology axis that integrates environmental signals, such as diet and the gut microbiome to homeostatically regulate haematopoiesis and training of immune responses, which become dysregulated during (obesity-accelerated) ageing. Of note, we find sexual dimorphisms in the decline in bone health, and associated dysregulation of haematopoiesis and consequent peripheral immune responses, during obesity-accelerated ageing, highlighting the importance of developing sex-specific anti-ageing strategies. Related to this, ES-62 protects trabecular bone structure, maintaining bone marrow (BM) niches that counter the ageing-associated decline in haematopoietic stem cell (HSC) functionality highlighted by a bias towards myeloid lineages, in male but not female, HCD-fed mice. This is evidenced by the ability of ES-62 to suppress the adipocyte and megakaryocyte bias and correspondingly promote increases in B lymphocytes in the BM. Furthermore, the consequent prevention of ageing-associated myeloid/lymphoid skewing is associated with reduced accumulation of inflammatory CD11c+ macrophages and IL-1ß in adipose tissue, disrupting the perpetuation of inflammation-driven dysregulation of haematopoiesis during obesity-accelerated ageing in male HCD-fed mice. Finally, we report the ability of small drug-like molecule analogues of ES-62 to mimic some of its key actions, particularly in strongly protecting trabecular bone structure, highlighting the translational potential of these studies.


Subject(s)
Helminth Proteins , Helminths , Aging , Animals , Anti-Inflammatory Agents , Disease Models, Animal , Female , Helminths/metabolism , Humans , Inflammation , Male , Mice , Mice, Inbred C57BL , Obesity
6.
PLoS Pathog ; 17(11): e1010069, 2021 11.
Article in English | MEDLINE | ID: mdl-34748611

ABSTRACT

ES-62 is the major secreted protein of the parasitic filarial nematode, Acanthocheilonema viteae. The molecule exists as a large tetramer (MW, ~240kD), which possesses immunomodulatory properties by virtue of multiple phosphorylcholine (PC) moieties attached to N-type glycans. By suppressing inflammatory immune responses, ES-62 can prevent disease development in certain mouse models of allergic and autoimmune conditions, including joint pathology in collagen-induced arthritis (CIA), a model of rheumatoid arthritis (RA). Such protection is associated with functional suppression of "pathogenic" hyper-responsive synovial fibroblasts (SFs), which exhibit an aggressive inflammatory and bone-damaging phenotype induced by their epigenetic rewiring in response to the inflammatory microenvironment of the arthritic joint. Critically, exposure to ES-62 in vivo induces a stably-imprinted CIA-SF phenotype that exhibits functional responses more typical of healthy, Naïve-SFs. Consistent with this, ES-62 "rewiring" of SFs away from the hyper-responsive phenotype is associated with suppression of ERK activation, STAT3 activation and miR-155 upregulation, signals widely associated with SF pathogenesis. Surprisingly however, DNA methylome analysis of Naïve-, CIA- and ES-62-CIA-SF cohorts reveals that rather than simply preventing pathogenic rewiring of SFs, ES-62 induces further changes in DNA methylation under the inflammatory conditions pertaining in the inflamed joint, including targeting genes associated with ciliogenesis, to programme a novel "resolving" CIA-SF phenotype. In addition to introducing a previously unsuspected aspect of ES-62's mechanism of action, such unique behaviour signposts the potential for developing DNA methylation signatures predictive of pathogenesis and its resolution and hence, candidate mechanisms by which novel therapeutic interventions could prevent SFs from perpetuating joint inflammation and destruction in RA. Pertinent to these translational aspects of ES-62-behavior, small molecule analogues (SMAs) based on ES-62's active PC-moieties mimic the rewiring of SFs as well as the protection against joint disease in CIA afforded by the parasitic worm product.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/prevention & control , Epigenesis, Genetic , Fibroblasts/metabolism , Helminth Proteins/pharmacology , Inflammation/prevention & control , Synoviocytes/metabolism , Acanthocheilonema/metabolism , Animals , Arthritis, Experimental/etiology , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Cells, Cultured , DNA Methylation , Fibroblasts/drug effects , Fibroblasts/immunology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred DBA , Synoviocytes/drug effects , Synoviocytes/immunology
7.
Front Immunol ; 12: 809896, 2021.
Article in English | MEDLINE | ID: mdl-35095899

ABSTRACT

The guanine nucleotide exchange factor cytohesin-2 (ARNO) is a major activator of the small GTPase ARF6 that has been shown to play an important role(s) in cell adhesion, migration and cytoskeleton reorganization in various cell types and models of disease. Interestingly, dysregulated cell migration, in tandem with hyper-inflammatory responses, is one of the hallmarks associated with activated synovial fibroblasts (SFs) during chronic inflammatory joint diseases, like rheumatoid arthritis. The role of ARNO in this process has previously been unexplored but we hypothesized that the pro-inflammatory milieu of inflamed joints locally induces activation of ARNO-mediated pathways in SFs, promoting an invasive cell phenotype that ultimately leads to bone and cartilage damage. Thus, we used small interference RNA to investigate the impact of ARNO on the pathological migration and inflammatory responses of murine SFs, revealing a fully functional ARNO-ARF6 pathway which can be rapidly activated by IL-1ß. Such signalling promotes cell migration and formation of focal adhesions. Unexpectedly, ARNO was also shown to modulate SF-inflammatory responses, dictating their precise cytokine and chemokine expression profile. Our results uncover a novel role for ARNO in SF-dependent inflammation, that potentially links pathogenic migration with initiation of local joint inflammation, offering new approaches for targeting the fibroblast compartment in chronic arthritis and joint disease.


Subject(s)
Cell Movement/genetics , Fibroblasts/metabolism , GTPase-Activating Proteins/genetics , Immunomodulation/genetics , Synovial Membrane/cytology , ADP-Ribosylation Factor 6/metabolism , Animals , Biomarkers , Cytokines/metabolism , GTPase-Activating Proteins/metabolism , Gene Expression , Interleukin-1beta/metabolism , Mice , Phosphorylation , RNA, Small Interfering/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction
8.
PLoS Pathog ; 16(3): e1008391, 2020 03.
Article in English | MEDLINE | ID: mdl-32163524

ABSTRACT

Improvements in hygiene and health management have driven significant increases in human lifespan over the last 50 years. Frustratingly however, this extension of lifespan has not been matched by equivalent improvements in late-life health, not least due to the global pandemic in type-2 diabetes, obesity and cardiovascular disease, all ageing-associated conditions exacerbated and accelerated by widespread adoption of the high calorie Western diet (HCD). Recently, evidence has begun to emerge that parasitic worm infection might protect against such ageing-associated co-morbidities, as a serendipitous side-effect of their evolution of pro-survival, anti-inflammatory mechanisms. As a novel therapeutic strategy, we have therefore investigated the potential of ES-62, an anti-inflammatory secreted product of the filarial nematode Acanthocheilonema viteae, to improve healthspan (the period of life before diseases of ageing appear) by targeting the chronic inflammation that drives metabolic dysregulation underpinning ageing-induced ill-health. We administered ES-62 subcutaneously (at a dose of 1 µg/week) to C57BL/6J mice undergoing HCD-accelerated ageing throughout their lifespan, while subjecting the animals to analysis of 120 immunometabolic responses at various time-points. ES-62 improved a number of inflammatory parameters, but markedly, a range of pathophysiological, metabolic and microbiome parameters of ageing were also successfully targeted. Notably, ES-62-mediated promotion of healthspan in male and female HCD-mice was associated with different mechanisms and reflecting this, machine learning modelling identified sex-specific signatures predictive of ES-62 action against HCD-accelerated ageing. Remarkably, ES-62 substantially increased the median survival of male HCD-mice. This was not the case with female animals and unexpectedly, this difference between the two sexes could not be explained in terms of suppression of the chronic inflammation driving ageing, as ES-62 tended to be more effective in reducing this in female mice. Rather, the difference appeared to be associated with ES-62's additional ability to preferentially promote a healthier gut-metabolic tissue axis in male animals.


Subject(s)
Acanthocheilonema/immunology , Acanthocheilonemiasis/immunology , Diet, Western/adverse effects , Helminth Proteins/immunology , Longevity/immunology , Models, Immunological , Animals , Female , Male , Mice
9.
Mol Biochem Parasitol ; 234: 111232, 2019 12.
Article in English | MEDLINE | ID: mdl-31634505

ABSTRACT

One of the most rapidly increasing human public health problems is obesity, whose sequelae like type-2 diabetes, represent continuously worsening, life-long conditions. Over the last 15 years, data have begun to emerge from human and more frequently, mouse studies, that support the idea that parasitic worm infection can protect against this condition. We have therefore investigated the potential of two synthetic small molecule analogues (SMAs) of the anti-inflammatory Acanthocheilonema viteae product ES-62, to protect against metabolic dysfunction in a C57BL/6 J mouse model of high calorie diet-induced obesity. We found weekly subcutaneous administration of the SMAs in combination (1 µg of each), starting one week before continuous exposure to high calorie diet (HCD), decreased fasting glucose levels and reversed the impaired glucose clearance observed in male mice, when measured at approximately 7 and 13 weeks after exposure to HCD. Fasting glucose levels were also-reduced in male mice fed a HCD for some 38 weeks when given SMA-treatment 13 weeks after the start of HCD, indicating an SMA-therapeutic potential. For the most part, protective effects were not observed in female mice. SMA treatment also conferred protection against each of reduced ileum villus length and liver fibrosis, but more prominently in female mice. Previous studies in mice indicate that protection against metabolic dysfunction is usually associated with polarisation of the immune system towards a type-2/anti-inflammatory direction but our attempts to correlate improved metabolic parameters with such changes were unsuccessful. Further analysis will therefore be required to define mechanism of action. Nevertheless, overall our data clearly show the potential of the drug-like SMAs as a preventative or treatment for metabolic dysregulation associated with obesity.


Subject(s)
Helminth Proteins/immunology , Homeostasis/drug effects , Immunologic Factors/administration & dosage , Metabolic Syndrome/prevention & control , Obesity/prevention & control , Animals , Blood Glucose/analysis , Blood Glucose/metabolism , Disease Models, Animal , Energy Intake , Female , Helminth Proteins/chemistry , Humans , Immunologic Factors/chemistry , Injections, Subcutaneous , Liver/drug effects , Liver/metabolism , Male , Metabolic Syndrome/blood , Metabolic Syndrome/etiology , Metabolic Syndrome/metabolism , Mice , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/immunology , Obesity/blood , Obesity/etiology , Obesity/metabolism , Sex Factors
10.
Nat Commun ; 10(1): 1554, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30952846

ABSTRACT

The human immune system has evolved in the context of our colonisation by bacteria, viruses, fungi and parasitic helminths. Reflecting this, the rapid eradication of pathogens appears to have resulted in reduced microbiome diversity and generation of chronically activated immune systems, presaging the recent rise of allergic, autoimmune and metabolic disorders. Certainly, gastrointestinal helminths can protect against gut and lung mucosa inflammatory conditions by modulating the microbiome and suppressing the chronic inflammation associated with dysbiosis. Here, we employ ES-62, an immunomodulator secreted by tissue-dwelling Acanthocheilonema viteae to show that helminth-modulation of the gut microbiome does not require live infection with gastrointestinal-based worms nor is protection restricted to mucosal diseases. Specifically, subcutaneous administration of this defined immunomodulator affords protection against joint disease in collagen-induced arthritis, a mouse model of rheumatoid arthritis, which is associated with normalisation of gut microbiota and prevention of loss of intestinal barrier integrity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Arthritis, Experimental/drug therapy , Gastrointestinal Microbiome/drug effects , Helminth Proteins/therapeutic use , Animals , Arthritis, Experimental/immunology , Helminth Proteins/pharmacology , Immunomodulation , Male , Mice
11.
Molecules ; 23(10)2018 Oct 17.
Article in English | MEDLINE | ID: mdl-30336585

ABSTRACT

Parasitic helminths and their isolated secreted products show promise as novel treatments for allergic and autoimmune conditions in humans. Foremost amongst the secreted products is ES-62, a glycoprotein derived from Acanthocheilonema viteae, a filarial nematode parasite of gerbils, which is anti-inflammatory by virtue of covalently-attached phosphorylcholine (PC) moieties. ES-62 has been found to protect against disease in mouse models of rheumatoid arthritis, systemic lupus erythematosus, and airway hyper-responsiveness. Furthermore, novel PC-based synthetic small molecule analogues (SMAs) of ES-62 have recently been demonstrated to show similar anti-inflammatory properties to the parent molecule. In spite of these successes, we now show that ES-62 and its SMAs are unable to provide protection in mouse models of certain autoimmune conditions where other helminth species or their secreted products can prevent disease development, namely type I diabetes, multiple sclerosis and inflammatory bowel disease. We speculate on the reasons underlying ES-62's failures in these conditions and how the negative data generated may help us to further understand ES-62's mechanism of action.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Helminth Proteins/administration & dosage , Inflammatory Bowel Diseases/drug therapy , Multiple Sclerosis/drug therapy , Acanthocheilonema/chemistry , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Helminth Proteins/chemistry , Helminths/chemistry , Humans , Inflammatory Bowel Diseases/pathology , Mice , Multiple Sclerosis/pathology
12.
Front Immunol ; 9: 1016, 2018.
Article in English | MEDLINE | ID: mdl-29867986

ABSTRACT

The immunomodulatory actions of parasitic helminth excretory-secretory (ES) products that serendipitously protect against development of chronic inflammatory disorders are well established: however, knowledge of the interaction between ES products and the host musculoskeletal system in such diseases is limited. In this study, we have focused on ES-62, a glycoprotein secreted by the rodent filarial nematode Acanthocheilonema viteae that is immunomodulatory by virtue of covalently attached phosphorylcholine (PC) moieties, and also two synthetic drug-like PC-based small molecule analogues (SMAs) that mimic ES-62's immunomodulatory activity. We have previously shown that each of these molecules prevents development of pathology in collagen-induced arthritis (CIA), a model of the musculoskeletal disease rheumatoid arthritis (RA) and reflecting this, we now report that ES-62 and its SMAs, modify bone remodeling by altering bone marrow progenitors and thus impacting on osteoclastogenesis. Consistent with this, we find that these molecules inhibit functional osteoclast differentiation in vitro. Furthermore, this appears to be achieved by induction of anti-oxidant response gene expression, thereby resulting in reduction of the reactive oxygen species production that is necessary for the increased osteoclastogenesis witnessed in musculoskeletal diseases like RA.


Subject(s)
Arthritis, Experimental/prevention & control , Helminth Proteins/pharmacology , Immunologic Factors/pharmacology , Osteogenesis/drug effects , Acanthocheilonema/chemistry , Animals , Male , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism
13.
Acta Trop ; 185: 212-218, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29802846

ABSTRACT

Parasitic worms are receiving much attention as a potential new therapeutic approach to treating autoimmune and allergic conditions but concerns remain regarding their safety. As an alternative strategy, we have focused on the use of defined parasitic worm products and recently taken this one step further by designing drug-like small molecule analogues of one such product, ES-62, which is anti-inflammatory by virtue of covalently attached phosphorylcholine moieties. Previously, we have shown that ES-62 mimics are efficacious in protecting against disease in mouse models of rheumatoid arthritis, systemic lupus erythematosus and skin and lung allergy. Given the potential role of chronic inflammation in fibrosis, in the present study we have focused our attention on lung fibrosis, a debilitating condition for which there is no cure and which in spite of treatment slowly gets worse over time. Two mouse models of fibrosis - bleomycin-induced and LPS-induced - in which roles for inflammation have been implicated were adopted. Four ES-62 analogues were tested - 11a and 12b, previously shown to be active in mouse models of allergic and autoimmune disease and 16b and AIK-29/62 both of which are structurally related to 11a. All four compounds were found to significantly reduce disease development in both fibrosis models, as shown by histopathological analysis of lung tissue, indicating their potential as treatments for this condition.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Helminth Proteins/therapeutic use , Pulmonary Fibrosis/drug therapy , Animals , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL
14.
Sci Rep ; 8(1): 4497, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29540770

ABSTRACT

ES-62 is a secreted parasitic worm-derived immunomodulator that exhibits therapeutic potential in allergy by downregulating aberrant MyD88 signalling to normalise the inflammatory phenotype and mast cell responses. IL-33 plays an important role in driving mast cell responses and promoting type-2 allergic inflammation, particularly with respect to asthma, via MyD88-integrated crosstalk amongst the IL-33 receptor (ST2), TLR4 and FcεRI. We have now investigated whether ES-62 targets this pathogenic network by subverting ST2-signalling, specifically by characterising how the functional outcomes of crosstalk amongst ST2, TLR4 and FcεRI are modulated by the worm product in wild type and ST2-deficient mast cells. This analysis showed that whilst ES-62 inhibits IL-33/ST2 signalling, the precise functional modulation observed varies with receptor usage and/or mast cell phenotype. Thus, whilst ES-62's harnessing of the capacity of ST2 to sequester MyD88 appears sufficient to mediate its inhibitory effects in peritoneal-derived serosal mast cells, downregulation of MyD88 expression appears to be required to dampen the higher levels of cytokine production typically released by bone marrow-derived mucosal mast cells.


Subject(s)
Helminth Proteins/pharmacology , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Receptors, IgE/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Cell Degranulation/drug effects , Cell Degranulation/immunology , Cytokines/metabolism , Immunomodulation , Inflammation Mediators/metabolism , Interleukin-1 Receptor-Like 1 Protein/genetics , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Knockout , Protein Binding
15.
Sci Rep ; 8(1): 2123, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29391452

ABSTRACT

ES-62 is a protein secreted by the parasitic worm Acanthocheilonema viteae that is anti-inflammatory by virtue of covalently attached phosphorylcholine. Previously we have reported that drug-like Small Molecule Analogues (SMAs) of its phosphorylcholine moiety can mimic ES-62 in protecting against disease development in certain mouse models of autoimmune and allergic conditions, due to them causing partial degradation of the TLR/IL-1R adaptor MyD88. We have now taken a molecular modelling approach to investigating the mechanism underlying this effect and this predicts that the SMAs interact directly with the MyD88 TIR domain. Further support for this is provided by assay of LPS-induced MyD88/NF-κB-driven secreted alkaline phosphatase (SEAP) reporter activity in commercially-available stably transfected (TLR4-MD2-NF-κB-SEAP) HEK293 cells, as SMA12b-mediated inhibition of such SEAP activity is blocked by its pre-incubation with recombinant MyD88-TIR domain. Direct binding of SMA12b to the TIR domain is also shown to inhibit homo-dimerization of the adaptor, an event that can explain the observed degradation of the adaptor and inhibition of subsequent downstream signalling. Thus, these new data identify initial events by which drug-like ES-62 SMAs, which we also demonstrate are able to inhibit cytokine production by human cells, homeostatically maintain "safe" levels of MyD88 signalling.


Subject(s)
Acanthocheilonema/metabolism , Anti-Inflammatory Agents/pharmacology , Helminth Proteins/pharmacology , Inflammation/drug therapy , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , Acanthocheilonema/growth & development , HEK293 Cells , Humans , Inflammation/metabolism , Inflammation/pathology , Myeloid Differentiation Factor 88/genetics , NF-kappa B/genetics , Protein Domains , Tumor Necrosis Factor-alpha
16.
Trends Parasitol ; 33(9): 694-705, 2017 09.
Article in English | MEDLINE | ID: mdl-28606411

ABSTRACT

There has been increasing recognition that the alarming surge in allergy and autoimmunity in the industrialised and developing worlds shadows the rapid eradication of pathogens, such as parasitic helminths. Appreciation of this has fuelled an explosion in research investigating the therapeutic potential of these worms. This review considers the current state-of-play with a particular focus on exciting recent advances in the identification of potential novel targets for immunomodulation that can be exploited therapeutically. Furthermore, we contemplate the prospects for designing worm-derived immunotherapies for an ever-widening range of inflammatory diseases, including, for example, obesity, cardiovascular disease, and ageing as well as neurodevelopmental disorders like autism.


Subject(s)
Helminthiasis/immunology , Helminths/immunology , Immunomodulation , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , Humans , Hypersensitivity/immunology , Hypersensitivity/prevention & control , Immunotherapy/trends
17.
Sci Rep ; 7(1): 1704, 2017 05 10.
Article in English | MEDLINE | ID: mdl-28490801

ABSTRACT

ES-62, a glycoprotein secreted by the parasitic filarial nematode Acanthocheilonema viteae, subverts host immune responses towards anti-inflammatory phenotypes by virtue of covalently attached phosphorylcholine (PC). The PC dictates that ES-62 exhibits protection in murine models of inflammatory disease and hence a library of drug-like PC-based small molecule analogues (SMAs) was synthesised. Four sulfone-containing SMAs termed 11a, 11e, 11i and 12b were found to reduce mouse bone marrow-derived dendritic cell (DC) pathogen-associated molecular pattern (PAMP)-induced pro-inflammatory cytokine production, inhibit NF-κB p65 activation, and suppress LPS-induced up-regulation of CD40 and CD86. Active SMAs also resulted in a DC phenotype that exhibited reduced capacity to prime antigen (Ag)-specific IFN-γ production during co-culture with naïve transgenic TCR DO.11.10 T cells in vitro and reduced their ability, following adoptive transfer, to prime the expansion of Ag-specific T lymphocytes, specifically TH17 cells, in vivo. Consistent with this, mice receiving DCs treated with SMAs exhibited significantly reduced severity of collagen-induced arthritis and this was accompanied by a significant reduction in IL-17+ cells in the draining lymph nodes. Collectively, these studies indicate that drug-like compounds that target DCs can be designed from parasitic worm products and demonstrate the potential for ES-62 SMA-based DC therapy in inflammatory disease.


Subject(s)
Anthelmintics/pharmacology , Dendritic Cells/metabolism , Helminth Proteins/chemistry , Helminths/drug effects , Small Molecule Libraries/pharmacology , Animals , Anthelmintics/chemistry , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Bone Marrow Cells/metabolism , Cytokines/biosynthesis , Dendritic Cells/drug effects , Interleukin-17/metabolism , Interleukin-1beta/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/drug effects , Th17 Cells/immunology
18.
Biomed J ; 40(1): 9-22, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28411887

ABSTRACT

Christian de Duve first coined the expression "autophagy" during his seminal work on the discovery of lysosomes, which led to him being awarded the Nobel Prize in Physiology or Medicine in 1974. The term was adopted to distinguish degradation of intracellular components from the uptake and degradation of extracellular substances that he called "heterophagy". Studies until the 1990s were largely observational/morphological-based until in 1993 Yoshinori Oshumi described a genetic screen in yeast undergoing nitrogen deprivation that led to the isolation of autophagy-defective mutants now better known as ATG (AuTophaGy-related) genes. The screen identified mutants that fell into 15 complementation groups implying that at least 15 genes were involved in the regulation of autophagy in yeast undergoing nutrient deprivation, but today, 41 yeast ATG genes have been described and many (though not all) have orthologues in humans. Attempts to identify the genetic basis of autophagy led to an explosion in its research and it's not surprising that in 2016 Yoshinori Oshumi was awarded the Nobel Prize in Physiology or Medicine. Our aim here is not to exhaustively review the ever-expanding autophagy literature (>60 papers per week), but to celebrate Yoshinori Oshumi's Nobel Prize by highlighting just a few aspects that are not normally extensively covered. In an accompanying mini-review we address the role of autophagy in early-diverging eukaryote parasites that like yeast, lack lysosomes and so use a digestive vacuole to degrade autophagosome cargo and also discuss how parasitized host cells react to infection by subverting regulation of autophagy.


Subject(s)
Autophagy/physiology , Lysosomes/metabolism , Nobel Prize , Phagosomes/metabolism , Animals , Awards and Prizes , Humans , Medicine
19.
Sci Rep ; 6: 37276, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27869138

ABSTRACT

We have previously shown that ES-62, a phosphorylcholine (PC)-containing glycoprotein secreted by the parasitic filarial nematode Acanthocheilonema viteae targets dendritic cell (DC) responses, specifically by suppressing TLR4 signalling to inhibit Th1/Th17-driven inflammation. We have now investigated the molecular mechanisms underpinning such immunomodulation and show here that ES-62-mediated downregulation of protein kinase C-δ (PKC-δ), a TLR4-associated signalling mediator required for full activation of LPS-driven pro-inflammatory responses, is associated with induction of a low level of autophagic flux, as evidenced by upregulation and trafficking of p62 and LC3 and their consequent autophagolysosomal degradation. By contrast, the classical TLR4 ligand LPS, strongly upregulates p62 and LC3 expression but under such canonical TLR4 signalling this upregulation appears to reflect a block in autophagic flux, with these elements predominantly degraded in a proteasomal manner. These data are consistent with autophagic flux acting to homeostatically suppress proinflammatory DC responses and indeed, blocking of PKC-δ degradation by the autophagolysosomal inhibitors, E64d plus pepstatin A, results in abrogation of the ES-62-mediated suppression of LPS-driven release of IL-6, IL-12p70 and TNF-α by DCs. Thus, by harnessing this homeostatic regulatory mechanism, ES-62 can protect against aberrant inflammation, either to promote parasite survival or serendipitously, exhibit therapeutic potential in inflammatory disease.


Subject(s)
Dendritic Cells/drug effects , Dendritic Cells/metabolism , Helminth Proteins/pharmacology , Protein Kinase C-delta/metabolism , Toll-Like Receptors/metabolism , Animals , Autophagosomes/metabolism , Cytokines/metabolism , Lipopolysaccharides/pharmacology , Lysosomes/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Protein Kinase C-delta/genetics , Proteolysis/drug effects , Th1 Cells/drug effects , Th17 Cells/drug effects , Toll-Like Receptors/immunology , Tumor Necrosis Factor-alpha/metabolism
20.
Immunol Lett ; 168(1): 31-40, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26343793

ABSTRACT

ES-62, a glycoprotein secreted by the filarial nematode Acanthocheilonema viteae, has been shown to modulate the immune system through subversion of signal transduction pathways operating in various immune system cells. With respect to human bone marrow-derived mast cells (BMMCs), ES-62 was previously shown to inhibit FcϵRI-mediated mast cell functional responses such as degranulation and pro-inflammatory cytokine release through a mechanism involving the degradation of PKC-α. At the same time, it was noted that the worm product was able to degrade certain other PKC isoforms but the significance of this was uncertain. In this study, we have employed PKC isoform KO mice to investigate the role of PKC-α, -ß -ϵ, and -θ in mouse BMMCs in order to establish their involvement in mast cell-mediated responses and also, if their absence impacts on ES-62's activity. The data obtained support that in response to antigen cross-linking of IgE bound to FcϵRI, pro-inflammatory cytokine release is controlled in part by a partnership between one conventional and one novel isoform with PKC-α and -θ acting as positive regulators of IL-6 and TNF-α production, while PKC-ß and ϵ act as negative regulators of such cytokines. Furthermore, ES-62 appears to target certain other PKC isoforms in addition to PKC-α to inhibit cytokine release and this may enable it to more efficiently inhibit mast cell responses.


Subject(s)
Bone Marrow Cells/immunology , Helminth Proteins/immunology , Mast Cells/immunology , Protein Kinase C/immunology , Animals , Blotting, Western , Bone Marrow Cells/enzymology , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Helminth Proteins/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Isoenzymes/genetics , Isoenzymes/immunology , Isoenzymes/metabolism , Male , Mast Cells/enzymology , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase C beta/genetics , Protein Kinase C beta/immunology , Protein Kinase C beta/metabolism , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/immunology , Protein Kinase C-alpha/metabolism , Protein Kinase C-epsilon/genetics , Protein Kinase C-epsilon/immunology , Protein Kinase C-epsilon/metabolism , Receptors, IgE/immunology , Receptors, IgE/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...