Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 255
Filtrar
1.
Int J Mol Sci ; 25(13)2024 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-39000527

RESUMEN

Mast cells are essential immune cells involved in the host's defence against gastrointestinal nematodes. To evade the immune response, parasitic nematodes produce a variety of molecules. Galectin 1, produced by Teladorsagia circumcincta (Tci-gal-1), reduces mast cell degranulation and selectively regulates mediator production and release in an IgE-dependent manner. To uncover the activity of Tci-gal-1, we have examined the effect of the protein on gene expression, protein production, and apoptosis in activated basophilic leukaemia RBL-2H3 cells. Rat RBL-2H3 cells were activated with anti-DNP IgE and DNP-HSA, and then treated with Tci-gal-1. Microarray analysis was used to examine gene expression. The levels of several apoptosis-related molecules and cytokines were determined using antibody arrays and ELISA. Early and late apoptosis was evaluated cytometrically. Degranulation of cells was determined by a ß-hexosaminidase release assay. Treatment of activated RBL-2H3 cells with Tci-gal-1 resulted in inhibited apoptosis and decreased degranulation, although we did not detect significant changes in gene expression. The production of pro-apoptotic molecules, receptor for advanced glycation end products (RAGE) and Fas ligand (FasL), and the cytokines IL-9, IL-10, IL-13, TNF-α, and IL-2 was strongly inhibited. Tci-gal-1 modulates apoptosis, degranulation, and production of cytokines by activated RBL-2H3 cells without detectable influence on gene transcription. This parasite protein is crucial for modulation of the protective immune response and the inhibition of chronic inflammation driven by mast cell activity.


Asunto(s)
Apoptosis , Degranulación de la Célula , Inmunoglobulina E , Leucemia Basofílica Aguda , Animales , Ratas , Inmunoglobulina E/inmunología , Línea Celular Tumoral , Leucemia Basofílica Aguda/metabolismo , Leucemia Basofílica Aguda/inmunología , Leucemia Basofílica Aguda/patología , Mastocitos/inmunología , Mastocitos/metabolismo , Citocinas/metabolismo , Galectinas/metabolismo , Proteínas del Helminto/farmacología , Proteínas del Helminto/metabolismo , Galectina 1/metabolismo , Galectina 1/genética
2.
Immun Inflamm Dis ; 12(6): e1321, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38888451

RESUMEN

BACKGROUND: For decades, studies have demonstrated the anti-inflammatory potential of proteins secreted by helminths in allergies and asthma. Previous studies have demonstrated the immunomodulatory capabilities of Succinate Coenzyme A ligase beta-like protein (SUCLA-ß) derived from Trichinella spiralis, a crucial excretory product of this parasite. OBJECTIVE: To explore the therapeutic potential of SUCLA-ß in alleviating and controlling ovalbumin (OVA)-induced allergic asthma, as well as its influence on host immune modulation. METHODS: In this research, we utilized the rTs-SUCLA-ß protein derived from T. spiralis to investigate its potential in mitigating airway inflammation in a murine model of asthma induced by OVA sensitization/stimulation, both pre- and post-challenge. The treatment's efficacy was assessed by quantifying the extent of inflammation in the lungs. RESULTS: Treatment with rTs-SUCLA-ß demonstrated efficacy in ameliorating OVA-induced airway inflammation, as evidenced by a reduction in eosinophil infiltration, levels of OVA-specific Immunoglobulin E, interferon-γ, interleukin (IL)-9, and IL-17A, along with an elevation in IL-10. The equilibrium between Th17 and Treg cells plays a pivotal role in modulating the abundance of inflammatory cells within the organism, thereby ameliorating inflammation and alleviating symptoms associated with allergic asthma. CONCLUSIONS AND CLINICAL RELEVANCE: Our data revealed that T. spiralis-derived Ts-SUCLA-ß protein may inhibit the allergic airway inflammation by regulating host immune responses.


Asunto(s)
Asma , Proteínas del Helminto , Ovalbúmina , Trichinella spiralis , Trichinella spiralis/inmunología , Animales , Asma/inmunología , Asma/tratamiento farmacológico , Ratones , Ovalbúmina/inmunología , Proteínas del Helminto/inmunología , Proteínas del Helminto/farmacología , Ratones Endogámicos BALB C , Modelos Animales de Enfermedad , Femenino , Citocinas/metabolismo , Citocinas/inmunología , Inmunoglobulina E/inmunología , Pulmón/inmunología , Pulmón/parasitología , Pulmón/patología , Linfocitos T Reguladores/inmunología , Hipersensibilidad/inmunología , Hipersensibilidad/tratamiento farmacológico , Células Th17/inmunología
3.
Int J Mol Sci ; 25(12)2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38928413

RESUMEN

Rheumatoid arthritis (RA) is an autoimmune disease that significantly impacts quality of life by disrupting CD4+ T cell immune homeostasis. The identification of a low-side-effect drug for RA treatment is urgently needed. Our previous study suggests that Trichinella spiralis paramyosin (Ts-Pmy) has immunomodulatory effects, but its potential effect on CD4+ T cell response in RA remains unclear. In this study, we used a murine model to investigate the role of rTs-Pmy in regulating CD4+ T cell differentiation in collagen-induced arthritis (CIA). Additionally, we assessed the impact of rTs-Pmy on CD4+ T cell differentiation towards the Th1 and Th17 phenotypes, which are associated with inflammatory responses in arthritis, using in vitro assays. The results demonstrated that rTs-Pmy administration reduced arthritis severity by inhibiting Th1 and Th17 response while enhancing Treg response. Prophylactic administration of Ts-Pmy showed superior efficacy on CIA compared to therapeutic administration. Furthermore, in vitro assays demonstrated that rTs-Pmy could inhibit the differentiation of CD4+ T cells into Th1 and Th17 while inducing the production of Tregs, suggesting a potential mechanism underlying its therapeutic effects. This study suggests that Ts-Pmy may ameliorate CIA by restoring the immune balance of CD4+ T cells and provides new insights into the mechanism through which helminth-derived proteins exert their effects on autoimmune diseases.


Asunto(s)
Artritis Experimental , Linfocitos T CD4-Positivos , Diferenciación Celular , Células Th17 , Trichinella spiralis , Tropomiosina , Animales , Trichinella spiralis/inmunología , Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Experimental/tratamiento farmacológico , Ratones , Diferenciación Celular/efectos de los fármacos , Tropomiosina/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Células TH1/inmunología , Masculino , Proteínas del Helminto/farmacología , Proteínas del Helminto/uso terapéutico , Proteínas del Helminto/inmunología , Artritis Reumatoide/inmunología , Artritis Reumatoide/tratamiento farmacológico , Linfocitos T Reguladores/inmunología , Modelos Animales de Enfermedad , Ratones Endogámicos DBA
4.
Environ Res ; 251(Pt 1): 118534, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38395336

RESUMEN

TatD DNase, a key enzyme in vertebrates and invertebrates, plays a pivotal role in various physiological processes. Dugesia japonica (D. japonica), a flatworm species, has remarkable regenerative capabilities and possesses a simplified immune system. However, the existence and biological functions of TatD DNase in D. japonica require further investigation. Here, we obtained the open reading frame (ORF) of DjTatD and demonstrated its conservation. The three-dimensional structure of DjTatD revealed its active site and binding mechanism. To investigate its enzymological properties, we overexpressed, purified, and characterized recombinant DjTatD (rDjTatD). We observed that DjTatD was primarily expressed in the pharynx and its expression could be significantly challenged upon stimulation with lipopolysaccharide, peptidoglycan, gram-positive and gram-negative bacteria. RNA interference results indicated that both DjTatD and DjDN2s play a role in pharyngeal regeneration and may serve as functional complements to each other. Additionally, we found that rDjTatD and recombinant T7DjTatD effectively reduce biofilm formation regardless of their bacterial origin. Together, our results demonstrated that DjTatD may be involved in the planarian immune response and pharyngeal regeneration. Furthermore, after further optimization in the future, rDjTatD and T7DjTatD can be considered highly effective antibiofilm agents.


Asunto(s)
Biopelículas , Desoxirribonucleasas , Planarias , Animales , Planarias/genética , Planarias/fisiología , Planarias/enzimología , Biopelículas/efectos de los fármacos , Desoxirribonucleasas/metabolismo , Desoxirribonucleasas/genética , Desoxirribonucleasas/química , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Proteínas del Helminto/química , Proteínas del Helminto/farmacología , Secuencia de Aminoácidos
5.
Mar Drugs ; 20(10)2022 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-36286436

RESUMEN

The widespread resistance to antibiotics in pathogenic bacteria makes the development of a new generation of antimicrobials an urgent task. The development of new antibiotics must be accompanied by a comprehensive study of all of their biological activities in order to avoid adverse side-effects from their application. Some promising antibiotic prototypes derived from the structures of arenicins, antimicrobial peptides from the lugworm Arenicola marina, have been developed. Previously, we described the ability of natural arenicins -1 and -2 to modulate the human complement system activation in vitro. In this regard, it seems important to evaluate the effect of therapeutically promising arenicin analogues on complement activation. Here, we describe the complement-modulating activity of three such analogues, Ar-1[V8R], ALP1, and AA139. We found that the mode of action of Ar-1[V8R] and ALP1 on the complement was similar to that of natural arenicins, which can both activate and inhibit the complement, depending on the concentration. However, Ar-1[V8R] behaved predominantly as an inhibitor, showing only a moderate increase in C3a production in the alternative pathway model and no enhancement at all of the classical pathway of complement activation. In contrast, the action of ALP1 was characterized by a marked increase in the complement activation through the classical pathway in the concentration range of 2.5-20 µg/mL. At the same time, at higher concentrations (80-160 µg/mL), this peptide exhibited a complement inhibitory effect characteristic of the other arenicins. Peptide AA139, like other arenicins, exhibited an inhibitory effect on complement at a concentration of 160 µg/mL, but was much less pronounced. Overall, our results suggest that the effect on the complement system should be taken into account in the development of antibiotics based on arenicins.


Asunto(s)
Poliquetos , Animales , Humanos , Poliquetos/química , Péptidos Catiónicos Antimicrobianos/farmacología , Estudios Prospectivos , Proteínas del Helminto/farmacología , Antibacterianos/farmacología , Antibacterianos/química , Activación de Complemento
6.
Proc Natl Acad Sci U S A ; 119(36): e2202795119, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-36037362

RESUMEN

Parasitic helminth infections, while a major cause of neglected tropical disease burden, negatively correlate with the incidence of immune-mediated inflammatory diseases such as inflammatory bowel diseases (IBD). To evade expulsion, helminths have developed sophisticated mechanisms to regulate their host's immune responses. Controlled experimental human helminth infections have been assessed clinically for treating inflammatory conditions; however, such a radical therapeutic modality has challenges. An alternative approach is to harness the immunomodulatory properties within the worm's excretory-secretory (ES) complement, its secretome. Here, we report a biologics discovery and validation pipeline to generate and screen in vivo a recombinant cell-free secretome library of helminth-derived immunomodulatory proteins. We successfully expressed 78 recombinant ES proteins from gastrointestinal hookworms and screened the crude in vitro translation reactions for anti-IBD properties in a mouse model of acute colitis. After statistical filtering and ranking, 20 proteins conferred significant protection against various parameters of colitis. Lead candidates from distinct protein families, including annexins, transthyretins, nematode-specific retinol-binding proteins, and SCP/TAPS were identified. Representative proteins were produced in mammalian cells and further validated, including ex vivo suppression of inflammatory cytokine secretion by T cells from IBD patient colon biopsies. Proteins identified herein offer promise as novel, safe, and mechanistically differentiated biologics for treating the globally increasing burden of inflammatory diseases.


Asunto(s)
Antiinflamatorios , Productos Biológicos , Colitis , Proteínas del Helminto , Enfermedades Inflamatorias del Intestino , Animales , Antiinflamatorios/farmacología , Productos Biológicos/farmacología , Colitis/tratamiento farmacológico , Proteínas del Helminto/genética , Proteínas del Helminto/farmacología , Helmintos , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/parasitología , Ratones
7.
Biochim Biophys Acta Gen Subj ; 1866(8): 130156, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35523364

RESUMEN

Arenicin-3 is an amphipathic ß-hairpin antimicrobial peptide that is produced by the lugworm Arenicola marina. In this study, we have investigated the mechanism of action of arenicin-3 and an optimized synthetic analogue, AA139, by studying their effects on lipid bilayer model membranes and Escherichia coli bacterial cells. The results show that simple amino acid changes can lead to subtle variations in their interaction with membranes and therefore alter their pre-clinical potency, selectivity and toxicity. While the mechanism of action of arenicin-3 is primarily dependent on universal membrane permeabilization, our data suggest that the analogue AA139 relies on more specific binding and insertion properties to elicit its improved antibacterial activity and lower toxicity, as exemplified by greater selectivity between lipid composition when inserting into model membranes i.e. the N-terminus of AA139 seems to insert deeper into lipid bilayers than arenicin-3 does, with a clear distinction between zwitterionic and negatively charged lipid bilayer vesicles, and AA139 demonstrates a cytoplasmic permeabilization dose response profile that is consistent with its greater antibacterial potency against E. coli cells compared to arenicin-3.


Asunto(s)
Péptidos Catiónicos Antimicrobianos , Membrana Dobles de Lípidos , Antibacterianos/química , Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/química , Péptidos Antimicrobianos , Escherichia coli/metabolismo , Proteínas del Helminto/química , Proteínas del Helminto/farmacología , Membrana Dobles de Lípidos/metabolismo
8.
J Exp Med ; 219(1)2022 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-34779829

RESUMEN

Helminth parasites are adept manipulators of the immune system, using multiple strategies to evade the host type 2 response. In the intestinal niche, the epithelium is crucial for initiating type 2 immunity via tuft cells, which together with goblet cells expand dramatically in response to the type 2 cytokines IL-4 and IL-13. However, it is not known whether helminths modulate these epithelial cell populations. In vitro, using small intestinal organoids, we found that excretory/secretory products (HpES) from Heligmosomoides polygyrus blocked the effects of IL-4/13, inhibiting tuft and goblet cell gene expression and expansion, and inducing spheroid growth characteristic of fetal epithelium and homeostatic repair. Similar outcomes were seen in organoids exposed to parasite larvae. In vivo, H. polygyrus infection inhibited tuft cell responses to heterologous Nippostrongylus brasiliensis infection or succinate, and HpES also reduced succinate-stimulated tuft cell expansion. Our results demonstrate that helminth parasites reshape their intestinal environment in a novel strategy for undermining the host protective response.


Asunto(s)
Células Epiteliales/metabolismo , Células Caliciformes/metabolismo , Intestino Delgado/citología , Organoides/metabolismo , Infecciones por Strongylida/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Epiteliales/parasitología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células Caliciformes/parasitología , Proteínas del Helminto/metabolismo , Proteínas del Helminto/farmacología , Interacciones Huésped-Parásitos , Interleucina-13/farmacología , Interleucina-4/farmacología , Intestino Delgado/parasitología , Ratones Endogámicos C57BL , Nematospiroides dubius/metabolismo , Nematospiroides dubius/fisiología , Nippostrongylus/metabolismo , Nippostrongylus/fisiología , Organoides/citología , Organoides/parasitología , Infecciones por Strongylida/parasitología , Ácido Succínico/farmacología , Transcriptoma/efectos de los fármacos
9.
PLoS Pathog ; 17(11): e1010069, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34748611

RESUMEN

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.


Asunto(s)
Antiinflamatorios/farmacología , Artritis Experimental/prevención & control , Epigénesis Genética , Fibroblastos/metabolismo , Proteínas del Helminto/farmacología , Inflamación/prevención & control , Sinoviocitos/metabolismo , Acanthocheilonema/metabolismo , Animales , Artritis Experimental/etiología , Artritis Experimental/metabolismo , Artritis Experimental/patología , Células Cultivadas , Metilación de ADN , Fibroblastos/efectos de los fármacos , Fibroblastos/inmunología , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos DBA , Sinoviocitos/efectos de los fármacos , Sinoviocitos/inmunología
10.
Exp Parasitol ; 231: 108174, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34752732

RESUMEN

In a previous study we demonstrated that Fasciola hepatica fatty acid binding protein (Fh12) significantly suppress macrophage function by inhibiting IL-6, IL-1ß, tumor necrosis factor (TNF)-α and IL-12 production in TLR4-stimulated murine macrophages, an effect mediated through the signaling of CD14 co-receptor without affecting the viability of these cells. Given that dendritic cells (DCs) are immune cells that play a central role in the initiation of primary immune responses and that are the only antigen-presenting cells capable of stimulating naïve T-cells, in the present study we investigated the effect of Fh12 on DCs. We found that Fh12 exerts a strong suppressive effect on activation and function of DCs. However, in contrast to the effect observed on macrophages, Fh12 induces early and late apoptosis of DCs being this phenomenon dose-dependent and CD14-coreceptor independent. At low concentration Fh12 modulates the LPS-induced DCs maturation status by suppressing the MHC-II, and co-stimulatory molecules CD40 and CD80 surface expression together with the pro-inflammatory cytokines IL-12p70 and IL-6 production whereas increase the IL-10 levels. Besides, Fh12 decreased the ability of LPS-activated DCs to induce IFN-γ production against allogeneic splenocytes, while increasing IL-4 production. We have described for the first time the ability of Fh12 to modify selectively the viability of DCs by apoptosis induction. The selective diminution in DCs survival could be a F. hepatica strategy in order to prevent a host immune response during the earliest phases of infection.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Fasciola hepatica/química , Proteínas de Unión a Ácidos Grasos/farmacología , Proteínas del Helminto/farmacología , Macrófagos/efectos de los fármacos , Animales , Supervivencia Celular , Femenino , Citometría de Flujo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados
11.
Exp Parasitol ; 229: 108152, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34419413

RESUMEN

Dog roundworm (Toxocara canis) is the major causative agent of toxocarosis, a parasitic disease of both veterinary and medical importance. Knowledge gaps in fundamental and applied aspects hinder the control of this important zoonotic disease. To have a better understanding of Toxocara infection and host immune responses, mouse macrophages were exposed to excretory/secretory (ES) proteins released by adult worms of T. canis in vitro. The messenger RNA transcription and protein expression of nucleotide-binding oligomerization domain-containing protein 1 (NOD1), receptor interacting protein 2 (RIP2) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in macrophages were analysed using quantitative real-time PCR (qRT-PCR) and Western blot. The levels of tumour necrosis factor alpha (TNF-ɑ), interleukin-1 beta (IL-1ß) and IL-6 released by the stimulated macrophages were analysed using enzyme-linked immunosorbent assay. It was found that 20 µg/mL ES proteins of adult T. canis induced the expression of NOD1, RIP2 and NF-κB in mouse macrophages at both transcriptional and translational levels after 9 h of incubation in vitro. Incubation with 20 µg/mL ES proteins also modulated the production of pro-inflammatory cytokines TNF-ɑ, IL-1ß and IL-6 by the macrophages. Taken together, ES proteins of adult T. canis appeared to be able to affect the macrophage NOD1-RIP2-NF-κB signalling pathway, which might play a role in regulating the production of proinflammatory cytokines. Further investigation of these aspects should lead to a better understanding of immune recognition of and modulation by Toxocara canis in host animals.


Asunto(s)
Citocinas/biosíntesis , Proteínas del Helminto/metabolismo , Macrófagos Peritoneales/metabolismo , Toxocara canis/metabolismo , Animales , Western Blotting , Supervivencia Celular , Citocinas/metabolismo , Perros , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Femenino , Proteínas del Helminto/farmacología , Interleucina-1beta/biosíntesis , Interleucina-1beta/metabolismo , Interleucina-6/biosíntesis , Interleucina-6/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/parasitología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD1/metabolismo , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Toxocara canis/química , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo
12.
Korean J Parasitol ; 59(2): 173-178, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33951774

RESUMEN

The DM9 domain is a protein unit of 60-75 amino acids that has been first detected in the fruit fly Drosophila as a repeated motif of unknown function. Recent research on proteins carrying DM9 domains in the mosquito Anopheles gambiae and the oyster Crassostrea gigas indicated an association with the uptake of microbial organisms. Likewise, in the trematode Fasciola gigantica DM9-1 showed intracellular relocalization following microbial, heat and drug stress. In the present research, we show that FgDM9-1 is a lectin with a novel mannose-binding site that has been recently described for the protein CGL1 of Crassostrea gigas. This property allowed FgDM9-1 to agglutinate gram-positive and -negative bacteria with appropriate cell surface glycosylation patterns. Furthermore, FgDM9-1 caused hemagglutination across all ABO blood group phenotypes. It is speculated that the parenchymal located FgDM9-1 has a role in cellular processes that involve the transport of mannose-carrying molecules in the parenchymal cells of the parasite.


Asunto(s)
Proteínas del Helminto/farmacología , Lectina de Unión a Manosa/farmacología , Aglutinación , Secuencia de Aminoácidos , Animales , Bacterias/citología , Bacterias/efectos de los fármacos , Eritrocitos/citología , Eritrocitos/efectos de los fármacos , Fasciola/química , Fasciola/genética , Proteínas del Helminto/química , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Hemaglutinación/efectos de los fármacos , Humanos , Lectina de Unión a Manosa/química , Lectina de Unión a Manosa/genética , Lectina de Unión a Manosa/metabolismo , Alineación de Secuencia , Streptococcus/citología , Streptococcus/efectos de los fármacos
13.
Front Immunol ; 12: 615369, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33717104

RESUMEN

Helminth infections and their components have been shown to have the potential to modulate and attenuate immune responses. The objective of this study was to evaluate the potential protective effects of Clonorchis sinensis-derived protein (CSp) on ankylosing spondylitis (AS). Cytotoxicity of CSp at different doses was assessed by MTS and flow cytometry before performing experiments. Peripheral blood mononuclear cells (PBMCs) and synovial fluid mononuclear cells (SFMCs) were obtained from AS patients. Inflammatory cytokine-producing cells were analyzed using flow cytometry. The levels of INF- γ , IL-17A, TNF-α, and IL-6 were measured by enzyme-linked immunosorbent assay (ELISA). SKG mice were treated with CSp or vehicles. Inflammation and new bone formation were evaluated using immunohistochemistry, positron emission tomography (PET), and micro-computed tomography (CT). Treatment with CSp resulted in no reduced cell viability of PBMCs or SFMCs until 24 h. In experiments culturing PBMCs and SFMCs, the frequencies of IFN- γ and IL-17A producing cells were significantly reduced after CSp treatment. In the SKG mouse model, CSp treatment significantly suppressed arthritis, enthesitis, and enteritis. Micro-CT analysis of hind paw revealed reduced new bone formation in CSp-treated mice than in vehicle-treated mice. We provide the first evidence demonstrating that CSp can ameliorate clinical signs and cytokine derangements in AS. In addition, such CSp treatment could reduce the new bone formation of AS.


Asunto(s)
Antiinflamatorios/farmacología , Clonorchis sinensis/fisiología , Proteínas del Helminto/farmacología , Osteogénesis/efectos de los fármacos , Espondilitis Anquilosante/tratamiento farmacológico , Espondilitis Anquilosante/metabolismo , Adolescente , Adulto , Animales , Presentación de Antígeno/inmunología , Antígenos Helmínticos/inmunología , Biomarcadores , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunohistoquímica , Mediadores de Inflamación/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Masculino , Ratones , Tomografía Computarizada por Tomografía de Emisión de Positrones , Espondilitis Anquilosante/diagnóstico , Espondilitis Anquilosante/etiología , Microtomografía por Rayos X , Adulto Joven
14.
Mol Med Rep ; 23(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33576450

RESUMEN

Abnormal osteoclastic activation and secretion of cysteine proteinases result in excessive bone resorption, which is one of the primary factors in the development of bone metabolic disorders, such as rheumatoid arthritis and osteoporosis. Mammalian cystatins have been demonstrated to restrain osteoclastic bone resorption and to alleviate severe osteolytic destruction via blocking the activity of cysteine proteinases. However, the specific effects of parasite cystatins on the formation and function of osteoclasts remain unclear. The purpose of the current study was to explore the effects of cystatins from Schistosoma japonicum (Sj­Cys) on macrophage colony­stimulating factor (M­CSF) and receptor activator of NF­κB ligand (RANKL)­induced osteoclast differentiation, as well as the underlying molecular mechanisms. Recombinant Sj­Cys (rSj­Cys) dose­dependently restrained osteoclast formation, with a half­maximal inhibitory concentration (IC50) value of 0.3 µM, and suppressed osteoclastic bone resorptive capability in vitro. The findings were based on tartrate resistant acid phosphatase (TRAP) staining and bone resorption assays, respectively. However, the cell viability assay showed that the repression of rSj­Cys on osteoclast formation did not depend on effects on cell viability or apoptosis. Based on the results of reverse transcription­quantitative PCR and western blot analysis, it was found that rSj­Cys downregulated the expression levels of osteoclastogenesis­related genes and proteins, by interfering with M­CSF and RANKL­induced NF­κB signaling and downstream transcription factors during early­phase osteoclastogenesis. Overall, the results of the present study revealed that rSj­Cys exerted an inhibitory role in osteoclast differentiation and could be a prospective biotherapeutic candidate for the treatment and prevention of bone metabolic disorders.


Asunto(s)
Cistatinas/farmacología , Proteínas del Helminto/farmacología , FN-kappa B/metabolismo , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Resorción Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Cistatinas/genética , Proteínas del Helminto/genética , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Osteoclastos/citología , Osteoclastos/metabolismo , Ligando RANK/farmacología , Células RAW 264.7 , Proteínas Recombinantes/farmacología , Schistosoma japonicum/genética , Schistosoma japonicum/metabolismo
15.
FASEB J ; 35(2): e21331, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33476078

RESUMEN

Type 2 immunity plays an essential role in the maintenance of metabolic homeostasis and its disruption during obesity promotes meta-inflammation and insulin resistance. Infection with the helminth parasite Schistosoma mansoni and treatment with its soluble egg antigens (SEA) induce a type 2 immune response in metabolic organs and improve insulin sensitivity and glucose tolerance in obese mice, yet, a causal relationship remains unproven. Here, we investigated the effects and underlying mechanisms of the T2 ribonuclease omega-1 (ω1), one of the major S mansoni immunomodulatory glycoproteins, on metabolic homeostasis. We show that treatment of obese mice with plant-produced recombinant ω1, harboring similar glycan motifs as present on the native molecule, decreased body fat mass, and improved systemic insulin sensitivity and glucose tolerance in a time- and dose-dependent manner. This effect was associated with an increase in white adipose tissue (WAT) type 2 T helper cells, eosinophils, and alternatively activated macrophages, without affecting type 2 innate lymphoid cells. In contrast to SEA, the metabolic effects of ω1 were still observed in obese STAT6-deficient mice with impaired type 2 immunity, indicating that its metabolic effects are independent of the type 2 immune response. Instead, we found that ω1 inhibited food intake, without affecting locomotor activity, WAT thermogenic capacity or whole-body energy expenditure, an effect also occurring in leptin receptor-deficient obese and hyperphagic db/db mice. Altogether, we demonstrate that while the helminth glycoprotein ω1 can induce type 2 immunity, it improves whole-body metabolic homeostasis in obese mice by inhibiting food intake via a STAT6-independent mechanism.


Asunto(s)
Ingestión de Alimentos , Endorribonucleasas/uso terapéutico , Glicoproteínas/uso terapéutico , Proteínas del Helminto/uso terapéutico , Obesidad/tratamiento farmacológico , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Células Cultivadas , Endorribonucleasas/farmacología , Glicoproteínas/farmacología , Proteínas del Helminto/farmacología , Locomoción , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Schistosoma mansoni/enzimología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Termogénesis , Nicotiana/genética , Nicotiana/metabolismo
16.
Mol Biochem Parasitol ; 242: 111351, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33428949

RESUMEN

The genus Echinococcus of cestode parasites includes important pathogens of humans and livestock animals. Transcriptomic and genomic studies on E. granulosus and E. multilocularis uncovered striking expansion of monodomain Kunitz proteins. This expansion is accompanied by the specialization of some family members away from the ancestral protease inhibition function to fulfill cation channel blockade functions. Since cation channels are involved in immune processes, we tested the effects on macrophage physiology of two E. granulosus Kunitz-type inhibitors of voltage-activated cation channels (Kv) that are close paralogs. Both inhibitors, EgKU-1 and EgKU-4, inhibited production of the Th1/Th17 cytokine subunit IL-12/23p40 by macrophages stimulated with the TLR4 agonist LPS. In addition, EgKU-4 but not EgKU-1 inhibited production of the inflammatory cytokine IL-6. These activities were not displayed by EgKU-3, a family member that is a protease inhibitor without known activity on cation channels. EgKU-4 potently inhibited macrophage proliferation in response to M-CSF, whereas EgKU-1 displayed similar activity but with much lower potency, similar to EgKU-3. We discuss structural differences, including a heavily cationic C-terminal extension present in EgKU-4 but not in EgKU-1, that may explain the differential activities of the two close paralogs.


Asunto(s)
Echinococcus granulosus/química , Proteínas del Helminto/farmacología , Interleucina-12/antagonistas & inhibidores , Interleucina-6/antagonistas & inhibidores , Macrófagos/efectos de los fármacos , Proteínas Inhibidoras de Proteinasas Secretoras/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica , Proteínas del Helminto/aislamiento & purificación , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-12/genética , Interleucina-12/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Canal de Potasio Kv1.3/genética , Canal de Potasio Kv1.3/inmunología , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/citología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Cultivo Primario de Células , Proteínas Inhibidoras de Proteinasas Secretoras/aislamiento & purificación , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
17.
Front Immunol ; 11: 605288, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33304354

RESUMEN

Schistosomiasis threatens 800 million people worldwide. Chronic pathology manifests as hepatosplenomegaly, and intestinal schistosomiasis caused by Schistosoma mansoni can lead to liver fibrosis, cirrhosis, and blood in the stool. To assist the only FDA-approved drug, praziquantel, in parasite elimination, the development of a vaccine would be of high value. S. mansoni Cathepsin B (SmCB) is a well-documented vaccine target for intestinal schistosomiasis. Herein, we test the increased efficacy and immunogenicity of SmCB when combined with sulfated lactosyl archaeol (SLA) archaeosomes or AddaVax™ (a squalene based oil-in-water emulsion). Both vaccine formulations resulted in robust humoral and cell mediated immune responses. Impressively, both formulations were able to reduce parasite burden greater than 40% (WHO standard), with AddaVax™ reaching 86.8%. Additionally, SmCB with both adjuvants were able to reduce granuloma size and the amount of larval parasite hatched from feces, which would reduce transmission. Our data support SmCB as a target for S. mansoni vaccination; especially when used in an adjuvanted formulation.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antígenos Arqueales/farmacología , Catepsina B/farmacología , Proteínas del Helminto/farmacología , Lípidos/farmacología , Polisorbatos/farmacología , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/prevención & control , Escualeno/farmacología , Vacunas Sintéticas/farmacología , Animales , Anticuerpos/sangre , Catepsina B/inmunología , Células Cultivadas , Citocinas/metabolismo , Composición de Medicamentos , Femenino , Proteínas del Helminto/inmunología , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Inmunización , Inmunogenicidad Vacunal , Ratones Endogámicos C57BL , Recuento de Huevos de Parásitos , Schistosoma mansoni/enzimología , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/parasitología , Caracoles , Vacunas Sintéticas/inmunología
18.
Mar Drugs ; 18(12)2020 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-33321960

RESUMEN

Antimicrobial peptides (AMPs) are not only cytotoxic towards host pathogens or cancer cells but also are able to act as immunomodulators. It was shown that some human and non-human AMPs can interact with complement proteins and thereby modulate complement activity. Thus, AMPs could be considered as the base for complement-targeted therapeutics development. Arenicins from the sea polychaete Arenicola marina, the classical example of peptides with a ß-hairpin structure stabilized by a disulfide bond, were shown earlier to be among the most prospective regulators. Here, we investigate the link between arenicins' structure and their antimicrobial, hemolytic and complement-modulating activities using the derivative Ar-1-(C/A) without a disulfide bond. Despite the absence of this bond, the peptide retains all important functional activities and also appears less hemolytic in comparison with the natural forms. These findings could help to investigate new complement drugs for regulation using arenicin derivatives.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Activación de Complemento/efectos de los fármacos , Inactivadores del Complemento/farmacología , Proteínas del Helminto/farmacología , Animales , Antibacterianos/química , Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/toxicidad , Inactivadores del Complemento/química , Inactivadores del Complemento/toxicidad , Eritrocitos/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Proteínas del Helminto/química , Proteínas del Helminto/toxicidad , Hemólisis/efectos de los fármacos , Humanos , Listeria monocytogenes/efectos de los fármacos , Listeria monocytogenes/crecimiento & desarrollo , Conformación Proteica , Conejos , Oveja Doméstica , Relación Estructura-Actividad
19.
Mol Pain ; 16: 1744806920970099, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33342372

RESUMEN

The transient receptor potential cation channel subfamily V member 1 (TRPV1) receptor is an important mediator of nociception and its expression is enriched in nociceptive neurons. TRPV1 signaling has been implicated in bladder pain and is a potential analgesic target. Resiniferatoxin is the most potent known agonist of TRPV1. Acute exposure of the rat bladder to resiniferatoxin has been demonstrated to result in pain-related freezing and licking behaviors that are alleviated by virally encoded IL-4. The interleukin-4-inducing principle of Schistosoma mansoni eggs (IPSE) is a powerful inducer of IL-4 secretion, and is also known to alter host cell transcription through a nuclear localization sequence-based mechanism. We previously reported that IPSE ameliorates ifosfamide-induced bladder pain in an IL-4- and nuclear localization sequence-dependent manner. We hypothesized that pre-administration of IPSE to resiniferatoxin-challenged mice would dampen pain-related behaviors. IPSE indeed lessened resiniferatoxin-triggered freezing behaviors in mice. This was a nuclear localization sequence-dependent phenomenon, since administration of a nuclear localization sequence mutant version of IPSE abrogated IPSE's analgesic effect. In contrast, IPSE's analgesic effect did not seem IL-4-dependent, since use of anti-IL-4 antibody in mice given both IPSE and resiniferatoxin did not significantly affect freezing behaviors. RNA-Seq analysis of resiniferatoxin- and IPSE-exposed bladders revealed differential expression of TNF/NF-κb-related signaling pathway genes. In vitro testing of IPSE uptake by urothelial cells and TRPV1-expressing neuronal cells showed uptake by both cell types. Thus, IPSE's nuclear localization sequence-dependent therapeutic effects on TRPV1-mediated bladder pain may act on TRPV1-expressing neurons and/or may rely upon urothelial mechanisms.


Asunto(s)
Diterpenos/efectos adversos , Proteínas del Huevo/uso terapéutico , Proteínas del Helminto/uso terapéutico , Interacciones Huésped-Parásitos/inmunología , Factores Inmunológicos/uso terapéutico , Dolor/tratamiento farmacológico , Parásitos/química , Vejiga Urinaria/patología , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proteínas del Huevo/farmacología , Endocitosis/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas del Helminto/farmacología , Humanos , Factores Inmunológicos/farmacología , Interleucina-4/metabolismo , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Señales de Localización Nuclear/metabolismo , Dolor/genética , Análisis de Componente Principal , Transporte de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Vejiga Urinaria/efectos de los fármacos , Urotelio/metabolismo
20.
Sci Rep ; 10(1): 20283, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-33219293

RESUMEN

Tolerogenic dendritic cells (tolDCs) are central players in the maintenance of immune tolerance and thereby have been identified as the most favourable candidates for cell therapy of autoimmune diseases. We have recently shown that excretory-secretory products (ES L1) released by Trichinella spiralis larvae induce stable human tolDCs in vitro via Toll-like receptor 2 (TLR2) and TLR4. However, engagement of these receptors did not fully explain the tolerogenic profile of DCs. Here, we observed for the first time that dendritic cell-specific ICAM-3 grabbing non-integrin (DC-SIGN) interacts with highly glycosylated ES L1 and contributes to the generation of ES L1-induced tolDCs. Blocking DC-SIGN interfered with the ES L1-induced higher expression of CD40 and CCR7 and the production of IL-10 and TGF-ß by DCs. The cooperation of TLR2, TLR4 and DC-SIGN receptors is of importance for the capacity of DCs to prime T cell response toward Th2 and to induce expansion of CD4+CD25+Foxp3+ T cells, as well as for the production of IL-10 and TGF-ß by these cells. Overall, these results indicate that induction of tolDCs by ES L1 involves engagement of multiple pattern recognition receptors namely, TLR2, TLR4 and DC-SIGN.


Asunto(s)
Antígenos Helmínticos/farmacología , Moléculas de Adhesión Celular/metabolismo , Células Dendríticas/efectos de los fármacos , Proteínas del Helminto/farmacología , Tolerancia Inmunológica/efectos de los fármacos , Lectinas Tipo C/metabolismo , Receptores de Superficie Celular/metabolismo , Trichinella spiralis/inmunología , Animales , Antígenos Helmínticos/aislamiento & purificación , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Voluntarios Sanos , Proteínas del Helminto/aislamiento & purificación , Humanos , Larva/inmunología , Larva/metabolismo , Masculino , Cultivo Primario de Células , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...