Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.405
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
PLoS Pathog ; 20(8): e1012479, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39178325

RESUMO

Alveolar echinococcosis (AE) is a highly lethal helminth infection. Current chemotherapeutic strategies for AE primarily involve the use of benzimidazoles (BZs) such as mebendazole (MDZ) and albendazole (ABZ), which exhibit limited efficacy. In a previous study, the vaccine of recombinant Echinococcus granulosus P29 (rEgP29) showed significant immunoprotection against E. granulosus in both mice and sheep. In the current study, we utilized hybridoma technology to generate five monoclonal antibodies (mAbs) against P29, among which 4G10F4 mAb exhibited the highest antigen-specific binding capacity. This mAb was selected for further investigation of anti-AE therapy, both in vivo and in vitro. In vitro, 4G10F4 inhibited a noteworthy inhibition of E. multilocularis protoscoleces and primary cells viability through complement-dependent cytotoxicity (CDC) mechanism. In vivo, two experiments were conducted. In the first experiment, mice were intraperitoneally injected with Em protoscoleces, and subsequently treated with 4G10F4 mAb (2.5/5/10 mg/kg) at 12 weeks postinfection once per week for 8 times via tail vein injection. Mice that were treated with 4G10F4 mAb only in dosage of 5mg/kg exhibited a significant lower mean parasite burden (0.89±0.97 g) compared to isotype mAb treated control mice (2.21±1.30 g). In the second experiment, mice were infected through hepatic portal vein and treated with 4G10F4 mAb (5mg/kg) at one week after surgery once per week for 8 times. The numbers of hepatic metacestode lesions of the 4G10F4 treatment group were significantly lower in comparison to the isotype control group. Pathological analysis revealed severe disruption of the inner structure of the metacestode in both experiments, particularly affecting the germinal and laminated layers, resulting in the transformation into infertile vesicles after treatment with 4G10F4. In addition, the safety of 4G10F4 for AE treatment was confirmed through assessment of mouse weight and evaluation of liver and kidney function. This study presents antigen-specific monoclonal antibody immunotherapy as a promising therapeutic approach against E. multilocularis induced AE.


Assuntos
Anticorpos Monoclonais , Equinococose , Animais , Equinococose/tratamento farmacológico , Equinococose/imunologia , Anticorpos Monoclonais/farmacologia , Camundongos , Proteínas de Helminto/imunologia , Proteínas de Helminto/farmacologia , Camundongos Endogâmicos BALB C , Echinococcus multilocularis/imunologia , Echinococcus multilocularis/efeitos dos fármacos , Feminino , Echinococcus granulosus/imunologia , Ovinos , Anticorpos Anti-Helmínticos/imunologia , Antígenos de Helmintos/imunologia
2.
PLoS Pathog ; 20(5): e1012268, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38814989

RESUMO

The eggs of the blood fluke Schistosoma mansoni are the main cause of the clinical manifestations of chronic schistosomiasis. After laying, the egg "winners" attach to the endothelium of the mesenteric vein and, after a period of development, induce the growth of a small granuloma, which facilitates their passage to the intestinal lumen. Egg "losers" carried by the bloodstream to non-specific tissues also undergo full development and induce large granuloma formation, but their life ends there. Although these trapped eggs represent a dead end in the parasite life cycle, the vast majority of studies attempting to describe the biology of the S. mansoni eggs have studied these liver-trapped "losers" instead of migrating intestinal "winners". This raises the fundamental question of how these eggs differ. With robust comparative transcriptomic analysis performed on S. mansoni eggs isolated 7 weeks post infection, we show that gene expression is critically dependent on tissue localization, both in the early and late stages of development. While mitochondrial genes and venom allergen-like proteins are significantly upregulated in mature intestinal eggs, well-described egg immunomodulators IPSE/alpha-1 and omega-1, together with micro-exon genes, are predominantly expressed in liver eggs. In addition, several proteases and protease inhibitors previously implicated in egg-host interactions display clear tissue-specific gene expression patterns. These major differences in gene expression could be then reflected in the observed different ability of liver and intestinal soluble egg antigens to elicit host immune responses and in the shorter viability of miracidia hatched from liver eggs. Our comparative analysis provides a new perspective on the biology of parasite's eggs in the context of their development and tissue localization. These findings could contribute to a broader and more accurate understanding of parasite eggs interactions with the host, which have historically been often restricted to liver eggs and sometimes inaccurately generalized.


Assuntos
Fígado , Schistosoma mansoni , Esquistossomose mansoni , Animais , Schistosoma mansoni/imunologia , Schistosoma mansoni/genética , Fígado/parasitologia , Fígado/imunologia , Fígado/metabolismo , Esquistossomose mansoni/imunologia , Esquistossomose mansoni/parasitologia , Camundongos , Óvulo/metabolismo , Óvulo/imunologia , Intestinos/parasitologia , Intestinos/imunologia , Antígenos de Helmintos/imunologia , Proteínas de Helminto/genética , Proteínas de Helminto/metabolismo , Proteínas de Helminto/imunologia , Feminino , Proteínas do Ovo
3.
Immunity ; 47(4): 739-751.e5, 2017 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-29045903

RESUMO

Infection by helminth parasites is associated with amelioration of allergic reactivity, but mechanistic insights into this association are lacking. Products secreted by the mouse parasite Heligmosomoides polygyrus suppress type 2 (allergic) immune responses through interference in the interleukin-33 (IL-33) pathway. Here, we identified H. polygyrus Alarmin Release Inhibitor (HpARI), an IL-33-suppressive 26-kDa protein, containing three predicted complement control protein (CCP) modules. In vivo, recombinant HpARI abrogated IL-33, group 2 innate lymphoid cell (ILC2) and eosinophilic responses to Alternaria allergen administration, and diminished eosinophilic responses to Nippostrongylus brasiliensis, increasing parasite burden. HpARI bound directly to both mouse and human IL-33 (in the cytokine's activated state) and also to nuclear DNA via its N-terminal CCP module pair (CCP1/2), tethering active IL-33 within necrotic cells, preventing its release, and forestalling initiation of type 2 allergic responses. Thus, HpARI employs a novel molecular strategy to suppress type 2 immunity in both infection and allergy.


Assuntos
Proteínas de Helminto/imunologia , Interleucina-33/imunologia , Nematospiroides dubius/imunologia , Infecções por Strongylida/imunologia , Alérgenos/imunologia , Alternaria/imunologia , Sequência de Aminoácidos , Animais , Western Blotting , Eosinófilos/imunologia , Proteínas de Helminto/genética , Proteínas de Helminto/metabolismo , Interações Hospedeiro-Parasita/imunologia , Humanos , Imunidade Inata/imunologia , Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-33/genética , Interleucina-33/metabolismo , Linfócitos/imunologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nematospiroides dubius/genética , Nematospiroides dubius/metabolismo , Ligação Proteica/imunologia , Receptores de Interleucina/imunologia , Receptores de Interleucina/metabolismo , Homologia de Sequência de Aminoácidos , Infecções por Strongylida/metabolismo , Infecções por Strongylida/parasitologia
4.
PLoS Pathog ; 18(1): e1010226, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35007288

RESUMO

The complement system is a first-line innate host immune defence against invading pathogens. It is activated via three pathways, termed Classical, Lectin and Alternative, which are mediated by antibodies, carbohydrate arrays or microbial liposaccharides, respectively. The three complement pathways converge in the formation of C3-convertase followed by the assembly of a lethal pore-like structure, the membrane attack complex (MAC), on the pathogen surface. We found that the infectious stage of the helminth parasite Fasciola hepatica, the newly excysted juvenile (NEJ), is resistant to the damaging effects of complement. Despite being coated with mannosylated proteins, the main initiator of the Lectin pathway, the mannose binding lectin (MBL), does not bind to the surface of live NEJ. In addition, we found that recombinantly expressed serine protease inhibitors secreted by NEJ (rFhSrp1 and rFhSrp2) selectively prevent activation of the complement via the Lectin pathway. Our experiments demonstrate that rFhSrp1 and rFhSrp2 inhibit native and recombinant MBL-associated serine proteases (MASPs), impairing the primary step that mediates C3b and C4b deposition on the NEJ surface. Indeed, immunofluorescence studies show that MBL, C3b, C4b or MAC are not deposited on the surface of NEJ incubated in normal human serum. Taken together, our findings uncover new means by which a helminth parasite prevents the activation of the Lectin complement pathway to become refractory to killing via this host response, in spite of presenting an assortment of glycans on their surface.


Assuntos
Proteínas do Sistema Complemento/imunologia , Fasciola hepatica/imunologia , Proteínas de Helminto/imunologia , Lectina de Ligação a Manose/imunologia , Serina Proteases Associadas a Proteína de Ligação a Manose/imunologia , Animais , Proteínas de Helminto/metabolismo , Humanos , Imunidade Inata/imunologia , Lectina de Ligação a Manose/metabolismo , Serina Proteases Associadas a Proteína de Ligação a Manose/metabolismo , Serpinas/imunologia , Serpinas/metabolismo
5.
Int J Mol Sci ; 25(12)2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38928413

RESUMO

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.


Assuntos
Artrite Experimental , Linfócitos T CD4-Positivos , Diferenciação Celular , Células Th17 , Trichinella spiralis , Tropomiosina , Animais , Trichinella spiralis/imunologia , Artrite Experimental/imunologia , Artrite Experimental/patologia , Artrite Experimental/tratamento farmacológico , Camundongos , Diferenciação Celular/efeitos dos fármacos , Tropomiosina/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Células Th1/imunologia , Masculino , Proteínas de Helminto/farmacologia , Proteínas de Helminto/uso terapêutico , Proteínas de Helminto/imunologia , Artrite Reumatoide/imunologia , Artrite Reumatoide/tratamento farmacológico , Linfócitos T Reguladores/imunologia , Modelos Animais de Doenças , Camundongos Endogâmicos DBA
6.
J Biol Chem ; 298(6): 101994, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35500648

RESUMO

The mouse intestinal helminth Heligmosomoides polygyrus modulates host immune responses by secreting a transforming growth factor (TGF)-ß mimic (TGM), to expand the population of Foxp3+ Tregs. TGM comprises five complement control protein (CCP)-like domains, designated D1-D5. Though lacking homology to TGF-ß, TGM binds directly to the TGF-ß receptors TßRI and TßRII and stimulates the differentiation of naïve T-cells into Tregs. However, the molecular determinants of binding are unclear. Here, we used surface plasmon resonance, isothermal calorimetry, NMR spectroscopy, and mutagenesis to investigate how TGM binds the TGF-ß receptors. We demonstrate that binding is modular, with D1-D2 binding to TßRI and D3 binding to TßRII. D1-D2 and D3 were further shown to compete with TGF-ß(TßRII)2 and TGF-ß for binding to TßRI and TßRII, respectively. The solution structure of TGM-D3 revealed that TGM adopts a CCP-like fold but is also modified to allow the C-terminal strand to diverge, leading to an expansion of the domain and opening potential interaction surfaces. TGM-D3 also incorporates a long structurally ordered hypervariable loop, adding further potential interaction sites. Through NMR shift perturbations and binding studies of TGM-D3 and TßRII variants, TGM-D3 was shown to occupy the same site of TßRII as bound by TGF-ß using both a novel interaction surface and the hypervariable loop. These results, together with the identification of other secreted CCP-like proteins with immunomodulatory activity in H. polygyrus, suggest that TGM is part of a larger family of evolutionarily plastic parasite effector molecules that mediate novel interactions with their host.


Assuntos
Proteínas de Helminto , Interações Hospedeiro-Patógeno , Nematospiroides dubius , Receptores de Fatores de Crescimento Transformadores beta , Fator de Crescimento Transformador beta , Animais , Evolução Biológica , Proteínas de Helminto/genética , Proteínas de Helminto/imunologia , Interações Hospedeiro-Patógeno/imunologia , Camundongos , Nematospiroides dubius/classificação , Nematospiroides dubius/genética , Nematospiroides dubius/imunologia , Nematospiroides dubius/metabolismo , Ligação Proteica , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta/metabolismo
7.
PLoS Pathog ; 17(12): e1010064, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34969052

RESUMO

Schistosomes are long lived, intravascular parasitic platyhelminths that infect >200 million people globally. The molecular mechanisms used by these blood flukes to dampen host immune responses are described in this review. Adult worms express a collection of host-interactive tegumental ectoenzymes that can cleave host signaling molecules such as the "alarmin" ATP (cleaved by SmATPDase1), the platelet activator ADP (SmATPDase1, SmNPP5), and can convert AMP into the anti-inflammatory mediator adenosine (SmAP). SmAP can additionally cleave the lipid immunomodulator sphingosine-1-phosphate and the proinflammatory anionic polymer, polyP. In addition, the worms release a barrage of proteins (e.g., SmCB1, SjHSP70, cyclophilin A) that can impinge on immune cell function. Parasite eggs also release their own immunoregulatory proteins (e.g., IPSE/α1, omega1, SmCKBP) as do invasive cercariae (e.g., Sm16, Sj16). Some schistosome glycans (e.g., LNFPIII, LNnT) and lipids (e.g., Lyso-PS, LPC), produced by several life stages, likewise affect immune cell responses. The parasites not only produce eicosanoids (e.g., PGE2, PGD2-that can be anti-inflammatory) but can also induce host cells to release these metabolites. Finally, the worms release extracellular vesicles (EVs) containing microRNAs, and these too have been shown to skew host cell metabolism. Thus, schistosomes employ an array of biomolecules-protein, lipid, glycan, nucleic acid, and more, to bend host biochemistry to their liking. Many of the listed molecules have been individually shown capable of inducing aspects of the polarized Th2 response seen following infection (with the generation of regulatory T cells (Tregs), regulatory B cells (Bregs) and anti-inflammatory, alternatively activated (M2) macrophages). Precisely how host cells integrate the impact of these myriad parasite products following natural infection is not known. Several of the schistosome immunomodulators described here are in development as novel therapeutics against autoimmune, inflammatory, and other, nonparasitic, diseases.


Assuntos
Proteínas de Helminto/imunologia , Fatores Imunológicos/imunologia , Schistosoma/imunologia , Esquistossomose/imunologia , Animais , Humanos
8.
PLoS Pathog ; 17(3): e1009337, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33651853

RESUMO

The establishment of type 2 responses driven by allergic sensitization prior to exposure to helminth parasites has demonstrated how tissue-specific responses can protect against migrating larval stages, but, as a consequence, allow for immune-mediated, parasite/allergy-associated morbidity. In this way, whether helminth cross-reacting allergen-specific antibodies are produced and play a role during the helminth infection, or exacerbate the allergic outcome awaits elucidation. Thus, the main objective of the study was to investigate whether house dust mite (HDM) sensitization triggers allergen-specific antibodies that interact with Ascaris antigens and mediate antibody-dependent deleterious effects on these parasites as well as, to assess the capacity of cross-reactive helminth proteins to trigger allergic inflammation in house dust mite presensitized mice. Here, we show that the sensitization with HDM-extract drives marked IgE and IgG1 antibody responses that cross-react with Ascaris larval antigens. Proteomic analysis of Ascaris larval antigens recognized by these HDM-specific antibodies identified Ascaris tropomyosin and enolase as the 2 major HDM homologues based on high sequence and structural similarity. Moreover, the helminth tropomyosin could drive Type-2 associated pulmonary inflammation similar to HDM following HDM tropomyosin sensitization. The HDM-triggered IgE cross-reactive antibodies were found to be functional as they mediated immediate hypersensitivity responses in skin testing. Finally, we demonstrated that HDM sensitization in either B cells or FcγRIII alpha-chain deficient mice indicated that the allergen driven cell-mediated larval killing is not antibody-dependent. Taken together, our data suggest that aeroallergen sensitization drives helminth reactive antibodies through molecular and structural similarity between HDM and Ascaris antigens suggesting that cross-reactive immune responses help drive allergic inflammation.


Assuntos
Poeira/imunologia , Hipersensibilidade/imunologia , Pyroglyphidae/imunologia , Animais , Antígenos de Dermatophagoides/imunologia , Proteínas de Helminto/imunologia , Imunoglobulina E/imunologia , Imunoglobulina G/imunologia , Camundongos , Proteômica
9.
Mol Cell Proteomics ; 20: 100055, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33581320

RESUMO

Paramphistomosis, caused by the rumen fluke, Calicophoron daubneyi, is a parasitic infection of ruminant livestock, which has seen a rapid rise in prevalence throughout Western Europe in recent years. After ingestion of metacercariae (parasite cysts) by the mammalian host, newly excysted juveniles (NEJs) emerge and invade the duodenal submucosa, which causes significant pathology in heavy infections. The immature flukes then migrate upward, along the gastrointestinal tract, and enter the rumen where they mature and begin to produce eggs. Despite their emergence, and sporadic outbreaks of acute disease, we know little about the molecular mechanisms used by C. daubneyi to establish infection, acquire nutrients, and avoid the host immune response. Here, transcriptome analysis of four intramammalian life-cycle stages, integrated with secretome analysis of the NEJ and adult parasites (responsible for acute and chronic diseases, respectively), revealed how the expression and secretion of selected families of virulence factors and immunomodulators are regulated in accordance with fluke development and migration. Our data show that while a family of cathepsins B with varying S2 subsite residues (indicating distinct substrate specificities) is differentially secreted by NEJs and adult flukes, cathepsins L and F are secreted in low abundance by NEJs only. We found that C. daubneyi has an expanded family of aspartic peptidases, which is upregulated in adult worms, although they are under-represented in the secretome. The most abundant proteins in adult fluke secretions were helminth defense molecules that likely establish an immune environment permissive to fluke survival and/or neutralize pathogen-associated molecular patterns such as bacterial lipopolysaccharide in the microbiome-rich rumen. The distinct collection of molecules secreted by C. daubneyi allowed the development of the first coproantigen-based ELISA for paramphistomosis which, importantly, did not recognize antigens from other helminths commonly found as coinfections with rumen fluke.


Assuntos
Proteínas de Helminto/genética , Proteínas de Helminto/metabolismo , Paramphistomatidae/genética , Paramphistomatidae/metabolismo , Animais , Antígenos de Helmintos/genética , Antígenos de Helmintos/imunologia , Antígenos de Helmintos/metabolismo , Bovinos , Cisteína Proteases/genética , Cisteína Proteases/metabolismo , Fezes/parasitologia , Proteínas de Helminto/imunologia , Estágios do Ciclo de Vida , Paramphistomatidae/crescimento & desenvolvimento , Rúmen/parasitologia , Secretoma , Transcriptoma , Infecções por Trematódeos/diagnóstico , Infecções por Trematódeos/imunologia , Infecções por Trematódeos/parasitologia
10.
Proc Natl Acad Sci U S A ; 117(32): 19299-19309, 2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32737161

RESUMO

Schistosomes are parasitic flatworms that cause schistosomiasis, a neglected tropical disease affecting over 200 million people. Schistosomes develop multiple body plans while navigating their complex life cycle, which involves two different hosts: a mammalian definitive host and a molluscan intermediate host. Their survival and propagation depend upon proliferation and differentiation of stem cells necessary for parasite homeostasis and reproduction. Infective larvae released from snails carry a handful of stem cells that serve as the likely source of new tissues as the parasite adapts to life inside the mammalian host; however, the role of these stem cells during this critical life cycle stage remains unclear. Here, we characterize stem cell fates during early intramammalian development. Surprisingly, we find that the esophageal gland, an accessory organ of the digestive tract, develops before the rest of the digestive system is formed and blood feeding is initiated, suggesting a role in processes beyond nutrient uptake. To explore such a role, we examine schistosomes that lack the esophageal gland due to knockdown of a forkhead-box transcription factor, Sm-foxA, which blocks development and maintenance of the esophageal gland, without affecting the development of other somatic tissues. Intriguingly, schistosomes lacking the esophageal gland die after transplantation into naive mice, but survive in immunodeficient mice lacking B cells. We show that parasites lacking the esophageal gland are unable to lyse ingested immune cells within the esophagus before passing them into the gut. These results unveil an immune-evasion mechanism mediated by the esophageal gland, which is essential for schistosome survival and pathogenesis.


Assuntos
Esôfago/parasitologia , Evasão da Resposta Imune , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Animais , Esôfago/imunologia , Feminino , Proteínas de Helminto/genética , Proteínas de Helminto/imunologia , Interações Hospedeiro-Parasita , Humanos , Estágios do Ciclo de Vida , Masculino , Camundongos , Schistosoma mansoni/genética , Schistosoma mansoni/crescimento & desenvolvimento , Esquistossomose mansoni/parasitologia , Esquistossomose mansoni/fisiopatologia
11.
PLoS Pathog ; 16(3): e1008391, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32163524

RESUMO

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.


Assuntos
Acanthocheilonema/imunologia , Acantoqueilonemíase/imunologia , Dieta Ocidental/efeitos adversos , Proteínas de Helminto/imunologia , Longevidade/imunologia , Modelos Imunológicos , Animais , Feminino , Masculino , Camundongos
12.
PLoS Pathog ; 16(5): e1008508, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32407385

RESUMO

Parasitic helminths have coevolved with humans over millennia, intricately refining and developing an array of mechanisms to suppress or skew the host's immune system, thereby promoting their long-term survival. Some helminths, such as hookworms, cause little to no overt pathology when present in modest numbers and may even confer benefits to their human host. To exploit this evolutionary phenomenon, clinical trials of human helminth infection have been established and assessed for safety and efficacy for a range of immune dysfunction diseases and have yielded mixed outcomes. Studies of live helminth therapy in mice and larger animals have convincingly shown that helminths and their excretory/secretory products possess anti-inflammatory drug-like properties and represent an untapped pharmacopeia. These anti-inflammatory moieties include extracellular vesicles, proteins, glycans, post-translational modifications, and various metabolites. Although the concept of helminth-inspired therapies holds promise, it also presents a challenge to the drug development community, which is generally unfamiliar with foreign biologics that do not behave like antibodies. Identification and characterization of helminth molecules and vesicles and the molecular pathways they target in the host present a unique opportunity to develop tailored drugs inspired by nature that are efficacious, safe, and have minimal immunogenicity. Even so, much work remains to mine and assess this out-of-the-box therapeutic modality. Industry-based organizations need to consider long-haul investments aimed at unraveling and exploiting unique and differentiated mechanisms of action as opposed to toe-dipping entries with an eye on rapid and profitable turnarounds.


Assuntos
Proteínas de Helminto/imunologia , Helmintíase/imunologia , Helmintos/imunologia , Imunomodulação , Animais , Helmintíase/patologia , Helmintíase/terapia , Helmintos/patogenicidade , Humanos
13.
PLoS Pathog ; 16(4): e1008465, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32271834

RESUMO

Roundworm parasite infections are a major cause of human and livestock disease worldwide and a threat to global food security. Disease control currently relies on anthelmintic drugs to which roundworms are becoming increasingly resistant. An alternative approach is control by vaccination and 'hidden antigens', components of the worm gut not encountered by the infected host, have been exploited to produce Barbervax, the first commercial vaccine for a gut dwelling nematode of any host. Here we present the structure of H-gal-GP, a hidden antigen from Haemonchus contortus, the Barber's Pole worm, and a major component of Barbervax. We demonstrate its novel architecture, subunit composition and topology, flexibility and heterogeneity using cryo-electron microscopy, mass spectrometry, and modelling. Importantly, we demonstrate that complexes with the same architecture are present in other Strongylid roundworm parasites including human hookworm. This suggests a common ancestry and the potential for development of a unified hidden antigen vaccine.


Assuntos
Endopeptidases/metabolismo , Endopeptidases/ultraestrutura , Haemonchus/imunologia , Proteínas de Helminto/metabolismo , Proteínas de Helminto/ultraestrutura , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/ultraestrutura , Animais , Anti-Helmínticos/farmacologia , Anticorpos Anti-Helmínticos , Antígenos de Helmintos/imunologia , Microscopia Crioeletrônica , Endopeptidases/imunologia , Haemonchus/patogenicidade , Proteínas de Helminto/imunologia , Glicoproteínas de Membrana/imunologia , Parasitos , Vacinação , Vacinas/imunologia
14.
Clin Exp Allergy ; 51(5): 703-715, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33512036

RESUMO

BACKGROUND: Serum inhibition of allergen-specific IgE has been associated with competing IgG4 and non-specific polyclonal IgE. In allergen immunotherapy, beneficial responses have been associated with high IgG4/IgE ratios. Helminths potentiate antibody class switching to IgG4 and stimulate polyclonal IgE synthesis; therefore, we hypothesized a role for helminth-associated IgG4 and total IgE in protection against atopic sensitization and clinical allergy (asthma) in tropical low-income countries. METHODS: Among community residents of Ugandan rural Schistosoma mansoni (Sm)-endemic islands and a mainland urban setting with lower helminth exposure, and among urban asthmatic schoolchildren and non-asthmatic controls, we measured total, Schistosoma adult worm antigen (SWA)-specific, Schistosoma egg antigen (SEA)-specific and allergen (house dust mite [HDM] and German cockroach)-specific IgE and IgG4 by ImmunoCAP® and/or ELISA. We assessed associations between these antibody profiles and current Sm infection, the rural-urban environment, HDM and cockroach skin prick test (SPT) reactivity, and asthma. RESULTS: Total IgE, total IgG4 and SWA-, SEA- and allergen-specific IgE and IgG4 levels were significantly higher in the rural, compared to the urban setting. In both community settings, both Sm infection and SPT reactivity were positively associated with allergen-specific and total IgE responses. SPT reactivity was inversely associated with Schistosoma-specific IgG4, allergen-specific IgG4/IgE ratios and total IgE/allergen-specific IgE ratios. Asthmatic schoolchildren, compared with non-asthmatic controls, had significantly higher levels of total and allergen-specific IgE, but lower ratios of allergen-specific IgG4/IgE and total IgE/allergen-specific IgE. CONCLUSIONS AND CLINICAL RELEVANCE: Our immuno-epidemiological data support the hypothesis that the IgG4-IgE balance and the total IgE-allergen-specific IgE balance are more important than absolute total, helminth- or allergen-specific antibody levels in inhibition of allergies in the tropics.


Assuntos
Antígenos de Dermatophagoides/imunologia , Asma/imunologia , Proteínas de Helminto/imunologia , Hipersensibilidade/imunologia , Imunoglobulina E/imunologia , Imunoglobulina G/imunologia , Proteínas de Insetos/imunologia , Adolescente , Alérgenos/imunologia , Animais , Asma/epidemiologia , Blattellidae , Criança , Pré-Escolar , Feminino , Humanos , Hipersensibilidade/epidemiologia , Masculino , População Rural , Schistosoma mansoni , Testes Cutâneos , Uganda/epidemiologia , População Urbana
15.
BMC Microbiol ; 21(1): 5, 2021 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-33407120

RESUMO

BACKGROUND: During the last two decades research on animal filarial parasites, especially Onchocerca ochengi, infecting cattle in savanna areas of Africa revealed that O. ochengi as an animal model has biological features that are similar to those of O. volvulus, the aetiological agent of human onchocerciasis. There is, however, a paucity of biochemical, immunological and pathological data for O. ochengi. Galectins can be generated by parasites and their hosts. They are multifunctional molecules affecting the interaction between filarial parasites and their mammalian hosts including immune responses. This study characterized O. ochengi galectin, verified its immunologenicity and established its immune reactivity and that of Onchocerca volvulus galectin. RESULTS: The phylogenetic analysis showed the high degree of identity between the identified O. ochengi and the O. volvulus galectin-1 (ß-galactoside-binding protein-1) consisting only in one exchange of alanine for serine. O. ochengi galectin induced IgG antibodies during 28 days after immunization of Wistar rats. IgG from O. ochengi-infected cattle and O. volvulus-infected humans cross-reacted with the corresponding galectins. Under the applied experimental conditions in a cell proliferation test, O. ochengi galectin failed to significantly stimulate peripheral blood mononuclear cells (PBMCs) from O. ochengi-infected cattle, regardless of their parasite load. CONCLUSION: An O. ochengi galectin gene was identified and the recombinantly expressed protein was immunogenic. IgG from Onchocerca-infected humans and cattle showed similar cross-reaction with both respective galectins. The present findings reflect the phylogenetic relationship between the two parasites and endorse the appropriateness of the cattle O. ochengi model for O. volvulus infection research.


Assuntos
Galectinas/administração & dosagem , Galectinas/genética , Imunoglobulina G/sangue , Leucócitos Mononucleares/imunologia , Onchocerca/imunologia , Animais , Bovinos , Clonagem Molecular/métodos , Feminino , Galectinas/imunologia , Perfilação da Expressão Gênica , Proteínas de Helminto/administração & dosagem , Proteínas de Helminto/genética , Proteínas de Helminto/imunologia , Humanos , Imunização , Leucócitos Mononucleares/parasitologia , Onchocerca/genética , Filogenia , Ratos , Ratos Wistar , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Análise de Sequência de DNA
16.
Biologicals ; 72: 18-26, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34229924

RESUMO

Coinfection of Leishmania with bacteria, viruses, protozoans, and nematodes alter the immune system of the host, thereby influencing the disease outcomes. Here, we have determined the immunogenic property and protective efficacy of the cross-reactive molecule HSP60 of filarial parasite B. malayi against the L. donovani in BALB/c mice. Parasitological parameters results showed a significant decrease in the parasite burden (~59%; P < 0.001) and also a substantial increase in the delayed-type hypersensitivity (DTH) response (P < 0.001) in mice immunized with 10 µg of rBmHSP60. Protection against L. donovani in mice immunized with rBmHSP60 resulted from activation of the T cells, which is characterized by higher levels of nitric oxide (NO) production, enhanced cell proliferation, higher levels (expression and release) of IFN- γ, TNF- α, and IL-12, also, higher production of IgG and IgG2a antibodies. This strong Th1 immune response creates an inflammatory domain for L. donovani and protects the host from VL.


Assuntos
Brugia Malayi , Chaperonina 60/imunologia , Proteção Cruzada , Proteínas de Helminto/imunologia , Leishmaniose Visceral , Animais , Citocinas/imunologia , Imunidade Celular , Leishmania donovani , Leishmaniose Visceral/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/imunologia , Células Th1/imunologia
17.
Parasitol Res ; 120(1): 173-185, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33079271

RESUMO

A vaccine is an important method to control schistosomiasis. Molecules related to lung-stage schistosomulum are considered potential vaccine candidates. We previously showed that glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and cathepsin L3 (CL3) displayed differential expression in the lung-stage schistosomula of Schistosoma japonicum cocultured with host cells. In the present study, we prepared the two proteins and detected the protective effects of SjGAPDH by immunizing mice with this protein alone and in combination with SjCL3 with or without Freund's adjuvant. Then, we investigated the possible mechanisms underlying S. japonicum infection. The results showed that vaccination of adjuvanted SjGAPDH decreased the worm burden (37.8%) and egg load (38.1%), and the combination of adjuvanted SjGAPDH and SjCL3 further decreased the worm burden (65.6%) and egg load (70.9%) during Schistosoma japonicum infection. However, the immunization of a combination of adjuvant-free SjGAPDH and SjCL3 displayed a lower protective effect (< 15%) than those of the adjuvanted SjCL3, the adjuvanted SjGAPDH, and a combination of adjuvanted SjGAPDH and SjCL3. Flow cytometric results showed that the frequency of regulatory T cells (Tregs) was lower (P < 0.05) in the group with adjuvanted SjGAPDH and SjCL3 (2.61%) than the remaining groups. The enzyme-linked immunosorbent assay (ELISA) results indicated that except for the uninfected and infected control groups, the remaining groups displayed a Th1-type shift in immune responses. These results showed the immunization of SjGAPDH resulted in partial protection (approximately 38%); inoculation with a combination of SjCL3 and SjGAPDH in Freund's adjuvant resulted in a high immunoprotective effect (> 65%) against Schistosoma japonicum infection in mice, which was possibly caused by the reduced percentage of Tregs and a Th1-type shift in immune responses; and SjCL3 has no adjuvant-like effect, dissimilar to SmCL3.


Assuntos
Catepsinas/imunologia , Gliceraldeído-3-Fosfato Desidrogenases/imunologia , Schistosoma japonicum/imunologia , Esquistossomose Japônica/prevenção & controle , Vacinas/imunologia , Animais , Catepsinas/administração & dosagem , Feminino , Gliceraldeído-3-Fosfato Desidrogenases/administração & dosagem , Proteínas de Helminto/administração & dosagem , Proteínas de Helminto/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T Reguladores/imunologia , Vacinação , Vacinas/administração & dosagem
18.
Int J Mol Sci ; 22(9)2021 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-33946385

RESUMO

Radopholus similis is a migratory endoparasitic nematode that is extremely harmful to host plants. Venom allergen-like proteins (VAPs) are members of the cysteine-rich secretory protein family that are widely present in plants and animals. In this study, we cloned a VAP gene from R. similis, designated as RsVAP. RsVAP contains an open reading frame of 1089 bp encoding 362 amino acids. RsVAP is specifically expressed in the esophageal gland, and the expression levels of RsVAP are significantly higher in juveniles than in other life stages of R. similis. This expression pattern of RsVAP was consistent with the biological characteristics of juveniles of R. similis, which have the ability of infection and are the main infection stages of R. similis. The pathogenicity and reproduction rate of R. similis in tomato was significantly attenuated after RsVAP was silenced. In tobacco leaves transiently expressing RsVAP, the pathogen-associated molecular pattern-triggered immunity (PTI) induced by a bacterial flagellin fragment (flg22) was inhibited, while the cell death induced by two sets of immune elicitors (BAX and Gpa2/RBP-1) was repressed. The RsVAP-interacting, ras-related protein RABA1d (LeRabA1d) was identified in tomato hosts by yeast two-hybrid and co-immunoprecipitation assays. RsVAP may interact with LeRabA1d to affect the host defense response, which in turn facilitates nematode infection. This study provides the first evidence for the inhibition of plant defense response by a VAP from migratory plant-parasitic nematodes, and, for the first time, the target protein of R. similis in its host was identified.


Assuntos
Proteínas de Helminto/imunologia , Nicotiana/imunologia , Doenças das Plantas/imunologia , Imunidade Vegetal , Solanum lycopersicum/imunologia , Tylenchida/imunologia , Animais , Interações Hospedeiro-Parasita , Solanum lycopersicum/parasitologia , Doenças das Plantas/parasitologia , Nicotiana/parasitologia , Tylenchida/fisiologia
19.
PLoS Pathog ; 14(10): e1007300, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30335852

RESUMO

Despite causing considerable damage to host tissue at the onset of parasitism, invasive helminths establish remarkably persistent infections in both animals and plants. Secretions released by these obligate parasites during host invasion are thought to be crucial for their persistence in infection. Helminth secretions are complex mixtures of molecules, most of which have unknown molecular targets and functions in host cells or tissues. Although the habitats of animal- and plant-parasitic helminths are very distinct, their secretions share the presence of a structurally conserved group of proteins called venom allergen-like proteins (VALs). Helminths abundantly secrete VALs during several stages of parasitism while inflicting extensive damage to host tissue. The tight association between the secretion of VALs and the onset of parasitism has triggered a particular interest in this group of proteins, as improved knowledge on their biological functions may assist in designing novel protection strategies against parasites in humans, livestock, and important food crops.


Assuntos
Alérgenos/imunologia , Produtos Agrícolas/imunologia , Proteínas de Helminto/imunologia , Helmintos/imunologia , Interações Hospedeiro-Parasita/imunologia , Infecções por Nematoides/parasitologia , Peçonhas/imunologia , Animais , Infecções por Nematoides/imunologia
20.
Parasite Immunol ; 42(11): e12777, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32681576

RESUMO

AIM: Identification of a 29 kDa heat stress protein in filarial parasite Setaria cervi and evaluation of its diagnostic potential against lymphatic filariasis. METHODS AND RESULTS: The Heat shock proteins (HSPs) were induced in filarial parasite S cervi by incubated at 42°C for 2 hours. The 10% SDS-PAGE of cytosolic extract showed several over-expressed bands. The MALDI-LC/MS analysis of 29 kDa band showed 100% similarity with Bm14-3-3 like protein 2. Multiple sequence alignment of Bm14-3-3 like protein 2 sequence with W bancrofti, Caenorhabditis elegans; Loa loa and Homo sapiens showed 100%, 86%, 83% and 78%, sequence similarity respectively. The antigenic efficacy of Sc14-3-3 protein was evaluated with different filarial sera using ELISA which showed cross-reactivity in order to Endemic Normal (EN) < Microfilaraemic (MF) < Chronic(CH) with IgG1 and EN < CH < MF in IgG4 ELISA. IgG1- and IgG4-specific immunoblotting with CH and MF sera further explicated its specific antigenic cross-reactivity. CONCLUSION: A 29 kDa heat shock protein of S cervi was identified as 14-3-3 protein having 100% homology to human filarial parasite B malayi. It showed strong reactivity with IgG1 and IgG4 subclass antibodies of W bancrofti-infected human sera suggesting that 14-3-3 protein could be used as a vaccine/ diagnostic marker.


Assuntos
Anticorpos Anti-Helmínticos/imunologia , Filariose Linfática/diagnóstico , Proteínas de Choque Térmico/imunologia , Imunoglobulina G/imunologia , Setaria (Nematoide)/imunologia , Wuchereria bancrofti/imunologia , Sequência de Aminoácidos , Animais , Biomarcadores/análise , Reações Cruzadas , Filariose Linfática/imunologia , Filariose Linfática/parasitologia , Feminino , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Proteínas de Helminto/genética , Proteínas de Helminto/imunologia , Proteínas de Helminto/metabolismo , Humanos , Alinhamento de Sequência , Setaria (Nematoide)/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA