Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 251
Filtrar
1.
Mar Drugs ; 20(10)2022 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-36286436

RESUMO

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.


Assuntos
Poliquetos , Animais , Humanos , Poliquetos/química , Peptídeos Catiônicos Antimicrobianos/farmacologia , Estudos Prospectivos , Proteínas de Helminto/farmacologia , Antibacterianos/farmacologia , Antibacterianos/química , Ativação do Complemento
2.
Proc Natl Acad Sci U S A ; 119(36): e2202795119, 2022 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-36037362

RESUMO

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.


Assuntos
Anti-Inflamatórios , Produtos Biológicos , Colite , Proteínas de Helminto , Doenças Inflamatórias Intestinais , Animais , Anti-Inflamatórios/farmacologia , Produtos Biológicos/farmacologia , Colite/tratamento farmacológico , Proteínas de Helminto/genética , Proteínas de Helminto/farmacologia , Helmintos , Humanos , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/parasitologia , Camundongos
3.
Biochim Biophys Acta Gen Subj ; 1866(8): 130156, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35523364

RESUMO

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.


Assuntos
Peptídeos Catiônicos Antimicrobianos , Bicamadas Lipídicas , Antibacterianos/química , Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Antimicrobianos , Escherichia coli/metabolismo , Proteínas de Helminto/química , Proteínas de Helminto/farmacologia , Bicamadas Lipídicas/metabolismo
4.
J Exp Med ; 219(1)2022 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-34779829

RESUMO

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.


Assuntos
Células Epiteliais/metabolismo , Células Caliciformes/metabolismo , Intestino Delgado/citologia , Organoides/metabolismo , Infecções por Strongylida/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Epiteliais/parasitologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Células Caliciformes/parasitologia , Proteínas de Helminto/metabolismo , Proteínas de Helminto/farmacologia , Interações Hospedeiro-Parasita , Interleucina-13/farmacologia , Interleucina-4/farmacologia , Intestino Delgado/parasitologia , Camundongos Endogâmicos C57BL , Nematospiroides dubius/metabolismo , Nematospiroides dubius/fisiologia , Nippostrongylus/metabolismo , Nippostrongylus/fisiologia , Organoides/citologia , Organoides/parasitologia , Infecções por Strongylida/parasitologia , Ácido Succínico/farmacologia , Transcriptoma/efeitos dos fármacos
5.
PLoS Pathog ; 17(11): e1010069, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34748611

RESUMO

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.


Assuntos
Anti-Inflamatórios/farmacologia , Artrite Experimental/prevenção & controle , Epigênese Genética , Fibroblastos/metabolismo , Proteínas de Helminto/farmacologia , Inflamação/prevenção & controle , Sinoviócitos/metabolismo , Acanthocheilonema/metabolismo , Animais , Artrite Experimental/etiologia , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Células Cultivadas , Metilação de DNA , Fibroblastos/efeitos dos fármacos , Fibroblastos/imunologia , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Sinoviócitos/efeitos dos fármacos , Sinoviócitos/imunologia
6.
Exp Parasitol ; 231: 108174, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34752732

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Fasciola hepatica/química , Proteínas de Ligação a Ácido Graxo/farmacologia , Proteínas de Helminto/farmacologia , Macrófagos/efeitos dos fármacos , Animais , Sobrevivência Celular , Feminino , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout
7.
Exp Parasitol ; 229: 108152, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34419413

RESUMO

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.


Assuntos
Citocinas/biossíntese , Proteínas de Helminto/metabolismo , Macrófagos Peritoneais/metabolismo , Toxocara canis/metabolismo , Animais , Western Blotting , Sobrevivência Celular , Citocinas/metabolismo , Cães , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Feminino , Proteínas de Helminto/farmacologia , Interleucina-1beta/biossíntese , Interleucina-1beta/metabolismo , Interleucina-6/biossíntese , Interleucina-6/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/metabolismo , Toxocara canis/química , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/metabolismo
8.
Korean J Parasitol ; 59(2): 173-178, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33951774

RESUMO

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.


Assuntos
Proteínas de Helminto/farmacologia , Lectina de Ligação a Manose/farmacologia , Aglutinação , Sequência de Aminoácidos , Animais , Bactérias/citologia , Bactérias/efeitos dos fármacos , Eritrócitos/citologia , Eritrócitos/efeitos dos fármacos , Fasciola/química , Fasciola/genética , Proteínas de Helminto/química , Proteínas de Helminto/genética , Proteínas de Helminto/metabolismo , Hemaglutinação/efeitos dos fármacos , Humanos , Lectina de Ligação a Manose/química , Lectina de Ligação a Manose/genética , Lectina de Ligação a Manose/metabolismo , Alinhamento de Sequência , Streptococcus/citologia , Streptococcus/efeitos dos fármacos
9.
Front Immunol ; 12: 615369, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33717104

RESUMO

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.


Assuntos
Anti-Inflamatórios/farmacologia , Clonorchis sinensis/fisiologia , Proteínas de Helminto/farmacologia , Osteogênese/efeitos dos fármacos , Espondilite Anquilosante/tratamento farmacológico , Espondilite Anquilosante/metabolismo , Adolescente , Adulto , Animais , Apresentação de Antígeno/imunologia , Antígenos de Helmintos/imunologia , Biomarcadores , Sobrevivência Celular/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Mediadores da Inflamação/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Masculino , Camundongos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Espondilite Anquilosante/diagnóstico , Espondilite Anquilosante/etiologia , Microtomografia por Raio-X , Adulto Jovem
10.
Mol Med Rep ; 23(4)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33576450

RESUMO

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.


Assuntos
Cistatinas/farmacologia , Proteínas de Helminto/farmacologia , NF-kappa B/metabolismo , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Reabsorção Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Cistatinas/genética , Proteínas de Helminto/genética , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Osteoclastos/citologia , Osteoclastos/metabolismo , Ligante RANK/farmacologia , Células RAW 264.7 , Proteínas Recombinantes/farmacologia , Schistosoma japonicum/genética , Schistosoma japonicum/metabolismo
11.
Mol Biochem Parasitol ; 242: 111351, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33428949

RESUMO

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.


Assuntos
Echinococcus granulosus/química , Proteínas de Helminto/farmacologia , Interleucina-12/antagonistas & inibidores , Interleucina-6/antagonistas & inibidores , Macrófagos/efeitos dos fármacos , Proteínas Secretadas Inibidoras de Proteinases/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Regulação da Expressão Gênica , Proteínas de Helminto/isolamento & purificação , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-12/genética , Interleucina-12/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.3/imunologia , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Cultura Primária de Células , Proteínas Secretadas Inibidoras de Proteinases/isolamento & purificação , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia
12.
FASEB J ; 35(2): e21331, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33476078

RESUMO

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.


Assuntos
Ingestão de Alimentos , Endorribonucleases/uso terapêutico , Glicoproteínas/uso terapêutico , Proteínas de Helminto/uso terapêutico , Obesidade/tratamento farmacológico , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Células Cultivadas , Endorribonucleases/farmacologia , Glicoproteínas/farmacologia , Proteínas de Helminto/farmacologia , Locomoção , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Schistosoma mansoni/enzimologia , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Termogênese , Nicotiana/genética , Nicotiana/metabolismo
13.
Mol Pain ; 16: 1744806920970099, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33342372

RESUMO

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.


Assuntos
Diterpenos/efeitos adversos , Proteínas do Ovo/uso terapêutico , Proteínas de Helminto/uso terapêutico , Interações Hospedeiro-Parasita/imunologia , Fatores Imunológicos/uso terapêutico , Dor/tratamento farmacológico , Parasitos/química , Bexiga Urinária/patologia , Animais , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Proteínas do Ovo/farmacologia , Endocitose/efeitos dos fármacos , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Helminto/farmacologia , Humanos , Fatores Imunológicos/farmacologia , Interleucina-4/metabolismo , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Sinais de Localização Nuclear/metabolismo , Dor/genética , Análise de Componente Principal , Transporte Proteico/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Bexiga Urinária/efeitos dos fármacos , Urotélio/metabolismo
14.
Front Immunol ; 11: 605288, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33304354

RESUMO

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.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antígenos Arqueais/farmacologia , Catepsina B/farmacologia , Proteínas de Helminto/farmacologia , Lipídeos/farmacologia , Polissorbatos/farmacologia , Schistosoma mansoni/imunologia , Esquistossomose mansoni/prevenção & controle , Esqualeno/farmacologia , Vacinas Sintéticas/farmacologia , Animais , Anticorpos/sangue , Catepsina B/imunologia , Células Cultivadas , Citocinas/metabolismo , Composição de Medicamentos , Feminino , Proteínas de Helminto/imunologia , Imunidade Celular/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Imunização , Imunogenicidade da Vacina , Camundongos Endogâmicos C57BL , Contagem de Ovos de Parasitas , Schistosoma mansoni/enzimologia , Esquistossomose mansoni/imunologia , Esquistossomose mansoni/parasitologia , Caramujos , Vacinas Sintéticas/imunologia
15.
Mar Drugs ; 18(12)2020 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-33321960

RESUMO

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.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Ativação do Complemento/efeitos dos fármacos , Inativadores do Complemento/farmacologia , Proteínas de Helminto/farmacologia , Animais , Antibacterianos/química , Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/toxicidade , Inativadores do Complemento/química , Inativadores do Complemento/toxicidade , Eritrócitos/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Proteínas de Helminto/química , Proteínas de Helminto/toxicidade , Hemólise/efeitos dos fármacos , Humanos , Listeria monocytogenes/efeitos dos fármacos , Listeria monocytogenes/crescimento & desenvolvimento , Conformação Proteica , Coelhos , Carneiro Doméstico , Relação Estrutura-Atividade
16.
Front Immunol ; 11: 2087, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33193292

RESUMO

Fasciola hepatica is helminth parasite found around the world that causes fasciolosis, a chronic disease affecting mainly cattle, sheep, and occasionally humans. Triclabendazole is the drug of choice to treat this parasite. However, the continuous use of this drug has led to the development of parasite resistance and, consequently, the limitation of its effectiveness. Hence, vaccination appears as an attractive option to develop. In this work, we evaluated the potential of F. hepatica Kunitz-type molecule (FhKTM) as an antigen formulated with a liquid crystal nanostructure formed by self-assembly of 6-O-ascorbyl palmitate ester (Coa-ASC16) and the synthetic oligodeoxynucleotide containing unmethylated cytosine-guanine motifs (CpG-ODN) during an experimental model of fasciolosis in mice, and we further dissected the immune response associated with host protection. Our results showed that immunization of mice with FhKTM/CpG-ODN/Coa-ASC16 induces protection against F. hepatica challenge by preventing liver damage and improving survival after F. hepatica infection. FhKTM/CpG-ODN/Coa-ASC16-immunized mice elicited potent IFN-γ and IL-17A with high levels of antigen-specific IgG1, IgG2a, and IgA serum antibodies. Strikingly, IL-17A blockade during infection decreased IgG2a and IgA antibody levels as well as IFN-γ production, leading to an increase in mortality of vaccinated mice. The present study highlights the potential of a new vaccine formulation to improve control and help the eradication of F. hepatica infection, with potential applications for natural hosts such as cattle and sheep.


Assuntos
Anticorpos Anti-Helmínticos/imunologia , Fasciola hepatica/imunologia , Fasciolíase/prevenção & controle , Proteínas de Helminto/farmacologia , Interferon gama/imunologia , Interleucina-17/imunologia , Vacinas/farmacologia , Animais , Fasciolíase/imunologia , Feminino , Proteínas de Helminto/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Vacinas/imunologia
17.
Sci Rep ; 10(1): 20283, 2020 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-33219293

RESUMO

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.


Assuntos
Antígenos de Helmintos/farmacologia , Moléculas de Adesão Celular/metabolismo , Células Dendríticas/efeitos dos fármacos , Proteínas de Helminto/farmacologia , Tolerância Imunológica/efeitos dos fármacos , Lectinas Tipo C/metabolismo , Receptores de Superfície Celular/metabolismo , Trichinella spiralis/imunologia , Animais , Antígenos de Helmintos/isolamento & purificação , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Voluntários Saudáveis , Proteínas de Helminto/isolamento & purificação , Humanos , Larva/imunologia , Larva/metabolismo , Masculino , Cultura Primária de Células , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo
18.
Folia Parasitol (Praha) ; 672020 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-33082302

RESUMO

The elastase, which belongs to the serine protease family, hydrolyses various proteins and may be involved in the parasite invasion. In this study, complete sequence of elastase-1 (TsE) the nematode Trichinella spiralis (Owen, 1835) was cloned into the plasmid pcDNA3.1 as TsE DNA vaccine. After intramuscular vaccination, serum anti-Trichinella antibodies (IgG and subclass IgG1/IgG2a, and IgA), total and specific intestinal mucosal sIgA in mice vaccinated with pcDNA3.1/TsE were measured by ELISA. The results showed that vaccination with pcDNA3.1/TsE induced a systemic humoral immune response (high levels of serum IgG and subclass IgG1/IgG2a and IgA) and local intestinal mucosal immune responses (high levels of TsE-specific sIgA). Vaccination of mice with TsE DNA vaccine also triggered a systemic and local concomitant Th1/Th2 response, as demonstrated by significant elevation of Th1 (IFN-γ and IL-2) / Th2 (IL-4 and IL-10) cytokine levels after the spleen, mesenteric lymph node and Peyer's patch cells from vaccinated mice were stimulated with recombinant TsE (rTsE). The vaccination of mice with pcDNA3.1/TsE displayed a 17% reduction of intestinal adult worms and a 39% reduction of muscle larvae. Our results indicated that TsE DNA vaccine elicited a systemic concomitant Th1/Th2 response and an enteral local sIgA response, and produced a partial protection against infection with T. spiralis. The TsE may be regarded as a potential candidate vaccine target against Trichinella infection. The oral polyvalent vaccines should be developed to improve the protective efficacy of anti-Trichinella vaccines.


Assuntos
Proteínas de Helminto/imunologia , Elastase Pancreática/imunologia , Trichinella spiralis/imunologia , Triquinelose/imunologia , Vacinação , Vacinas de DNA/administração & dosagem , Animais , Proteínas de Helminto/administração & dosagem , Proteínas de Helminto/farmacologia , Camundongos , Elastase Pancreática/administração & dosagem , Elastase Pancreática/farmacologia , Triquinelose/parasitologia
19.
Parasitol Res ; 119(11): 3705-3718, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32901341

RESUMO

Excretory-secretory products (ESPs) of parasitic helminths are well known to exert immunostimulation and immunomodulation in hosts. Immune regulation plays a key role in anti-tumour therapy. The present study explored the anti-tumour effect of ESPs released by Angiostrongylus cantonensis. In Hepa1-6 mouse tumour models, ESPs significantly reduced tumour growth. Tumour-bearing mice treated with ESPs had significantly higher CD3+, CD4+, and CD8+ T cell counts than those treated with Freund's adjuvant. In vitro, human hepatocarcinoma HepG2 cells, human lung cancer A549 cells, and normal human liver HL-7702 cells were co-incubated with ESPs for 24 h and 48 h. ESPs significantly accelerated HepG2 apoptosis but had no inhibitory effect on the proliferation of A549 and HL-7702 cells. Apoptotic HepG2 cells displayed condensed nuclei, apoptotic bodies, and swollen endoplasmic reticulum (ER). Expression of the endoplasmic reticulum stress (ERS)-related factors activating transcription factor 6 (ATF6) and C/EBP-homologous protein (CHOP) in HepG2 cells increased with increasing ESP concentration and treatment time. Calreticulin (CRT) is a key effector protein of ESPs, and recombinant calreticulin (rCRT) was produced in BL21 Escherichia coli (E. coli). In contrast to ESPs, rCRT markedly reduced the proliferation of HepG2 cells. The expression levels of ATF6 and CHOP in HepG2 cells treated with 30 µg/mL rCRT significantly increased at 48 h. Notably, these findings synergistically suggest that ESPs and rCRT are promising candidates for anti-tumour immunotherapy.


Assuntos
Angiostrongylus cantonensis/metabolismo , Fatores Imunológicos/uso terapêutico , Neoplasias/tratamento farmacológico , Células A549 , Angiostrongylus cantonensis/genética , Animais , Apoptose/efeitos dos fármacos , Calreticulina/genética , Calreticulina/metabolismo , Calreticulina/farmacologia , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Helminto/genética , Proteínas de Helminto/metabolismo , Proteínas de Helminto/farmacologia , Células Hep G2 , Humanos , Fatores Imunológicos/metabolismo , Fatores Imunológicos/farmacologia , Camundongos , Neoplasias/patologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia
20.
Exp Parasitol ; 218: 107983, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32861680

RESUMO

Trichinella spiralis (T. spiralis) muscle larvae (ML) excretory/secretory products (ESPs) are antitumor substances extracted from the culture medium of T. spiralis ML. The ESPs inhibit tumor growth and induce tumor cell apoptosis. To explore the effects of these products on the non-small-cell lung cancer (NSCLC) line A549, logarithmically growing A549 cells were co-cultured with different concentrations of T. spiralis ML ESPs for 24, 36 and 48 h. Our results showed that T. spiralis ML ESPs significantly inhibited A549 cells proliferation, which was dose-and time-dependent. To evaluate the inhibition by T. spiralis ML ESPs of the growth of A549 cells, we assayed their apoptosis and cell-cycle distribution by flow cytometry (FCM). To determine whether ESPs induced apoptosis of A549 cells via the mitochondrial pathway, we evaluated the levels of mitochondrion-related factors by Western blotting. The FCM indicated a clear trend toward apoptosis of A549 cells co-cultured with ESPs for 24 h. The cells were blocked in S-phase. Western blotting revealed that the expression levels of the genes encoding Bax, caspase-3, and caspase-9 increased (compared to a control group), and the Bcl-2 gene expression level decreased. Our results suggest that T. spiralis ML ESPs induce apoptosis of the NSCLC line A549 via the mitochondrial pathway; the cells become arrested in S-phase. This may explain the antineoplastic activity of T. spiralis ML ESPs.


Assuntos
Células A549/efeitos dos fármacos , Antígenos de Helmintos/farmacologia , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proteínas de Helminto/farmacologia , Fase S/efeitos dos fármacos , Trichinella spiralis/química , Células A549/citologia , Análise de Variância , Animais , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/patologia , Proliferação de Células/efeitos dos fármacos , Meios de Cultura Livres de Soro , Feminino , Humanos , Larva/química , Camundongos , Trichinella spiralis/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...