Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
1.
J Immunol Res ; 2021: 6234836, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34869783

RESUMEN

Immunomodulation of airway hyperreactivity by excretory-secretory (ES) products of the first larval stage (L1) of the gastrointestinal nematode Trichuris suis is reported by us and others. Here, we aimed to identify the proteins accounting for the modulatory effects of the T. suis L1 ES proteins and studied six selected T. suis L1 proteins for their immunomodulatory efficacy in a murine OVA-induced allergic airway disease model. In particular, an enzymatically active T. suis chitinase mediated amelioration of clinical signs of airway hyperreactivity, primarily associated with suppression of eosinophil recruitment into the lung, the associated chemokines, and increased numbers of RELMα + interstitial lung macrophages. While there is no indication of T. suis chitinase directly interfering with dendritic cell activation or antigen presentation to CD4 T cells, treatment of allergic mice with the worm chitinase influenced the hosts' own chitinase activity in the inflamed lung. The three-dimensional structure of the T. suis chitinase as determined by high-resolution X-ray crystallography revealed high similarities to mouse acidic mammalian chitinase (AMCase) but a unique ability of T. suis chitinase to form dimers. Our data indicate that the structural similarities between the parasite and host chitinase contribute to the disease-ameliorating effect of the helminth-derived chitinase on allergic lung inflammation.


Asunto(s)
Quitinasas/ultraestructura , Eosinofilia/tratamiento farmacológico , Proteínas del Helminto/administración & dosificación , Agentes Inmunomoduladores/administración & dosificación , Hipersensibilidad Respiratoria/tratamiento farmacológico , Animales , Líquido del Lavado Bronquioalveolar , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Eosinofilia/diagnóstico , Eosinofilia/inmunología , Eosinofilia/patología , Femenino , Proteínas del Helminto/ultraestructura , Interacciones Huésped-Parásitos/inmunología , Humanos , Pulmón/efectos de los fármacos , Pulmón/inmunología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Ratones , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Hipersensibilidad Respiratoria/diagnóstico , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/patología , Trichuris/enzimología
2.
Microbiol Spectr ; 9(3): e0191021, 2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34937173

RESUMEN

Due to their phylogenetic proximity to humans, nonhuman primates (NHPs) are considered an adequate choice for a basic and preclinical model of sepsis. Gram-negative bacteria are the primary causative of sepsis. During infection, bacteria continuously release the potent toxin lipopolysaccharide (LPS) into the bloodstream, which triggers an uncontrolled systemic inflammatory response leading to death. Our previous research has demonstrated in vitro and in vivo using a mouse model of septic shock that Fh15, a recombinant variant of the Fasciola hepatica fatty acid binding protein, acts as an antagonist of Toll-like receptor 4 (TLR4) suppressing the LPS-induced proinflammatory cytokine storm. The present communication is a proof-of concept study aimed to demonstrate that a low-dose of Fh15 suppresses the cytokine storm and other inflammatory markers during the early phase of sepsis induced in rhesus macaques by intravenous (i.v.) infusion with lethal doses of live Escherichia coli. Fh15 was administered as an isotonic infusion 30 min prior to the bacterial infusion. Among the novel findings reported in this communication, Fh15 (i) significantly prevented bacteremia, suppressed LPS levels in plasma, and the production of C-reactive protein and procalcitonin, which are key signatures of inflammation and bacterial infection, respectively; (ii) reduced the production of proinflammatory cytokines; and (iii) increased innate immune cell populations in blood, which suggests a role in promoting a prolonged steady state in rhesus macaques even in the presence of inflammatory stimuli. This report is the first to demonstrate that a F. hepatica-derived molecule possesses potential as an anti-inflammatory drug against sepsis in an NHP model. IMPORTANCE Sepsis caused by Gram-negative bacteria affects 1.7 million adults annually in the United States and is one of the most important causes of death at intensive care units. Although the effective use of antibiotics has resulted in improved prognosis of sepsis, the pathological and deathly effects have been attributed to the persistent inflammatory cascade. There is a present need to develop anti-inflammatory agents that can suppress or neutralize the inflammatory responses and prevent the lethal consequences of sepsis. We demonstrated here that a small molecule of 14.5 kDa can suppress the bacteremia, endotoxemia, and many other inflammatory markers in an acute Gram-negative sepsis rhesus macaque model. These results reinforce the notion that Fh15 constitutes an excellent candidate for drug development against sepsis.


Asunto(s)
Antiinflamatorios/administración & dosificación , Bacteriemia/tratamiento farmacológico , Fasciola hepatica/metabolismo , Proteínas de Unión a Ácidos Grasos/administración & dosificación , Bacterias Gramnegativas/fisiología , Proteínas del Helminto/administración & dosificación , Animales , Antiinflamatorios/metabolismo , Bacteriemia/genética , Bacteriemia/inmunología , Bacteriemia/microbiología , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Fasciola hepatica/química , Fasciola hepatica/genética , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Bacterias Gramnegativas/clasificación , Bacterias Gramnegativas/genética , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Humanos , Macaca mulatta , Masculino , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
3.
PLoS Negl Trop Dis ; 15(11): e0009981, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34793443

RESUMEN

Extracellular Vesicles (EVs) are an integral component of cellular/organismal communication and have been found in the excreted/secreted (ES) products of both protozoan and metazoan parasites. Within the blood fluke schistosomes, EVs have been isolated from egg, schistosomula, and adult lifecycle stages. However, the role(s) that EVs have in shaping aspects of parasite biology and/or manipulating host interactions is poorly defined. Herein, we characterise the most abundant EV-enriched protein in Schistosoma mansoni tissue-migrating schistosomula (Schistosoma mansoni Larval Extracellular Vesicle protein 1 (SmLEV1)). Comparative sequence analysis demonstrates that lev1 orthologs are found in all published Schistosoma genomes, yet homologs are not found outside of the Schistosomatidae. Lifecycle expression analyses collectively reveal that smlev1 transcription peaks in cercariae, is male biased in adults, and is processed by alternative splicing in intra-mammalian lifecycle stages. Immunohistochemistry of cercariae using a polyclonal anti-recombinant SmLEV1 antiserum localises this protein to the pre-acetabular gland, with some disperse localisation to the surface of the parasite. S. mansoni-infected Ugandan fishermen exhibit a strong IgG1 response against SmLEV1 (dropping significantly after praziquantel treatment), with 11% of the cohort exhibiting an IgE response and minimal levels of detectable antigen-specific IgG4. Furthermore, mice vaccinated with rSmLEV1 show a slightly reduced parasite burden upon challenge infection and significantly reduced granuloma volumes, compared with control animals. Collectively, these results describe SmLEV1 as a Schistosomatidae-specific, EV-enriched immunogen. Further investigations are now necessary to uncover the full extent of SmLEV1's role in shaping schistosome EV function and definitive host relationships.


Asunto(s)
Cercarias/inmunología , Vesículas Extracelulares/inmunología , Proteínas del Helminto/inmunología , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/parasitología , Adolescente , Adulto , Secuencia de Aminoácidos , Animales , Antihelmínticos/administración & dosificación , Anticuerpos Antihelmínticos/inmunología , Cercarias/genética , Cercarias/crecimiento & desarrollo , Niño , Estudios de Cohortes , Vesículas Extracelulares/genética , Femenino , Proteínas del Helminto/administración & dosificación , Proteínas del Helminto/química , Proteínas del Helminto/genética , Humanos , Inmunogenicidad Vacunal , Inmunoglobulina E/inmunología , Inmunoglobulina G/inmunología , Masculino , Ratones , Persona de Mediana Edad , Praziquantel/administración & dosificación , Schistosoma mansoni/química , Schistosoma mansoni/genética , Schistosoma mansoni/crecimiento & desarrollo , Esquistosomiasis mansoni/tratamiento farmacológico , Esquistosomiasis mansoni/inmunología , Alineación de Secuencia , Vacunas/administración & dosificación , Vacunas/genética , Vacunas/inmunología , Adulto Joven
4.
PLoS Negl Trop Dis ; 15(10): e0009865, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34699522

RESUMEN

BACKGROUND: Trichinellosis is a serious zoonotic disease distributed around the world. It is needed to develop a safe, effective and feasible anti-Trichinella vaccine for prevention and control of trichinellosis. The aim of this study was to construct a recombinant Lactobacillus plantarum encoding Trichinella spiralis inorganic pyrophosphatase (TsPPase) and investigate its immune protective effects against T. spiralis infection. METHODOLOGY/PRINCIPAL FINDINGS: The growth of recombinant L. plantarum was not affected by TsPPase/pSIP409-pgsA' plasmid, and the recombinant plasmid was inherited stably in bacteria. Western blot and immunofluorescence assay (IFA) indicated that the rTsPPase was expressed on the surface of recombinant L. plantarum. Oral vaccination with rTsPPase induced higher levels of specific serum IgG, IgG1, IgG2a and mucosal secretory IgA (sIgA) in BALB/c mice. ELISA analysis revealed that the levels of IFN-γ and IL-4 released from spleen, mesenteric lymph nodes and Peyer's patches were evidently increased at 2-4 weeks following vaccination, compared to MRS (De Man, Rogosa, Sharpe) medium control group (P < 0.05). Immunization of mice with rTsPPase exhibited a 67.18, 54.78 and 51.91% reduction of intestinal infective larvae, adult worms and muscle larvae at 24 hours post infection (hpi), 6 days post infection (dpi) and 35 dpi, respectively (P < 0.05), and the larval molting and development was significantly inhibited by 45.45% at 24 hpi, compared to the MRS group. CONCLUSIONS: TsPPase plays a crucial role in T. spiralis molting and development, oral vaccination with rTsPPase induced a significant local mucosal sIgA response and systemic Th1/Th2 immune response, and immune protection against T. spiralis infection in BALB/c mice.


Asunto(s)
Proteínas del Helminto/administración & dosificación , Pirofosfatasa Inorgánica/administración & dosificación , Lactobacillus plantarum/genética , Trichinella spiralis/inmunología , Triquinelosis/prevención & control , Vacunas/administración & dosificación , Administración Oral , Animales , Anticuerpos Antihelmínticos/inmunología , Femenino , Proteínas del Helminto/genética , Proteínas del Helminto/inmunología , Humanos , Inmunoglobulina G/inmunología , Pirofosfatasa Inorgánica/genética , Pirofosfatasa Inorgánica/inmunología , Lactobacillus plantarum/metabolismo , Ratones , Ratones Endogámicos BALB C , Trichinella spiralis/enzimología , Trichinella spiralis/genética , Triquinelosis/inmunología , Triquinelosis/parasitología , Vacunación , Vacunas/genética , Vacunas/inmunología
5.
Iran Biomed J ; 25(4): 284-96, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34217159

RESUMEN

Background: Cystic echinococcosis is a zoonotic parasitic infection caused by Echinococcus granulosus worldwide and is associated with economic losses among livestock animals. EG95 is an immunogenic antigen from the E. granulosus. Lactococcus lactis has been prested as a safe vehicle for antigen delivery. The goal of this study was to design a novel L. lactis strain displaying EG95 as a vaccine delivery system. Methods: The eg95 encoding gene fragment fused to the M6 anchoring protein was cloned into the pNZ7021 vector, and L. lactis NZ9000 displaying recombinant EG95 was constructed. The expression of an approximately 32-kDa EG95 protein was confirmed by Western blotting and immunofluorescence analysis. The immune responses were evaluated in BALB/c mice immunized orally and subcutaneously with the live and killed recombinant L. lactis, respectively. Results: Total IgG level in mice immunized with heat-killed recombinant L. lactis (pNZ7021-eg95) significantly increased compared to the control group. Mucosal IgA was significantly higher in mice received live recombinant L. lactis (pNZ7021-eg95) compared to the control mice. Splenic lymphocytes from immunized mice represented the high levels of IFN-γ and the low-levels of IL-4 and IL-10. Conclusion: Our results indicate that immunization with EG95-expressing L. lactis can induce both specific humoral and cellular immune responses in mice.


Asunto(s)
Antígenos Helmínticos/inmunología , Echinococcus granulosus/inmunología , Proteínas del Helminto/inmunología , Inmunidad Humoral/inmunología , Fenómenos Inmunogenéticos/fisiología , Lactococcus lactis/inmunología , Animales , Antígenos Helmínticos/administración & dosificación , Femenino , Proteínas del Helminto/administración & dosificación , Inmunidad Humoral/efectos de los fármacos , Fenómenos Inmunogenéticos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes/inmunología
6.
Transl Res ; 232: 88-102, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33676036

RESUMEN

The symbiotic relationships shared between humans and their gastrointestinal parasites present opportunities to discover novel therapies for inflammatory diseases. A prime example of this phenomenon is the interaction of humans and roundworms such as the hookworm, Necator americanus. Epidemiological observations, animal studies and clinical trials using experimental human hookworm infection show that hookworms can suppress inflammation in a safe and well-tolerated way, and that the key to their immunomodulatory properties lies within their secreted proteome. Herein we describe the identification of 2 netrin domain-containing proteins from the N. americanus secretome, and explore their potential in treating intestinal inflammation in mouse models of ulcerative colitis. One of these proteins, subsequently named Na-AIP-1, was effective at suppressing disease when administered prophylactically in the acute TNBS-induced model of colitis. This protective effect was validated in the more robust CD4 T cell transfer model of chronic colitis, where prophylactic Na-AIP-1 reduced T-cell-dependent type-1 cytokine responses in the intestine and the associated intestinal pathology. Mechanistic studies revealed that depletion of CD11c+ cells abrogated the protective anticolitic effect of Na-AIP-1. Next generation sequencing of colon tissue in the T-cell transfer model of colitis revealed that Na-AIP-1 induced a transcriptomic profile associated with the downregulation of metabolic and signaling pathways involved in type-1 inflammation, notably TNF. Finally, co-culture of Na-AIP-1 with a human monocyte-derived M1 macrophage cell line resulted in significantly reduced secretion of TNF. Na-AIP-1 is now a candidate for clinical development as a novel therapeutic for the treatment of human inflammatory bowel diseases.


Asunto(s)
Antiinflamatorios/administración & dosificación , Linfocitos T CD4-Positivos/inmunología , Colitis Ulcerosa/prevención & control , Proteínas del Helminto/administración & dosificación , Necator americanus/química , Netrinas/administración & dosificación , Animales , Linfocitos T CD4-Positivos/trasplante , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/inmunología , Modelos Animales de Enfermedad , Femenino , Proteínas del Helminto/química , Proteínas del Helminto/genética , Infecciones por Uncinaria/metabolismo , Humanos , Masculino , Inhibidores de la Metaloproteinasa de la Matriz/química , Ratones Endogámicos C57BL , Ratones Noqueados , Netrinas/análisis , Proteínas Recombinantes/administración & dosificación
7.
BMC Microbiol ; 21(1): 5, 2021 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-33407120

RESUMEN

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.


Asunto(s)
Galectinas/administración & dosificación , Galectinas/genética , Inmunoglobulina G/sangre , Leucocitos Mononucleares/inmunología , Onchocerca/inmunología , Animales , Bovinos , Clonación Molecular/métodos , Femenino , Galectinas/inmunología , Perfilación de la Expresión Génica , Proteínas del Helminto/administración & dosificación , Proteínas del Helminto/genética , Proteínas del Helminto/inmunología , Humanos , Inmunización , Leucocitos Mononucleares/parasitología , Onchocerca/genética , Filogenia , Ratas , Ratas Wistar , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Análisis de Secuencia de ADN
8.
Parasitol Res ; 120(1): 173-185, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33079271

RESUMEN

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.


Asunto(s)
Catepsinas/inmunología , Gliceraldehído-3-Fosfato Deshidrogenasas/inmunología , Schistosoma japonicum/inmunología , Esquistosomiasis Japónica/prevención & control , Vacunas/inmunología , Animales , Catepsinas/administración & dosificación , Femenino , Gliceraldehído-3-Fosfato Deshidrogenasas/administración & dosificación , Proteínas del Helminto/administración & dosificación , Proteínas del Helminto/inmunología , Ratones , Ratones Endogámicos BALB C , Linfocitos T Reguladores/inmunología , Vacunación , Vacunas/administración & dosificación
9.
Front Immunol ; 11: 2106, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33013887

RESUMEN

The same mechanisms that enable host defense against helminths also drive allergic inflammation. This suggests that pathomechanisms of allergic diseases represent evolutionary old responses against helminth parasites and that studying antihelminth immunity may provide insights into pathomechanisms of asthma. However, helminths have developed an intricate array of immunoregulatory mechanisms to modulate type 2 immune mechanisms. This has led to the hypothesis that the lack of helminth infection may contribute to the rise in allergic sensitization in modern societies. Indeed, the anti-inflammatory potential of helminth (worm) parasites and their products in allergy and asthma has been recognized for decades. As helminth infections bring about multiple undesired effects including an increased susceptibility to other infections, intended helminth infection is not a feasible approach to broadly prevent or treat allergic asthma. Thus, the development of new helminth-based biopharmaceutics may represent a safer approach of harnessing type 2-suppressive effects of helminths. However, progress regarding the mechanisms and molecules that are employed by helminths to modulate allergic inflammation has been relatively recent. The scavenging of alarmins and the modulation of lipid mediator pathways and macrophage function by helminth proteins have been identified as important immunoregulatory mechanisms targeting innate immunity in asthma and allergy. In addition, by regulating the activation of dendritic cells and by promoting regulatory T-cell responses, helminth proteins can counterregulate the adaptive T helper 2 cell response that drives allergic inflammation. Despite these insights, important open questions remain to be addressed before helminth molecules can be used for the prevention and treatment of asthma and other allergic diseases.


Asunto(s)
Asma/inmunología , Helmintiasis/inmunología , Interacciones Huésped-Parásitos/inmunología , Hipersensibilidad/inmunología , Modelos Inmunológicos , Inmunidad Adaptativa , Alarminas/metabolismo , Animales , Asma/epidemiología , Asma/terapia , Evolución Biológica , Comorbilidad , Células Dendríticas/inmunología , Proteínas del Helminto/administración & dosificación , Proteínas del Helminto/fisiología , Proteínas del Helminto/uso terapéutico , Helmintiasis/epidemiología , Helmintiasis/parasitología , Helmintos/fisiología , Humanos , Hipersensibilidad/epidemiología , Hipersensibilidad/terapia , Inmunidad Celular , Inmunidad Innata , Inmunomodulación , Inflamación , Activación de Macrófagos , Ratones , Modelos Animales , Ratas , Subgrupos de Linfocitos T/inmunología , Terapia con Helmintos
10.
Folia Parasitol (Praha) ; 672020 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-33082302

RESUMEN

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.


Asunto(s)
Proteínas del Helminto/inmunología , Elastasa Pancreática/inmunología , Trichinella spiralis/inmunología , Triquinelosis/inmunología , Vacunación , Vacunas de ADN/administración & dosificación , Animales , Proteínas del Helminto/administración & dosificación , Proteínas del Helminto/farmacología , Ratones , Elastasa Pancreática/administración & dosificación , Elastasa Pancreática/farmacología , Triquinelosis/parasitología
11.
Acta Parasitol ; 65(3): 652-660, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32240492

RESUMEN

PURPOSE: Schistosomiasis is a disease that afflicts over 220 million people worldwide. To date, there is no vaccine against schistosomiasis and chemotherapy relies basically on a single drug, praziquantel. The current study was undertaken to investigate the therapeutic effects of monophosphoryl lipid A (MPLA) as an adjuvant in soluble egg antigen (SEA)-vaccinated and Schistosoma mansoni-infected mice. METHODS: Mice were divided into two groups of uninfected and Schistosoma mansoni infected. The two groups were treated differently with MPLA, SEA and praziquantel. Study parameters included parasitological, immunological and biochemical parameters. RESULTS: Parasitological parameters revealed that intraperitoneal injection of MPLA into SEA-vaccinated and S. mansoni-infected mice was effective in reducing the worm and egg burden, granuloma count and diameter as well as the total area of infection in their livers versus SEA-untreated but infected ones. In addition, MPLA showed ameliorative action on the elevated liver oxidative stress marker, including malondialdehyde (MDA) and a decrease in the level of the antioxidant enzymes, reduced glutathione (GSH) and catalase (CAT) which may have a role in the liver damage and fibrosis due to S. mansoni infection. CONCLUSION: Treatment with MPLA has multi-functions in attenuating the deleterious impacts of S. mansoni infection in mice livers. Its effects are mediated through a reduction of ova count, worm burden, granuloma diameter and amelioration of antioxidant defense systems, and liver function biomarkers.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Lípido A/análogos & derivados , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/prevención & control , Receptores Toll-Like/agonistas , Animales , Antihelmínticos/uso terapéutico , Antígenos Helmínticos/administración & dosificación , Antígenos Helmínticos/inmunología , Proteínas del Helminto/administración & dosificación , Proteínas del Helminto/inmunología , Lípido A/administración & dosificación , Lípido A/inmunología , Masculino , Ratones , Praziquantel/uso terapéutico , Schistosoma mansoni , Vacunación
12.
Vet Res ; 51(1): 43, 2020 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-32169101

RESUMEN

Trichinella spiralis is an important foodborne parasitic nematode that represents an enormous threat to the food safety of pork meat. The development of a preventive vaccine is valuable for the prevention and control of Trichinella infection in domestic pigs to ensure pork safety. Elastase is a trypsin-like serine protease that hydrolyzes the host's diverse tissue components and participates in parasite penetration, and it might be a novel vaccine target molecule. The aim of this study was to assess the protective immunity produced by vaccination with a novel Trichinella spiralis elastase-1 (TsE) in a mouse model. The results demonstrate that subcutaneous vaccination of mice with rTsE elicited a systemic humoral response (high levels of serum IgG and subclass IgG1/IgG2a and IgA) and significant local enteral mucosal sIgA responses. Anti-rTsE IgG recognized the native TsE at the cuticle, stichosome of intestinal infective larvae and adult worm (AW), and intrauterine embryos of female AW. The rTsE vaccination also produced a systemic and local mixed Th1/Th2 response, as demonstrated by clear elevation levels of Th1 cytokines (IFN-γ, IL-2) and Th2 cytokines (IL-4, IL-10) after spleen, mesenteric lymph node and Peyer's patch cells from immunized mice were stimulated with rTsE. The immunized mice exhibited a 52.19% reduction in enteral AW and a 64.06% reduction in muscle larvae after challenge infection. The immune response triggered by rTsE vaccination protected enteral mucosa from larval intrusion, suppressed larval development and reduced female fecundity. The results indicate that TsE may represent a novel target molecule for anti-T. spiralis vaccines.


Asunto(s)
Proteínas del Helminto/farmacología , Inmunidad Humoral , Elastasa Pancreática/farmacología , Trichinella spiralis/efectos de los fármacos , Triquinelosis/prevención & control , Vacunación/veterinaria , Animales , Femenino , Fertilidad , Proteínas del Helminto/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Elastasa Pancreática/administración & dosificación , Trichinella spiralis/fisiología , Triquinelosis/parasitología
13.
Sci Rep ; 10(1): 1055, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31974398

RESUMEN

Controlled infection with intestinal nematodes has therapeutic potential for preventing the symptoms of allergic and autoimmune diseases. Here, we engineered larvae of the filarial nematode Litomosoides sigmodontis as a vaccine strategy to induce adaptive immunity against a foreign, crosslinked protein, chicken egg ovalbumin (OVA), in the absence of an external adjuvant. The acylation of filarial proteins with fluorescent probes or biotin was not immediately detrimental to larval movement and survival, which died 3 to 5 days later. At least some of the labeled and skin-inoculated filariae migrated through lymphatic vessels to draining lymph nodes. The immunization potential of OVA-biotin-filariae was compared to that of an OVA-bound nanoparticulate carrier co-delivered with a CpG adjuvant in a typical vaccination scheme. Production of IFNγ and TNFα by restimulated CD4+ cells but not CD8+ confirmed the specific ability of filariae to stimulate CD4+ T cells. This alternative method of immunization exploits the intrinsic adjuvancy of the attenuated nematode carrier and has the potential to shift the vaccination immune response towards cellular immunity.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Hipersensibilidad al Huevo/inmunología , Filarioidea/inmunología , Larva/inmunología , Ovalbúmina/inmunología , Inmunidad Adaptativa , Animales , Linfocitos T CD4-Positivos/inmunología , Pollos , Hipersensibilidad al Huevo/etiología , Filarioidea/genética , Proteínas del Helminto/administración & dosificación , Proteínas del Helminto/genética , Proteínas del Helminto/inmunología , Humanos , Inmunización , Larva/genética , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/administración & dosificación , Ovalbúmina/efectos adversos , Ovalbúmina/química
14.
Parasitol Res ; 119(1): 203-214, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31845020

RESUMEN

Infection with helminth parasites or the administration of their antigens can prevent or attenuate autoimmune diseases. To date, the specific molecules that prime the amelioration are only limited. In this study, recombinant Schistosoma japonicum cystatin (rSjcystatin) and fructose-1,6-bisphosphate aldolase (rSjFBPA) were administered to female NOD mice via intraperitoneal (i.p.) injection to characterize the immunological response by the recombinant proteins. We have shown that the administration of rSjcystatin or rSjFBPA significantly reduced the diabetes incidence and ameliorated the severity of type 1 diabetes mellitus (T1DM). Disease attenuation was associated with suppressed interferon-gamma (IFN-γ) production in autoreactive T cells and with a switch to the production of Th2 cytokines. Following rSjcystatin or rSjFBPA injection, regulatory T cells (Tregs) were remarkably increased, which was accompanied by increased expression of interleukin-10 (IL-10) and transforming growth factor beta (TGF-ß). Our study suggests that helminth-derived proteins may be useful in strategies to limit pathology by promoting the Th2 response and upregulating Tregs during the inflammatory tissue-damage process in T1DM.


Asunto(s)
Cistatinas/administración & dosificación , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Fructosa-Bifosfato Aldolasa/administración & dosificación , Proteínas del Helminto/administración & dosificación , Factores Inmunológicos/administración & dosificación , Schistosoma japonicum/enzimología , Animales , Cistatinas/genética , Cistatinas/metabolismo , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Femenino , Fructosa-Bifosfato Aldolasa/genética , Fructosa-Bifosfato Aldolasa/metabolismo , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Factores Inmunológicos/genética , Factores Inmunológicos/metabolismo , Ratones , Ratones Endogámicos NOD , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Linfocitos T Reguladores/inmunología
15.
Parasit Vectors ; 12(1): 457, 2019 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-31547847

RESUMEN

BACKGROUND: Potent granulomatous inflammation responses induced by schistosome eggs and resultant fibrosis are the primary causes of morbidity in schistosomiasis. Recombinant Sj16 (rSj16), a 16-kDa protein of Schistosoma japonicum produced in Escherichia coli, has been demonstrated to have novel immunoregulatory effects in vivo and in vitro. Thus, this study investigated the anti-inflammatory and anti-fibrotic effects of rSj16 treatment in S. japonicum-infected mice and demonstrated the immune modulation between the schistosome and the host. METHODS: Schistosoma japonicum infected mice were treated with the rSj16 protein and Sj16 peptide at different time points post-infection to assess their efficacy at the optimal time point. Sj16 peptide and/or Praziquantel (PZQ) treatments were initiated at week 5 post-infection to compare the therapeutic efficacy of each regimen. Hepatic granulomatous inflammation, fibrosis and cytokine production (pro-inflammatory, Th1, Th2, Th17 and regulatory cytokines IL-10) were detected. Moreover, M2 macrophages were measured to illuminate the mechanisms of Sj16. RESULTS: The rSj16 protein and Sj16 peptide had significant protective effects in S. japonicum-infected mice, as shown by decreased granuloma formation, areas of collagen deposition and inhibition of pro-inflammatory Th1, Th2 and Th17 cytokine production. These protective activities were more obvious when animals were treated with either the Sj16 protein or peptide at early stages post-infection. Interestingly, the combined treatment of PZQ and Sj16 was more effective and upregulated IL-10 production than administration of PZQ alone in infected mice. Furthermore, the Sj16 treatment alleviated the pathological effects associated with activated M2 macrophages. CONCLUSIONS: This study demonstrates the anti-inflammatory and anti-fibrotic effects of rSj16 in schistosomiasis. Therefore, the combination of rSj16 with PZQ could be a viable and promising therapeutic strategy for schistosomiasis. In addition, this investigation provides additional information on schistosome-mediated immune modulation and host-parasite interactions.


Asunto(s)
Granuloma/prevención & control , Proteínas del Helminto/metabolismo , Factores Inmunológicos/metabolismo , Hígado/patología , Macrófagos/inmunología , Esquistosomiasis Japónica/patología , Animales , Antihelmínticos/administración & dosificación , Modelos Animales de Enfermedad , Fibrosis/patología , Fibrosis/prevención & control , Granuloma/patología , Proteínas del Helminto/administración & dosificación , Factores Inmunológicos/administración & dosificación , Inflamación/patología , Inflamación/prevención & control , Ratones , Praziquantel/administración & dosificación , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo , Resultado del Tratamiento
16.
Int J Cancer ; 145(11): 3126-3139, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31407335

RESUMEN

Inflammation is currently considered a hallmark of cancer and plays a decisive role in different stages of tumorigenesis, including initiation, promotion, progression, metastasis and resistance to antitumor therapies. Colorectal cancer is a disease widely associated with local chronic inflammation. Additionally, extrinsic factors such as infection may beneficially or detrimentally alter cancer progression. Several reports have noted the ability of various parasitic infections to modulate cancer development, favoring tumor progression in many cases and inhibiting tumorigenesis in others. The aim of our study was to determine the effects of excreted/secreted products of the helminth Taenia crassiceps (TcES) as a treatment in a murine model of colitis-associated colon cancer (CAC). Here, we found that after inducing CAC, treatment with TcES was able to reduce inflammatory cytokines such as IL-1ß, TNF-α, IL-33 and IL-17 and significantly attenuate colon tumorigenesis. This effect was associated with the inhibition of signal transducer and activator of transcription 3 and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) phosphorylation. Furthermore, we determined that TcES interfered with LPS-induced NF-κB p65 activation in human colonic epithelial cell lines in a Raf-1 proto-oncogene-dependent manner. Moreover, in three-dimensional cultures, TcES promoted reorganization of the actin cytoskeleton, altering cell morphology and forming colonospheres, features associated with a low grade of aggressiveness. Our study demonstrates a remarkable effect of helminth-derived molecules on suppressing ongoing colorectal cancer by downregulating proinflammatory and protumorigenic signaling pathways.


Asunto(s)
Antiinflamatorios/administración & dosificación , Azoximetano/efectos adversos , Colitis/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Proteínas del Helminto/administración & dosificación , Taenia/metabolismo , Animales , Antiinflamatorios/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colitis/inducido químicamente , Colitis/complicaciones , Neoplasias del Colon/etiología , Modelos Animales de Enfermedad , Femenino , Proteínas del Helminto/farmacología , Humanos , Interleucina-1beta/metabolismo , Interleucina-33/metabolismo , Ratones , FN-kappa B/metabolismo , Fosforilación , Proto-Oncogenes Mas , Factor de Transcripción STAT3/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
17.
Am J Physiol Renal Physiol ; 316(6): F1133-F1140, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30785353

RESUMEN

Chemotherapy-induced hemorrhagic cystitis is characterized by bladder pain and voiding dysfunction caused by hemorrhage and inflammation. Novel therapeutic options to treat hemorrhagic cystitis are needed. We previously reported that systemic administration of the Schistosomiasis hematobium-derived protein H-IPSEH06 (IL-4-inducing principle from Schistosoma mansoni eggs) is superior to three doses of MESNA in alleviating hemorrhagic cystitis (Mbanefo EC, Le L, Pennington LF, Odegaard JI, Jardetzky TS, Alouffi A, Falcone FH, Hsieh MH. FASEB J 32: 4408-4419, 2018). Based on prior reports by others on S. mansoni IPSE (M-IPSE) and additional work by our group, we reasoned that H-IPSE mediates its effects on hemorrhagic cystitis by binding IgE on basophils and inducing IL-4 expression, promoting urothelial proliferation, and translocating to the nucleus to modulate expression of genes implicated in relieving bladder dysfunction. We speculated that local bladder injection of the S. hematobium IPSE ortholog IPSEH03, hereafter called H-IPSEH03, might be more efficacious in preventing hemorrhagic cystitis compared with systemic administration of IPSEH06. We report that H-IPSEH03, like M-IPSE and H-IPSEH06, activates IgE-bearing basophils in a nuclear factor of activated T-cells reporter assay, indicating activation of the cytokine pathway. Furthermore, H-IPSEH03 attenuates ifosfamide-induced increases in bladder wet weight in an IL-4-dependent fashion. H-IPSEH03 relieves hemorrhagic cystitis-associated allodynia and modulates voiding patterns in mice. Finally, H-IPSEH03 drives increased urothelial cell proliferation, suggesting that IPSE induces bladder repair mechanisms. Taken together, H-IPSEH03 may be a potential novel therapeutic to treat hemorrhagic cystitis by basophil activation, attenuation of allodynia, and promotion of urothelial cell proliferation.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Cistitis/prevención & control , Proteínas del Huevo/administración & dosificación , Proteínas del Helminto/administración & dosificación , Hemorragia/prevención & control , Factores Inmunológicos/administración & dosificación , Vejiga Urinaria/efectos de los fármacos , Urotelio/efectos de los fármacos , Administración Intravesical , Animales , Basófilos/efectos de los fármacos , Basófilos/inmunología , Basófilos/metabolismo , Línea Celular , Cistitis/inducido químicamente , Cistitis/inmunología , Cistitis/metabolismo , Modelos Animales de Enfermedad , Femenino , Hemorragia/inducido químicamente , Hemorragia/inmunología , Hemorragia/metabolismo , Humanos , Ifosfamida , Inmunoglobulina E/inmunología , Inmunoglobulina E/metabolismo , Inyecciones Intravenosas , Interleucina-4/inmunología , Interleucina-4/metabolismo , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/inmunología , Factores de Transcripción NFATC/metabolismo , Transducción de Señal , Vejiga Urinaria/inmunología , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Urodinámica/efectos de los fármacos , Urotelio/inmunología , Urotelio/metabolismo , Urotelio/patología
18.
J Helminthol ; 94: e26, 2019 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-30702051

RESUMEN

The tegument of schistosomes is a source of many potential anti-Schistosoma vaccine molecules. This work aimed to assess the protective effects of the adult Schistosoma mansoni tegument treated (TT) with sub-curative praziquantel (PZQ), whether in vivo (in vivo TT) or in vitro (in vitro TT), in murine schistosomiasis. In vitro TT and in vivo TT showed great similarity, and they differed from untreated tegument antigen (Teg) in terms of quantity and quality of protein bands on SDS-PAGE. Two immunization trials were performed, each with 50 mice, divided randomly into five groups of 10 mice each: (1) uninfected control mice (UC), (2) infected mice given phosphate buffer saline + adjuvant (PBS + adjuvant), (3) infected, Teg vaccinated, (4) infected, in vivo TT vaccinated, and (5) infected, in vitro TT vaccinated. All the immunizations with antigens induced mixed Th1/Th2 immune responses, as indicated by significantly high (P < 0.001) specific IgG2a and IgG1 levels, with Th1 predominating, as shown by a diminished IgG1/IgG2a ratio, as well as a high serum concentration of IFN-γ, an absence of IL-4 and increased IL-10. In vitro TT gave the most pronounced response. With respect to reduction of total worm burden, relative to PBS + adjuvant mice, in vitro TT achieved the highest significant (P < 0.001) results, followed by in vivo TT and Teg (51.8-57.04%, 44.6-50.2% and 35.2-39.3%, respectively). In scanning electron microscopy studies, all the tested antigens caused tegumental changes in adult worms, with the worst occurring with in vitro TT, such as retracted ventral sucker, an effect on the gynaecophoric canal, and changes to tubercles. In conclusion, in vitro TT, which is cheap to prepare, could be a potential vaccine against S. mansoni.


Asunto(s)
Antihelmínticos/administración & dosificación , Proteínas del Helminto/inmunología , Praziquantel/administración & dosificación , Schistosoma mansoni/efectos de los fármacos , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/prevención & control , Animales , Anticuerpos Antihelmínticos/inmunología , Femenino , Proteínas del Helminto/administración & dosificación , Proteínas del Helminto/genética , Humanos , Inmunización , Interferón gamma/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Masculino , Ratones , Schistosoma mansoni/genética , Esquistosomiasis mansoni/genética , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/parasitología , Células TH1/inmunología , Células Th2/inmunología
19.
mSphere ; 3(6)2018 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-30567900

RESUMEN

Sepsis caused by Gram-negative bacteria is the consequence of an unrestrained infection that continuously releases lipopolysaccharide (LPS) into the bloodstream, which triggers an uncontrolled systemic inflammatory response leading to multiorgan failure and death. After scrutinizing the immune modulation exerted by a recombinant Fasciola hepatica fatty acid binding protein termed Fh15, our group demonstrated that addition of Fh15 to murine macrophages 1 h prior to LPS stimulation significantly suppresses the expression of proinflammatory cytokines tumor necrosis factor alpha (TNF-α) and interleukin-1ß (IL1-ß). The present study aimed to demonstrate that Fh15 could exert a similar anti-inflammatory effect in vivo using a mouse model of septic shock. Among the novel findings reported in this article, (i) Fh15 suppressed numerous serum proinflammatory cytokines/chemokines when injected intraperitoneally 1 h after exposure of animals to lethal doses of LPS, (ii) concurrently, Fh15 increased the population of large peritoneal macrophages (LPMs) in the peritoneal cavity (PerC) of LPS-injected animals, and (iii) Fh15 downregulated the expression on spleen macrophages of CD38, a cell surface ectoenzyme with a critical role during inflammation. These findings present the first evidence that the recombinant parasitic antigen Fh15 is an excellent modulator of the PerC cell content and in vivo macrophage activation, endorsing Fh15's potential as a drug candidate against sepsis-related inflammatory response.IMPORTANCE Sepsis is a potentially life-threatening complication of an infection. Sepsis is mostly the consequence of systemic bacterial infections leading to exacerbated activation of immune cells by bacterial products, resulting in enhanced release of inflammatory mediators. Lipopolysaccharide (LPS), the major component of the outer membrane of Gram-negative bacteria, is a critical factor in the pathogenesis of sepsis, which is sensed by Toll-like receptor 4 (TLR4). The scientific community highly pursues the development of antagonists capable of blocking the cytokine storm by blocking TLR4. We report here that a recombinant molecule of 14.5 kDa belonging to the Fasciola hepatica fatty acid binding protein (Fh15) is capable of significantly suppressing the LPS-induced cytokine storm in a mouse model of septic shock when administered by the intraperitoneal route 1 h after a lethal LPS injection. These results suggest that Fh15 is an excellent candidate for drug development against endotoxemia.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Citocinas/metabolismo , Proteínas de Unión a Ácidos Grasos/administración & dosificación , Proteínas del Helminto/administración & dosificación , Lipopolisacáridos/inmunología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/fisiología , Choque Séptico/patología , ADP-Ribosil Ciclasa 1/análisis , Animales , Células Cultivadas , Citosol/química , Modelos Animales de Enfermedad , Ácidos Grasos/análisis , Inyecciones Intravenosas , Glicoproteínas de Membrana/análisis , Ratones , Proteínas Recombinantes/administración & dosificación , Bazo/inmunología
20.
Molecules ; 23(10)2018 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-30336585

RESUMEN

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


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Proteínas del Helminto/administración & dosificación , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Esclerosis Múltiple/tratamiento farmacológico , Acanthocheilonema/química , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/química , Diabetes Mellitus Tipo 1/patología , Modelos Animales de Enfermedad , Proteínas del Helminto/química , Helmintos/química , Humanos , Enfermedades Inflamatorias del Intestino/patología , Ratones , Esclerosis Múltiple/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA