Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Exp Parasitol ; 238: 108267, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35550886

RESUMEN

BACKGROUND: Human ascariasis is one of the most prevalent neglected tropical diseases worldwide. The immune response during human ascariasis is characterized by Th2 polarization and a mixed Th2/Th17 response during the pathogenesis of experimental larval ascariasis. Cytokines and other pro-inflammatory mediators, such as nitric oxide (NO), are involved in helminthic infections. However, the role of NO in ascariasis remains unclear. OBJECTIVES: Given the importance of NO in inflammation, we aimed to determine the immunological and histopathological alterations in the livers of C57BL/6 iNOS-/- mice during A. suum infection. METHODS: In this study, parasitic load was evaluated in the livers of wild type C57BL/6 and C57BL/6 iNOS-/- mice infected with A. suum. Histopathological and morphometric analyses and analysis of serum cytokines via Cytometric Bead Array were performed, and the activity of eosinophil peroxidase and myeloperoxidase of neutrophils in the tissues were determined. RESULTS: The results showed that NO is important for controlling parasitic load during infection by A. suum. C57BL/6iNOS-/- mice showed reduced inflammatory processes and less tissue damage during liver larval migration of A. suum, which is associated with a reduction in serum levels of pro-inflammatory cytokines. CONCLUSIONS: We demonstrated that NO is a crucial inflammatory molecule during Ascaris sp. infection and controls the establishment of the parasite and the development of the host immune response in the liver.


Asunto(s)
Ascariasis , Ascaris suum , Parásitos , Animales , Ascariasis/parasitología , Citocinas , Inflamación , Hígado/parasitología , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico
2.
Infect Immun ; 90(2): e0059521, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-34807734

RESUMEN

Ascariasis is a neglected tropical disease that is widespread in the world and has important socioeconomic impacts. The presence of various stages of worm development in the pulmonary and intestinal mucosae induces a humoral and cellular immune response. However, although there is much evidence of the protective role of mucosal immunity against various pathogens, including helminths, there is still a gap in the knowledge about the immune response and the mechanisms of action that are involved in protection against diseases, especially in the initial phase of ascariasis. Thus, the aim of this study was to evaluate the kinetic aspects of the immune parasitological parameters in intestinal and pulmonary mucosae in male mice with early ascariasis. Therefore, two mouse strains that showed different susceptibilities to ascariasis (BALB/c and C57BL/6J) when experimentally infected with 2,500 infective eggs of Ascaris suum from time point 0 were examined: the immune parasitological parameters were evaluated each 2 days after infection over a period of 12 days. The results were suggestive of a synergetic action of intestinal and pulmonary secretory IgA (S-IgA) contributing to protection against early ascariasis by reducing the amount of migrating larvae as well as the influx of leukocytes in the lung and the consequent impairment of pulmonary capacity.


Asunto(s)
Ascariasis , Ascaris suum , Parásitos , Neumonía , Enfermedades de los Porcinos , Animales , Ascaris suum/genética , Antecedentes Genéticos , Inmunoglobulina A Secretora , Masculino , Ratones , Ratones Endogámicos C57BL , Porcinos
3.
PLoS Pathog ; 17(11): e1010067, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34784389

RESUMEN

Human ascariasis is the most prevalent but neglected tropical disease in the world, affecting approximately 450 million people. The initial phase of Ascaris infection is marked by larval migration from the host's organs, causing mechanical injuries followed by an intense local inflammatory response, which is characterized mainly by neutrophil and eosinophil infiltration, especially in the lungs. During the pulmonary phase, the lesions induced by larval migration and excessive immune responses contribute to tissue remodeling marked by fibrosis and lung dysfunction. In this study, we investigated the relationship between SIgA levels and eosinophils. We found that TLR2 and TLR4 signaling induces eosinophils and promotes SIgA production during Ascaris suum infection. Therefore, control of parasite burden during the pulmonary phase of ascariasis involves eosinophil influx and subsequent promotion of SIgA levels. In addition, we also demonstrate that eosinophils also participate in the process of tissue remodeling after lung injury caused by larval migration, contributing to pulmonary fibrosis and dysfunction in re-infected mice. In conclusion, we postulate that eosinophils play a central role in mediating host innate and humoral immune responses by controlling parasite burden, tissue inflammation, and remodeling during Ascaris suum infection. Furthermore, we suggest that the use of probiotics can induce eosinophilia and SIgA production and contribute to controlling parasite burden and morbidity of helminthic diseases with pulmonary cycles.


Asunto(s)
Ascariasis/inmunología , Ascaris suum/inmunología , Eosinófilos/fisiología , Inmunoglobulina A Secretora/metabolismo , Neumonía/prevención & control , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Ascariasis/metabolismo , Ascariasis/parasitología , Femenino , Inmunoglobulina A Secretora/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neumonía/inmunología , Neumonía/parasitología , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética
4.
Malar J ; 20(1): 296, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-34210332

RESUMEN

BACKGROUND: Ascariasis and malaria are highly prevalent parasitic diseases in tropical regions and often have overlapping endemic areas, contributing to high morbidity and mortality rates in areas with poor sanitary conditions. Several studies have previously aimed to correlate the effects of Ascaris-Plasmodium coinfections but have obtained contradictory and inconclusive results. Therefore, the present study aimed to investigate parasitological and immunopathological aspects of the lung during murine experimental concomitant coinfection by Plasmodium berghei and Ascaris suum during larvae ascariasis. METHODS: C57BL/6J mice were inoculated with 1 × 104 P. berghei strain NK65-NY-infected red blood cells (iRBCs) intraperitoneally and/or 2500 embryonated eggs of A. suum by oral gavage. P. berghei parasitaemia, morbidity and the survival rate were assessed. On the seventh day postinfection (dpi), A. suum lung burden analysis; bronchoalveolar lavage (BAL); histopathology; NAG, MPO and EPO activity measurements; haematological analysis; and respiratory mechanics analysis were performed. The concentrations of interleukin (IL)-1ß, IL-12/IL-23p40, IL-6, IL-4, IL-33, IL-13, IL-5, IL-10, IL-17A, IFN-γ, TNF and TGF-ß were assayed by sandwich ELISA. RESULTS: Animals coinfected with P. berghei and A. suum show decreased production of type 1, 2, and 17 and regulatory cytokines; low leukocyte recruitment in the tissue; increased cellularity in the circulation; and low levels of NAG, MPO and EPO activity that lead to an increase in larvae migration, as shown by the decrease in larvae recovered in the lung parenchyma and increase in larvae recovered in the airway. This situation leads to severe airway haemorrhage and, consequently, an impairment respiratory function that leads to high morbidity and early mortality. CONCLUSIONS: This study demonstrates that the Ascaris-Plasmodium interaction is harmful to the host and suggests that this coinfection may potentiate Ascaris-associated pathology by dampening the Ascaris-specific immune response, resulting in the early death of affected animals.


Asunto(s)
Ascariasis , Coinfección , Regulación hacia Abajo/inmunología , Inmunidad Innata/genética , Malaria , Animales , Ascariasis/inmunología , Ascariasis/parasitología , Ascariasis/patología , Ascaris suum/genética , Ascaris suum/fisiología , Coinfección/inmunología , Coinfección/parasitología , Coinfección/patología , Regulación de la Expresión Génica , Pulmón/patología , Malaria/inmunología , Malaria/parasitología , Malaria/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei/fisiología
5.
PLoS Negl Trop Dis ; 15(7): e0009639, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34324507

RESUMEN

Toxocariasis is a neglected disease that affects people around the world. Humans become infected by accidental ingestion of eggs containing Toxocara canis infective larvae, which upon reaching the intestine, hatch, penetrate the mucosa and migrate to various tissues such as liver, lungs and brain. Studies have indicated that Th2 response is the main immune defense mechanism against toxocariasis, however, there are still few studies related to this response, mainly the IL-33/ST2 pathway. Some studies have reported an increase in IL-33 during helminth infections, including T. canis. By binding to its ST2 receptor, IL-33 stimulating the Th2 polarized immune cell and cytokine responses. Thus, we aimed to investigate the role of the IL-33/ST2 pathway in the context of T. canis larval migration and the immunological and pathophysiological aspects of the infection in the liver, lungs and brain from Wild-Type (WT) BALB/c background and genetically deficient mice for the ST2 receptor (ST2-/-). The most important findings revealed that the IL-33/ST2 pathway is involved in eosinophilia, hepatic and cerebral parasitic burden, and induces the formation of granulomas related to tissue damage and pulmonary dysfunction. However, ST2-/- mice, the immune response was skewed to Th1/Th17 type than Th2, that enhanced the control of parasite burden related to IgG2a levels, tissue macrophages infiltration and reduced lung dysfunction. Collectively, our results demonstrate that the Th2 immune response triggered by IL-33/ST2 pathway mediates susceptibility to T. canis, related to parasitic burden, eosinophilia and granuloma formation in which consequently contributes to tissue inflammation and injury.


Asunto(s)
Eosinófilos/fisiología , Inflamación/inmunología , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/metabolismo , Toxocara canis , Toxocariasis/inmunología , Animales , Femenino , Regulación de la Expresión Génica , Proteína 1 Similar al Receptor de Interleucina-1/genética , Interleucina-33/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Células Th2/fisiología , Toxocariasis/patología
6.
PLoS Negl Trop Dis ; 15(7): e0009613, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34314435

RESUMEN

Chagas disease, caused by the parasite Trypanosoma cruzi, is considered endemic in more than 20 countries but lacks both an approved vaccine and limited treatment for its chronic stage. Chronic infection is most harmful to human health because of long-term parasitic infection of the heart. Here we show that immunization with a virus-like particle vaccine displaying a high density of the immunogenic α-Gal trisaccharide (Qß-αGal) induced several beneficial effects concerning acute and chronic T. cruzi infection in α1,3-galactosyltransferase knockout mice. Approximately 60% of these animals were protected from initial infection with high parasite loads. Vaccinated animals also produced high anti-αGal IgG antibody titers, improved IFN-γ and IL-12 cytokine production, and controlled parasitemia in the acute phase at 8 days post-infection (dpi) for the Y strain and 22 dpi for the Colombian strain. In the chronic stage of infection (36 and 190 dpi, respectively), all of the vaccinated group survived, showing significantly decreased heart inflammation and clearance of amastigote nests from the heart tissue.


Asunto(s)
Cardiomiopatía Chagásica/prevención & control , Corazón/parasitología , Vacunas Antiprotozoos/inmunología , Trypanosoma cruzi , Animales , Anticuerpos Antiprotozoarios/sangre , Cardiomiopatía Chagásica/parasitología , Cardiomiopatía Chagásica/patología , Citocinas/genética , Citocinas/metabolismo , Femenino , Regulación de la Expresión Génica/inmunología , Inmunoglobulina G/sangre , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/parasitología , Ratones , Ratones Endogámicos C57BL , Parasitemia , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
7.
Front Immunol ; 12: 788185, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34992603

RESUMEN

Control of human ascariasis, the most prevalent neglected tropical disease globally affecting 450 million people, mostly relies on mass drug administration of anthelmintics. However, chemotherapy alone is not efficient due to the high re-infection rate for people who live in the endemic area. The development of a vaccine that reduces the intensity of infection and maintains lower morbidity should be the primary target for infection control. Previously, our group demonstrated that immunization with crude Ascaris antigens in mice induced an IgG-mediated protective response with significant worm reduction. Here, we aimed to develop a multipeptide chimera vaccine based on conserved B-cell epitopes predicted from 17 common helminth proteomes using a bioinformatics algorithm. More than 480 B-cell epitopes were identified that are conserved in all 17 helminths. The Ascaris-specific epitopes were selected based on their reactivity to the pooled sera of mice immunized with Ascaris crude antigens or infected three times with A. suum infective eggs. The top 35 peptides with the strongest reactivity to Ascaris immune serum were selected to construct a chimeric antigen connected in sequence based on conformation. This chimera, called ASCVac-1, was produced as a soluble recombinant protein in an Escherichia coli expression system and, formulated with MPLA, was used to immunize mice. Mice immunized with ASCVac-1/MPLA showed around 50% reduced larvae production in the lungs after being challenged with A. suum infective eggs, along with significantly reduced inflammation and lung tissue/function damage. The reduced parasite count and pathology in infected lungs were associated with strong Th2 immune responses characterized by the high titers of antigen-specific IgG and its subclasses (IgG1, IgG2a, and IgG3) in the sera and significantly increased IL-4, IL-5, IL-13 levels in lung tissues. The reduced IL-33 titers and stimulated eosinophils were also observed in lung tissues and may also contribute to the ASCVac-1-induced protection. Taken together, the preclinical trial with ASCVac-1 chimera in a mouse model demonstrated its significant vaccine efficacy associated with strong IgG-based Th2 responses, without IgE induction, thus reducing the risk of an allergic response. All results suggest that the multiepitope-based ASCVac-1 chimera is a promising vaccine candidate against Ascaris sp. infections.


Asunto(s)
Antígenos Helmínticos/administración & dosificación , Ascariasis/prevención & control , Ascaris suum/inmunología , Enfermedades Desatendidas/prevención & control , Vacunas Antiprotozoos/administración & dosificación , Animales , Antígenos Helmínticos/inmunología , Ascariasis/inmunología , Ascariasis/parasitología , Ascariasis/patología , Ascaris suum/aislamiento & purificación , Femenino , Humanos , Pulmón/inmunología , Pulmón/parasitología , Pulmón/patología , Ratones , Enfermedades Desatendidas/inmunología , Enfermedades Desatendidas/parasitología , Enfermedades Desatendidas/patología , Vacunas Antiprotozoos/inmunología , Células Th2/inmunología , Eficacia de las Vacunas , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología
8.
PLoS Negl Trop Dis ; 13(11): e0007896, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31765381

RESUMEN

Ascariasis is considered the most neglected tropical disease, and is a major problem for the public health system. However, idiopathic pulmonary fibrosis (IPF) is a result of chronic extracellular deposition of matrix in the pulmonary parenchyma, and thickening of the alveolar septa, which reduces alveolar gas exchange. Considering the high rates of ascariasis and pulmonary fibrosis, we believe that these two diseases may co-exist and possibly lead to comorbidities. We therefore investigated the mechanisms involved in comorbidity of Ascaris suum (A. suum) infection, which could interfere with the progression of pulmonary fibrosis. In addition, we evaluated whether a previous lung fibrosis could interfere with the pulmonary cycle of A. suum in mice. The most important findings related to comorbidity in which A. suum infection exacerbated pulmonary and liver injury, inflammation and dysfunction, but did not promote excessive fibrosis in mice during the investigated comorbidity period. Interestingly, we found that pulmonary fibrosis did not alter the parasite cycle that transmigrated preferentially through preserved but not fibrotic areas of the lungs. Collectively, our results demonstrate that A. suum infection leads to comorbidity, and contributes to the aggravation of pulmonary dysfunction during pulmonary fibrosis, which also leads to significant liver injury and inflammation, without changing the A. suum cycle in the lungs.


Asunto(s)
Ascariasis/complicaciones , Ascariasis/patología , Hepatopatías/patología , Fibrosis Pulmonar/complicaciones , Fibrosis Pulmonar/patología , Animales , Ascaris suum/aislamiento & purificación , Modelos Animales de Enfermedad , Femenino , Inflamación/patología , Pulmón/parasitología , Pulmón/patología , Ratones Endogámicos C57BL
9.
Front Immunol ; 9: 2535, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30473693

RESUMEN

Human ascariasis has a global and cosmopolitan distribution, and has been characterized as the most prevalent neglected tropical disease worldwide. The development of a preventive vaccine is highly desirable to complement current measures required for this parasitic infection control and to reduce chronic childhood morbidities. In the present study, we describe the mechanism of protection elicited by a preventive vaccine against ascariasis. Vaccine efficacy was evaluated after immunization with three different Ascaris suum antigen extracts formulated with monophosphoryl lipid A (MPLA) as an adjuvant: crude extract of adult worm (ExAD); crude extract of adult worm cuticle (CUT); and crude extract of infective larvae (L3) (ExL3). Immunogenicity elicited by immunization was assessed by measuring antibody responses, cytokine production, and influx of tissue inflammatory cells. Vaccine efficacy was evaluated by measuring the reductions in the numbers of larvae in the lungs of immunized BALB/c mice that were challenged with A. suum eggs. Moreover, lung physiology and functionality were tested by spirometry to determine clinical efficacy. Finally, the role of host antibody mediated protection was determined by passive transfer of serum from immunized mice. Significant reductions in the total number of migrating larvae were observed in mice immunized with ExL3 61% (p < 0.001), CUT 59% (p < 0.001), and ExAD 51% (p < 0.01) antigens in comparison with non-immunized mice. For the Ascaris antigen-specific IgG antibody levels, a significant and progressive increase was observed with each round of immunization, in association with a marked increase of IgG1 and IgG3 subclasses. Moreover, a significant increase in concentration of IL-5 and IL-10 (pre-challenge) in the blood and IL-10 in the lung tissue (post-challenge) was induced by CUT immunization. Finally, ExL3 and CUT-immunized mice showed a marked improvement in lung pathology and tissue fibrosis as well as reduced pulmonary dysfunction induced by Ascaris challenge, when compared to non-immunized mice. Moreover, the passive transfer of specific IgG antibodies from ExL3, CUT, and ExAD elicited a protective response in naïve mice, with significant reductions in parasite burdens in lungs of 65, 64, and 64%, respectively. Taken together, these studies indicated that IgG antibodies contribute to protective immunity.


Asunto(s)
Ascaris suum/inmunología , Inmunoglobulina G/inmunología , Sustancias Protectoras/farmacología , Adyuvantes Inmunológicos/farmacología , Animales , Anticuerpos Antihelmínticos/inmunología , Antígenos Helmínticos/inmunología , Ascariasis/inmunología , Ascariasis/parasitología , Femenino , Inmunidad/efectos de los fármacos , Inmunidad/inmunología , Inmunización/métodos , Interleucina-10/inmunología , Larva/inmunología , Pulmón/inmunología , Pulmón/parasitología , Masculino , Ratones , Ratones Endogámicos BALB C , Porcinos/inmunología , Porcinos/parasitología , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/parasitología , Vacunación/métodos , Vacunas/inmunología
10.
PLoS Negl Trop Dis ; 10(1): e0004382, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26814713

RESUMEN

Ascaris spp. infection affects 800 million people worldwide, and half of the world population is currently at risk of infection. Recurrent reinfection in humans is mostly due to the simplicity of the parasite life cycle, but the impact of multiple exposures to the biology of the infection and the consequences to the host's homeostasis are poorly understood. In this context, single and multiple exposures in mice were performed in order to characterize the parasitological, histopathological, tissue functional and immunological aspects of experimental larval ascariasis. The most important findings revealed that reinfected mice presented a significant reduction of parasite burden in the lung and an increase in the cellularity in the bronchoalveolar lavage (BAL) associated with a robust granulocytic pulmonary inflammation, leading to a severe impairment of respiratory function. Moreover, the multiple exposures to Ascaris elicited an increased number of circulating inflammatory cells as well as production of higher levels of systemic cytokines, mainly IL-4, IL-5, IL-6, IL-10, IL-17A and TNF-α when compared to single-infected animals. Taken together, our results suggest the intense pulmonary inflammation associated with a polarized systemic Th2/Th17 immune response are crucial to control larval migration after multiple exposures to Ascaris.


Asunto(s)
Ascariasis/inmunología , Ascaris suum/inmunología , Células Th17/inmunología , Células Th2/inmunología , Animales , Ascariasis/parasitología , Ascaris suum/fisiología , Citocinas/inmunología , Femenino , Humanos , Pulmón/inmunología , Pulmón/parasitología , Masculino , Ratones , Ratones Endogámicos BALB C
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA