Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Nat Microbiol ; 9(1): 120-135, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38066332

ABSTRACT

Oxidative stress triggers ferroptosis, a form of cellular necrosis characterized by iron-dependent lipid peroxidation, and has been implicated in Mycobacterium tuberculosis (Mtb) pathogenesis. We investigated whether Bach1, a transcription factor that represses multiple antioxidant genes, regulates host resistance to Mtb. We found that BACH1 expression is associated clinically with active pulmonary tuberculosis. Bach1 deletion in Mtb-infected mice increased glutathione levels and Gpx4 expression that inhibit lipid peroxidation. Bach1-/- macrophages exhibited increased resistance to Mtb-induced cell death, while Mtb-infected Bach1-deficient mice displayed reduced bacterial loads, pulmonary necrosis and lipid peroxidation concurrent with increased survival. Single-cell RNA-seq analysis of lungs from Mtb-infected Bach1-/- mice revealed an enrichment of genes associated with ferroptosis suppression. Bach1 depletion in Mtb-infected B6.Sst1S mice that display human-like necrotic lung pathology also markedly reduced necrosis and increased host resistance. These findings identify Bach1 as a key regulator of cellular and tissue necrosis and host resistance in Mtb infection.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis, Pulmonary , Tuberculosis , Animals , Mice , Basic-Leucine Zipper Transcription Factors/genetics , Macrophages/microbiology , Mycobacterium tuberculosis/genetics , Necrosis , Tuberculosis/microbiology , Tuberculosis, Pulmonary/genetics
2.
Exp Parasitol ; 238: 108267, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35550886

ABSTRACT

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.


Subject(s)
Ascariasis , Ascaris suum , Parasites , Animals , Ascariasis/parasitology , Cytokines , Inflammation , Liver/parasitology , Mice , Mice, Inbred C57BL , Nitric Oxide
3.
PLoS Pathog ; 17(11): e1010067, 2021 11.
Article in English | MEDLINE | ID: mdl-34784389

ABSTRACT

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.


Subject(s)
Ascariasis/immunology , Ascaris suum/immunology , Eosinophils/physiology , Immunoglobulin A, Secretory/metabolism , Pneumonia/prevention & control , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , Ascariasis/metabolism , Ascariasis/parasitology , Female , Immunoglobulin A, Secretory/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/parasitology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics
4.
PLoS Pathog ; 17(3): e1009337, 2021 03.
Article in English | MEDLINE | ID: mdl-33651853

ABSTRACT

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


Subject(s)
Dust/immunology , Hypersensitivity/immunology , Pyroglyphidae/immunology , Animals , Antigens, Dermatophagoides/immunology , Helminth Proteins/immunology , Immunoglobulin E/immunology , Immunoglobulin G/immunology , Mice , Proteomics
5.
Acta Trop ; 197: 105049, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31158344

ABSTRACT

The Kato-Katz (KK) technique is the mainstay mapping tool for the diagnosis of Schistosoma mansoni infection, despite showing poor sensitivity in cases of low-intensity infections. As an alternative, a rapid point-of-care circulating cathodic antigen diagnostic test (POC-CCA) has been commercially developed that involves a simple urine assay to detect S. mansoni, rather than a stool-based parasitological examination. Although POC-CCA has proven to be a more sensitive test than KK, it is not yet clear how to interpret discordant results between the two tests, particularly for situations in which the KK result is positive and the POC-CCA result is negative. Thus, the objective of this study was to evaluate the degree of diagnostic variability between different POC-CCA batches with respect to results obtained with KK. For this purpose, we collected urine and stool samples of school-aged children from areas of low and moderate endemicity in Brazil, and compared different POC-CCA batches results with those of KK-positive individuals. We found a statistically significant difference between the results obtained from various POC-CCA batches using the same urine samples, regardless of the degree of endemicity and the intensity of infection in positive KK samples. In addition, there was poor agreement between the KK and POC-CCA results in some batches of the rapid test, resulting in false negatives. These findings raise concerns around quality control checks of POC-CCA, especially in light of the high cost and increasing reliance on this new diagnostic method as control programs move towards a goal of elimination.


Subject(s)
Antigens, Helminth/immunology , Antigens, Helminth/urine , Point-of-Care Systems , Schistosoma mansoni/immunology , Schistosoma mansoni/isolation & purification , Schistosomiasis mansoni/parasitology , Animals , Brazil , Child , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/urine , Sensitivity and Specificity
6.
Vaccine ; 37(8): 1038-1045, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30685251

ABSTRACT

INTRODUCTION: Malaria transmission blocking vaccines (TBV) are innovative approaches that aim to induce immunity in humans against Plasmodium during mosquito stage, neutralizing the capacity of the infected vectors to transmit malaria. Pfs230D1-EPA/Alhydrogel®, a promising protein-protein conjugate malaria TBV, is currently being tested in human clinical trials in areas where P. falciparum malaria is coendemic with helminth parasites. Helminths are complex metazoans that share the master capacity to downregulate the host immune response towards themselves and also to bystander antigens, including vaccines. However, it is not known whether the activity of a protein-based malaria TBV may be affected by a chronic helminth infection. METHODS: Using an experimental murine model for a chronic helminth infection (Heligmosomoides polygyrus bakeri - Hpb), we evaluated whether prior infection alters the activity of Pfs230D1-EPA/Alhydrogel® TBV in mice. RESULTS: After establishment of a chronic infection, characterized by a marked increase of parasite antigen-specific IgG1, IgA and IgE antibody responses, concomitant with an increase of systemic IL-10, IL-5 and IL-6 levels, the Hpb-infected mice were immunized with Pfs230D1-EPA/Alhydrogel® and the vaccine-specific immune response was compared with that in non-infected immunized mice. TBV immunizations induced an elevated vaccine specific-antibody response, however Pfs230D1 specific-IgG levels were similar between infected and uninfected mice at days 15, 25 and 35 post-vaccination. Absolute numbers of Pfs230D1-activated B cells generated in response to the vaccine were also similar among the vaccinated groups. Finally, vaccine activity assessed by reduction of oocyst number in P. falciparum infected mosquitoes was similar between Hpb-infected and immunized mice with non-infected immunized mice. CONCLUSION: Pfs230D1-EPA/Alhydrogel® efficacy is not impaired by a chronic helminth infection in mice.


Subject(s)
Aluminum Hydroxide/immunology , Antigens, Protozoan/immunology , Carrier Proteins/immunology , Helminths/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Protozoan Proteins/immunology , Animals , Antibodies, Protozoan/immunology , Antigens/immunology , Immunization/methods , Male , Mice , Mice, Inbred BALB C , Plasmodium falciparum/immunology , Vaccination/methods , Vaccines, Conjugate/immunology
7.
Front Immunol ; 9: 2535, 2018.
Article in English | MEDLINE | ID: mdl-30473693

ABSTRACT

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.


Subject(s)
Ascaris suum/immunology , Immunoglobulin G/immunology , Protective Agents/pharmacology , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Helminth/immunology , Antigens, Helminth/immunology , Ascariasis/immunology , Ascariasis/parasitology , Female , Immunity/drug effects , Immunity/immunology , Immunization/methods , Interleukin-10/immunology , Larva/immunology , Lung/immunology , Lung/parasitology , Male , Mice , Mice, Inbred BALB C , Swine/immunology , Swine/parasitology , Swine Diseases/immunology , Swine Diseases/parasitology , Vaccination/methods , Vaccines/immunology
9.
Front Immunol, v. 9, 2535, 2018
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: bud-2606

ABSTRACT

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.

10.
Front. Immunol. ; 9: 2535, 2018.
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: but-ib15682

ABSTRACT

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.

11.
BMC Infect Dis ; 17(1): 253, 2017 04 08.
Article in English | MEDLINE | ID: mdl-28390393

ABSTRACT

BACKGROUND: While the macrophage polarization is well characterized in helminth infections, the natural heterogeneity of monocytes with multiple cell phenotypes might influence the outcome of neglected diseases, such hookworm infection. Here, we report the profile of monocytes in human hookworm infections as a model to study the regulatory subpopulation of monocytes in helminth infections. METHODS: Blood samples were collected from 19 Necator americanus-infected individuals and 13 healthy individuals. Peripheral blood mononuclear cells (PBMCs) were isolated, and immunophenotyping was conducted by flow cytometry. The expressions of genes encoding human nitric oxide synthase (iNOS), interleukin 4 (IL-4), arginase-1 (Arg-1) and glyceraldehyde 3-phosphate dehydrogenase were quantified by qPCR. Plasma levels of IL-4 were determined by sandwich ELISA. Unpaired t-tests or Mann-Whitney tests were used depending on the data distribution. RESULTS: Hookworm infected individuals (HWI) showed a significant increase in the number of monocytes/mm3 (555.2 ± 191.0) compared to that of the non-infected (NI) individuals (120.4 ± 44.7) (p < 0.0001). While the frequencies of CD14+IL-10+ and CD14+IL-12+ cells were significantly reduced in the HWI compared to NI group (p = 0.0289 and p < 0.0001, respectively), the ratio between IL-10/IL-12 producing monocytes was significantly elevated in HWI (p = 0.0004), indicating the potential regulatory activity of these cells. Measurement of IL-4 levels and gene expression of IL-4 and Arg-1 (highly expressed in alternatively activated macrophages) revealed no significant differences between the NI and HWI groups. Interestingly, individuals from the HWI group had higher expression of the iNOS gene (associated with a regulatory profile) (20.27 ± 2.97) compared to the NI group (11.28 ± 1.18, p = 0.0409). Finally, individuals from the HWI group had a significantly higher frequency of CD206+CD23+IL-10+ (7.57 ± 1.96) cells compared to individuals from the NI group (0.35 ± 0.09) (p < 0.001), suggesting that activated monocytes are a potential source of regulatory cytokines during hookworm infection. CONCLUSIONS: Natural hookworm infection induces a high frequency of circulating monocytes that present a regulatory profile and promote the downmodulation of the proinflammatory response, which may contribute to prolonged survival of the parasite in the host.


Subject(s)
Hookworm Infections/immunology , Monocytes/immunology , Adult , Aged , Animals , Arginase/genetics , Cytokines/metabolism , Female , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Humans , Immunophenotyping , Interleukin-10/metabolism , Interleukin-12/metabolism , Interleukin-4/metabolism , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Nitric Oxide Synthase Type II/genetics , Peptide Fragments/genetics
12.
Int J Parasitol ; 47(1): 1-10, 2017 01.
Article in English | MEDLINE | ID: mdl-28003150

ABSTRACT

The aim of this work was to elucidate the immunopathological mechanisms of how helminths may influence the course of a viral infection, using a murine model. Severe virulence, a relevant increase in the virus titres in the lung and a higher mortality rate were observed in Ascaris and Vaccinia virus (VACV) co-infected mice, compared with VACV mono-infected mice. Immunopathological analysis suggested that the ablation of CD8+ T cells, the marked reduction of circulating CD4+ T cells producing IFN-γ, and the robust pulmonary inflammation were associated with the increase of morbidity/mortality in co-infection and subsequently with the negative impact of concomitant pulmonary ascariasis and respiratory VACV infection for the host. On the other hand, when evaluating the impact of the co-infection on the parasitic burden, co-infected mice presented a marked decrease in the total number of migrating Ascaris lung-stage larvae in comparison with Ascaris mono-infection. Taken together, our major findings suggest that Ascaris and VACV co-infection may potentiate the virus-associated pathology by the downmodulation of the VACV-specific immune response. Moreover, this study provides new evidence of how helminth parasites may influence the course of a coincident viral infection.


Subject(s)
Ascariasis/virology , Ascaris/immunology , Coinfection/immunology , Pneumonia/parasitology , Vaccinia virus/immunology , Vaccinia/etiology , Animals , Ascariasis/immunology , CD8-Positive T-Lymphocytes/immunology , Coinfection/parasitology , Coinfection/virology , Cytokines/immunology , Disease Models, Animal , Female , Interferon-gamma/immunology , Larva/parasitology , Lung/immunology , Lung/parasitology , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Pneumonia/immunology , Pneumonia/virology , Swine , Vaccinia/immunology , Vaccinia/pathology , Vaccinia/virology , Viral Load
13.
PLoS Negl Trop Dis ; 10(1): e0004382, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26814713

ABSTRACT

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.


Subject(s)
Ascariasis/immunology , Ascaris suum/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Animals , Ascariasis/parasitology , Ascaris suum/physiology , Cytokines/immunology , Female , Humans , Lung/immunology , Lung/parasitology , Male , Mice , Mice, Inbred BALB C
14.
Trop Med Int Health ; 20(12): 1787-96, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26426162

ABSTRACT

OBJECTIVES: To identify immunodominant antigens of Toxocara canis recognised by Toxocara-infected sera as recombinant reagents for immunodiagnosis of toxocariasis. METHODS: Pooled sera from human cases of toxocariasis were used to identify immunodominant antigens by immunoscreening a T. canis larval expression cDNA library. The positive clones were sequenced to reveal the identity of the antigens. The recombinant proteins were expressed in E. coli and then used to confirm their immunoreaction with sera of humans with toxocariasis. Two chosen antigens were also used to differentiate Toxocara infection from other helminth infections in mice. RESULTS: Eleven antigens with immunodiagnostic potential were identified, including two C-type lectins (CTLs) that reacted strongly with the Toxocara-positive serum pool. The first CTL (Tc-CTL-1) is the same as TES-32, previously identified as a major immunodominant component of TES; the second CTL (Tc-CTL-2) is a novel C-type lectin sharing 83% amino acid sequence identity within the functional domain of Tc-CTL-1. The E. coli-expressed recombinant Tc-CTL-1 was strongly recognised by the Toxocara-positive serum pool or sera from animals experimentally infected with T. canis. Reactivity with recombinant Tc-CTL-1 was higher when the unreduced protein was used in an enzyme-linked immunosorbent assay (ELISA), dot-blot assay or Western blot test compared to the protein under reduced condition. Both recombinant Tc-CTL-1- and Tc-CTL-2-based ELISAs were able to differentiate T. canis infection from other helminth infections in experimentally infected mice. CONCLUSIONS: Both Tc-CTL-1 and Tc-CTL-2 were able to differentiate Toxocara infection from other helminth infections and could potentially be used as sensitive and specific immunodiagnostic antigens.


Subject(s)
Antigens, Helminth/immunology , Immunodominant Epitopes , Toxocara canis/immunology , Toxocariasis/diagnosis , Amino Acid Sequence , Animals , Antibodies, Helminth/blood , Blotting, Western , Clinical Laboratory Techniques , DNA, Complementary , Enzyme-Linked Immunosorbent Assay , Escherichia coli , Helminthiasis/diagnosis , Helminthiasis/immunology , Humans , Larva , Lectins/immunology , Mice, Inbred C57BL , Recombinant Proteins/immunology , Toxocariasis/immunology
15.
Parasit Vectors ; 8: 354, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26135397

ABSTRACT

BACKGROUND: Nematodes of the genus Toxocara are cosmopolitan roundworms frequently found in dogs and cats. Toxocara spp. can accidentally infect humans and cause a zoonosis called human toxocariasis, which is characterized by visceral, ocular or cerebral migration of larval stages of the parasite, without completing its life cycle. In general, chronic nematode infections induce a polarized TH2 immune response. However, during the initial phase of infection, a strong pro-inflammatory response is part of the immunological profile and might determine the outcome and/or pathology of the infection. METHODS: Parasitological aspects and histopathology during larval migration were evaluated after early T. canis experimental infection of BALB/c mice, which were inoculated via the intra-gastric route with a single dose of 1000 fully embryonated eggs. Innate immune responses and systemic cytokine patterns (TH1, TH2, TH17 and regulatory cytokines) were determined at different times after experimental challenge by sandwich ELISA. RESULTS: We found that experimental infection with T. canis induced a mix of innate inflammatory/TH17/TH2 responses during early infection, with a predominance of the latter. The TH2 response was evidenced by significant increases in cytokines such as IL-4, IL-5, IL-13 and IL-33, in addition to increasing levels of IL-6 and IL-17. No significant increases were observed for IL-10, TNF-α or IFN-γ levels. In parallel, parasitological analysis clearly revealed the pattern of larval migration through the mouse organs, starting from the liver in the first 24 h of infection, reaching the peak in the lungs on the 3rd day of infection and finally being found numerously in the brain after 5 days of infection. Peripheral leukocytosis, characterized by early neutrophilia and subsequent eosinophilia, was remarkable during early infection. The tissue damage induced by larvae was evidenced by histopathological analysis of the organs at different time points of infection. In all of the affected organs, larval migration induced intense inflammatory infiltrate and hemorrhage. CONCLUSION: In conclusion, these new insights into early T. canis infection in mice presented here enabled a better understanding of the immunopathological events that might also occur during human toxocariasis, thus contributing to future strategies of diagnosis and control.


Subject(s)
Toxocara canis/physiology , Toxocariasis/immunology , Toxocariasis/parasitology , Animals , Disease Models, Animal , Female , Humans , Interleukin-17/immunology , Interleukin-4/immunology , Interleukin-5/immunology , Male , Mice , Mice, Inbred BALB C , Th17 Cells/immunology , Th2 Cells/immunology , Toxocariasis/pathology
16.
BMC Infect Dis ; 15: 35, 2015 Jan 31.
Article in English | MEDLINE | ID: mdl-25636730

ABSTRACT

BACKGROUND: For a long time, the role of CD8(+) T cells in blood-stage malaria was not considered important because erythrocytes do not express major histocompatibility complex (MHC) class I proteins. While recent evidences suggest that CD8(+) T cells may play an important role during the erythrocytic phase of infection by eliminating parasites, CD8(+) T cells might also contribute to modulate the host response through production of regulatory cytokines. Thus, the role of CD8(+) T cells during blood-stage malaria is unclear. Here, we report the phenotypic profiling of CD8(+) T cells subsets from patients with uncomplicated symptomatic P. vivax malaria. METHODS: Blood samples were collected from 20 Plasmodium vivax-infected individuals and 12 healthy individuals. Immunophenotyping was conducted by flow cytometry. Plasma levels of IFN-γ, TNF-α and IL-10 were determined by ELISA/CBA. Unpaired t-test or Mann-Whitney test was used depending on the data distribution. RESULTS: P. vivax-infected subjects had lower percentages and absolute numbers of CD8(+)CD45RA(+) and CD8(+)CD45RO(+) T cells when compared to uninfected individuals (p ≤ 0.0002). A significantly lower absolute number of circulating CD8(+)CD45(+)CCR7(+) cells (p = 0.002) was observed in P. vivax-infected individuals indicating that infection reduces the number of central memory T cells. Cytokine expression was significantly reduced in the naïve T cells from infected individuals compared with negative controls, as shown by lower numbers of IFN-γ(+) (p = 0.001), TNF-α(+) (p < 0.0001) and IL-10(+) (p < 0.0001) CD8(+) T cells. Despite the reduction in the number of CD8(+) memory T cells producing IFN-γ (p < 0.0001), P. vivax-infected individuals demonstrated a significant increase in memory CD8(+)TNF-α(+) (p = 0.016) and CD8(+)IL-10(+) (p = 0.004) cells. Positive correlations were observed between absolute numbers of CD8(+)IL-10(+) and numbers of CD8(+)IFN-γ(+) (p < 0.001) and CD8(+)TNF-α(+) T cells (p ≤ 0.0001). Finally, an increase in the plasma levels of TNF-α (p = 0.017) and IL-10 (p = 0.006) and a decrease in the IFN-γ plasma level (p <0.0001) were observed in the P. vivax-infected individuals. CONCLUSIONS: P. vivax infection reduces the numbers of different subsets of CD8(+) T cells, particularly the memory cells, during blood-stage of infection and enhances the number of CD8(+) memory T cells expressing IL-10, which positively correlates with the number of cells expressing TNF-α and IFN-γ.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Malaria, Vivax/immunology , Plasmodium vivax/immunology , Adult , Aged , Blood Cell Count , Case-Control Studies , Female , Flow Cytometry , Humans , Malaria, Vivax/blood , Male , Middle Aged , Phenotype , Young Adult
17.
Int J Parasitol ; 43(9): 697-706, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23665127

ABSTRACT

Studies related to the immunobiological aspects of an Ascaris spp. infection are still scarce, especially those that aim to elucidate the early events of the immune response. In this study, we demonstrated a novel standardized method for early experimental Ascaris infection, providing additional information about the infectivity of eggs embryonated in vitro as well as the influence of host age on development of the infection. Finally, we characterised the immunopathology of early infection, focusing on the tissue and systemic cytokine profiles and the histopathology of infection in the lungs of BALB/c mice. Our results demonstrated that the highest egg infectivity occurred on the 100th and 200th days of in vitro embryonation and that 8 week-old BALB/c mice were more susceptible to infection than 16 week-old mice. Ascaris-infected mice showed an early, significant level of IL-5 production in the lungs 4 days p.i., followed by an increase in the level of neutrophils in the inflammatory infiltrate at 8 days p.i, which was correlated with the peak of larval migration in the tissue and a significant level of IL-6 production. The inflammatory infiltrate in the lungs was gradually replaced by mononuclear cells and eosinophils on the 10th and 12th days p.i., respectively, and an increase in TNF levels was observed. The downmodulation of systemic TCD4(+) cell numbers might suggest that T cell hyporesponsiveness was induced by the Ascaris spp. larvae, contributing to safeguarding parasite survival during larval migration. Taken together, the novel aspects of Ascaris infection presented here enabled a better understanding of the immunopathological events during larval migration, providing insight for further studies focused on immunisation and immunoprophylatic assays.


Subject(s)
Ascariasis/immunology , Ascariasis/parasitology , Ascaris suum , Aging , Animals , Ascariasis/pathology , Intestines/parasitology , Liver/parasitology , Lung/parasitology , Male , Mice , Mice, Inbred BALB C , Ovum
18.
Front Biosci (Elite Ed) ; 5(2): 662-75, 2013 01 01.
Article in English | MEDLINE | ID: mdl-23277021

ABSTRACT

While several mechanisms of immunoregulation have been demonstrated for hookworm and other neglected tropical infections, the influence of apoptosis in the immunomodulation of hookworm infection is still poorly understood. In this study, we demonstrate the cytotoxic and pro-apoptotic activity of hookworm antigens in Jurkat T cells, mesenteric lymph nodes lymphocytes of healthy and hookworm-infected hamsters and during human natural infection. Our results showed that in vitrostimulation of Jurkat T cells with antigens induces a significant decrease of cell viability leading to a relevant increase of apoptotic cells. Similar results were also observed in experimental conditions, for both healthy and hookworm-infected hamsters` lymphocytes. Flow cytometric analysis demonstrated that hookworm-infected patients presented a significant increase of CD4+, CD8+, and CD19+lymphocytes in early and/or late apoptosis when compared with non-infected individuals. The downmodulation of TNF receptors, as well as the up-regulation of the pro-apoptotic genes belonging to the BCL-2 and P53 families, suggest that hookworm antigens induced apoptosis by an intrinsic mitochondrial pathway, acting as a sophisticated strategy to safeguard parasite long-term survival in their hosts.


Subject(s)
Antigens, Helminth/immunology , Apoptosis/immunology , Hookworm Infections/immunology , Immunomodulation/immunology , Lymph Nodes/chemistry , Mesentery/cytology , Animals , Annexin A5 , Brazil , Cell Proliferation , Cricetinae , Flow Cytometry , Humans , Jurkat Cells , Microscopy, Confocal , Propidium
19.
Exp Parasitol ; 133(1): 80-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23142084

ABSTRACT

Corticosteroids and cyclosporine A (CsA) are important clinical immunosuppressive drugs used in the maintenance of organ transplants and in suppressing undesired autoimmune or allergic immune responses. To study the effect of CsA and prednisolone on the course of an Ancylostoma ceylanicum infection, hamsters were treated with commercially available prednisolone or CsA. For both drugs, half the recommended dose was sufficient to inhibit the proliferation of more than 70% of hamster lymph node cells. There was no difference in the recovery of adult worms; however, animals treated with prednisolone presented with low egg counts in the feces. Infection with A. ceylanicum resulted in an increase in specific antibodies against adult worm antigens, but hamsters treated with either drug presented with lower IgG titers. We observed that A. ceylanicum infection caused peripheral cellular immune suppression, which is characterized by a reduction in the total white cell count, neutropenia and lymphopenia. We also observed a lymphoplasmacytic pattern and few eosinophils in the mucosal inflammatory infiltrate for all the animals. The animals treated with prednisolone showed changes in the architecture of the intestine, including the loss of the mucosa, intense congestion and inflammation. In spleen, we observed hyperplasia of white pulp in all infected animals; in addition, there was a loss of tissue architecture in the animals treated with prednisolone. In conclusion, this work shows that an A. ceylanicum infection leads to acute peripheral cellular immune suppression in hamsters but not humoral immune suppression and that CsA treatment does not interfere with the process of infection. However, prednisolone treatment causes intestinal injury, what could hamper the parasite attachment to the intestinal wall, and as a result affects copulation and, consequently, decreases the number of eggs eliminated in the feces. Moreover, the possibility that the drug can also be exerting an effect on female fertility should be considered.


Subject(s)
Ancylostomiasis/drug therapy , Cyclosporine/therapeutic use , Glucocorticoids/therapeutic use , Immunosuppressive Agents/therapeutic use , Prednisolone/therapeutic use , Ancylostomiasis/immunology , Animals , Cell Proliferation/drug effects , Cricetinae , Cyclosporine/pharmacology , Disease Models, Animal , Feces/parasitology , Female , Glucocorticoids/pharmacology , Immunoglobulin G/blood , Immunosuppressive Agents/pharmacology , Intestine, Small/parasitology , Intestine, Small/pathology , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mesentery , Mesocricetus , Parasite Egg Count , Prednisolone/pharmacology , Spleen/pathology
20.
Rev Bras Parasitol Vet ; 21(3): 283-6, 2012.
Article in English | MEDLINE | ID: mdl-23070440

ABSTRACT

The aim of this study was to evaluate the predatory activity of the fungus Duddingtonia flagrans (AC001) on infective larvae of Ancylostoma ceylanicum after gastrointestinal transit in hamsters. Twenty animals were used in the experiment, divided into two groups: a treated group (10 animals) and a control group (10 animals). In the group treated with D. flagrans, each animal received mycelium from the AC001 isolate, at an oral dose of 5 mg/25 g of live weight. To evaluate the predatory activity of the fungus, fecal samples were collected from the animals in both groups, at the times of 6, 8, 12, 24 and 36 hours after the treatment. Then, subsamples of 2 g of feces were placed in Petri dishes containing 2% water-agar (2% WA) culture medium and 1000 L3 of A. ceylanicum. Over the study period, the following percentage reductions were observed: 43.2% (6 hours), 30.8% (8 hours), 25.8% (12 hours), 30% (24 hours) and 11% (36 hours). The fungus D. flagrans presented predatory activity on the L3 of A. ceylanicum, after passing through the hamsters' gastrointestinal tract. It was therefore concluded that the fungus D. flagrans may be an alternative for biological control of the L3 of A. ceylanicum.


Subject(s)
Ancylostoma , Biological Control Agents , Duddingtonia , Animals , Cricetinae , Larva
SELECTION OF CITATIONS
SEARCH DETAIL
...