Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 23(1): 23-32, 2022 01.
Article in English | MEDLINE | ID: mdl-34937933

ABSTRACT

Systemic immune cell dynamics during coronavirus disease 2019 (COVID-19) are extensively documented, but these are less well studied in the (upper) respiratory tract, where severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replicates1-6. Here, we characterized nasal and systemic immune cells in individuals with COVID-19 who were hospitalized or convalescent and compared the immune cells to those seen in healthy donors. We observed increased nasal granulocytes, monocytes, CD11c+ natural killer (NK) cells and CD4+ T effector cells during acute COVID-19. The mucosal proinflammatory populations positively associated with peripheral blood human leukocyte antigen (HLA)-DRlow monocytes, CD38+PD1+CD4+ T effector (Teff) cells and plasmablasts. However, there was no general lymphopenia in nasal mucosa, unlike in peripheral blood. Moreover, nasal neutrophils negatively associated with oxygen saturation levels in blood. Following convalescence, nasal immune cells mostly normalized, except for CD127+ granulocytes and CD38+CD8+ tissue-resident memory T cells (TRM). SARS-CoV-2-specific CD8+ T cells persisted at least 2 months after viral clearance in the nasal mucosa, indicating that COVID-19 has both transient and long-term effects on upper respiratory tract immune responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Nasopharynx/immunology , Nose/cytology , Respiratory Mucosa/immunology , SARS-CoV-2/immunology , Antibodies, Viral/blood , COVID-19/immunology , COVID-19/pathology , Granulocytes/immunology , HLA-DR Antigens/metabolism , Humans , Killer Cells, Natural/immunology , Memory T Cells/immunology , Monocytes/immunology , Nasopharynx/cytology , Nasopharynx/virology , Neutrophils/immunology , Nose/immunology , Nose/virology , Prospective Studies , Respiratory Mucosa/cytology , Respiratory Mucosa/virology
3.
Immunity ; 49(5): 801-818, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30462997

ABSTRACT

Helminths are extraordinarily successful parasites due to their ability to modulate the host immune response. They have evolved a spectrum of immunomodulatory molecules that are now beginning to be defined, heralding a molecular revolution in parasite immunology. These discoveries have the potential both to transform our understanding of parasite adaptation to the host and to develop possible therapies for immune-mediated disease. In this review we will summarize the current state of the art in parasite immunomodulation and discuss perspectives on future areas for research and discovery.


Subject(s)
Helminthiasis/immunology , Helminths/immunology , Host-Parasite Interactions , Immunomodulation , Adaptive Immunity , Animals , Biological Evolution , Dendritic Cells/immunology , Dendritic Cells/metabolism , Helminthiasis/parasitology , Humans , Immunity, Innate , Macrophages/immunology , Macrophages/metabolism
4.
PLoS Pathog ; 16(9): e1008799, 2020 09.
Article in English | MEDLINE | ID: mdl-32898164

ABSTRACT

Professional antigen-presenting cells (APCs), like macrophages (Mϕs) and dendritic cells (DCs), are central players in the induction of natural and vaccine-induced immunity to malaria, yet very little is known about the interaction of SPZ with human APCs. Intradermal delivery of whole-sporozoite vaccines reduces their effectivity, possibly due to dermal immunoregulatory effects. Therefore, understanding these interactions could prove pivotal to malaria vaccination. We investigated human APC responses to recombinant circumsporozoite protein (recCSP), SPZ and anti-CSP opsonized SPZ both in monocyte derived MoDCs and MoMϕs. Both MoDCs and MoMϕs readily took up recCSP but did not change phenotype or function upon doing so. SPZ are preferentially phagocytosed by MoMϕs instead of DCs and phagocytosis greatly increased after opsonization. Subsequently MoMϕs show increased surface marker expression of activation markers as well as tolerogenic markers such as Programmed Death-Ligand 1 (PD-L1). Additionally they show reduced motility, produce interleukin 10 and suppressed interferon gamma (IFNγ) production by antigen specific CD8+ T cells. Importantly, we investigated phenotypic responses to SPZ in primary dermal APCs isolated from human skin explants, which respond similarly to their monocyte-derived counterparts. These findings are a first step in enhancing our understanding of pre-erythrocytic natural immunity and the pitfalls of intradermal vaccination-induced immunity.


Subject(s)
Antigen-Presenting Cells/immunology , Macrophages/immunology , Malaria/immunology , Plasmodium berghei/immunology , Protozoan Proteins/immunology , Skin/immunology , Sporozoites/immunology , Animals , Cells, Cultured , Female , Humans , Macrophages/parasitology , Malaria/parasitology , Mice , Skin/parasitology
5.
Lancet ; 396(10254): 854-866, 2020 09 19.
Article in English | MEDLINE | ID: mdl-32910907

ABSTRACT

Asthma is a complex disease that often starts in childhood. Genomic and environmental factors as well as aberrant immune maturation early in life can contribute to the onset of disease, with great disparity over time and geographical regions. Epidemiological studies have scrutinised environmental exposures and attempted to translate these exposures into prevention strategies. Some approaches for patients with asthma have been successful (eg, smoking ban, the Finnish Asthma Programme), and primary prevention of wheeze in pre-school children (age 0-5 years) by the supplementation of vitamin D or fish oil, or both, to pregnant women seems promising. Several recent prevention initiatives are based on strong asthma-protective environmental microbial exposures associated with traditional rural lifestyles. Preclinical studies with various bacterial lysates, bacterial and dietary metabolites, or helminthic compounds have yielded promising results that await translation into clinical practice. Given the immense societal and individual burden of asthma, there is an urgent need to further develop novel strategies to eradicate the disease.


Subject(s)
Asthma/epidemiology , Asthma/prevention & control , Primary Prevention/methods , Asthma/diet therapy , Asthma/immunology , Child, Preschool , Dietary Supplements/supply & distribution , Environmental Exposure/adverse effects , Female , Finland/epidemiology , Fish Oils/administration & dosage , Fish Oils/supply & distribution , Humans , Infant , Infant, Newborn , Male , Pregnancy , Prevalence , Protective Factors , Risk Factors , Smoking Prevention/methods , Vitamin D/administration & dosage , Vitamin D/supply & distribution
6.
PLoS Pathog ; 15(9): e1007924, 2019 09.
Article in English | MEDLINE | ID: mdl-31487324

ABSTRACT

Parasitic helminths evade, skew and dampen human immune responses through numerous mechanisms. Such effects will likely have consequences for HIV-1 transmission and disease progression. Here we analyzed the effects that soluble egg antigen (SEA) from Schistosoma mansoni had on modulating HIV-1 infection and cytokine/chemokine production in vitro. We determined that SEA, specifically through kappa-5, can potently bind to DC-SIGN and thereby blocks DC-SIGN mediated HIV-1 trans-infection (p<0.05) whilst not interfering with cis-infection. DCs exposed to SEA whilst maturing under Th2 promoting conditions, will upon co-culture with naïve T-cells induce a T-cell population that was less susceptible to HIV-1 R5 infection (p<0.05) compared to DCs unexposed to SEA, whereas HIV-1 X4 virus infection was unaffected. This was not observed for DCs exposed to SEA while maturing under Th1 or Th1/Th2 (Tmix) promoting conditions. All T-cell populations induced by SEA exposed DCs demonstrate a reduced capacity to produce IFN-γ and MIP-1ß. The infection profile of T-cells infected with HIV-1 R5 was not associated with down-modulation of CCR5 cell surface expression. We further show that DCs maturing under Tmix conditions exposed to plant recombinant omega-1 protein (rω-1), which demonstrates similar functions to natural ω-1, induced T-cell populations that were less sensitive for HIV-1 R5 infection (p<0.05), but not for X4 virus infection. This inhibition associated again with a reduction in IFN-γ and MIP-1ß expression, but additionally correlated with reduced CCR5 expression. We have shown that SEA parasite antigens and more specifically rω-1 can modulate HIV-1 infectivity with the potential to influence disease course in co-infected individuals.


Subject(s)
Antibodies, Helminth/immunology , Antigens, Helminth/immunology , Egg Proteins/immunology , HIV Infections/metabolism , Animals , Antibodies, Helminth/metabolism , Antigens, Helminth/metabolism , CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Egg Proteins/metabolism , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/immunology , Humans , Lymphocyte Activation , Receptors, CCR5/metabolism , Schistosoma mansoni/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Virus Replication/immunology
7.
J Infect Dis ; 222(12): 2103-2113, 2020 11 13.
Article in English | MEDLINE | ID: mdl-31844885

ABSTRACT

BACKGROUND: Although Schistosoma haematobium infection has been reported to be associated with alterations in immune function, in particular immune hyporesponsiveness, there have been only few studies that have used the approach of removing infection by drug treatment to establish this and to understand the underlying molecular mechanisms. METHODS: Schistosoma haematobium-infected schoolchildren were studied before and after praziquantel treatment and compared with uninfected controls. Cellular responses were characterized by cytokine production and flow cytometry, and in a subset of children RNA sequencing (RNA-Seq) transcriptome profiling was performed. RESULTS: Removal of S haematobium infection resulted in increased schistosome-specific cytokine responses that were negatively associated with CD4+CD25+FOXP3+ T-cells and accompanied by increased frequency of effector memory T-cells. Innate responses to Toll like receptor (TLR) ligation decreased with treatment and showed positive association with CD4+CD25+FOXP3+ T-cells. At the transcriptome level, schistosome infection was associated with enrichment in cell adhesion, whereas parasite removal was associated with a more quiescent profile. Further analysis indicated that alteration in cellular energy metabolism was associated with S haematobium infection and that the early growth response genes 2 and 3 (EGR 2 and EGR3), transcription factors that negatively regulate T-cell activation, may play a role in adaptive immune hyporesponsiveness. CONCLUSIONS: Using a longitudinal study design, we found contrasting effects of schistosome infection on innate and adaptive immune responses. Whereas the innate immune system appears more activated, the adaptive immunity is in a hyporesponsive state reflected in alterations in CD4+CD25+FOXP3+ T-cells, cellular metabolism, and transcription factors involved in anergy.


Subject(s)
Anthelmintics/therapeutic use , Cytokines/immunology , Praziquantel/therapeutic use , Schistosomiasis haematobia/immunology , Transcriptome , Adaptive Immunity , Animals , Child , Female , Flow Cytometry , Gabon/epidemiology , Humans , Immunity, Innate , Longitudinal Studies , Male , RNA-Seq , Schistosomiasis haematobia/drug therapy
8.
Eur J Immunol ; 49(8): 1226-1234, 2019 08.
Article in English | MEDLINE | ID: mdl-31099896

ABSTRACT

The helminth Schistosoma mansoni (S. mansoni) induces a network of regulatory immune cells, including interleukin (IL)-10-producing regulatory B cells (Bregs). However, the signals required for the development and activation of Bregs are not well characterized. Recent reports suggest that helminths induce type I interferons (IFN-I), and that IFN-I drive the development of Bregs in humans. We therefore assessed the role of IFN-I in the induction of Bregs by S. mansoni. Mice chronically infected with S. mansoni or i.v. injected with S. mansoni soluble egg antigen (SEA) developed a systemic IFN-I signature. Recombinant IFN-α enhanced IL-10 production by Bregs stimulated with S. mansoni SEA in vitro, while not activating Bregs by itself. IFN-I signaling also supported ex vivo IL-10 production by SEA-primed Bregs but was dispensable for activation of S. mansoni egg-induced Bregs in vivo. These data indicate that although IFN-I can serve as a coactivator for Breg IL-10 production, they are unlikely to participate in the development of Bregs in response to S. mansoni eggs.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Interferon Type I/metabolism , Schistosoma mansoni/physiology , Schistosomiasis mansoni/immunology , Animals , Antigens, Helminth/immunology , Cells, Cultured , Disease Models, Animal , Eggs , Female , Flow Cytometry , Humans , Interleukin-10/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Signal Transduction
9.
Eur Respir J ; 56(2)2020 08.
Article in English | MEDLINE | ID: mdl-32430415

ABSTRACT

Chronic respiratory diseases are highly prevalent worldwide and will continue to rise in the foreseeable future. Despite intensive efforts over recent decades, the development of novel and effective therapeutic approaches has been slow. However, there is new and increasing evidence that communities of micro-organisms in our body, the human microbiome, are crucially involved in the development and progression of chronic respiratory diseases. Understanding the detailed mechanisms underlying this cross-talk between host and microbiota is critical for development of microbiome- or host-targeted therapeutics and prevention strategies. Here we review and discuss the most recent knowledge on the continuous reciprocal interaction between the host and microbes in health and respiratory disease. Furthermore, we highlight promising developments in microbiome-based therapies and discuss the need to employ more holistic approaches of restoring both the pulmonary niche and the microbial community.


Subject(s)
Lung Diseases , Microbiota , Respiration Disorders , Respiratory Tract Diseases , Humans , Lung , Lung Diseases/therapy
10.
J Immunol ; 200(5): 1570-1579, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29352004

ABSTRACT

Epidemiological and experimental studies have shown that exposure to the gastric bacterium Helicobacter pylori, especially in early life, prevents the development of asthma. Recent mouse studies have shown that this protective effect does not require live bacteria and that treatment with an extract of H. pylori in neonates prevents the development of airway inflammation and goblet cell metaplasia. In the current study, the effect of administration of an extract of H. pylori was assessed in a therapeutic study design with application of the extract just prior to allergen challenge. C57BL/6 mice were sensitized and challenged with OVA or house dust mite. Treatment with H. pylori extract just prior to the challenge significantly reduced airway inflammation, as assessed in bronchoalveolar lavage fluid and lung tissue, and reduced airway remodeling, as assessed by goblet cell quantification. These effects were apparent in the OVA model and in the house dust mite model. Injection of H. pylori extract reduced the processing of allergen by dendritic cells in the lungs and mediastinal lymph node. Bone marrow-derived dendritic cells exposed to H. pylori extract were affected with regard to their ability to process Ag. These data show that application of H. pylori extract after sensitization effectively inhibits allergic airway disease.


Subject(s)
Allergens/immunology , Asthma/immunology , Helicobacter pylori/immunology , Hypersensitivity/immunology , Respiratory Hypersensitivity/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Dendritic Cells/immunology , Female , Goblet Cells/immunology , Inflammation/immunology , Lung/immunology , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Pyroglyphidae/immunology
11.
PLoS Pathog ; 13(7): e1006539, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28753651

ABSTRACT

Infection with the helminth Schistosoma (S.) mansoni drives the development of interleukin (IL)-10-producing regulatory B (Breg) cells in mice and man, which have the capacity to reduce experimental allergic airway inflammation and are thus of high therapeutic interest. However, both the involved antigen and cellular mechanisms that drive Breg cell development remain to be elucidated. Therefore, we investigated whether S. mansoni soluble egg antigens (SEA) directly interact with B cells to enhance their regulatory potential, or act indirectly on B cells via SEA-modulated macrophage subsets. Intraperitoneal injections of S. mansoni eggs or SEA significantly upregulated IL-10 and CD86 expression by marginal zone B cells. Both B cells as well as macrophages of the splenic marginal zone efficiently bound SEA in vivo, but macrophages were dispensable for Breg cell induction as shown by macrophage depletion with clodronate liposomes. SEA was internalized into acidic cell compartments of B cells and induced a 3-fold increase of IL-10, which was dependent on endosomal acidification and was further enhanced by CD40 ligation. IPSE/alpha-1, one of the major antigens in SEA, was also capable of inducing IL-10 in naïve B cells, which was reproduced by tobacco plant-derived recombinant IPSE. Other major schistosomal antigens, omega-1 and kappa-5, had no effect. SEA depleted of IPSE/alpha-1 was still able to induce Breg cells indicating that SEA contains more Breg cell-inducing components. Importantly, SEA- and IPSE-induced Breg cells triggered regulatory T cell development in vitro. SEA and recombinant IPSE/alpha-1 also induced IL-10 production in human CD1d+ B cells. In conclusion, the mechanism of S. mansoni-induced Breg cell development involves a direct targeting of B cells by SEA components such as the secretory glycoprotein IPSE/alpha-1.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Egg Proteins/immunology , Helminth Proteins/immunology , Ovum/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Animals , Antigens, Helminth/genetics , Antigens, Helminth/immunology , Egg Proteins/genetics , Female , Helminth Proteins/genetics , Humans , Interleukin-10/immunology , Mice , Mice, Inbred C57BL , Schistosoma mansoni/genetics , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/parasitology
13.
Allergy ; 74(1): 40-52, 2019 01.
Article in English | MEDLINE | ID: mdl-30276826

ABSTRACT

Current data indicate that the "bronchiolitis" diagnosis comprises more than one condition. Clinically, pathophysiologically, and even genetically three main clusters of patients can be identified among children suffering from severe bronchiolitis (or first wheezing episode): (a) respiratory syncytial virus (RSV)-induced bronchiolitis, characterized by young age of the patient, mechanical obstruction of the airways due to mucus and cell debris, and increased risk of recurrent wheezing. For this illness, an effective prophylactic RSV-specific monoclonal antibody is available; (b) rhinovirus-induced wheezing, associated with atopic predisposition of the patient and high risk of subsequent asthma development, which may, however, be reversed with systemic corticosteroids in those with severe illness; and (c) wheeze due to other viruses, characteristically likely to be less frequent and severe. Clinically, it is important to distinguish between these partially overlapping patient groups as they are likely to respond to different treatments. It appears that the first episode of severe bronchiolitis in under 2-year-old children is a critical event and an important opportunity for designing secondary prevention strategies for asthma. As data have shown bronchiolitis cannot simply be diagnosed using a certain cutoff age, but instead, as we suggest, using the viral etiology as the differentiating factor.


Subject(s)
Bronchiolitis/diagnosis , Bronchiolitis/drug therapy , Adrenal Cortex Hormones/therapeutic use , Antibodies, Monoclonal/therapeutic use , Bronchiolitis/etiology , Bronchiolitis/virology , Child , Child, Preschool , Humans , Respiratory Sounds/etiology , Respiratory Syncytial Viruses , Rhinovirus
15.
Int Arch Allergy Immunol ; 180(1): 1-9, 2019.
Article in English | MEDLINE | ID: mdl-31242493

ABSTRACT

BACKGROUND: An inverse relation between Helicobacter pylori infection and asthma has been shown in epidemiological studies. Infection with H. pylori, or application of an extract of it before or after sensitization, inhibits allergic airway disease in mice. OBJECTIVES: The aim of this study was to investigate the effect of an extract of H. pylori on allergic airway disease induced by repeated allergen exposure in mice that were sensitized and challenged prior to extract application. METHOD: C57BL/6 mice were intranasally (i.n.) sensitized and challenged with house dust mite (HDM). After a minimum of 4 weeks, mice received the H. pylori extract intraperitoneally and were rechallenged i.n. with HDM. Allergen-specific antibodies were measured by ELISA. Cells present in the bronchoalveolar lavage fluid and dendritic cell (DC) subsets in the lung tissue were analyzed by flow cytometry. Tissue inflammation and goblet cell hyperplasia were assessed by histology. Cells of the mediastinal lymph node (mLN) were isolated and in vitro restimulated with HDM or H. pylori extract. RESULTS: Treatment with H. pylori extract before rechallenge reduced allergen-specific IgE, the DC numbers in the tissue, and goblet cell hyperplasia. Cells isolated from mLN of mice treated with the extract produced significantly more IL-10 and IL-17 after in vitro restimulation with HDM. mLN cells of H. pylori-treated mice that were re-exposed to the H. pylori extract produced significantly more interferon gamma. CONCLUSIONS: An extract of H. pylori is effective in reducing mucus production and various features of inflammation in HDM rechallenged mice.


Subject(s)
Allergens/immunology , Antigens, Bacterial/immunology , Goblet Cells/pathology , Helicobacter Infections/complications , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/pathology , Animals , Biomarkers , Biopsy , Cytokines/metabolism , Environmental Exposure , Female , Helicobacter Infections/microbiology , Hyperplasia , Immunoglobulin E/immunology , Immunoglobulin G/immunology , Immunophenotyping , Mice , Pyroglyphidae/immunology
16.
Parasite Immunol ; 41(6): e12574, 2019 06.
Article in English | MEDLINE | ID: mdl-30043455

ABSTRACT

Type 2 immune responses are most commonly associated with allergy and helminth parasite infections. Since the discovery of Th1 and Th2 immune responses more than 30 years ago, models of both allergic disease and helminth infections have been useful in characterizing the development, effector mechanisms and pathological consequences of type 2 immune responses. The observation that some helminth infections negatively correlate with allergic and inflammatory disease led to a large field of research into parasite immunomodulation. However, it is worth noting that helminth parasites are not always benign infections, and that helminth immunomodulation can have stimulatory as well as suppressive effects on allergic responses. In this review, we will discuss how parasitic infections change host responses, the consequences for bystander immunity and how this interaction influences clinical symptoms of allergy.


Subject(s)
Helminthiasis/immunology , Helminths/immunology , Hypersensitivity/immunology , Animals , Helminthiasis/parasitology , Helminths/genetics , Humans , Hypersensitivity/parasitology , Hypersensitivity/therapy , Immunomodulation
17.
J Infect Dis ; 218(7): 1142-1146, 2018 08 24.
Article in English | MEDLINE | ID: mdl-29905805

ABSTRACT

To accelerate the development of novel vaccines for schistosomiasis, we set out to develop a human model for Schistosoma mansoni infection in healthy volunteers. During natural infections, female schistosomes produce eggs that give rise to morbidity. Therefore, we produced single-sex, male Schistosoma mansoni cercariae for human infection without egg production and associated pathology. Cercariae were produced in their intermediate snail hosts in accordance with the principles of good manufacturing practice (GMP). The application of GMP principles to an unconventional production process is a showcase for the controlled production of complex live challenge material in the European Union or under Food and Drug Administration guidance.


Subject(s)
Schistosoma mansoni/immunology , Schistosomiasis mansoni/prevention & control , Schistosomiasis/prevention & control , Snails/parasitology , Animals , Cercaria , Humans , Male , Schistosomiasis/parasitology , Schistosomiasis mansoni/parasitology
18.
PLoS Pathog ; 12(6): e1005687, 2016 06.
Article in English | MEDLINE | ID: mdl-27304615

ABSTRACT

B-cells not only produce immunoglobulins and present antigens to T-cells, but also additional key roles in the immune system. Current knowledge on the role of B-cells in infections caused by intracellular bacteria is fragmentary and contradictory. We therefore analysed the phenotypical and functional properties of B-cells during infection and disease caused by Mycobacterium tuberculosis (Mtb), the bacillus causing tuberculosis (TB), and included individuals with latent TB infection (LTBI), active TB, individuals treated successfully for TB, and healthy controls. Patients with active or treated TB disease had an increased proportion of antibodies reactive with mycobacteria. Patients with active TB had reduced circulating B-cell frequencies, whereas only minor increases in B-cells were detected in the lungs of individuals deceased from TB. Both active TB patients and individuals with LTBI had increased relative fractions of B-cells with an atypical phenotype. Importantly, these B-cells displayed impaired proliferation, immunoglobulin- and cytokine- production. These defects disappeared upon successful treatment. Moreover, T-cell activity was strongest in individuals successfully treated for TB, compared to active TB patients and LTBI subjects, and was dependent on the presence of functionally competent B-cells as shown by cellular depletion experiments. Thus, our results reveal that general B-cell function is impaired during active TB and LTBI, and that this B-cell dysfunction compromises cellular host immunity during Mtb infection. These new insights may provide novel strategies for correcting Mtb infection-induced immune dysfunction towards restored protective immunity.


Subject(s)
B-Lymphocytes/immunology , Tuberculosis/drug therapy , Tuberculosis/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunohistochemistry , Latent Tuberculosis/immunology , Mycobacterium tuberculosis/immunology , Phenotype
19.
Parasite Immunol ; 40(10): e12579, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30107039

ABSTRACT

Chronic helminth infection with Schistosoma (S.) mansoni protects against allergic airway inflammation (AAI) in mice and is associated with reduced Th2 responses to inhaled allergens in humans, despite the presence of schistosome-specific Th2 immunity. Schistosome eggs strongly induce type 2 immunity and allow to study the dynamics of Th2 versus regulatory responses in the absence of worms. Treatment with isolated S. mansoni eggs by i.p. injection prior to induction of AAI to ovalbumin (OVA)/alum led to significantly reduced AAI as assessed by less BAL and lung eosinophilia, less cellular influx into lung tissue, less OVA-specific Th2 cytokines in lungs and lung-draining mediastinal lymph nodes and less circulating allergen-specific IgG1 and IgE antibodies. While OVA-specific Th2 responses were inhibited, treatment induced a strong systemic Th2 response to the eggs. The protective effect of S. mansoni eggs was unaltered in µMT mice lacking mature (B2) B cells and unaffected by Treg cell depletion using anti-CD25 blocking antibodies during egg treatment and allergic sensitization. Notably, prophylactic egg treatment resulted in a reduced influx of pro-inflammatory, monocyte-derived dendritic cells into lung tissue of allergic mice following challenge. Altogether, S. mansoni eggs can protect against the development of AAI, despite strong egg-specific Th2 responses.


Subject(s)
Antibodies, Protozoan/blood , Asthma/prevention & control , Ovum/immunology , Schistosoma mansoni/immunology , Allergens/immunology , Alum Compounds/pharmacology , Animals , Antibodies, Protozoan/immunology , Asthma/immunology , Cytokines/immunology , Disease Models, Animal , Eosinophilia/prevention & control , Female , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Inflammation/pathology , Interleukin-2 Receptor alpha Subunit , Lung/immunology , Lung/parasitology , Lung/pathology , Mice, Inbred C57BL , Ovalbumin/immunology , Ovalbumin/pharmacology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL