Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 19(4): 386-396, 2018 04.
Article in English | MEDLINE | ID: mdl-29556002

ABSTRACT

Live attenuated vaccines are generally highly efficacious and often superior to inactivated vaccines, yet the underlying mechanisms of this remain largely unclear. Here we identify recognition of microbial viability as a potent stimulus for follicular helper T cell (TFH cell) differentiation and vaccine responses. Antigen-presenting cells (APCs) distinguished viable bacteria from dead bacteria through Toll-like receptor 8 (TLR8)-dependent detection of bacterial RNA. In contrast to dead bacteria and other TLR ligands, live bacteria, bacterial RNA and synthetic TLR8 agonists induced a specific cytokine profile in human and porcine APCs, thereby promoting TFH cell differentiation. In domestic pigs, immunization with a live bacterial vaccine induced robust TFH cell and antibody responses, but immunization with its heat-killed counterpart did not. Finally, a hypermorphic TLR8 polymorphism was associated with protective immunity elicited by vaccination with bacillus Calmette-Guérin (BCG) in a human cohort. We have thus identified TLR8 as an important driver of TFH cell differentiation and a promising target for TFH cell-skewing vaccine adjuvants.


Subject(s)
Lymphocyte Activation/immunology , Microbial Viability/immunology , T-Lymphocytes, Helper-Inducer/immunology , Toll-Like Receptor 8/immunology , Vaccines, Attenuated/immunology , Adult , Animals , Antibody Formation/immunology , Cell Differentiation/immunology , Female , Humans , Male , Swine
2.
Eur J Immunol ; 53(5): e2250237, 2023 05.
Article in English | MEDLINE | ID: mdl-36781418

ABSTRACT

Parasitic nematodes infect more than 1 billion people in the global south. The development of effective antihelminthic vaccines is a crucial tool for their future elimination. Protective immune responses to nematodes depend on Gata3+ Th2 cells, which can also be induced by nematode-released products. Whether these nematode products induce antigen-specific long-lived memory T cells and thereby confer protection against a challenge infection is not known yet. Hence, we set out to characterize the formation of memory Th2 cells induced by immunization with Heligmosomoides polygyrus excretory-secretory (HES) products, infection-induced versus immunization-induced recall responses to a challenge infection, and whether HES-induced memory T cells show protective properties following adoptive transfer. Our results show that 8 weeks postimmunization, HES induces long-lived functional memory Th2 cells at the site of immunization in the peritoneal cavity. Following a H. polygyrus challenge infection, HES-immunized mice display MHC-II-dependent antigen-specific Th2 cytokine responses in the gut-draining lymph nodes, comparable to those induced by a prior natural infection. Moreover, adoptive transfer of sorted memory CD4+ T cells from HES-immunized donors reduces female worm fecundity following a challenge H. polygyrus infection in recipient mice, highlighting a protective role for immunization-induced memory T cells.


Subject(s)
Nematoda , Nematospiroides dubius , Strongylida Infections , Mice , Female , Animals , Th2 Cells , Immunization , Cytokines , Vaccination , Mice, Inbred BALB C
3.
Parasite Immunol ; 45(4): e12957, 2023 04.
Article in English | MEDLINE | ID: mdl-36396405

ABSTRACT

Antibiotic treatment can lead to elimination of both pathogenic bacteria and beneficial commensals, as well as to altered host immune responses. Here, we investigated the influence of prolonged antibiotic treatment (Abx) on effector, memory and recall Th2 immune responses during the primary infection, memory phase and secondary infection with the small intestinal nematode Heligmosomoides polygyrus. Abx treatment significantly reduced gut bacterial loads, but neither worm burdens, nor worm fecundity in primary infection were affected, only worm burdens in secondary infection were elevated in Abx treated mice. Abx mice displayed trends for elevated effector and memory Th2 responses during primary infection, but overall frequencies of Th2 cells in the siLP, PEC, mLN and in the spleen were similar between Abx treated and untreated groups. Gata3+ effector and memory Th2 cytokine responses also remained unimpaired by prolonged Abx treatment. Similarly, the energy production and defence mechanisms of the host tissue and the parasite depicted by NAD(P)H fluorescence lifetime imaging (FLIM) did not change by the prolonged use of antibiotics. We show evidence that the host Th2 response to intestinal nematodes, as well as host and parasite metabolic pathways are robust and remain unimpaired by host microbiota abrogation.


Subject(s)
Coinfection , Microbiota , Nematoda , Nematospiroides dubius , Strongylida Infections , Animals , Mice , Cytokines/metabolism , Th2 Cells
4.
Eur J Immunol ; 50(11): 1712-1728, 2020 11.
Article in English | MEDLINE | ID: mdl-32558930

ABSTRACT

Pulmonary mucosal immune response is critical for preventing opportunistic Aspergillus fumigatus infections. Although fungus-specific CD4+ T cells in blood are described to reflect the actual host-pathogen interaction status, little is known about Aspergillus-specific pulmonary T-cell responses. Here, we exploit the domestic pig as human-relevant large animal model and introduce antigen-specific T-cell enrichment in pigs to address Aspergillus-specific T cells in the lung compared to peripheral blood. In healthy, environmentally Aspergillus-exposed pigs, the fungus-specific T cells are detectable in blood in similar frequencies as observed in healthy humans and exhibit a Th1 phenotype. Exposing pigs to 106 cfu/m3 conidia induces a long-lasting accumulation of Aspergillus-specific Th1 cells locally in the lung and also systemically. Temporary immunosuppression during Aspergillus-exposure showed a drastic reduction in the lung-infiltrating antifungal T-cell responses more than 2 weeks after abrogation of the suppressive treatment. This was reflected in blood, but to a much lesser extent. In conclusion, by using the human-relevant large animal model the pig, this study highlights that the blood clearly reflects the mucosal fungal-specific T-cell reactivity in environmentally exposed as well as experimentally exposed healthy pigs. But, immunosuppression significantly impacts the mucosal site in contrast to the initial systemic immune response.


Subject(s)
Antifungal Agents/immunology , Aspergillus fumigatus/immunology , Aspergillus/immunology , Sus scrofa/immunology , Animals , Disease Models, Animal , Host-Pathogen Interactions/immunology , Humans , Lung/immunology , Spores, Fungal/immunology , Swine , Th1 Cells/immunology
5.
Parasite Immunol ; 43(3): e12791, 2021 03.
Article in English | MEDLINE | ID: mdl-32918307

ABSTRACT

AIMS: IgA and Th17 responses are pivotal for the control of Giardia infections. Eosinophils support IgA class switching, the survival of intestinal IgA+ plasma cells at steady state and can control Th17 activity in the small intestine. To see whether eosinophils regulate adaptive immune responses during giardiasis, we investigated Giardia muris infections in wild-type BALB/c and eosinophil-deficient ∆dblGATA-1 mice. METHODS AND RESULTS: Infected ∆dblGATA-1 mice did not differ markedly in parasite control from wild-type mice. Confirming previous studies, naive ∆dblGATA-1 mice displayed diminished IgA+ B cell frequencies in Peyer's patches. However, IgA class switching and intestinal IgA secretion in response to G muris infection were comparable in wild-type BALB/c and ∆dblGATA-1 mice. Both strains displayed similarly low intestinal Th17 responses, accompanied by a mild expansion of type 3 innate lymphoid cells (ILC3). CONCLUSIONS: Contrasting previous reports on overt small intestinal Th17 activity in eosinophil-deficient mice, IL-17A production is kept in check in the absence of eosinophils during Giardia infection. Suboptimal homeostatic IgA responses in the absence of eosinophils are transiently fostered in infected mice and the maintenance of IgA+ plasma cells appears to be restored during persisting Giardia infection.


Subject(s)
Antibodies, Protozoan/immunology , Eosinophils/immunology , Giardia/immunology , Giardiasis/immunology , Immunoglobulin A/immunology , Th17 Cells/immunology , Animals , B-Lymphocytes/immunology , Female , Immunity, Innate , Intestine, Small/immunology , Intestines/immunology , Mice , Mice, Inbred BALB C
6.
Int J Mol Sci ; 22(16)2021 Aug 14.
Article in English | MEDLINE | ID: mdl-34445445

ABSTRACT

Ascariasis is a global health problem for humans and animals. Adult Ascaris nematodes are long-lived in the host intestine where they interact with host cells as well as members of the microbiota resulting in chronic infections. Nematode interactions with host cells and the microbial environment are prominently mediated by parasite-secreted proteins and peptides possessing immunomodulatory and antimicrobial activities. Previously, we discovered the C-type lectin protein AsCTL-42 in the secreted products of adult Ascaris worms. Here we tested recombinant AsCTL-42 for its ability to interact with bacterial and host cells. We found that AsCTL-42 lacks bactericidal activity but neutralized bacterial cells without killing them. Treatment of bacterial cells with AsCTL-42 reduced invasion of intestinal epithelial cells by Salmonella. Furthermore, AsCTL-42 interacted with host myeloid C-type lectin receptors. Thus, AsCTL-42 is a parasite protein involved in the triad relationship between Ascaris, host cells, and the microbiota.


Subject(s)
Ascaris suum/metabolism , Host-Parasite Interactions , Intestinal Mucosa/metabolism , Lectins, C-Type/metabolism , Lectins/metabolism , Salmonella , Animals , Ascariasis/metabolism , Ascariasis/microbiology , Ascaris suum/microbiology , Ascaris suum/physiology , Cell Line , Lectins/physiology , Recombinant Proteins , Sus scrofa/microbiology , Sus scrofa/parasitology
7.
Eur J Immunol ; 48(12): 1958-1964, 2018 12.
Article in English | MEDLINE | ID: mdl-30267404

ABSTRACT

T-helper type 2 (Th2) responses are central to the control of helminth infections, but sensitive to opposing cytokine signals favoring Th1 priming. We previously reported on GATA-3+ T-bet+ Th2/1 hybrid cell differentiation in helminth mono-infections, resulting in a substantial proportion of cells coproducing IFN-γ next to Th2 cytokines. Here, we demonstrate Th2/1 cells as the major source of parasite-specific IFN-γ production in acute and chronic infections with the enteric nematode Heligmosomoides polygyrus. Th2/1 cells differentiated from naive precursors and accumulated in spleen and intestine of infected mice, resulting in increased systemic and mucosal IFN-γ production. IFN-γ supplementation early during infection supported Th2/1 differentiation, associated with elevated parasite fecundity and the maintenance of high worm burdens in the chronic stage of infection, whereas mice lacking IFN-γ signals generated poor Th2/1 responses and restricted parasite fecundity more efficiently. These findings suggest that Th2/1 hybrid responses take part in immune regulation during helminth infection and restrain effective anti-helminth immunity.


Subject(s)
Interferon-gamma/metabolism , Intestines/immunology , Nematospiroides dubius/physiology , Spleen/immunology , Strongylida Infections/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Interferon-gamma/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen/parasitology
8.
J Neuroinflammation ; 16(1): 159, 2019 Jul 29.
Article in English | MEDLINE | ID: mdl-31352901

ABSTRACT

BACKGROUND: It has become increasingly evident that the immune and nervous systems are closely intertwined, relying on one another during regular homeostatic conditions. Prolonged states of imbalance between neural and immune homeostasis, such as chronic neuroinflammation, are associated with a higher risk for neural damage. Toxoplasma gondii is a highly successful neurotropic parasite causing persistent subclinical neuroinflammation, which is associated with psychiatric and neurodegenerative disorders. Little is known, however, by what means neuroinflammation and the associated neural impairment can be modulated by peripheral inflammatory processes. METHODS: Expression of immune and synapse-associated genes was assessed via quantitative real-time PCR to investigate how T. gondii infection-induced chronic neuroinflammation and associated neuronal alterations can be reshaped by a subsequent acute intestinal nematode co-infection. Immune cell subsets were characterized via flow cytometry in the brain of infected mice. Sulfadiazine and interferon-γ-neutralizing antibody were applied to subdue neuroinflammation. RESULTS: Neuroinflammation induced by T. gondii infection of mice was associated with increased microglia activation, recruitment of immune cells into the brain exhibiting Th1 effector functions, and enhanced production of Th1 and pro-inflammatory molecules (IFN-γ, iNOS, IL-12, TNF, IL-6, and IL-1ß) following co-infection with Heligmosomoides polygyrus. The accelerated cerebral Th1 immune response resulted in enhanced T. gondii removal but exacerbated the inflammation-related decrease of synapse-associated gene expression. Synaptic proteins EAAT2 and GABAAα1, which are involved in the excitation/inhibition balance in the CNS, were affected in particular. These synaptic alterations were partially recovered by reducing neuroinflammation indirectly via antiparasitic treatment and especially by application of IFN-γ-neutralizing antibody. Impaired iNOS expression following IFN-γ neutralization directly affected EAAT2 and GABAAα1 signaling, thus contributing to the microglial regulation of neurons. Besides, reduced CD36, TREM2, and C1qa gene expression points toward inflammation induced synaptic pruning as a fundamental mechanism. CONCLUSION: Our results suggest that neuroimmune responses following chronic T. gondii infection can be modulated by acute enteric nematode co-infection. While consecutive co-infection promotes parasite elimination in the CNS, it also adversely affects gene expression of synaptic proteins, via an IFN-γ-dependent manner.


Subject(s)
Brain/metabolism , Interferon-gamma/metabolism , Microglia/metabolism , Neurons/metabolism , Strongylida Infections/metabolism , Toxoplasmosis/metabolism , Animals , Brain/parasitology , Coinfection , Macrophage Activation/physiology , Mice , Microglia/parasitology , Nematospiroides dubius , Neurons/parasitology , Synapses/metabolism , Synapses/parasitology , Toxoplasma
9.
J Immunol ; 199(12): 3952-3958, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29109124

ABSTRACT

The vitamin D receptor participates in the control of IgE class-switch recombination in B cells. The physiologic vitamin D receptor agonist, 1,25(OH)2D3 (calcitriol), is synthesized by the essential enzyme 25-hydroxyvitamin D3-1α-hydroxylase (CYP27B1), which can be expressed by activated immune cells. The role of endogenous calcitriol synthesis for the regulation of IgE has not been proven. In this study, we investigated IgE-responses in Cyp27b1-knockout (KO) mice following sensitization to OVA or intestinal infection with Heligmosomoides polygyrus Specific Igs and plasmablasts were determined by ELISA and ELISpot, Cyp27b1 expression was measured by quantitative PCR. The data show elevated specific IgE and IgG1 concentrations in the blood of OVA-sensitized Cyp27b1-KO mice compared with wild-type littermates (+898 and +219%). Accordingly, more OVA-specific IgG1-secreting cells are present in spleen and fewer in the bone marrow of Cyp27b1-KO mice. Ag-specific mechanisms are suggested as the leucopoiesis is in general unchanged and activated murine B and T lymphocytes express Cyp27b1 Accordingly, elevated specific IgE concentrations in the blood of sensitized T cell-specific Cyp27b1-KO mice support a lymphocyte-driven mechanism. In an independent IgE-inducing model, i.e., intestinal infection with H. polygyrus, we validated the increase of total and specific IgE concentrations of Cyp27b1-KO compared with wild-type mice, but not those of IgG1 or IgA. We conclude that endogenous calcitriol has an impact on the regulation of IgE in vivo. Our data provide genetic evidence supporting previous preclinical and clinical findings and suggest that vitamin D deficiency not only promotes bone diseases but also type I sensitization.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/physiology , Calcitriol/immunology , Immunoglobulin Class Switching , Immunoglobulin E/blood , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/deficiency , Animals , B-Lymphocytes/immunology , Bone Marrow/immunology , Calcitriol/biosynthesis , Calcitriol/deficiency , Female , Helminthiasis, Animal/immunology , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Intestinal Diseases, Parasitic/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Nematospiroides dubius/immunology , Organ Specificity , Ovalbumin/immunology , Receptors, Calcitriol/physiology , Spleen/immunology , T-Lymphocytes/immunology , Vitamin D Deficiency/immunology
10.
Eur J Immunol ; 47(2): 353-363, 2017 02.
Article in English | MEDLINE | ID: mdl-27861815

ABSTRACT

Immunity to intestinal nematodes requires CD4⁺ Th2-cell responses, including IL-4 and IL-13 production. Chronic infection with intestinal nematodes leads to downregulation of these responses, and few functional T helper (Th) 2 cells are detected in secondary lymphoid organs in the chronic phase or after abrogation of infection. Here, we show with a natural murine infection with Heligmosomoides polygyrus that highly functional memory Th2 cells persist in the lamina propria and in addition in the peritoneal cavity (PC) after abrogation of infection. While both tissue-resident memory (TRM ) populations proliferate in situ and express IL-4, IL-5, and IL-13 upon TCR-dependent stimulation, only peritoneal memory cells express high levels of the IL-33 receptor and produce IL-5 and IL-13 upon TCR-independent stimulation with IL-33 and IL-7. Most importantly, PC-derived TRM cells are able to mediate anti-helminthic effects by decreasing the fecundity of female worms upon transfer into recipient mice. These results show that nonlymphoid compartments can serve as reservoirs for Th2 memory cells, and furthermore that innate effector function of Th2 memory cells is restricted to CD4⁺ memory T cells residing in the PC.


Subject(s)
Intestines/immunology , Nematospiroides dubius/immunology , Strongylida Infections/immunology , Th2 Cells/immunology , Animals , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Female , Immunity, Innate , Immunologic Memory , Interleukin-1 Receptor-Like 1 Protein/metabolism , Intestines/parasitology , Mice , Mice, Inbred C57BL , Mucous Membrane/pathology , Peritoneal Cavity/pathology , Th2 Cells/parasitology
11.
PLoS Pathog ; 12(4): e1005480, 2016 04.
Article in English | MEDLINE | ID: mdl-27101372

ABSTRACT

Helminths have evolved numerous pathways to prevent their expulsion or elimination from the host to ensure long-term survival. During infection, they target numerous host cells, including macrophages, to induce an alternatively activated phenotype, which aids elimination of infection, tissue repair, and wound healing. Multiple animal-based studies have demonstrated a significant reduction or complete reversal of disease by helminth infection, treatment with helminth products, or helminth-modulated macrophages in models of allergy, autoimmunity, and sepsis. Experimental studies of macrophage and helminth therapies are being translated into clinical benefits for patients undergoing transplantation and those with multiple sclerosis. Thus, helminths or helminth-modulated macrophages present great possibilities as therapeutic applications for inflammatory diseases in humans. Macrophage-based helminth therapies and the underlying mechanisms of their therapeutic or curative effects represent an under-researched area with the potential to open new avenues of treatment. This review explores the application of helminth-modulated macrophages as a new therapy for inflammatory diseases.


Subject(s)
Inflammation/immunology , Macrophages/immunology , Therapy with Helminths/methods , Animals , Autoimmune Diseases/immunology , Helminths/immunology , Humans
12.
Crit Rev Immunol ; 36(3): 193-238, 2016.
Article in English | MEDLINE | ID: mdl-28008805

ABSTRACT

Eosinophil numbers are highly elevated during helminth infections and a range of allergic and inflammatory disorders, but eosinophils are also present in several tissues in the absence of infection. Indeed, new findings demonstrate that eosinophils may be involved in events as diverse as glucose metabolism, mammary gland development, intestinal health, tissue remodeling, thymic selection, and B-cell survival. Although eosinophils often correlate with pathological parameters during conditions such as inflammatory bowel disease and asthma, the evidence for their contribution to tissue pathology remains controversial. Recent research suggests that eosinophils may have additional roles in these settings that are related to control and resolution of inflammation. Controversy also surrounds the involvement of eosinophils in anti-helminth immunity. Their assumed role in fighting parasites has increasingly been questioned, particularly as a result of data from studies of eosinophil-ablated mouse strains in which either no or only very moderate effects on helminth survival has been reported. Helminths are masters of immune regulation, but whether they actively suppress eosinophil function has rarely been considered. Thus, the purpose of this review is threefold: (1) to summarize our knowledge of the wide range of functions of eosinophils during homeostasis, (2) to discuss the role of eosinophil during inflammation and the recent discovery of eosinophils as mediators of inflammatory resolution, and (3) to summarize data on the effect of eosinophils on helminth infections and discuss the possibility of helminth-mediated modulation of eosinophils.


Subject(s)
Eosinophils/physiology , Helminthiasis/immunology , Inflammation/immunology , Adipose Tissue/metabolism , Animals , Breast/growth & development , Female , Gastrointestinal Microbiome , Humans , Wound Healing
13.
J Immunol ; 194(4): 1555-64, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25589067

ABSTRACT

Immunomodulation is a common feature of chronic helminth infections and mainly attributed to the secretion of bioactive molecules, which target and modify host immune cells. In this study, we show that the helminth immunomodulator AvCystatin, a cysteine protease inhibitor, induces a novel regulatory macrophage (Mreg; AvCystatin-Mreg), which is sufficient to mitigate major parameters of allergic airway inflammation and colitis in mice. A single adoptive transfer of AvCystatin-Mreg before allergen challenge suppressed allergen-specific IgE levels, the influx of eosinophils into the airways, local and systemic Th2 cytokine levels, and mucus production in lung bronchioles of mice, whereas increasing local and systemic IL-10 production by CD4(+) T cells. Moreover, a single administration of AvCystatin-Mreg during experimentally induced colitis strikingly reduced intestinal pathology. Phenotyping of AvCystatin-Mreg revealed increased expression of a distinct group of genes including LIGHT, sphingosine kinase 1, CCL1, arginase-1, and costimulatory molecules, CD16/32, ICAM-1, as well as PD-L1 and PD-L2. In cocultures with dendritic cells and CD4(+) T cells, AvCystatin-Mreg strongly induced the production of IL-10 in a cell-contact-independent manner. Collectively, our data identify a specific suppressive macrophage population induced by a single parasite immunomodulator, which protects against mucosal inflammation.


Subject(s)
Antigens, Helminth/immunology , CD4-Positive T-Lymphocytes/immunology , Immunity, Mucosal/immunology , Inflammation/prevention & control , Macrophages/immunology , Acanthocheilonema/immunology , Adoptive Transfer , Animals , Antigens, Helminth/pharmacology , Colitis/immunology , Colitis/prevention & control , Disease Models, Animal , Female , Immunosuppressive Agents/immunology , Immunosuppressive Agents/pharmacology , Inflammation/immunology , Interleukin-10/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/prevention & control
14.
Proc Natl Acad Sci U S A ; 111(48): E5169-77, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25404305

ABSTRACT

Basophils orchestrate protection against reinfections with gastrointestinal helminths and ticks, but the underlying mechanisms remain elusive. We investigated the role of Fc receptors on basophils, the antibody isotypes IgG1 and IgE, and basophil-derived IL-4/IL-13 during challenge infections with Heligmosomoides polygyrus and Nippostrongylus brasiliensis. Using mixed bone marrow chimeras, we found that activating Fc receptors on basophils were required for protective immunity but not for regulation of basophil homeostasis. Furthermore, rapid worm expulsion was impaired in IgE-deficient but not in IgG1-deficient mice. Basophils promoted the recruitment of other effector cells into the small intestine and induced expression of the antihelminthic proteins resistin-like molecule ß and mucin 5ac. Selective deletion of IL-4/IL-13 in basophils resulted in impaired worm expulsion. Collectively, our results indicate that IgE-mediated activation of basophils and the release of basophil-derived IL-4/IL-13 are critical steps in protective immunity against helminths. Therefore, development of effective vaccines against helminths should consider boosting the IL-4/IgE/basophil axis of the immune system.


Subject(s)
Basophils/immunology , Cytokines/immunology , Gastrointestinal Tract/immunology , Heligmosomatoidea/immunology , Immunoglobulin E/immunology , Strongylida Infections/immunology , Animals , Basophils/metabolism , Basophils/parasitology , Blotting, Western , Cytokines/metabolism , Flow Cytometry , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/parasitology , Heligmosomatoidea/physiology , Host-Parasite Interactions/immunology , Immunoglobulin E/genetics , Immunoglobulin E/metabolism , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-13/metabolism , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-4/metabolism , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/parasitology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nematospiroides dubius/immunology , Nematospiroides dubius/physiology , Nippostrongylus/immunology , Nippostrongylus/physiology , Receptors, Fc/genetics , Receptors, Fc/immunology , Receptors, Fc/metabolism , Strongylida Infections/metabolism , Strongylida Infections/parasitology , Th2 Cells/immunology , Th2 Cells/metabolism
15.
Infect Immun ; 84(5): 1274-1286, 2016 05.
Article in English | MEDLINE | ID: mdl-26883594

ABSTRACT

Small rodents serve as reservoir hosts for tick-borne pathogens, such as the spirochetes causing Lyme disease. Whether natural coinfections with other macroparasites alter the success of tick feeding, antitick immunity, and the host's reservoir competence for tick-borne pathogens remains to be determined. In a parasitological survey of wild mice in Berlin, Germany, approximately 40% of Ixodes ricinus-infested animals simultaneously harbored a nematode of the genus Heligmosomoides We therefore aimed to analyze the immunological impact of the nematode/tick coinfection as well as its effect on the tick-borne pathogen Borrelia afzelii Hosts experimentally coinfected with Heligmosomoides polygyrus and larval/nymphal I. ricinus ticks developed substantially stronger systemic type 2 T helper cell (Th2) responses, on the basis of the levels of GATA-3 and interleukin-13 expression, than mice infected with a single pathogen. During repeated larval infestations, however, anti-tick Th2 reactivity and an observed partial immunity to tick feeding were unaffected by concurrent nematode infections. Importantly, the strong systemic Th2 immune response in coinfected mice did not affect susceptibility to tick-borne B. afzelii An observed trend for decreased local and systemic Th1 reactivity against B. afzelii in coinfected mice did not result in a higher spirochete burden, nor did it facilitate bacterial dissemination or induce signs of immunopathology. Hence, this study indicates that strong systemic Th2 responses in nematode/tick-coinfected house mice do not affect the success of tick feeding and the control of the causative agent of Lyme disease.


Subject(s)
Coinfection/pathology , Disease Susceptibility , Lyme Disease/pathology , Nematode Infections/pathology , Tick Infestations/pathology , Animals , Borrelia burgdorferi Group/immunology , Borrelia burgdorferi Group/isolation & purification , Coinfection/microbiology , Coinfection/parasitology , Disease Models, Animal , Female , Ixodes/immunology , Ixodes/microbiology , Lyme Disease/complications , Lyme Disease/immunology , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Nematode Infections/complications , Nematode Infections/immunology , Nematospiroides dubius/growth & development , Nematospiroides dubius/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Tick Infestations/complications
16.
PLoS Biol ; 11(8): e1001633, 2013.
Article in English | MEDLINE | ID: mdl-23976880

ABSTRACT

Differentiated T helper (Th) cell lineages are thought to emerge from alternative cell fate decisions. However, recent studies indicated that differentiated Th cells can adopt mixed phenotypes during secondary immunological challenges. Here we show that natural primary immune responses against parasites generate bifunctional Th1 and Th2 hybrid cells that co-express the lineage-specifying transcription factors T-bet and GATA-3 and co-produce Th1 and Th2 cytokines. The integration of Th1-promoting interferon (IFN)-γ and interleukin (IL)-12 signals together with Th2-favoring IL-4 signals commits naive Th cells directly and homogeneously to the hybrid Th1/2 phenotype. Specifically, IFN-γ signals are essential for T-bet(+)GATA-3(+) cells to develop in vitro and in vivo by breaking the dominance of IL-4 over IL-12 signals. The hybrid Th1/2 phenotype is stably maintained in memory cells in vivo for months. It resists reprogramming into classic Th1 or Th2 cells by Th1- or Th2-promoting stimuli, which rather induce quantitative modulations of the combined Th1 and Th2 programs without abolishing either. The hybrid phenotype is associated with intermediate manifestations of both Th1 and Th2 cell properties. Consistently, hybrid Th1/2 cells support inflammatory type-1 and type-2 immune responses but cause less immunopathology than Th1 and Th2 cells, respectively. Thus, we propose the self-limitation of effector T cells based on the stable cell-intrinsic balance of two opposing differentiation programs as a novel concept of how the immune system can prevent excessive inflammation.


Subject(s)
GATA3 Transcription Factor/metabolism , Schistosoma mansoni/immunology , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Cells, Cultured , Female , Flow Cytometry , Mice , Mice, Inbred BALB C , Schistosoma mansoni/pathogenicity
17.
Neuroimmunomodulation ; 23(2): 109-21, 2016.
Article in English | MEDLINE | ID: mdl-27088850

ABSTRACT

OBJECTIVE: The aim of this study was to examine whether the natural protease inhibitor Av-cystatin (rAv17) of the parasitic nematode Acanthocheilonema viteae exerts anti-inflammatory effects in an in vitro model of lipopolysaccharide (LPS)-activated microglia. METHODS: Primary microglia were harvested from the brains of 2-day-old Wistar rats and cultured with or without rAv17 (250 nM). After 6 and 24 h the release of nitric oxide (Griess reagent) and TNF-α (ELISA) was measured in the supernatant. Real-time PCR was performed after 2, 6 and 24 h of culture to measure the mRNA expression of IL-1ß, IL-6, TNF-α, COX-2, iNOS and IL-10. To address the involved signaling pathways, nuclear NF-x0138;B translocation was visualized by immunocytochemistry. Morphological changes of microglia were analyzed by Coomassie blue staining. Differences between groups were calculated using one-way ANOVA with Bonferroni's post hoc test. RESULTS: Morphological analysis indicated that LPS-induced microglial transformation towards an amoeboid morphology is inhibited by rAv17. Av-cystatin caused a time-dependent downregulation of proinflammatory cytokines, iNOS and COX-2 mRNA expression, respectively. This was paralleled by an upregulated expression of IL-10 in resting as well as in LPS-stimulated microglia. Av-cystatin reduced the release of NO and TNF-α in the culture supernatant. Immunocytochemical staining demonstrated an attenuated translocation of NF-x0138;B by Av-cystatin in response to LPS. In addition, Western blot analysis revealed a rAv17-dependent reduction of the LPS-induced ERK1/2-pathway activation. CONCLUSION: The parasite-derived secretion product Av-cystatin inhibits proinflammatory mechanisms of LPS-induced microglia with IL-10, a potential key mediator.


Subject(s)
Acanthocheilonema , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Microglia/metabolism , Phenotype , Protease Inhibitors/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Helminths , Inflammation Mediators/antagonists & inhibitors , Microglia/drug effects , Protease Inhibitors/isolation & purification , Rats , Rats, Wistar
18.
Exp Cell Res ; 330(2): 346-357, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25149900

ABSTRACT

Glioblastoma cells exhibit highly invasive behavior whose mechanisms are not yet fully understood. The present study explores the relationship between the invasion capacity of 5 glioblastoma cell lines differing in p53 and PTEN status, expression of mTOR and several other marker proteins involved in cell invasion, actin cytoskeleton organization and cell morphology. We found that two glioblastoma lines mutated in both p53 and PTEN genes (U373-MG and SNB19) exhibited the highest invasion rates through the Matrigel or collagen matrix. In DK-MG (p53wt/PTENwt) and GaMG (p53mut/PTENwt) cells, F-actin mainly occurred in the numerous stress fibers spanning the cytoplasm, whereas U87-MG (p53wt/PTENmut), U373-MG and SNB19 (both p53mut/PTENmut) cells preferentially expressed F-actin in filopodia and lamellipodia. Scanning electron microscopy confirmed the abundant filopodia and lamellipodia in the PTEN mutated cell lines. Interestingly, the gene profiling analysis revealed two clusters of cell lines, corresponding to the most (U373-MG and SNB19, i.e. p53 and PTEN mutated cells) and less invasive phenotypes. The results of this study might shed new light on the mechanisms of glioblastoma invasion.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , PTEN Phosphohydrolase/metabolism , Tumor Suppressor Protein p53/metabolism , Actin Cytoskeleton , Actins/biosynthesis , Benzothiazoles/pharmacology , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Humans , MAP Kinase Signaling System/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/biosynthesis , Proto-Oncogene Proteins c-akt/biosynthesis , RNA Interference , RNA, Small Interfering , TOR Serine-Threonine Kinases/biosynthesis , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
19.
Mol Ther ; 22(10): 1730-40, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24985163

ABSTRACT

New treatment strategies for inflammatory bowel disease are needed and parasitic nematode infections or application of helminth components improve clinical and experimental gut inflammation. We genetically modified the probiotic bacterium Escherichia coli Nissle 1917 to secrete the powerful nematode immunomodulator cystatin in the gut. This treatment was tested in a murine colitis model and on post-weaning intestinal inflammation in pigs, an outbred model with a gastrointestinal system similar to humans. Application of the transgenic probiotic significantly decreased intestinal inflammation in murine acute colitis, associated with increased frequencies of Foxp3(+) Tregs, suppressed local interleukin (IL)-6 and IL-17A production, decreased macrophage inflammatory protein-1α/ß, monocyte chemoattractant protein -1/3, and regulated upon activation, normal T-cell expressed, and secreted expression and fewer inflammatory macrophages in the colon. High dosages of the transgenic probiotic were well tolerated by post-weaning piglets. Despite being recognized by T cells, secreted cystatin did not lead to changes in cytokine expression or macrophage activation in the colon. However, colon transepithelial resistance and barrier function were significantly improved in pigs receiving the transgenic probotic and post-weaning colon inflammation was reduced. Thus, the anti-inflammatory efficiency of a probiotic can be improved by a nematode-derived immunoregulatory transgene. This treatment regimen should be further investigated as a potential therapeutic option for inflammatory bowel disease.


Subject(s)
Gastroenteritis/therapy , Immunologic Factors/biosynthesis , Immunologic Factors/genetics , Probiotics/metabolism , Probiotics/therapeutic use , Animals , Chemokines/genetics , Chemokines/metabolism , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Colitis/therapy , Cystatins/biosynthesis , Cystatins/genetics , Cystatins/immunology , Disease Models, Animal , Escherichia coli/genetics , Escherichia coli/metabolism , Gastroenteritis/immunology , Gastroenteritis/metabolism , Gastroenteritis/parasitology , Gene Expression , Immunologic Factors/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Male , Mice , Probiotics/administration & dosage , Probiotics/adverse effects , Swine
20.
Proc Natl Acad Sci U S A ; 109(17): 6644-9, 2012 Apr 24.
Article in English | MEDLINE | ID: mdl-22493240

ABSTRACT

Mast cells (MCs) are potent inflammatory cells that are distributed throughout mucosal barrier tissues and respond rapidly to pathogenic stimuli. During helminth infections, MCs play an important role as late-stage effectors. However, it is currently unknown whether MCs contribute to the early innate events that determine the priming of adaptive immunity. MC-deficient mouse strains and mice treated with the MC stabilizing agent cromolyn sodium had dramatically reduced Th2 priming and type 2 cytokine production and harbored increased parasite burdens following infection with gastrointestinal helminths (Heligmosomoides polygyrus bakeri and Trichuris muris). In addition, early production of the tissue-derived cytokines IL-25, IL-33, and thymic stromal lymphopoietin (TSLP) was significantly diminished in MC-deficient mice and resulted in decreased numbers of infection-elicited IL-25-dependent (Lin(-)CD45(-))CD34(+)Sca-1(+) progenitors, which produced type 2 cytokines and could be differentiated into mast cells ex vivo. Finally, repair of MC deficiency increased production of IL-25, IL-33, and TSLP, restored progenitor cell numbers and Th2 priming, and reduced parasite burden. Our data reveal an innate IgE-independent role for MCs in orchestrating type 2 immune responses via the regulation of IL-25, IL-33, and TSLP.


Subject(s)
Cytokines/immunology , Helminths/immunology , Mast Cells/immunology , Th2 Cells/immunology , Animals , Cytokines/biosynthesis , Intestines/parasitology , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL