Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 730
1.
Sci Rep ; 14(1): 10773, 2024 05 10.
Article En | MEDLINE | ID: mdl-38730262

The developing brain is vulnerable to maternal bacterial and viral infections which induce strong inflammatory responses in the mother that are mimicked in the offspring brain, resulting in irreversible neurodevelopmental defects, and associated cognitive and behavioural impairments. In contrast, infection during pregnancy and lactation with the immunoregulatory murine intestinal nematode, Heligmosomoides bakeri, upregulates expression of genes associated with long-term potentiation (LTP) of synaptic networks in the brain of neonatal uninfected offspring, and enhances spatial memory in uninfected juvenile offspring. As the hippocampus is involved in spatial navigation and sensitive to immune events during development, here we assessed hippocampal gene expression, LTP, and neuroimmunity in 3-week-old uninfected offspring born to H. bakeri infected mothers. Further, as maternal immunity shapes the developing immune system, we assessed the impact of maternal H. bakeri infection on the ability of offspring to resist direct infection. In response to maternal infection, we found an enhanced propensity to induce LTP at Schaffer collateral synapses, consistent with RNA-seq data indicating accelerated development of glutamatergic synapses in uninfected offspring, relative to those from uninfected mothers. Hippocampal RNA-seq analysis of offspring of infected mothers revealed increased expression of genes associated with neurogenesis, gliogenesis, and myelination. Furthermore, maternal infection improved resistance to direct infection of H. bakeri in offspring, correlated with transfer of parasite-specific IgG1 to their serum. Hippocampal immunohistochemistry and gene expression suggest Th2/Treg biased neuroimmunity in offspring, recapitulating peripheral immunoregulation of H. bakeri infected mothers. These findings indicate maternal H. bakeri infection during pregnancy and lactation alters peripheral and neural immunity in uninfected offspring, in a manner that accelerates neural maturation to promote hippocampal LTP, and upregulates the expression of genes associated with neurogenesis, gliogenesis, and myelination.


Hippocampus , Neuronal Plasticity , Animals , Female , Hippocampus/metabolism , Hippocampus/parasitology , Pregnancy , Mice , Nematode Infections/immunology , Nematode Infections/parasitology , Long-Term Potentiation , Prenatal Exposure Delayed Effects/immunology , Strongylida Infections/immunology , Strongylida Infections/parasitology , Male , Neuroimmunomodulation
2.
Front Immunol ; 14: 1130933, 2023.
Article En | MEDLINE | ID: mdl-37063913

The initiation of type 2 immune responses at mucosal barriers is regulated by rapidly secreted cytokines called alarmins. The alarmins IL-33, IL-25 and TSLP are mainly secreted by stromal and epithelial cells in tissues and were linked to chronic inflammatory diseases, such as allergic lung inflammation, or to resistance against worm infections. Receptors for alarmins are expressed by a variety of immune cells, including group 2 innate lymphoid cells (ILC2s), an early source of the type 2 cytokines, such as IL-5 and IL-13, which have been linked to atopic diseases and anti-worm immunity as well. However, the precise contribution of the IL-33 receptor signals for ILC2 activation still needs to be completed due to limitations in targeting genes in ILC2. Using the newly established Nmur1 iCre-eGFP mouse model, we obtained specific conditional genetic ablation of the IL-33 receptor subunit ST2 in ILC2s. ST2-deficient ILC2s were unresponsive to IL-33 but not to stimulation with the alarmin IL-25. As a result of defective ST2 signals, ILC2s produced limited amounts of IL-5 and IL-13 and failed to support eosinophil homeostasis. Further, ST2-deficient ILC2s were unable to expand and promote the recruitment of eosinophils during allergic lung inflammation provoked by papain administration. During infection with Nippostrongylus brasiliensis, ILC2-intrinsic ST2 signals were required to mount an effective type 2 immune response against the parasite leading to higher susceptibility against worm infection in conditional knockout mice. Therefore, this study argues for a non-redundant role of cell-intrinsic ST2 signals triggering proper activation of ILC2 for initiation of type 2 immunity.


Interleukin-1 Receptor-Like 1 Protein , Pulmonary Eosinophilia , Strongylida Infections , Animals , Mice , Alarmins , Cytokines/immunology , Immunity, Innate , Interleukin-1 Receptor-Like 1 Protein/immunology , Interleukin-13 , Interleukin-33 , Interleukin-5 , Lymphocytes , Pulmonary Eosinophilia/immunology , Nippostrongylus , Strongylida Infections/immunology
3.
Elife ; 122023 02 08.
Article En | MEDLINE | ID: mdl-36753434

The myeloid C-type lectin receptor (CLR) MINCLE senses the mycobacterial cell wall component trehalose-6,6'-dimycolate (TDM). Recently, we found that IL-4 downregulates MINCLE expression in macrophages. IL-4 is a hallmark cytokine in helminth infections, which appear to increase the risk for mycobacterial infection and active tuberculosis. Here, we investigated functional consequences of IL-4 and helminth infection on MINCLE-driven macrophage activation and Th1/Th17 adjuvanticity. IL-4 inhibited MINCLE and cytokine induction after macrophage infection with Mycobacterium bovis bacille Calmette-Guerin (BCG). Infection of mice with BCG upregulated MINCLE on myeloid cells, which was inhibited by IL-4 plasmid injection and by infection with the nematode Nippostrongylus brasiliensis in monocytes. To determine the impact of helminth infection on MINCLE-dependent immune responses, we vaccinated mice with a recombinant protein together with the MINCLE ligand trehalose-6,6-dibehenate (TDB) as adjuvant. Concurrent infection with N. brasiliensis or with Schistosoma mansoni promoted T cell-derived IL-4 production and suppressed Th1/Th17 differentiation in the spleen. In contrast, helminth infection did not reduce Th1/Th17 induction by TDB in draining peripheral lymph nodes, where IL-4 levels were unaltered. Upon use of the TLR4-dependent adjuvant G3D6A, N. brasiliensis infection impaired selectively the induction of splenic antigen-specific Th1 but not of Th17 cells. Inhibition of MINCLE-dependent Th1/Th17 responses in mice infected with N. brasiliensis was dependent on IL-4/IL-13. Thus, helminth infection attenuated the Th17 response to MINCLE-dependent immunization in an organ- and adjuvant-specific manner via the Th2 cytokines IL-4/IL-13. Taken together, our results demonstrate downregulation of MINCLE expression on monocytes and macrophages by IL-4 as a possible mechanism of thwarted Th17 vaccination responses by underlying helminth infection.


Interleukin-4 , Lectins, C-Type , Membrane Proteins , Strongylida Infections , Animals , Mice , Adjuvants, Immunologic , BCG Vaccine , Cytokines/immunology , Interleukin-13 , Interleukin-4/immunology , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Macrophages/immunology , Mycobacterium bovis , Th1 Cells , Th17 Cells/immunology , Membrane Proteins/metabolism , Nippostrongylus , Strongylida Infections/immunology
4.
Eur J Immunol ; 52(12): 1925-1933, 2022 12.
Article En | MEDLINE | ID: mdl-36116042

Infection of mice with Nippostrongylus brasiliensis (Nb) serves as a model for human hookworm infection affecting about 600 million people world-wide. Expulsion of Nb from the intestine requires IL-13-mediated mucus secretion from goblet cells and activation of smooth muscles cells. Type 2 innate lymphoid cells (ILC2s) are a major cellular source of IL-13 but it remains unclear whether IL-13 secretion from ILC2s is required for Nb expulsion. Here, we compared the immune response to Nb infection in mixed bone marrow chimeras with wild-type or IL-4/IL-13-deficient ILC2s. ILC2-derived IL-4/IL-13 was required for recruitment of eosinophils to the lung but had no influence of systemic eosinophil levels. In the small intestine, goblet cell hyperplasia and tuft cell accumulation was largely dependent on IL-4/IL-13 secretion from ILC2s. This further translated to higher eggs counts and impaired worm expulsion in mice with IL-4/IL-13-deficient ILC2s. Overall, we demonstrate that ILC2s constitute a non-redundant source of IL-4/IL-13 required for protective immunity against primary Nb infection.


Immunity, Innate , Lymphocytes , Strongylida Infections , Animals , Mice , Interleukin-13 , Interleukin-4 , Nippostrongylus , Strongylida Infections/immunology
5.
Cell Rep ; 38(2): 110215, 2022 01 11.
Article En | MEDLINE | ID: mdl-35021079

Macrophages are known to mediate anti-helminth responses, but it remains uncertain which subsets are involved or how macrophages actually kill helminths. Here, we show rapid monocyte recruitment to the lung after infection with the nematode parasite Nippostrongylus brasiliensis. In this inflamed tissue microenvironment, these monocytes differentiate into an alveolar macrophage (AM)-like phenotype, expressing both SiglecF and CD11c, surround invading parasitic larvae, and preferentially kill parasites in vitro. Monocyte-derived AMs (Mo-AMs) express type 2-associated markers and show a distinct remodeling of the chromatin landscape relative to tissue-derived AMs (TD-AMs). In particular, they express high amounts of arginase-1 (Arg1), which we demonstrate mediates helminth killing through L-arginine depletion. These studies indicate that recruited monocytes are selectively programmed in the pulmonary environment to express AM markers and an anti-helminth phenotype.


Lung/immunology , Macrophages, Alveolar/immunology , Strongylida Infections/immunology , Animals , Arginase/metabolism , Cell Differentiation , Cytokines , Female , Lung/parasitology , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Nippostrongylus , Strongylida Infections/parasitology
6.
Mucosal Immunol ; 15(1): 74-83, 2022 01.
Article En | MEDLINE | ID: mdl-34420044

Macrophages are a heterogeneous population of innate immune cells that are often divided into two major subsets: classically activated, typically pro-inflammatory (M1) macrophages that mediate host defense, and alternatively activated, tolerance-inducing (M2) macrophages that exert homeostatic and tissue-regenerative functions. Disturbed macrophage function/differentiation results either in inadequate, excessive immune activation or in a failure to induce efficient protective immune responses against pathogens. Loss-of-function variants in protein tyrosine phosphatase non-receptor type 2 (PTPN2) are associated with chronic inflammatory disorders, but the effect of macrophage-intrinsic PTPN2 loss is still poorly understood. Here we report that PTPN2-deficient macrophages fail to acquire an alternatively activated/M2 phenotype. This was the consequence of reduced IL-6 receptor expression and a failure to induce IL-4 receptor in response to IL-6, resulting in an inability to respond to the key M2-inducing cytokine IL-4. Ultimately, failure to adequately respond to IL-6 and IL-4 resulted in increased levels of M1 macrophage marker expression in vitro and exacerbated lung inflammation upon infection with Nippostrongylus brasiliensis in vivo. These results demonstrate that PTPN2 loss interferes with the ability of macrophages to adequately respond to inflammatory stimuli and might explain the increased susceptibility of PTPN2 loss-of-function carriers to developing inflammatory diseases.


Inflammation/immunology , Lung/immunology , Macrophages/immunology , Nippostrongylus/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Strongylida Infections/immunology , Animals , Cell Differentiation , Gene Knockdown Techniques , Humans , Interleukin-4/metabolism , Lung/parasitology , Mice , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , THP-1 Cells , Th1 Cells/immunology , Th2 Cells/immunology
7.
J Nutr Biochem ; 100: 108887, 2022 02.
Article En | MEDLINE | ID: mdl-34655757

Phytonutrients such as cinnamaldehyde (CA) have been studied for their effects on metabolic diseases, but their influence on mucosal inflammation and immunity to enteric infection are not well documented. Here, we show that consumption of CA in mice significantly down-regulates transcriptional pathways connected to inflammation in the small intestine, and alters T-cell populations in mesenteric lymph nodes. During infection with the enteric helminth Heligomosomoides polygyrus, CA treatment attenuated infection-induced changes in biological pathways connected to cell cycle and mitotic activity, and tended to reduce worm burdens. Mechanistically, CA did not appear to exert activity through a prebiotic effect, as CA treatment did not significantly change the composition of the gut microbiota. Instead, in vitro experiments showed that CA directly induced xenobiotic metabolizing pathways in intestinal epithelial cells and suppressed endotoxin-induced inflammatory responses in macrophages. Collectively, our results show that CA down-regulates inflammatory pathways in the intestinal mucosa and can limit the pathological response to enteric infection. These properties appear to be largely independent of the gut microbiota, and instead connected to the ability of CA to induce antioxidant pathways in intestinal cells. Our results encourage further investigation into the use of CA and related phytonutrients as functional food components to promote intestinal health in humans and animals.


Acrolein/analogs & derivatives , Dietary Supplements , Inflammation/immunology , Intestine, Small/metabolism , Phytochemicals/administration & dosage , Strongylida Infections/immunology , Acrolein/administration & dosage , Acrolein/pharmacology , Animals , Cells, Cultured , Female , Gastrointestinal Microbiome , Immunity, Mucosal , Inflammation/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Lymph Nodes/immunology , Macrophages/drug effects , Macrophages/immunology , Metabolic Networks and Pathways/drug effects , Mice , Mice, Inbred C57BL , Nematospiroides dubius , Phytochemicals/pharmacology , T-Lymphocytes/immunology , Transcription, Genetic , Transcriptome , Xenobiotics/metabolism
8.
Science ; 374(6568): eabe6723, 2021 Nov 05.
Article En | MEDLINE | ID: mdl-34735226

A diverse group of antimicrobial proteins (AMPs) helps protect the mammalian intestine from varied microbial challenges. We show that small proline-rich protein 2A (SPRR2A) is an intestinal antibacterial protein that is phylogenetically unrelated to previously discovered mammalian AMPs. In this study, SPRR2A was expressed in Paneth cells and goblet cells and selectively killed Gram-positive bacteria by disrupting their membranes. SPRR2A shaped intestinal microbiota composition, restricted bacterial association with the intestinal surface, and protected against Listeria monocytogenes infection. SPRR2A differed from other intestinal AMPs in that it was induced by type 2 cytokines produced during helminth infection. Moreover, SPRR2A protected against helminth-induced bacterial invasion of intestinal tissue. Thus, SPRR2A is a distinctive AMP triggered by type 2 immunity that protects the intestinal barrier during helminth infection.


Cornified Envelope Proline-Rich Proteins/metabolism , Gastrointestinal Microbiome , Gram-Positive Bacteria/physiology , Intestinal Mucosa/metabolism , Intestines/microbiology , Nematospiroides dubius , Strongylida Infections/immunology , Animals , Bacterial Load , Cell Membrane/metabolism , Cell Membrane Permeability , Cornified Envelope Proline-Rich Proteins/genetics , Cytokines/metabolism , Disease Susceptibility , Goblet Cells/metabolism , Humans , Immunity, Innate , Intestinal Mucosa/microbiology , Listeria monocytogenes/physiology , Listeriosis/microbiology , Mice , Microbial Viability , Paneth Cells/metabolism , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Strongylida Infections/metabolism , Strongylida Infections/microbiology
9.
Nature ; 599(7883): 125-130, 2021 11.
Article En | MEDLINE | ID: mdl-34671159

Tissue maintenance and repair depend on the integrated activity of multiple cell types1. Whereas the contributions of epithelial2,3, immune4,5 and stromal cells6,7 in intestinal tissue integrity are well understood, the role of intrinsic neuroglia networks remains largely unknown. Here we uncover important roles of enteric glial cells (EGCs) in intestinal homeostasis, immunity and tissue repair. We demonstrate that infection of mice with Heligmosomoides polygyrus leads to enteric gliosis and the upregulation of an interferon gamma (IFNγ) gene signature. IFNγ-dependent gene modules were also induced in EGCs from patients with inflammatory bowel disease8. Single-cell transcriptomics analysis of the tunica muscularis showed that glia-specific abrogation of IFNγ signalling leads to tissue-wide activation of pro-inflammatory transcriptional programs. Furthermore, disruption of the IFNγ-EGC signalling axis enhanced the inflammatory and granulomatous response of the tunica muscularis to helminths. Mechanistically, we show that the upregulation of Cxcl10 is an early immediate response of EGCs to IFNγ signalling and provide evidence that this chemokine and the downstream amplification of IFNγ signalling in the tunica muscularis are required for a measured inflammatory response to helminths and resolution of the granulomatous pathology. Our study demonstrates that IFNγ signalling in enteric glia is central to intestinal homeostasis and reveals critical roles of the IFNγ-EGC-CXCL10 axis in immune response and tissue repair after infectious challenge.


Homeostasis , Intestines/immunology , Intestines/physiology , Neuroglia/immunology , Neuroglia/physiology , Regeneration , Adventitia/immunology , Adventitia/parasitology , Animals , Chemokine CXCL10/immunology , Duodenum/immunology , Duodenum/parasitology , Duodenum/pathology , Duodenum/physiology , Female , Gliosis , Homeostasis/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/immunology , Intestines/parasitology , Intestines/pathology , Male , Mice , Nematospiroides dubius/immunology , Nematospiroides dubius/pathogenicity , Signal Transduction/immunology , Strongylida Infections/immunology , Strongylida Infections/parasitology , Strongylida Infections/pathology
10.
Front Immunol ; 12: 711907, 2021.
Article En | MEDLINE | ID: mdl-34484215

Group 2 innate lymphoid cells (ILC2s) are early effectors of mucosal type 2 immunity, producing cytokines such as interleukin (IL)-13 to mediate responses to helminth infection and allergen-induced inflammation. ILC2s are also present in lymph nodes (LNs) and can express molecules required for antigen presentation, but to date there are limited data on their dynamic behaviour. We used a CD2/IL-13 dual fluorescent reporter mouse for in vivo imaging of ILC2s and Th2 T cells in real time following a type 2 priming helminth infection or egg injection. After helminth challenge, we found that ILC2s were the main source of IL-13 in lymphoid organs (Peyer's patches and peripheral LNs), and were located in T cell areas. Intravital imaging demonstrated an increase in IL-13+ ILC2 size and movement following helminth infection, but reduced duration of interactions with T cells compared with those in homeostasis. In contrast, in the intestinal mucosa, we observed an increase in ILC2-T cell interactions post-infection, including some of prolonged duration, as well as increased IL-13+ ILC2 movement. These data suggest that ILC2 activation enhances cell motility, with the potential to increase the area of distribution of cytokines to optimise the early generation of type 2 responses. The prolonged ILC2 interactions with T cells within the intestinal mucosa are consistent with the conclusion that contact-based T cell activation may occur within inflamed tissues rather than lymphoid organs. Our findings have important implications for our understanding of the in vivo biology of ILC2s and the way in which these cells facilitate adaptive immune responses.


Intestinal Diseases, Parasitic/immunology , Lymphocyte Subsets/immunology , Nippostrongylus , Schistosomiasis mansoni/immunology , Strongylida Infections/immunology , Th2 Cells/immunology , Animals , Genes, Reporter , Interleukin-13/analysis , Intestinal Mucosa/immunology , Intestine, Small/immunology , Intestine, Small/parasitology , Intravital Microscopy , Lymphocyte Count , Lymphocyte Subsets/chemistry , Mice , Organ Specificity , Specific Pathogen-Free Organisms , Th2 Cells/chemistry
11.
Nat Metab ; 3(9): 1150-1162, 2021 09.
Article En | MEDLINE | ID: mdl-34531575

Macrophages exhibit a spectrum of activation states ranging from classical to alternative activation1. Alternatively, activated macrophages are involved in diverse pathophysiological processes such as confining tissue parasites2, improving insulin sensitivity3 or promoting an immune-tolerant microenvironment that facilitates tumour growth and metastasis4. Recently, the metabolic regulation of macrophage function has come into focus as both the classical and alternative activation programmes require specific regulated metabolic reprogramming5. While most of the studies regarding immunometabolism have focussed on the catabolic pathways activated to provide energy, little is known about the anabolic pathways mediating macrophage alternative activation. In this study, we show that the anabolic transcription factor sterol regulatory element binding protein 1 (SREBP1) is activated in response to the canonical T helper 2 cell cytokine interleukin-4 to trigger the de novo lipogenesis (DNL) programme, as a necessary step for macrophage alternative activation. Mechanistically, DNL consumes NADPH, partitioning it away from cellular antioxidant defences and raising reactive oxygen species levels. Reactive oxygen species serves as a second messenger, signalling sufficient DNL, and promoting macrophage alternative activation. The pathophysiological relevance of this mechanism is validated by showing that SREBP1/DNL is essential for macrophage alternative activation in vivo in a helminth infection model.


Antioxidants/metabolism , Fatty Acids/biosynthesis , Macrophages/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Dexamethasone/pharmacology , Humans , Interleukin-4/pharmacology , Lipopolysaccharides/pharmacology , Macrophage Activation , Macrophages/drug effects , Mice , Mice, Knockout , Nippostrongylus/isolation & purification , Nippostrongylus/pathogenicity , RAW 264.7 Cells , Sequence Analysis, RNA/methods , Strongylida Infections/immunology , Strongylida Infections/parasitology , Up-Regulation
12.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article En | MEDLINE | ID: mdl-34290141

"Taste-like" tuft cells in the intestine trigger type 2 immunity in response to worm infection. The secretion of interleukin-13 (IL-13) from type 2 innate lymphoid cells (ILC2) represents a key step in the tuft cell-ILC2 cell-intestinal epithelial cell circuit that drives the clearance of worms from the gut via type 2 immune responses. Hallmark features of type 2 responses include tissue remodeling, such as tuft and goblet cell expansion, and villus atrophy, yet it remains unclear if additional molecular changes in the gut epithelium facilitate the clearance of worms from the gut. Using gut organoids, we demonstrated that IL-4 and IL-13, two type 2 cytokines with similar functions, not only induced the classical type 2 responses (e.g., tuft cell expansion) but also drastically up-regulated the expression of gasdermin C genes (Gsdmcs). Using an in vivo worm-induced type 2 immunity model, we confirmed the up-regulation of Gsdmcs in Nippostrongylus brasiliensis-infected wild-type C57BL/6 mice. Consistent with gasdermin family members being principal effectors of pyroptosis, overexpression of Gsdmc2 in human embryonic kidney 293 (HEK293) cells triggered pyroptosis and lytic cell death. Moreover, in intestinal organoids treated with IL-4 or IL-13, or in wild-type mice infected with N. brasiliensis, lytic cell death increased, which may account for villus atrophy observed in worm-infected mice. Thus, we propose that the up-regulated Gsdmc family may be major effectors for type 2 responses in the gut and that Gsdmc-mediated pyroptosis may provide a conduit for the release of antiparasitic factors from enterocytes to facilitate the clearance of worms.


Cell Death , DNA-Binding Proteins/metabolism , Enterocytes/pathology , Immunity, Innate/immunology , Intestine, Small/pathology , Strongylida Infections/complications , Th2 Cells/immunology , Animals , Cell Proliferation , DNA-Binding Proteins/genetics , Enterocytes/immunology , Enterocytes/metabolism , Enterocytes/parasitology , Female , Interleukin-13/metabolism , Interleukin-4/metabolism , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/parasitology , Male , Mice , Mice, Inbred C57BL , Nippostrongylus/physiology , Signal Transduction , Strongylida Infections/immunology , Strongylida Infections/metabolism , Strongylida Infections/parasitology
13.
Indian J Tuberc ; 68(3): 340-349, 2021 Jul.
Article En | MEDLINE | ID: mdl-34099199

BACKGROUND: Tuberculosis is a chronic infection caused by Mycobacterium tuberculosis (M.tb), which needs proper macrophage activation for control. It has been debated whether the co-infection with helminth will affect the immune response to mycobacterial infection. OBJECTIVE: To determine the effect of sequential co-infection of Heligmosomoides polygyrus (H.pg) nematodes and M.tb on T cell responses, macrophages polarization and lung histopathological changes. METHOD: This study used 49 mice divided into 7 treatment groups, with different sequence of infection of M.tb via inhalation and H.pg via oral ingestion for 8 and 16 weeks. T cells response in the lung, intestine, and peripheral blood were determined by flow cytometry. Cytokines (IL-4, IFN-γ, TGB-ß1, and IL-10) were measured in peripheral blood using ELISA. Lung macrophage polarization were determined by the expression of iNOS (M1) or Arginase 1 (M2). Mycobacterial count were done in lung tissue. Lung histopathology were measured using Dorman's semiquantitative score assessing peribronchiolitis, perivasculitis, alveolitis, and granuloma formation. RESULT: M.tb infection induced Th1 response and M1 macrophage polarization, while H.pg infection induced Th2 and M2 polarization. In sequential co-infection, the final polarization of macrophage was dictated by the sequence of co-infection. However, all groups with M.tb infection showed the same degree of mycobacterial count in lung tissues and lung tissue histopathological changes. CONCLUSION: Sequential co-infection of H.pg and M.tb induces different T cell response which leads to different macrophage polarization in lung tissue. Helminth infection induced M2 lung macrophage polarization, but did not cause different mycobacterial count nor lung histopathological changes.


Lung , Macrophage Activation/immunology , Macrophages, Alveolar/immunology , Mycobacterium tuberculosis/immunology , Nematospiroides dubius/immunology , Strongylida Infections , Tuberculosis , Animals , Cell Count , Cell Polarity/immunology , Coinfection/immunology , Coinfection/microbiology , Coinfection/parasitology , Cytokines/blood , Disease Models, Animal , Immunity, Cellular , Lung/immunology , Lung/pathology , Mice , Strongylida Infections/immunology , Strongylida Infections/parasitology , T-Lymphocytes/immunology , Tuberculosis/immunology , Tuberculosis/microbiology
14.
Sci Immunol ; 6(60)2021 06 11.
Article En | MEDLINE | ID: mdl-34117110

Effective antiviral immunity requires generation of T and B lymphocytes expressing the transcription factor T-bet, a regulator of type 1 inflammatory responses. Using T-bet expression as an endogenous marker for cells participating in a type 1 response, we report coordinated interactions of T-bet-expressing T and B lymphocytes on the basis of their dynamic colocalization at the T cell zone and B follicle boundary (T-B boundary) and germinal centers (GCs) during lung influenza infection. We demonstrate that the assembly of this circuit takes place in distinct anatomical niches within the draining lymph node, guided by CXCR3 that enables positioning of TH1 cells at the T-B boundary. The encounter of B and TH1 cells at the T-B boundary enables IFN-γ produced by the latter to induce IgG2c class switching. Within GCs, T-bet+ TFH cells represent a specialized stable sublineage required for GC growth but dispensable for IgG2c class switching. Our studies show that during respiratory viral infection, T-bet-expressing T and B lymphocytes form a circuit assembled in a spatiotemporally controlled manner that acts as a functional unit enabling a robust and coherent humoral response tailored for optimal antiviral immunity.


B-Lymphocytes/immunology , Immunity, Humoral , Influenza, Human/immunology , T-Lymphocyte Subsets/immunology , Th1 Cells/immunology , Animals , B-Lymphocytes/metabolism , Cell Communication/immunology , Disease Models, Animal , Female , Germinal Center/cytology , Germinal Center/metabolism , Humans , Immunoglobulin Class Switching , Influenza A virus/immunology , Influenza, Human/pathology , Influenza, Human/virology , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lung/immunology , Lung/pathology , Lung/virology , Male , Mice , Mice, Transgenic , Nippostrongylus/immunology , Rats , Receptors, CXCR3/metabolism , Strongylida Infections/immunology , Strongylida Infections/parasitology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , T-Lymphocyte Subsets/metabolism , Th1 Cells/metabolism
15.
PLoS Pathog ; 17(6): e1009602, 2021 06.
Article En | MEDLINE | ID: mdl-34106992

The CD4+ T cell response is critical to host protection against helminth infection. How this response varies across different hosts and tissues remains an important gap in our understanding. Using IL-4-reporter mice to identify responding CD4+ T cells to Nippostrongylus brasiliensis infection, T cell receptor sequencing paired with novel clustering algorithms revealed a broadly reactive and clonally diverse CD4+ T cell response. While the most prevalent clones and clonotypes exhibited some tissue selectivity, most were observed to reside in both the lung and lung-draining lymph nodes. Antigen-reactivity of the broader repertoires was predicted to be shared across both tissues and individual mice. Transcriptome, trajectory, and chromatin accessibility analysis of lung and lymph-node repertoires revealed three unique but related populations of responding IL-4+ CD4+ T cells consistent with T follicular helper, T helper 2, and a transitional population sharing similarity with both populations. The shared antigen reactivity of lymph node and lung repertoires combined with the adoption of tissue-specific gene programs allows for the pairing of cellular and humoral responses critical to the orchestration of anti-helminth immunity.


CD4-Positive T-Lymphocytes/immunology , Strongylida Infections/immunology , Animals , Lung/immunology , Lymph Nodes/immunology , Mice , Nippostrongylus , Receptors, Antigen, T-Cell, alpha-beta/immunology , Single-Cell Analysis
16.
Cannabis Cannabinoid Res ; 6(3): 242-252, 2021 06.
Article En | MEDLINE | ID: mdl-33998896

Introduction: Over 1 billion humans carry infectious helminth parasites that can lead to chronic comorbidities such as anemia and growth retardation in children. Helminths induce a T-helper type 2 (Th2) immune response in the host and can cause severe tissue damage and fibrosis if chronic. We recently reported that mice infected with the soil-transmitted helminth, Nippostrongylus brasiliensis, displayed elevated levels of endocannabinoids (eCBs) in the lung and intestine. eCBs are lipid-signaling molecules that control inflammation; however, their function in infection is not well defined. Materials and Methods: A combination of pharmacological approaches and genetic mouse models was used to investigate roles for the eCB system in inflammatory responses and lung injury in mice during parasitic infection with N. brasiliensis. Results: Hemorrhaging of lung tissue in mice infected with N. brasiliensis was exacerbated by inhibiting peripheral cannabinoid receptor subtype-1 (CB1Rs) with the peripherally restricted CB1R antagonist, AM6545. In addition, these mice exhibited an increase in nonfunctional alveolar space and prolonged airway eosinophilia compared to vehicle-treated infected mice. In contrast to mice treated with AM6545, infected cannabinoid receptor subtype-2-null mice (Cnr2-/-) did not display any changes in these parameters compared to wild-type mice. Conclusions: Roles for the eCB system in Th2 immune responses are not well understood; however, increases in its activity in response to infection suggest an immunomodulatory role. Moreover, these findings suggest a role for eCB signaling at CB1Rs but not cannabinoid receptor subtypes-2 in the resolution of Th2 inflammatory responses, which become host destructive over time.


Endocannabinoids/immunology , Lung/pathology , Nippostrongylus/immunology , Receptor, Cannabinoid, CB1/immunology , Strongylida Infections/immunology , Animals , Eosinophilia , Hemorrhage , Lung/immunology , Lung/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Morpholines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/deficiency , Th2 Cells/immunology
17.
Front Immunol ; 12: 627638, 2021.
Article En | MEDLINE | ID: mdl-33936040

Background: Tuberculosis (TB) is still a major challenge for humankind. Because regions with the highest incidence also have a high prevalence of helminthiasis and nutritional scarcity, we wanted to understand the impact of these on TB progression. Methods: We have developed an experimental murine model for active TB in C3HeB/FeJ, coinfected with Trichuris muris and Heligmosomoides polygyrus nematodes, and exposed to an environmental mycobacterium (M. manresensis) and intermittent fasting. Cause-effect relationships among these factors were explored with Partial Least Squares Path modelling (PLSPM). Results: Previous parasitization had a major anti-inflammatory effect and reduced systemic levels of ADA, haptoglobin, local pulmonary levels of IL-1ß, IL-6, TNF-α, CXCL-1, CXCL-5 and IL-10. Oral administration of heat-killed M. manresensis resulted in a similar outcome. Both interventions diminished pulmonary pathology and bacillary load, but intermittent food deprivation reduced this protective effect increasing stress and inflammation. The PLSPM revealed nematodes might have protective effects against TB progression. Conclusions: Significantly higher cortisol levels in food-deprivation groups showed it is a stressful condition, which might explain its deleterious effect. This highlights the impact of food security on TB eradication policies and the need to prioritize food supply over deworming activities.


Coinfection , Food Deprivation , Helminthiasis/parasitology , Intestinal Diseases, Parasitic/parasitology , Lung/microbiology , Mycobacterium tuberculosis/pathogenicity , Nematospiroides dubius/pathogenicity , Strongylida Infections/parasitology , Trichuriasis/parasitology , Trichuris/pathogenicity , Tuberculosis, Pulmonary/microbiology , Animal Nutritional Physiological Phenomena , Animals , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Female , Helminthiasis/immunology , Helminthiasis/metabolism , Host-Parasite Interactions , Inflammation Mediators/metabolism , Intestinal Diseases, Parasitic/immunology , Intestinal Diseases, Parasitic/metabolism , Lung/immunology , Lung/metabolism , Male , Mice, Inbred C3H , Mycobacterium tuberculosis/immunology , Nematospiroides dubius/immunology , Nutritional Status , Strongylida Infections/immunology , Strongylida Infections/metabolism , Trichuriasis/immunology , Trichuriasis/metabolism , Trichuris/immunology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/metabolism
18.
PLoS One ; 16(5): e0251233, 2021.
Article En | MEDLINE | ID: mdl-34003838

The transcription factor Rora has been shown to be important for the development of ILC2 and the regulation of ILC3, macrophages and Treg cells. Here we investigate the role of Rora across CD4+ T cells in general, but with an emphasis on Th2 cells, both in vitro as well as in the context of several in vivo type 2 infection models. We dissect the function of Rora using overexpression and a CD4-conditional Rora-knockout mouse, as well as a RORA-reporter mouse. We establish the importance of Rora in CD4+ T cells for controlling lung inflammation induced by Nippostrongylus brasiliensis infection, and have measured the effect on downstream genes using RNA-seq. Using a systematic stimulation screen of CD4+ T cells, coupled with RNA-seq, we identify upstream regulators of Rora, most importantly IL-33 and CCL7. Our data suggest that Rora is a negative regulator of the immune system, possibly through several downstream pathways, and is under control of the local microenvironment.


CD4-Positive T-Lymphocytes/immunology , Macrophages/immunology , Nuclear Receptor Subfamily 1, Group F, Member 1/immunology , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Pneumonia/immunology , Th2 Cells/immunology , Animals , Antigens, Helminth/immunology , Antigens, Helminth/metabolism , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression Regulation/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nippostrongylus/immunology , Pneumonia/parasitology , Pneumonia/pathology , Strongylida Infections/immunology , Strongylida Infections/parasitology
19.
Mol Neurobiol ; 58(8): 3968-3982, 2021 Aug.
Article En | MEDLINE | ID: mdl-33904019

Angiostrongylus cantonensis infection is a typical cause of eosinophilic encephalitis (EM), which has been reported to induce serious damage in the central nervous system. Both parasite and host factors contribute to the onset of EM, but the related immune-inflammation pathogenesis remains poorly characterised. An A. cantonensis infection model was generated through the infection of mice by gavage. Transmission electron microscopy and immunohistochemistry were used to assess the pathologic changes in the brain. The mRNA expression of inflammatory factors was tested using qRT-PCR. A combination of flow cytometry and western blotting was used to evaluate the alteration of leukocytes and related cytokines. A critical role of IL-17 was found by injecting IL-17A monoclonal antibody into naïve and A. cantonensis-infected mice. A. cantonensis larvae altered the immune homeostasis in the brain, leading to the destruction of myelin sheaths and activation of microglia and macrophage. During this process, IL-17A accumulation was observed, and IL-17RA was expressed in oligodendrocytes and microglia during the infection. Notably, γδ T cell was the major origin of IL-17A production induced by the parasite. After an IL-17A-neutralising antibody was applied, alterations in myelination and the state of the microglia/macrophage were discovered; the neurobehavioural scores of the mice also improved. Our study reveals one unrecognised impact of the γδ T cells in parasitic encephalopathy and emphasises that blocking IL-17A signalling can attenuate microglia and macrophage activation, thus reducing CNS demyelination and ameliorating the neurobehavioural deficit in A. cantonensis-infected mice.


Brain/metabolism , Demyelinating Diseases/metabolism , Interleukin-17/biosynthesis , Intraepithelial Lymphocytes/metabolism , Strongylida Infections/metabolism , Animals , Brain/immunology , Demyelinating Diseases/immunology , Intraepithelial Lymphocytes/immunology , Male , Mice , Mice, Inbred BALB C , Microglia/immunology , Microglia/metabolism , Strongylida Infections/immunology
20.
Sci Immunol ; 6(56)2021 02 12.
Article En | MEDLINE | ID: mdl-33579750

Microanatomical organization of innate immune cells within lymph nodes (LNs) is critical for the generation of adaptive responses. In particular, steady-state LN-resident dendritic cells (Res cDCs) are strategically localized to intercept lymph-draining antigens. Whether myeloid cell organization changes during inflammation and how that might affect the generation of immune responses are unknown. Here, we report that during type I, but not type II, inflammation after adjuvant immunization or viral infection, antigen-presenting Res cDCs undergo CCR7-dependent intranodal repositioning from the LN periphery into the T cell zone (TZ) to elicit T cell priming. Concurrently, inflammatory monocytes infiltrate the LNs via local blood vessels, enter the TZ, and cooperate with Res cDCs by providing polarizing cytokines to optimize T cell effector differentiation. Monocyte infiltration is nonuniform across LNs, generating distinct microenvironments with varied local innate cell composition. These spatial microdomains are associated with divergent early T cell effector programming, indicating that innate microenvironments within LNs play a critical role in regulating the quality and heterogeneity of T cell responses. Together, our findings reveal that dynamic modulation of innate cell microenvironments during type I inflammation leads to optimized generation of adaptive immune responses to vaccines and infections.


Cell Communication/immunology , Cellular Microenvironment/immunology , Lymph Nodes/immunology , Strongylida Infections/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Cell Movement/immunology , Dendritic Cells/immunology , Disease Models, Animal , Humans , Immunity, Innate , Inflammation/immunology , Inflammation/pathology , Lymph Nodes/cytology , Lymph Nodes/pathology , Lymphocyte Activation , Mice , Mice, Transgenic , Monocytes/immunology , Nippostrongylus/immunology , Strongylida Infections/parasitology
...