Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 284
Filtrar
1.
PLoS Pathog ; 20(8): e1011766, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39141685

RESUMO

Intestinal helminth infection triggers a type 2 immune response that promotes a 'weep-and sweep' response characterised by increased mucus secretion and intestinal hypermotility, which function to dislodge the worm from its intestinal habitat. Recent studies have discovered that several other pathogens cause intestinal dysmotility through major alterations to the immune and enteric nervous systems (ENS), and their interactions, within the gastrointestinal tract. However, the involvement of these systems has not been investigated for helminth infections. Eosinophils represent a key cell type recruited by the type 2 immune response and alter intestinal motility under steady-state conditions. Our study aimed to investigate whether altered intestinal motility driven by the murine hookworm, Nippostrongylus brasiliensis, infection involves eosinophils and how the ENS and smooth muscles of the gut are impacted. Eosinophil deficiency did not influence helminth-induced intestinal hypermotility and hypermotility did not involve gross structural or functional changes to the ENS. Hypermotility was instead associated with a dramatic increase in smooth muscle thickness and contractility, an observation that extended to another rodent nematode, Heligmosomoides polygyrus. In summary our data indicate that, in contrast to other pathogens, helminth-induced intestinal hypermotility is driven by largely by myogenic, rather than neurogenic, alterations with such changes occurring independently of eosinophils. (<300 words).


Assuntos
Sistema Nervoso Entérico , Eosinófilos , Motilidade Gastrointestinal , Músculo Liso , Nippostrongylus , Animais , Camundongos , Eosinófilos/imunologia , Músculo Liso/parasitologia , Sistema Nervoso Entérico/parasitologia , Sistema Nervoso Entérico/imunologia , Motilidade Gastrointestinal/fisiologia , Nematospiroides dubius/fisiologia , Nematospiroides dubius/imunologia , Infecções por Strongylida/imunologia , Infecções por Strongylida/parasitologia , Enteropatias Parasitárias/imunologia , Enteropatias Parasitárias/parasitologia , Helmintíase/imunologia , Helmintíase/parasitologia , Neurônios/parasitologia , Neurônios/metabolismo , Camundongos Endogâmicos C57BL
2.
Sci Transl Med ; 16(761): eado1941, 2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39167662

RESUMO

Although vaccines have reduced the burden of COVID-19, their efficacy in helminth infection-endemic areas is not well characterized. We evaluated the impact of infection by Heligmosomoides polygyrus bakeri (Hpb), a murine intestinal roundworm, on the efficacy of an mRNA vaccine targeting the Wuhan-1 spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in mice. Although immunization generated similar B cell responses in Hpb-infected and uninfected mice, polyfunctional CD4+ and CD8+ T cell responses were markedly reduced in Hpb-infected mice. Hpb-infected and mRNA-vaccinated mice were protected against the ancestral SARS-CoV-2 strain WA1/2020, but control of lung infection was diminished against an Omicron variant compared with animals immunized without Hpb infection. Helminth-mediated suppression of spike protein-specific CD8+ T cell responses occurred independently of signal transducer and activator of transcription 6 (STAT6) signaling, whereas blockade of interleukin-10 (IL-10) rescued vaccine-induced CD8+ T cell responses. Together, these data show that, in mice, intestinal helminth infection impaired vaccine-induced T cell responses through an IL-10 pathway, which compromised protection against antigenically drifted SARS-CoV-2 variants.


Assuntos
Linfócitos T CD8-Positivos , Vacinas contra COVID-19 , COVID-19 , Interleucina-10 , Nematospiroides dubius , SARS-CoV-2 , Animais , COVID-19/imunologia , COVID-19/prevenção & controle , SARS-CoV-2/imunologia , Camundongos , Nematospiroides dubius/imunologia , Vacinas contra COVID-19/imunologia , Interleucina-10/metabolismo , Linfócitos T CD8-Positivos/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Camundongos Endogâmicos C57BL , Feminino , Linfócitos T/imunologia , Infecções por Strongylida/imunologia , Fator de Transcrição STAT6/metabolismo
3.
PLoS One ; 19(7): e0292408, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38950025

RESUMO

Co-infections are a common reality but understanding how the immune system responds in this context is complex and can be unpredictable. Heligmosomoides bakeri (parasitic roundworm, previously Heligmosomoides polygyrus) and Toxoplasma gondii (protozoan parasite) are well studied organisms that stimulate a characteristic Th2 and Th1 response, respectively. Several studies have demonstrated reduced inflammatory cytokine responses in animals co-infected with such organisms. However, while general cytokine signatures have been examined, the impact of the different cytokine producing lymphocytes on parasite control/clearance is not fully understood. We investigated five different lymphocyte populations (NK, NKT, γδ T, CD4+ T and CD8+ T cells), five organs (small intestine, Peyer's patches, mesenteric lymph nodes, spleen and liver), and 4 cytokines (IFN©, IL-4, IL-10 and IL-13) at two different time points (days 5 and 10 post T. gondii infection). We found that co-infected animals had significantly higher mortality than either single infection. This was accompanied by transient and local changes in parasite loads and cytokine profiles. Despite the early changes in lymphocyte and cytokine profiles, severe intestinal pathology in co-infected mice likely contributed to early mortality due to significant damage by both parasites in the small intestine. Our work demonstrates the importance of taking a broad view during infection research, studying multiple cell types, organs/tissues and time points to link and/or uncouple immunological from pathological findings. Our results provide insights into how co-infection with parasites stimulating different arms of the immune system can lead to drastic changes in infection dynamics.


Assuntos
Coinfecção , Citocinas , Nematospiroides dubius , Toxoplasma , Animais , Coinfecção/imunologia , Coinfecção/parasitologia , Toxoplasma/imunologia , Camundongos , Citocinas/metabolismo , Nematospiroides dubius/imunologia , Infecções por Strongylida/imunologia , Infecções por Strongylida/parasitologia , Infecções por Strongylida/mortalidade , Toxoplasmose/imunologia , Toxoplasmose/mortalidade , Toxoplasmose/complicações , Feminino , Toxoplasmose Animal/imunologia , Toxoplasmose Animal/mortalidade , Toxoplasmose Animal/parasitologia , Baço/imunologia , Baço/patologia , Baço/parasitologia , Carga Parasitária , Tecido Linfoide/imunologia , Tecido Linfoide/patologia , Tecido Linfoide/parasitologia
4.
Nat Commun ; 15(1): 5226, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38890291

RESUMO

IL-33 plays a significant role in inflammation, allergy, and host defence against parasitic helminths. The model gastrointestinal nematode Heligmosomoides polygyrus bakeri secretes the Alarmin Release Inhibitor HpARI2, an effector protein that suppresses protective immune responses and asthma in its host by inhibiting IL-33 signalling. Here we reveal the structure of HpARI2 bound to mouse IL-33. HpARI2 contains three CCP-like domains, and we show that it contacts IL-33 primarily through the second and third of these. A large loop which emerges from CCP3 directly contacts IL-33 and structural comparison shows that this overlaps with the binding site on IL-33 for its receptor, ST2, preventing formation of a signalling complex. Truncations of HpARI2 which lack the large loop from CCP3 are not able to block IL-33-mediated signalling in a cell-based assay and in an in vivo female mouse model of asthma. This shows that direct competition between HpARI2 and ST2 is responsible for suppression of IL-33-dependent responses.


Assuntos
Asma , Proteínas de Helminto , Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-33 , Nematospiroides dubius , Animais , Interleucina-33/metabolismo , Interleucina-33/química , Nematospiroides dubius/imunologia , Proteínas de Helminto/metabolismo , Proteínas de Helminto/química , Proteínas de Helminto/imunologia , Camundongos , Feminino , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Asma/imunologia , Asma/metabolismo , Humanos , Transdução de Sinais , Infecções por Strongylida/imunologia , Infecções por Strongylida/parasitologia , Infecções por Strongylida/metabolismo , Ligação Proteica , Modelos Animais de Doenças , Sítios de Ligação , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
5.
Gut Microbes ; 16(1): 2370917, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38944838

RESUMO

Polyphenols are phytochemicals commonly found in plant-based diets which have demonstrated immunomodulatory and anti-inflammatory properties. However, the interplay between polyphenols and pathogens at mucosal barrier surfaces has not yet been elucidated in detail. Here, we show that proanthocyanidin (PAC) polyphenols interact with gut parasites to influence immune function and gut microbial-derived metabolites in mice. PAC intake inhibited mastocytosis during infection with the small intestinal roundworm Heligmosomoides polygyrus, and altered the host tissue transcriptome at the site of infection with the large intestinal whipworm Trichuris muris, with a notable enhancement of type-1 inflammatory and interferon-driven gene pathways. In the absence of infection, PAC intake promoted the expansion of Turicibacter within the gut microbiota, increased fecal short chain fatty acids, and enriched phenolic metabolites such as phenyl-γ-valerolactones in the cecum. However, these putatively beneficial effects were reduced in PAC-fed mice infected with T. muris, suggesting concomitant parasite infection can attenuate gut microbial-mediated PAC catabolism. Collectively, our results suggest an inter-relationship between a phytonutrient and infection, whereby PAC may augment parasite-induced inflammation (most prominently with the cecum dwelling T. muris), and infection may abrogate the beneficial effects of health-promoting phytochemicals.


Assuntos
Microbioma Gastrointestinal , Nematospiroides dubius , Polifenóis , Proantocianidinas , Tricuríase , Trichuris , Animais , Camundongos , Polifenóis/farmacologia , Polifenóis/metabolismo , Trichuris/metabolismo , Tricuríase/parasitologia , Tricuríase/imunologia , Nematospiroides dubius/imunologia , Proantocianidinas/metabolismo , Proantocianidinas/farmacologia , Camundongos Endogâmicos C57BL , Infecções por Strongylida/imunologia , Infecções por Strongylida/parasitologia , Infecções por Strongylida/metabolismo , Feminino , Bactérias/classificação , Bactérias/metabolismo , Bactérias/genética , Bactérias/isolamento & purificação , Fezes/parasitologia , Fezes/microbiologia
6.
Allergy ; 79(8): 2157-2172, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38924546

RESUMO

BACKGROUND: Respiratory syncytial virus (RSV) infection in infants is a major cause of viral bronchiolitis and hospitalisation. We have previously shown in a murine model that ongoing infection with the gut helminth Heligmosomoides polygyrus protects against RSV infection through type I interferon (IFN-I) dependent reduction of viral load. Yet, the cellular basis for this protection has remained elusive. Given that recruitment of mononuclear phagocytes to the lung is critical for early RSV infection control, we assessed their role in this coinfection model. METHODS: Mice were infected by oral gavage with H. polygyrus. Myeloid immune cell populations were assessed by flow cytometry in lung, blood and bone marrow throughout infection and after secondary infection with RSV. Monocyte numbers were depleted by anti-CCR2 antibody or increased by intravenous transfer of enriched monocytes. RESULTS: H. polygyrus infection induces bone marrow monopoiesis, increasing circulatory monocytes and lung mononuclear phagocytes in a IFN-I signalling dependent manner. This expansion causes enhanced lung mononuclear phagocyte counts early in RSV infection that may contribute to the reduction of RSV load. Depletion or supplementation of circulatory monocytes prior to RSV infection confirms that these are both necessary and sufficient for helminth induced antiviral protection. CONCLUSIONS: H. polygyrus infection induces systemic monocytosis contributing to elevated mononuclear phagocyte numbers in the lung. These cells are central to an anti-viral effect that reduces the peak viral load in RSV infection. Treatments to promote or modulate these cells may provide novel paths to control RSV infection in high risk individuals.


Assuntos
Modelos Animais de Doenças , Monócitos , Infecções por Vírus Respiratório Sincicial , Carga Viral , Animais , Infecções por Vírus Respiratório Sincicial/imunologia , Camundongos , Monócitos/imunologia , Nematospiroides dubius/imunologia , Pulmão/imunologia , Pulmão/virologia , Infecções por Strongylida/imunologia , Vírus Sinciciais Respiratórios/imunologia , Interferon Tipo I/metabolismo
7.
Immunity ; 57(6): 1260-1273.e7, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38744292

RESUMO

Upon parasitic helminth infection, activated intestinal tuft cells secrete interleukin-25 (IL-25), which initiates a type 2 immune response during which lamina propria type 2 innate lymphoid cells (ILC2s) produce IL-13. This causes epithelial remodeling, including tuft cell hyperplasia, the function of which is unknown. We identified a cholinergic effector function of tuft cells, which are the only epithelial cells that expressed choline acetyltransferase (ChAT). During parasite infection, mice with epithelial-specific deletion of ChAT had increased worm burden, fitness, and fecal egg counts, even though type 2 immune responses were comparable. Mechanistically, IL-13-amplified tuft cells release acetylcholine (ACh) into the gut lumen. Finally, we demonstrated a direct effect of ACh on worms, which reduced their fecundity via helminth-expressed muscarinic ACh receptors. Thus, tuft cells are sentinels in naive mice, and their amplification upon helminth infection provides an additional type 2 immune response effector function.


Assuntos
Acetilcolina , Mucosa Intestinal , Animais , Acetilcolina/metabolismo , Camundongos , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/parasitologia , Colina O-Acetiltransferase/metabolismo , Interleucina-13/metabolismo , Interleucina-13/imunologia , Camundongos Knockout , Camundongos Endogâmicos C57BL , Helmintíase/imunologia , Helmintíase/parasitologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Imunidade Inata , Nematospiroides dubius/imunologia , Células em Tufo
8.
Immunol Cell Biol ; 102(5): 396-406, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38648862

RESUMO

Increased permeability of the intestinal epithelial layer is linked to the pathogenesis and perpetuation of a wide range of intestinal and extra-intestinal diseases. Infecting humans with controlled doses of helminths, such as human hookworm (termed hookworm therapy), is proposed as a treatment for many of the same diseases. Helminths induce immunoregulatory changes in their host which could decrease epithelial permeability, which is highlighted as a potential mechanism through which helminths treat disease. Despite this, the influence of a chronic helminth infection on epithelial permeability remains unclear. This study uses the chronically infecting intestinal helminth Heligmosomoides polygyrus to reveal alterations in the expression of intestinal tight junction proteins and epithelial permeability during the infection course. In the acute infection phase (1 week postinfection), an increase in intestinal epithelial permeability is observed. Consistent with this finding, jejunal claudin-2 is upregulated and tricellulin is downregulated. By contrast, in the chronic infection phase (6 weeks postinfection), colonic claudin-1 is upregulated and epithelial permeability decreases. Importantly, this study also investigates changes in epithelial permeability in a small human cohort experimentally challenged with the human hookworm, Necator americanus. It demonstrates a trend toward small intestinal permeability increasing in the acute infection phase (8 weeks postinfection), and colonic and whole gut permeability decreasing in the chronic infection phase (24 weeks postinfection), suggesting a conserved epithelial response between humans and mice. In summary, our findings demonstrate dynamic changes in epithelial permeability during a chronic helminth infection and provide another plausible mechanism by which chronic helminth infections could be utilized to treat disease.


Assuntos
Mucosa Intestinal , Permeabilidade , Animais , Humanos , Mucosa Intestinal/parasitologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/imunologia , Doença Crônica , Nematospiroides dubius/imunologia , Camundongos , Necator americanus , Enteropatias Parasitárias/imunologia , Junções Íntimas/metabolismo , Proteínas de Junções Íntimas/metabolismo , Intestino Delgado/parasitologia , Intestino Delgado/imunologia , Feminino , Camundongos Endogâmicos C57BL , Masculino , Helmintíase/imunologia , Helmintíase/parasitologia , Necatoríase/imunologia , Proteína 2 com Domínio MARVEL/metabolismo
9.
Nature ; 607(7917): 142-148, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35732734

RESUMO

The divergence of the common dendritic cell progenitor1-3 (CDP) into the conventional type 1 and type 2 dendritic cell (cDC1 and cDC2, respectively) lineages4,5 is poorly understood. Some transcription factors act in the commitment of already specified progenitors-such as BATF3, which stabilizes Irf8 autoactivation at the +32 kb Irf8 enhancer4,6-but the mechanisms controlling the initial divergence of CDPs remain unknown. Here we report the transcriptional basis of CDP divergence and describe the first requirements for pre-cDC2 specification. Genetic epistasis analysis7 suggested that Nfil3 acts upstream of Id2, Batf3 and Zeb2 in cDC1 development but did not reveal its mechanism or targets. Analysis of newly generated NFIL3 reporter mice showed extremely transient NFIL3 expression during cDC1 specification. CUT&RUN and chromatin immunoprecipitation followed by sequencing identified endogenous NFIL3 binding in the -165 kb Zeb2 enhancer8 at three sites that also bind the CCAAT-enhancer-binding proteins C/EBPα and C/EBPß. In vivo mutational analysis using CRISPR-Cas9 targeting showed that these NFIL3-C/EBP sites are functionally redundant, with C/EBPs supporting and NFIL3 repressing Zeb2 expression at these sites. A triple mutation of all three NFIL3-C/EBP sites ablated Zeb2 expression in myeloid, but not lymphoid progenitors, causing the complete loss of pre-cDC2 specification and mature cDC2 development in vivo. These mice did not generate T helper 2 (TH2) cell responses against Heligmosomoides polygyrus infection, consistent with cDC2 supporting TH2 responses to helminths9-11. Thus, CDP divergence into cDC1 or cDC2 is controlled by competition between NFIL3 and C/EBPs at the -165 kb Zeb2 enhancer.


Assuntos
Diferenciação Celular , Células Dendríticas , Elementos Facilitadores Genéticos , Mutação , Homeobox 2 de Ligação a E-box com Dedos de Zinco , Animais , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Diferenciação Celular/genética , Células Dendríticas/classificação , Células Dendríticas/citologia , Células Dendríticas/patologia , Elementos Facilitadores Genéticos/genética , Epistasia Genética , Proteína 2 Inibidora de Diferenciação , Linfócitos/citologia , Camundongos , Células Mieloides/citologia , Nematospiroides dubius/imunologia , Proteínas Repressoras , Células Th2/citologia , Células Th2/imunologia , Homeobox 2 de Ligação a E-box com Dedos de Zinco/genética
10.
J Biol Chem ; 298(6): 101994, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35500648

RESUMO

The mouse intestinal helminth Heligmosomoides polygyrus modulates host immune responses by secreting a transforming growth factor (TGF)-ß mimic (TGM), to expand the population of Foxp3+ Tregs. TGM comprises five complement control protein (CCP)-like domains, designated D1-D5. Though lacking homology to TGF-ß, TGM binds directly to the TGF-ß receptors TßRI and TßRII and stimulates the differentiation of naïve T-cells into Tregs. However, the molecular determinants of binding are unclear. Here, we used surface plasmon resonance, isothermal calorimetry, NMR spectroscopy, and mutagenesis to investigate how TGM binds the TGF-ß receptors. We demonstrate that binding is modular, with D1-D2 binding to TßRI and D3 binding to TßRII. D1-D2 and D3 were further shown to compete with TGF-ß(TßRII)2 and TGF-ß for binding to TßRI and TßRII, respectively. The solution structure of TGM-D3 revealed that TGM adopts a CCP-like fold but is also modified to allow the C-terminal strand to diverge, leading to an expansion of the domain and opening potential interaction surfaces. TGM-D3 also incorporates a long structurally ordered hypervariable loop, adding further potential interaction sites. Through NMR shift perturbations and binding studies of TGM-D3 and TßRII variants, TGM-D3 was shown to occupy the same site of TßRII as bound by TGF-ß using both a novel interaction surface and the hypervariable loop. These results, together with the identification of other secreted CCP-like proteins with immunomodulatory activity in H. polygyrus, suggest that TGM is part of a larger family of evolutionarily plastic parasite effector molecules that mediate novel interactions with their host.


Assuntos
Proteínas de Helminto , Interações Hospedeiro-Patógeno , Nematospiroides dubius , Receptores de Fatores de Crescimento Transformadores beta , Fator de Crescimento Transformador beta , Animais , Evolução Biológica , Proteínas de Helminto/genética , Proteínas de Helminto/imunologia , Interações Hospedeiro-Patógeno/imunologia , Camundongos , Nematospiroides dubius/classificação , Nematospiroides dubius/genética , Nematospiroides dubius/imunologia , Nematospiroides dubius/metabolismo , Ligação Proteica , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta/metabolismo
11.
Nature ; 599(7883): 125-130, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34671159

RESUMO

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.


Assuntos
Homeostase , Intestinos/imunologia , Intestinos/fisiologia , Neuroglia/imunologia , Neuroglia/fisiologia , Regeneração , Túnica Adventícia/imunologia , Túnica Adventícia/parasitologia , Animais , Quimiocina CXCL10/imunologia , Duodeno/imunologia , Duodeno/parasitologia , Duodeno/patologia , Duodeno/fisiologia , Feminino , Gliose , Homeostase/imunologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Interferon gama/imunologia , Intestinos/parasitologia , Intestinos/patologia , Masculino , Camundongos , Nematospiroides dubius/imunologia , Nematospiroides dubius/patogenicidade , Transdução de Sinais/imunologia , Infecções por Strongylida/imunologia , Infecções por Strongylida/parasitologia , Infecções por Strongylida/patologia
12.
Indian J Tuberc ; 68(3): 340-349, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34099199

RESUMO

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.


Assuntos
Pulmão , Ativação de Macrófagos/imunologia , Macrófagos Alveolares/imunologia , Mycobacterium tuberculosis/imunologia , Nematospiroides dubius/imunologia , Infecções por Strongylida , Tuberculose , Animais , Contagem de Células , Polaridade Celular/imunologia , Coinfecção/imunologia , Coinfecção/microbiologia , Coinfecção/parasitologia , Citocinas/sangue , Modelos Animais de Doenças , Imunidade Celular , Pulmão/imunologia , Pulmão/patologia , Camundongos , Infecções por Strongylida/imunologia , Infecções por Strongylida/parasitologia , Linfócitos T/imunologia , Tuberculose/imunologia , Tuberculose/microbiologia
13.
Front Immunol ; 12: 627638, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33936040

RESUMO

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.


Assuntos
Coinfecção , Privação de Alimentos , Helmintíase/parasitologia , Enteropatias Parasitárias/parasitologia , Pulmão/microbiologia , Mycobacterium tuberculosis/patogenicidade , Nematospiroides dubius/patogenicidade , Infecções por Strongylida/parasitologia , Tricuríase/parasitologia , Trichuris/patogenicidade , Tuberculose Pulmonar/microbiologia , Fenômenos Fisiológicos da Nutrição Animal , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Feminino , Helmintíase/imunologia , Helmintíase/metabolismo , Interações Hospedeiro-Parasita , Mediadores da Inflamação/metabolismo , Enteropatias Parasitárias/imunologia , Enteropatias Parasitárias/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Masculino , Camundongos Endogâmicos C3H , Mycobacterium tuberculosis/imunologia , Nematospiroides dubius/imunologia , Estado Nutricional , Infecções por Strongylida/imunologia , Infecções por Strongylida/metabolismo , Tricuríase/imunologia , Tricuríase/metabolismo , Trichuris/imunologia , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/metabolismo
14.
Nat Immunol ; 22(2): 216-228, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33462454

RESUMO

CD4+ effector lymphocytes (Teff) are traditionally classified by the cytokines they produce. To determine the states that Teff cells actually adopt in frontline tissues in vivo, we applied single-cell transcriptome and chromatin analyses to colonic Teff cells in germ-free or conventional mice or in mice after challenge with a range of phenotypically biasing microbes. Unexpected subsets were marked by the expression of the interferon (IFN) signature or myeloid-specific transcripts, but transcriptome or chromatin structure could not resolve discrete clusters fitting classic helper T cell (TH) subsets. At baseline or at different times of infection, transcripts encoding cytokines or proteins commonly used as TH markers were distributed in a polarized continuum, which was functionally validated. Clones derived from single progenitors gave rise to both IFN-γ- and interleukin (IL)-17-producing cells. Most of the transcriptional variance was tied to the infecting agent, independent of the cytokines produced, and chromatin variance primarily reflected activities of activator protein (AP)-1 and IFN-regulatory factor (IRF) transcription factor (TF) families, not the canonical subset master regulators T-bet, GATA3 or RORγ.


Assuntos
Bactérias/patogenicidade , Infecções Bacterianas/microbiologia , Linfócitos T CD4-Positivos/microbiologia , Linfócitos T CD4-Positivos/parasitologia , Colo/microbiologia , Colo/parasitologia , Microbioma Gastrointestinal , Heligmosomatoidea/patogenicidade , Enteropatias Parasitárias/parasitologia , Animais , Bactérias/imunologia , Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Infecções Bacterianas/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Cromatina/genética , Cromatina/metabolismo , Citrobacter rodentium/imunologia , Citrobacter rodentium/patogenicidade , Colo/imunologia , Colo/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Heligmosomatoidea/imunologia , Interações Hospedeiro-Patógeno , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Enteropatias Parasitárias/genética , Enteropatias Parasitárias/imunologia , Enteropatias Parasitárias/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Nematospiroides dubius/imunologia , Nematospiroides dubius/patogenicidade , Nippostrongylus/imunologia , Nippostrongylus/patogenicidade , Fenótipo , Salmonella enterica/imunologia , Salmonella enterica/patogenicidade , Análise de Célula Única , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Transcriptoma
15.
Eur J Immunol ; 51(2): 433-444, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33067820

RESUMO

Infection with parasitic worms (helminths) alters host immune responses and can inhibit pathogenic inflammation. Helminth infection promotes a strong Th2 and T regulatory response while suppressing Th1 and Th17 function. Th2 responses are largely dependent on transcriptional programs directed by Stat6-signaling. We examined the importance of intact T cell Stat6 signaling on helminth-induced suppression of murine colitis that results from T cell transfer into immune-deficient mice. Colonization with the intestinal nematode Heligmosomoides polygyrus bakeri resolves WT T cell transfer colitis. However, if the transferred T cells lack intact Stat6 then helminth exposure failed to attenuate colitis or suppress MLN T cell IFN-γ or IL17 production. Loss of Stat6 signaling resulted in decreased IL10 and increased IFN-γ co-expression by IL-17+ T cells. We also transferred T cells from mice with constitutive T cell expression of activated Stat6 (Stat6VT). These mice developed a severe eosinophilic colitis that also was not attenuated by helminth infection. These results show that T cell expression of intact but regulated Stat6 signaling is required for helminth infection-associated regulation of pathogenic intestinal inflammation.


Assuntos
Colite/imunologia , Nematospiroides dubius/imunologia , Fator de Transcrição STAT6/imunologia , Transdução de Sinais/imunologia , Infecções por Strongylida/imunologia , Linfócitos T Reguladores/imunologia , Animais , Colite/parasitologia , Interferon gama/imunologia , Interleucina-10/imunologia , Interleucina-17/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Células Th1/imunologia , Células Th1/parasitologia , Células Th17/imunologia , Células Th17/parasitologia , Células Th2/imunologia , Células Th2/parasitologia
16.
Dev Comp Immunol ; 115: 103879, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33007336

RESUMO

The mouse intestinal parasite Heligmosomoides polygyrus demonstrates adaptation to the inflammatory milieu as a result of colitis induced by dextran sulphate sodium (DSS). Nematodes from mice with colitis had different effects on dendritic cells than nematodes from mice without colitis. Immature JAWSII cells pre-exposed to L4 stage H. polygyrus from DSS-treated mice were adoptively transferred to mice with induced colitis. After two days, a higher disease activity index, macroscopic damage score and colon histology score were observed. MLN T cells isolated nine days after transfer demonstrated proinflammatory IFN-γ and IL-17 production. Transfer of JAWSII stimulated with male or female L4 larvae from a control invasion resulted in a slight improvement of colitis; in addition, dendritic cells exposed to H. polygyrus female L4 larvae, provoked migration of CD8+CD25+ T cells from MLN to the colon. Nematodes from an inflammatory environment changed cytokine production by dendritic cells. Inflammatory milieu changing nematode immunomodulatory activity affects dendritic cell functions, which offers new insight into the helminth-host relationship.


Assuntos
Colite/terapia , Células Dendríticas/imunologia , Nematospiroides dubius/imunologia , Terapia com Helmintos/métodos , Transferência Adotiva , Animais , Movimento Celular/imunologia , Colite/induzido quimicamente , Colite/imunologia , Colite/patologia , Sulfato de Dextrana/administração & dosagem , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Feminino , Interações Hospedeiro-Parasita/imunologia , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Larva , Linfonodos/citologia , Linfonodos/imunologia , Masculino , Mesentério , Camundongos , Cultura Primária de Células , Fatores Sexuais , Linfócitos T/imunologia
17.
Sci Immunol ; 5(53)2020 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-33188058

RESUMO

Interleukin-33 (IL-33) is a pleiotropic cytokine that can promote type 2 inflammation but also drives immunoregulation through Foxp3+Treg expansion. How IL-33 is exported from cells to serve this dual role in immunosuppression and inflammation remains unclear. Here, we demonstrate that the biological consequences of IL-33 activity are dictated by its cellular source. Whereas IL-33 derived from epithelial cells stimulates group 2 innate lymphoid cell (ILC2)-driven type 2 immunity and parasite clearance, we report that IL-33 derived from myeloid antigen-presenting cells (APCs) suppresses host-protective inflammatory responses. Conditional deletion of IL-33 in CD11c-expressing cells resulted in lowered numbers of intestinal Foxp3+Treg cells that express the transcription factor GATA3 and the IL-33 receptor ST2, causing elevated IL-5 and IL-13 production and accelerated anti-helminth immunity. We demonstrate that cell-intrinsic IL-33 promoted mouse dendritic cells (DCs) to express the pore-forming protein perforin-2, which may function as a conduit on the plasma membrane facilitating IL-33 export. Lack of perforin-2 in DCs blocked the proliferative expansion of the ST2+Foxp3+Treg subset. We propose that perforin-2 can provide a plasma membrane conduit in DCs that promotes the export of IL-33, contributing to mucosal immunoregulation under steady-state and infectious conditions.


Assuntos
Células Dendríticas/imunologia , Interleucina-33/metabolismo , Proteínas de Membrana/metabolismo , Infecções por Strongylida/imunologia , Linfócitos T Reguladores/imunologia , Animais , Membrana Celular/metabolismo , Doença Crônica , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Tolerância Imunológica , Imunidade Inata , Imunidade nas Mucosas , Interleucina-33/análise , Interleucina-33/genética , Masculino , Camundongos , Camundongos Transgênicos , Mucosa Nasal/imunologia , Mucosa Nasal/patologia , Pólipos Nasais/imunologia , Pólipos Nasais/patologia , Nematospiroides dubius/imunologia , Nippostrongylus/imunologia , Proteínas Citotóxicas Formadoras de Poros , Rinite/imunologia , Rinite/patologia , Sinusite/imunologia , Sinusite/patologia , Infecções por Strongylida/parasitologia , Linfócitos T Reguladores/metabolismo
18.
Front Immunol ; 11: 1830, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33117327

RESUMO

Helminth parasites are effective in biasing Th2 immunity and inducing regulatory pathways that minimize excessive inflammation within their hosts, thus allowing chronic infection to occur whilst also suppressing bystander atopic or autoimmune diseases. Multiple sclerosis (MS) is a severe autoimmune disease characterized by inflammatory lesions within the central nervous system; there are very limited therapeutic options for the progressive forms of the disease and none are curative. Here, we used the experimental autoimmune encephalomyelitis (EAE) model to examine if the intestinal helminth Heligmosomoides polygyrus and its excretory/secretory products (HES) are able to suppress inflammatory disease. Mice infected with H. polygyrus at the time of immunization with the peptide used to induce EAE (myelin-oligodendrocyte glycoprotein, pMOG), showed a delay in the onset and peak severity of EAE disease, however, treatment with HES only showed a marginal delay in disease onset. Mice that received H. polygyrus 4 weeks prior to EAE induction were also not significantly protected. H. polygyrus secretes a known TGF-ß mimic (Hp-TGM) and simultaneous H. polygyrus infection with pMOG immunization led to a significant expansion of Tregs; however, administering the recombinant Hp-TGM to EAE mice failed to replicate the EAE protection seen during infection, indicating that this may not be central to the disease protecting mechanism. Mice infected with H. polygyrus also showed a systemic Th2 biasing, and restimulating splenocytes with pMOG showed release of pMOG-specific IL-4 as well as suppression of inflammatory IL-17A. Notably, a Th2-skewed response was found only in mice infected with H. polygyrus at the time of EAE induction and not those with a chronic infection. Furthermore, H. polygyrus failed to protect against disease in IL-4Rα-/- mice. Together these results indicate that the EAE disease protective mechanism of H. polygyrus is likely to be predominantly Th2 deviation, and further highlights Th2-biasing as a future therapeutic strategy for MS.


Assuntos
Antígenos de Helmintos/imunologia , Encefalomielite Autoimune Experimental/imunologia , Receptores de Superfície Celular/imunologia , Infecções por Strongylida/imunologia , Células Th2/imunologia , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nematospiroides dubius/imunologia
19.
Int J Mol Sci ; 21(21)2020 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-33105843

RESUMO

As the global incidences of colorectal cancer rises, there is a growing importance in understanding the interaction between external factors, such as common infections, on the initiation and progression of this disease. While certain helminth infections have been shown to alter the severity and risk of developing colitis-associated colorectal cancer, whether these parasites can directly affect colorectal cancer progression is unknown. Here, we made use of murine and human colorectal cancer cell lines to demonstrate that exposure to antigens derived from the gastrointestinal nematode Heligmosomoides polygyrus significantly reduced colorectal cancer cell proliferation in vitro. Using a range of approaches, we demonstrate that antigen-dependent reductions in cancer cell proliferation and viability are associated with increased expression of the critical cell cycle regulators p53 and p21. Interestingly, H. polygyrus-derived antigens significantly increased murine colorectal cancer cell migration, which was associated with an increased expression of the adherens junction protein ß-catenin, whereas the opposite was true for human colorectal cancer cells. Together, these findings demonstrate that antigens derived from a gastrointestinal nematode can significantly alter colorectal cancer cell behavior. Further in-depth analysis may reveal novel candidates for targeting and treating late-stage cancer.


Assuntos
Antígenos de Helmintos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Nematospiroides dubius/imunologia , Animais , Antígenos de Helmintos/imunologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Células HCT116 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Nematospiroides dubius/metabolismo , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Especificidade da Espécie , Proteína Supressora de Tumor p53/metabolismo , beta Catenina/metabolismo
20.
Arch Immunol Ther Exp (Warsz) ; 68(5): 30, 2020 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-32960348

RESUMO

An effective host immune system prevents the growth of most cancer cells. However, as intestinal nematodes are able to induce both immunotolerance and immunosuppression in the host, it is possible that their presence could allow co-occurring cancer cells to proliferate and metastasize. Our findings indicate that previous, subsequent or concurrent intestinal nematode infection affects the formation of lung metastatic nodules in mice experimentally infected with Heligmosomoides polygyrus. In addition, pre-infection with nematodes renders mice resistant to metastasis development in lungs, with the inoculated EL4 cancer cells being located mainly in mesenteric lymph nodes. The present paper discusses the nematode-induced mechanisms which may influence the metastatic process.


Assuntos
Helmintíase/imunologia , Enteropatias Parasitárias/imunologia , Neoplasias Pulmonares/secundário , Linfoma/imunologia , Linfoma/parasitologia , Nematospiroides dubius/imunologia , Animais , Modelos Animais de Doenças , Imunomodulação , Neoplasias Pulmonares/parasitologia , Linfoma/patologia , Masculino , Camundongos , Infecções por Nematoides/imunologia , Metástase Neoplásica , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...