Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mucosal Immunol ; 14(4): 912-922, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33846533

RESUMO

Acute exacerbations (AE) of asthma, remain one of the biggest concerns for patients living with asthma. As such, identifying the causes, the molecular mechanisms involved and new therapeutic interventions to prevent AE is a high priority. Immunity to intestinal helminths involves the reactivation of type-2 immune responses leading to smooth muscle contraction and mucus hypersecretion-physiological processes very similar to acute exacerbations in the airways following allergen exposure. In this study, we employed a murine model of intestinal helminth infection, using Heligmosomoides polygyrus, to identify miRNAs during active expulsion, as a system for the identification of miRNAs that may contribute to AE in the airways. Concomitant with type-2 immunity and expulsion of H. polygyrus, we identified miR-99a-5p, miR-148a-3p and miR-155-5p that were differentially regulated. Systemic inhibition of these miRNAs, alone or in combination, had minimal impact on expulsion of H. polygyrus, but inhibition of miR-99a-5p or miR-155-5p significantly reduced house dust mite (HDM)-driven acute inflammation, modelling human acute exacerbations. Immunological, pathological and transcriptional analysis identified that miR-155-5p or miR-99a-5p contribute significantly to HDM-driven AE and that transient inhibition of these miRNAs may provide relief from allergen-driven AE, without compromising anti-helminth immunity in the gut.


Assuntos
Alérgenos/imunologia , Asma/etiologia , Memória Imunológica , MicroRNAs/genética , Animais , Asma/metabolismo , Asma/patologia , Biomarcadores , Progressão da Doença , Suscetibilidade a Doenças , Perfilação da Expressão Gênica , Helmintíase Animal/complicações , Helmintíase Animal/imunologia , Helmintíase Animal/parasitologia , Interações Hospedeiro-Parasita , Imunidade Inata , Camundongos
2.
Viruses ; 11(2)2019 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-30791609

RESUMO

Viruses are a major threat to human health and economic well-being. In recent years Ebola, Zika, influenza, and chikungunya virus epidemics have raised awareness that infections can spread rapidly before vaccines or specific antagonists can be made available. Broad-spectrum antivirals are drugs with the potential to inhibit infection by viruses from different groups or families, which may be deployed during outbreaks when specific diagnostics, vaccines or directly acting antivirals are not available. While pathogen-directed approaches are generally effective against a few closely related viruses, targeting cellular pathways used by multiple viral agents can have broad-spectrum efficacy. Virus entry, particularly clathrin-mediated endocytosis, constitutes an attractive target as it is used by many viruses. Using a phenotypic screening strategy where the inhibitory activity of small molecules was sequentially tested against different viruses, we identified 12 compounds with broad-spectrum activity, and found a subset blocking viral internalisation and/or fusion. Importantly, we show that compounds identified with this approach can reduce viral replication in a mouse model of Zika infection. This work provides proof of concept that it is possible to identify broad-spectrum inhibitors by iterative phenotypic screenings, and that inhibition of host-pathways critical for viral life cycles can be an effective antiviral strategy.


Assuntos
Antivirais/isolamento & purificação , Antivirais/farmacologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas , Internalização do Vírus/efeitos dos fármacos , Vírus/efeitos dos fármacos , Animais , Células HeLa , Humanos , Concentração Inibidora 50 , Camundongos , RNA Viral/genética , Replicação Viral/efeitos dos fármacos , Zika virus/efeitos dos fármacos , Infecção por Zika virus/tratamento farmacológico
3.
Cell Host Microbe ; 22(4): 484-493.e5, 2017 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-29024642

RESUMO

Immunity to intestinal helminth infections has been well studied, but the mechanism of helminth killing prior to expulsion remains unclear. Here we identify epithelial-cell-derived phospholipase A2 group 1B (PLA2g1B) as a host-derived endogenous anthelmintic. PLA2g1B is elevated in resistant mice and is responsible for killing tissue-embedded larvae. Despite comparable activities of other essential type-2-dependent immune mechanisms, Pla2g1b-/- mice failed to expel the intestinal helminths Heligmosomoides polygyrus or Nippostrongylus brasiliensis. Expression of Pla2g1b by epithelial cells was dependent upon intestinal microbiota, adaptive immunity, and common-gamma chain-dependent signaling. Notably, Pla2g1b was downregulated in susceptible mice and inhibited by IL-4R-signaling in vitro, uncoupling parasite killing from expulsion mechanisms. Resistance was restored in Pla2g1b-/- mice by treating infective H. polygyrus L3 larvae with PLA2g1B, which reduced larval phospholipid abundance. These findings uncover epithelial-cell-derived Pla2g1b as an essential mediator of helminth killing, highlighting a previously overlooked mechanism of anti-helminth immunity.


Assuntos
Fosfolipases A2 do Grupo IB/imunologia , Mucosa Intestinal/imunologia , Nematospiroides dubius/imunologia , Nippostrongylus/imunologia , Fosfolipídeos/metabolismo , Infecções por Strongylida/imunologia , Imunidade Adaptativa , Animais , Microbioma Gastrointestinal/imunologia , Fosfolipases A2 do Grupo IB/genética , Mucosa Intestinal/citologia , Mucosa Intestinal/enzimologia , Larva/imunologia , Camundongos , Camundongos Knockout , Cultura Primária de Células
4.
J Exp Med ; 214(6): 1809-1826, 2017 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-28507062

RESUMO

Immunity to intestinal helminth infections requires the rapid activation of T helper 2 cells (Th2 cells). However, simultaneous expansion of CD4+Foxp3+ regulatory T cells (T reg cells) impedes protective responses, resulting in chronic infections. The ratio between T reg and effector T cells can therefore determine the outcome of infection. The redifferentiation of T reg cells into Th cells has been identified in hyperinflammatory diseases. In this study, we asked whether ex-T reg Th2 cells develop and contribute to type-2 immunity. Using multigene reporter and fate-reporter systems, we demonstrate that a significant proportion of Th2 cells derive from Foxp3+ cells after Heligmosomoides polygyrus infection and airway allergy. Ex-Foxp3 Th2 cells exhibit characteristic Th2 effector functions and provide immunity to H. polygyrus Through selective deletion of Il4ra on Foxp3+ cells, we further demonstrate IL-4 is required for the development of ex-Foxp3 Th2 cells. Collectively, our findings indicate that converting T reg cells into Th2 cells could concomitantly enhance Th2 cells and limit T reg cell-mediated suppression.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Imunidade , Interleucina-4/metabolismo , Intestinos/imunologia , Intestinos/parasitologia , Nematospiroides dubius/imunologia , Células Th2/imunologia , Transferência Adotiva , Animais , Polaridade Celular , Perfilação da Expressão Gênica , Imunidade/genética , Camundongos Endogâmicos C57BL , Receptores de Interleucina-4/metabolismo , Transdução de Sinais , Infecções por Strongylida/imunologia , Infecções por Strongylida/parasitologia , Linfócitos T Reguladores/imunologia
5.
J Allergy Clin Immunol ; 139(2): 655-666.e7, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27484038

RESUMO

BACKGROUND: The molecular and cellular pathways driving the pathogenesis of severe asthma are poorly defined. Tumor progression locus 2 (TPL-2) (COT, MAP3K8) kinase activates the MEK1/2-extracellular-signal regulated kinase 1/2 MAP kinase signaling pathway following Toll-like receptor, TNFR1, and IL-1R stimulation. OBJECTIVE: TPL-2 has been widely described as a critical regulator of inflammation, and we sought to investigate the role of TPL-2 in house dust mite (HDM)-mediated allergic airway inflammation. METHODS: A comparative analysis of wild-type and Map3k8-/- mice was conducted. Mixed bone marrow chimeras, conditional knockout mice, and adoptive transfer models were also used. Differential cell counts were performed on the bronchoalveolar lavage fluid, followed by histological analysis of lung sections. Flow cytometry and quantitative PCR was used to measure type 2 cytokines. ELISA was used to assess the production of IgE, type 2 cytokines, and Ccl24. RNA sequencing was used to characterize dendritic cell (DC) transcripts. RESULTS: TPL-2 deficiency led to exacerbated HDM-induced airway allergy, with increased airway and tissue eosinophilia, lung inflammation, and IL-4, IL-5, IL-13, and IgE production. Increased airway allergic responses in Map3k8-/- mice were not due to a cell-intrinsic role for TPL-2 in T cells, B cells, or LysM+ cells but due to a regulatory role for TPL-2 in DCs. TPL-2 inhibited Ccl24 expression in lung DCs, and blockade of Ccl24 prevented the exaggerated airway eosinophilia and lung inflammation in mice given HDM-pulsed Map3k8-/- DCs. CONCLUSIONS: TPL-2 regulates DC-derived Ccl24 production to prevent severe type 2 airway allergy in mice.


Assuntos
Asma/imunologia , Quimiocina CCL24/metabolismo , Células Dendríticas/imunologia , Eosinófilos/imunologia , Pulmão/imunologia , MAP Quinase Quinase Quinases/metabolismo , Pneumonia/imunologia , Proteínas Proto-Oncogênicas/metabolismo , Animais , Antígenos de Dermatophagoides/imunologia , Citocinas/metabolismo , Imunoglobulina E/sangue , MAP Quinase Quinase Quinases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas/genética , Pyroglyphidae/imunologia , Transdução de Sinais , Células Th2/imunologia
6.
Traffic ; 17(9): 997-1013, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27219333

RESUMO

Interferon inducible transmembrane proteins (IFITMs) are broad-spectrum antiviral factors. In cell culture the entry of many enveloped viruses, including orthomyxo-, flavi-, and filoviruses, is inhibited by IFITMs, though the mechanism(s) involved remain unclear and may vary between viruses. We demonstrate that Sindbis and Semliki Forest virus (SFV), which both use endocytosis and acid-induced membrane fusion in early endosomes to infect cells, are restricted by the early endosomal IFITM3. The late endosomal IFITM2 is less restrictive and the plasma membrane IFITM1 does not inhibit normal infection by either virus. IFITM3 inhibits release of the SFV capsid into the cytosol, without inhibiting binding, internalization, trafficking to endosomes or low pH-induced conformational changes in the envelope glycoprotein. Infection by SFV fusion at the cell surface was inhibited by IFITM1, but was equally inhibited by IFITM3. Furthermore, an IFITM3 mutant (Y20A) that is localized to the plasma membrane inhibited infection by cell surface fusion more potently than IFITM1. Together, these results indicate that IFITMs, in particular IFITM3, can restrict alphavirus infection by inhibiting viral fusion with cellular membranes. That IFITM3 can restrict SFV infection by fusion at the cell surface equivalently to IFITM1 suggests that IFITM3 has greater antiviral potency against SFV.


Assuntos
Antígenos de Diferenciação/metabolismo , Endossomos/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Ligação a RNA/metabolismo , Vírus da Floresta de Semliki/fisiologia , Sindbis virus/fisiologia , Células A549 , Infecções por Alphavirus/metabolismo , Infecções por Alphavirus/virologia , Antígenos de Diferenciação/genética , Endocitose/fisiologia , Endossomos/virologia , Humanos , Proteínas de Membrana/genética , Proteínas de Ligação a RNA/genética , Internalização do Vírus
7.
Proc Natl Acad Sci U S A ; 113(5): E568-76, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26787865

RESUMO

There is a paucity of new therapeutic targets to control allergic reactions and forestall the rising trend of allergic diseases. Although a variety of immune cells contribute to allergy, cytokine-secreting αß(+)CD4(+) T-helper 2 (TH2) cells orchestrate the type-2-driven immune response in a large proportion of atopic asthmatics. To identify previously unidentified putative targets in pathogenic TH2 cells, we performed in silico analyses of recently published transcriptional data from a wide variety of pathogenic TH cells [Okoye IS, et al. (2014) Proc Natl Acad Sci USA 111(30):E3081-E3090] and identified that transcription intermediary factor 1 regulator-alpha (Tif1α)/tripartite motif-containing 24 (Trim24) was predicted to be active in house dust mite (HDM)- and helminth-elicited Il4(gfp+)αß(+)CD4(+) TH2 cells but not in TH1, TH17, or Treg cells. Testing this prediction, we restricted Trim24 deficiency to T cells by using a mixed bone marrow chimera system and found that T-cell-intrinsic Trim24 is essential for HDM-mediated airway allergy and antihelminth immunity. Mechanistically, HDM-elicited Trim24(-/-) T cells have reduced expression of many TH2 cytokines and chemokines and were predicted to have compromised IL-1-regulated signaling. Following this prediction, we found that Trim24(-/-) T cells have reduced IL-1 receptor (IL-1R) expression, are refractory to IL-1ß-mediated activation in vitro and in vivo, and fail to respond to IL-1ß-exacerbated airway allergy. Collectively, these data identify a previously unappreciated Trim24-dependent requirement for IL-1R expression on TH2 cells and an important nonredundant role for T-cell-intrinsic Trim24 in TH2-mediated allergy and antihelminth immunity.


Assuntos
Hipersensibilidade/imunologia , Proteínas Nucleares/fisiologia , Receptores de Interleucina-1/metabolismo , Células Th2/imunologia , Fatores de Transcrição/fisiologia , Animais , Helmintos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Nucleares/genética , Células Th2/metabolismo , Fatores de Transcrição/genética
8.
PLoS Pathog ; 11(7): e1004994, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26147567

RESUMO

Parasitic helminths establish chronic infections in mammalian hosts. Helminth/Plasmodium co-infections occur frequently in endemic areas. However, it is unclear whether Plasmodium infections compromise anti-helminth immunity, contributing to the chronicity of infection. Immunity to Plasmodium or helminths requires divergent CD4+ T cell-driven responses, dominated by IFNγ or IL-4, respectively. Recent literature has indicated that Th cells, including Th2 cells, have phenotypic plasticity with the ability to produce non-lineage associated cytokines. Whether such plasticity occurs during co-infection is unclear. In this study, we observed reduced anti-helminth Th2 cell responses and compromised anti-helminth immunity during Heligmosomoides polygyrus and Plasmodium chabaudi co-infection. Using newly established triple cytokine reporter mice (Il4gfpIfngyfpIl17aFP635), we demonstrated that Il4gfp+ Th2 cells purified from in vitro cultures or isolated ex vivo from helminth-infected mice up-regulated IFNγ following adoptive transfer into Rag1-/- mice infected with P. chabaudi. Functionally, Th2 cells that up-regulated IFNγ were transcriptionally re-wired and protected recipient mice from high parasitemia. Mechanistically, TCR stimulation and responsiveness to IL-12 and IFNγ, but not type I IFN, was required for optimal IFNγ production by Th2 cells. Finally, blockade of IL-12 and IFNγ during co-infection partially preserved anti-helminth Th2 responses. In summary, this study demonstrates that Th2 cells retain substantial plasticity with the ability to produce IFNγ during Plasmodium infection. Consequently, co-infection with Plasmodium spp. may contribute to the chronicity of helminth infection by reducing anti-helminth Th2 cells and converting them into IFNγ-secreting cells.


Assuntos
Coinfecção/imunologia , Interferon gama/metabolismo , Interleucina-12/imunologia , Malária/imunologia , Infecções por Strongylida/imunologia , Células Th2/imunologia , Transferência Adotiva , Animais , Separação Celular , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Interferon gama/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Nematospiroides dubius/imunologia , Plasmodium chabaudi/imunologia , Reação em Cadeia da Polimerase
9.
PLoS One ; 9(8): e104341, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25105503

RESUMO

InterFeron Inducible TransMembrane proteins 1-3 (IFITM1, IFITM2 and IFITM3) are a family of proteins capable of inhibiting the cellular entry of numerous human and animal viruses. IFITM1-3 are unique amongst the currently described viral restriction factors in their apparent ability to block viral entry. This restrictive property is dependant on the localisation of the proteins to plasma and endosomal membranes, which constitute the main portals of viral entry into cells. The topology of the IFITM proteins within cell membranes is an unresolved aspect of their biology. Here we present data from immunofluorescence microscopy, protease cleavage, biotin-labelling and immuno-electron microscopy assays, showing that human IFITM1 has a membrane topology in which the N-terminal domain resides in the cytoplasm, and the C-terminal domain is extracellular. Furthermore, we provide evidence that this topology is conserved for all of the human interferon-induced IFITM proteins. This model is consistent with that recently proposed for murine IFITM3, but differs from that proposed for murine IFITM1.


Assuntos
Antígenos de Diferenciação/química , Antígenos de Diferenciação/ultraestrutura , Modelos Moleculares , Western Blotting , Linhagem Celular Tumoral , Primers do DNA/genética , Citometria de Fluxo , Imunofluorescência , Células HEK293 , Humanos , Processamento de Imagem Assistida por Computador , Microscopia Eletrônica de Transmissão , Conformação Proteica , Reação em Cadeia da Polimerase em Tempo Real
10.
Proc Natl Acad Sci U S A ; 111(30): E3081-90, 2014 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-25024218

RESUMO

Allergic diseases, orchestrated by hyperactive CD4(+) Th2 cells, are some of the most common global chronic diseases. Therapeutic intervention relies upon broad-scale corticosteroids with indiscriminate impact. To identify targets in pathogenic Th2 cells, we took a comprehensive approach to identify the microRNA (miRNA) and mRNA transcriptome of highly purified cytokine-expressing Th1, Th2, Th9, Th17, and Treg cells both generated in vitro and isolated ex vivo from allergy, infection, and autoimmune disease models. We report here that distinct regulatory miRNA networks operate to regulate Th2 cells in house dust mite-allergic or helminth-infected animals and in vitro Th2 cells, which are distinguishable from other T cells. We validated several miRNA (miR) candidates (miR-15a, miR-20b, miR-146a, miR-155, and miR-200c), which targeted a suite of dynamically regulated genes in Th2 cells. Through in-depth studies using miR-155(-/-) or miR-146a(-/-) T cells, we identified that T-cell-intrinsic miR-155 was required for type-2 immunity, in part through regulation of S1pr1, whereas T-cell-intrinsic miR-146a was required to prevent overt Th1/Th17 skewing. These data identify miR-155, but not miR-146a, as a potential therapeutic target to alleviate Th2-medited inflammation and allergy.


Assuntos
Helmintíase Animal/imunologia , Hipersensibilidade/imunologia , MicroRNAs/imunologia , Células Th2/imunologia , Animais , Perfilação da Expressão Gênica , Helmintíase Animal/genética , Helmintíase Animal/patologia , Hipersensibilidade/genética , Hipersensibilidade/patologia , Camundongos , Camundongos Knockout , MicroRNAs/genética , Pyroglyphidae/imunologia , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/imunologia , Receptores de Esfingosina-1-Fosfato , Células Th1/imunologia , Células Th1/patologia , Células Th17/imunologia , Células Th17/patologia , Células Th2/patologia
11.
Immunity ; 41(1): 89-103, 2014 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-25035954

RESUMO

Foxp3(+) T regulatory (Treg) cells prevent inflammatory disease but the mechanistic basis of suppression is not understood completely. Gene silencing by RNA interference can act in a cell-autonomous and non-cell-autonomous manner, providing mechanisms of intercellular regulation. Here, we demonstrate that non-cell-autonomous gene silencing, mediated by miRNA-containing exosomes, is a mechanism employed by Treg cells to suppress T-cell-mediated disease. Treg cells transferred microRNAs (miRNA) to various immune cells, including T helper 1 (Th1) cells, suppressing Th1 cell proliferation and cytokine secretion. Use of Dicer-deficient or Rab27a and Rab27b double-deficient Treg cells to disrupt miRNA biogenesis or the exosomal pathway, respectively, established a requirement for miRNAs and exosomes for Treg-cell-mediated suppression. Transcriptional analysis and miRNA inhibitor studies showed that exosome-mediated transfer of Let-7d from Treg cell to Th1 cells contributed to suppression and prevention of systemic disease. These studies reveal a mechanism of Treg-cell-mediated suppression mediated by miRNA-containing exosomes.


Assuntos
Exossomos/genética , MicroRNAs/imunologia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Animais , Antígenos CD19/imunologia , Linfócitos B/imunologia , Proliferação de Células , Citocinas/metabolismo , RNA Helicases DEAD-box/genética , Exossomos/imunologia , Exossomos/metabolismo , Feminino , Fatores de Transcrição Forkhead/imunologia , Transferência Genética Horizontal/genética , Inflamação/imunologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/biossíntese , MicroRNAs/genética , Interferência de RNA , Ribonuclease III/genética , Células Th17/imunologia , Proteínas rab de Ligação ao GTP/genética , Proteínas rab27 de Ligação ao GTP
13.
PLoS Pathog ; 9(6): e1003451, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23825948

RESUMO

A diverse suite of effector immune responses provide protection against various pathogens. However, the array of effector responses must be immunologically regulated to limit pathogen- and immune-associated damage. CD4(+)Foxp3(+) regulatory T cells (Treg) calibrate immune responses; however, how Treg cells adapt to control different effector responses is unclear. To investigate the molecular mechanism of Treg diversity we used whole genome expression profiling and next generation small RNA sequencing of Treg cells isolated from type-1 or type-2 inflamed tissue following Leishmania major or Schistosoma mansoni infection, respectively. In-silico analyses identified two miRNA "regulatory hubs" miR-10a and miR-182 as critical miRNAs in Th1- or Th2-associated Treg cells, respectively. Functionally and mechanistically, in-vitro and in-vivo systems identified that an IL-12/IFNγ axis regulated miR-10a and its putative transcription factor, Creb. Importantly, reduced miR-10a in Th1-associated Treg cells was critical for Treg function and controlled a suite of genes preventing IFNγ production. In contrast, IL-4 regulated miR-182 and cMaf in Th2-associed Treg cells, which mitigated IL-2 secretion, in part through repression of IL2-promoting genes. Together, this study indicates that CD4(+)Foxp3(+) cells can be shaped by local environmental factors, which orchestrate distinct miRNA pathways preserving Treg stability and suppressor function.


Assuntos
Leishmania major/imunologia , Leishmaniose Cutânea/imunologia , MicroRNAs/imunologia , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Linfócitos T Reguladores/imunologia , Animais , Citocinas/genética , Citocinas/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/parasitologia , Inflamação/patologia , Leishmaniose Cutânea/genética , Leishmaniose Cutânea/patologia , Camundongos , Camundongos Knockout , MicroRNAs/genética , Esquistossomose mansoni/genética , Esquistossomose mansoni/patologia , Linfócitos T Reguladores/patologia , Células Th1/imunologia , Células Th1/patologia , Células Th2/imunologia , Células Th2/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...