Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 153
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Microbiol Spectr ; 9(3): e0039921, 2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34878295

RESUMO

Escherichia coli K1 causes bacteremia and meningitis in human neonates. The K1 capsule, an α2,8-linked polysialic acid (PSA) homopolymer, is its essential virulence factor. PSA is usually partially modified by O-acetyl groups. It is known that O-acetylation alters the antigenicity of PSA, but its impact on the interactions between E. coli K1 and host cells is unclear. In this study, a phase variant was obtained by passage of E. coli K1 parent strain, which expressed a capsule with 44% O-acetylation whereas the capsule of the parent strain has only 3%. The variant strain showed significantly reduced adherence and invasion to macrophage-like cells in comparison to the parent strain. Furthermore, we found that O-acetylation of PSA enhanced the modulation of trafficking of E. coli-containing vacuoles (ECV), enabling them to avoid fusing with lysosomes in these cells. Intriguingly, by using quartz crystal microbalance, we demonstrated that the PSA purified from the parent strain interacted with human sialic acid-binding immunoglobulin-like lectins (Siglecs), including Siglec-5, Siglec-7, Siglec-11, and Siglec-14. However, O-acetylated PSA from the variant interacted much less and also suppressed the production of Siglec-mediated proinflammatory cytokines. The adherence of the parent strain to human macrophage-like cells was significantly blocked by monoclonal antibodies against Siglec-11 and Siglec-14. Furthermore, the variant strain caused increased bacteremia and higher lethality in neonatal mice compared to the parent strain. These data elucidate that O-acetylation of K1 capsule enables E. coli to escape from Siglec-mediated innate immunity and lysosomal degradation; therefore, it is a strategy used by E. coli K1 to regulate its virulence. IMPORTANCE Escherichia coli K1 is a leading cause of neonatal meningitis. The mortality and morbidity of this disease remain significantly high despite antibiotic therapy. One major limitation on advances in prevention and therapy for meningitis is an incomplete understanding of its pathogenesis. E. coli K1 is surrounded by PSA, which is observed to have high-frequency variation of O-acetyl modification. Here, we present an in-depth study of the function of O-acetylation in PSA at each stage of host-pathogen interaction. We found that a high level of O-acetylation significantly interfered with Siglec-mediated bacterial adherence to macrophage-like cells, and blunted the proinflammatory response. Furthermore, the O-acetylation of PSA modulated the trafficking of ECVs to prevent them from fusing with lysosomes, enabling them to escape degradation by lysozymes within these cells. Elucidating how subtle modification of the capsule enhances bacterial defenses against host innate immunity will enable the future development of effective drugs or vaccines against infection by E. coli K1.


Assuntos
Cápsulas Bacterianas/imunologia , Infecções por Escherichia coli/imunologia , Escherichia coli/imunologia , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/imunologia , Ácidos Siálicos/imunologia , Acetilação , Animais , Escherichia coli/genética , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Evasão da Resposta Imune , Imunidade Inata , Lisossomos/imunologia , Lisossomos/microbiologia , Masculino , Camundongos , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/genética , Vacúolos/imunologia , Vacúolos/microbiologia
2.
J Exp Med ; 218(11)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34670268

RESUMO

The intracellular parasite Toxoplasma gondii has long provided a tractable experimental system to investigate how the immune system deals with intracellular infections. This review highlights the advances in defining how this organism was first detected and the studies with T. gondii that contribute to our understanding of how the cytokine IFN-γ promotes control of vacuolar pathogens. In addition, the genetic tractability of this eukaryote organism has provided the foundation for studies into the diverse strategies that pathogens use to evade antimicrobial responses and now provides the opportunity to study the basis for latency. Thus, T. gondii remains a clinically relevant organism whose evolving interactions with the host immune system continue to teach lessons broadly relevant to host-pathogen interactions.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Toxoplasma/imunologia , Animais , Controle de Doenças Transmissíveis/métodos , Citocinas/imunologia , Humanos , Interferon gama/imunologia , Vacúolos/imunologia , Vacúolos/parasitologia
3.
Biosci Rep ; 41(9)2021 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-34558607

RESUMO

Vacuoles are membrane vesicles in eukaryotic cells, the digestive system of cells that break down substances absorbed outside the cell and digest the useless components of the cell itself. Researches on anticancer and intractable diseases using vacuoles are being actively conducted. The practical application of the present study to animals requires the determination of the biocompatibility of vacuole. In the present study, we evaluated the effects of vacuoles isolated from Saccharomyces cerevisiae in RAW 264.7 cells. This showed a significant increase in the production of nitric oxide (NO) produced by macrophage activity. Using Reactive Oxygen Species (ROS) assay, we identified that ROS is increased in a manner dependent on vacuole concentration. Western blot analysis showed that vacuole concentration-dependently increased protein levels of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2). Therefore, iNOS expression was stimulated to induce NO production. In addition, pro-inflammatory cytokines levels promoted, such as interleukin (IL) 6 (IL-6) and tumor necrosis factor (TNF) α (TNF-α). In summary, vacuoles activate the immune response of macrophages by promoting the production of immune-mediated transporters NO, ROS, and pro-inflammatory cytokines.


Assuntos
Mediadores da Inflamação/metabolismo , Ativação de Macrófagos , Macrófagos/imunologia , Saccharomyces cerevisiae/imunologia , Vacúolos/imunologia , Animais , Ciclo-Oxigenase 2/metabolismo , Citocinas/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Células RAW 264.7 , Espécies Reativas de Oxigênio/metabolismo , Saccharomyces cerevisiae/metabolismo , Vacúolos/metabolismo , Vacúolos/transplante
5.
PLoS Pathog ; 16(8): e1008327, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32853276

RESUMO

Host resistance to Toxoplasma gondii relies on CD8 T cell IFNγ responses, which if modulated by the host or parasite could influence chronic infection and parasite transmission between hosts. Since host-parasite interactions that govern this response are not fully elucidated, we investigated requirements for eliciting naïve CD8 T cell IFNγ responses to a vacuolar resident antigen of T. gondii, TGD057. Naïve TGD057 antigen-specific CD8 T cells (T57) were isolated from transnuclear mice and responded to parasite-infected bone marrow-derived macrophages (BMDMs) in an antigen-dependent manner, first by producing IL-2 and then IFNγ. T57 IFNγ responses to TGD057 were independent of the parasite's protein export machinery ASP5 and MYR1. Instead, host immunity pathways downstream of the regulatory Immunity-Related GTPases (IRG), including partial dependence on Guanylate-Binding Proteins, are required. Multiple T. gondii ROP5 isoforms and allele types, including 'avirulent' ROP5A from clade A and D parasite strains, were able to suppress CD8 T cell IFNγ responses to parasite-infected BMDMs. Phenotypic variance between clades B, C, D, F, and A strains suggest T57 IFNγ differentiation occurs independently of parasite virulence or any known IRG-ROP5 interaction. Consistent with this, removal of ROP5 is not enough to elicit maximal CD8 T cell IFNγ production to parasite-infected cells. Instead, macrophage expression of the pathogen sensors, NLRP3 and to a large extent NLRP1, were absolute requirements. Other members of the conventional inflammasome cascade are only partially required, as revealed by decreased but not abrogated T57 IFNγ responses to parasite-infected ASC, caspase-1/11, and gasdermin D deficient cells. Moreover, IFNγ production was only partially reduced in the absence of IL-12, IL-18 or IL-1R signaling. In summary, T. gondii effectors and host machinery that modulate parasitophorous vacuolar membranes, as well as NLR-dependent but inflammasome-independent pathways, determine the full commitment of CD8 T cells IFNγ responses to a vacuolar antigen.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Inflamassomos/imunologia , Interferon gama/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteínas de Protozoários/metabolismo , Transdução de Sinais , Toxoplasma/imunologia , Toxoplasmose Animal/imunologia , Animais , Linfócitos T CD8-Positivos/parasitologia , Feminino , Macrófagos/imunologia , Macrófagos/parasitologia , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteínas de Protozoários/genética , Toxoplasmose Animal/parasitologia , Vacúolos/imunologia , Vacúolos/metabolismo , Vacúolos/parasitologia , Virulência/imunologia
6.
Cell Rep ; 32(6): 108008, 2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32783936

RESUMO

Interferon-inducible guanylate-binding proteins (GBPs) promote cell-intrinsic defense through host cell death. GBPs target pathogens and pathogen-containing vacuoles and promote membrane disruption for release of microbial molecules that activate inflammasomes. GBP1 mediates pyroptosis or atypical apoptosis of Salmonella Typhimurium (STm)- or Toxoplasma gondii (Tg)- infected human macrophages, respectively. The pathogen-proximal detection-mechanisms of GBP1 remain poorly understood, as humans lack functional immunity-related GTPases (IRGs) that assist murine Gbps. Here, we establish that GBP1 promotes the lysis of Tg-containing vacuoles and parasite plasma membranes, releasing Tg-DNA. In contrast, we show GBP1 targets cytosolic STm and recruits caspase-4 to the bacterial surface for its activation by lipopolysaccharide (LPS), but does not contribute to bacterial vacuole escape. Caspase-1 cleaves and inactivates GBP1, and a cleavage-deficient GBP1D192E mutant increases caspase-4-driven pyroptosis due to the absence of feedback inhibition. Our studies elucidate microbe-specific roles of GBP1 in infection detection and its triggering of the assembly of divergent caspase signaling platforms.


Assuntos
Caspases/imunologia , Proteínas de Ligação ao GTP/imunologia , Salmonella typhimurium/imunologia , Toxoplasma/imunologia , Morte Celular/imunologia , Células HEK293 , Humanos , Inflamassomos/imunologia , Interferon gama/farmacologia , Ligantes , Infecções por Salmonella/imunologia , Infecções por Salmonella/microbiologia , Células THP-1 , Toxoplasma/genética , Toxoplasmose/imunologia , Toxoplasmose/microbiologia , Vacúolos/imunologia
7.
Infect Immun ; 88(8)2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32482642

RESUMO

Xenophagy targets intracellular pathogens for destruction by the host autophagy pathway. Ubiquitin chains are conjugated to xenophagic targets and recruit multiple autophagy adaptors. The intracellular pathogen Legionella pneumophila resides in a vacuole that is ubiquitinated; however, this pathogen avoids xenophagic detection. Here, the mechanisms by which L. pneumophila can prevent the host xenophagy pathway from targeting the vacuole in which it resides were examined. Ubiquitin-labeled vacuoles containing L. pneumophila failed to recruit autophagy adaptors by a process that was independent of RavZ function. Coinfection studies were conducted using a strain of Listeria monocytogenes that served as a robust xenophagic target. Legionella pneumophila infection blocked xenophagic targeting of L. monocytogenes by a RavZ-dependent mechanism. Importantly, when coinfection studies were conducted with a RavZ-deficient strain of L. pneumophila, L. monocytogenes was targeted by the host xenophagy system but vacuoles containing L. pneumophila avoided targeting. Enhanced adaptor recruitment to the vacuole was observed by using a strain of L. pneumophila in which all of the effector proteins in the SidE family were deleted; however, this strain was still not targeted by the host autophagy pathway. Thus, there are at least two pathways by which L. pneumophila can disrupt xenophagic targeting of the vacuole in which it resides. One mechanism involves global disruption of the host autophagy machinery by the effector protein RavZ. A second cis-acting mechanism prevents the binding of autophagy adaptors to the ubiquitin-decorated surface of the L. pneumophila-containing vacuole.


Assuntos
Proteínas de Bactérias/genética , Interações Hospedeiro-Patógeno/genética , Legionella pneumophila/genética , Macrófagos/microbiologia , Sistemas de Secreção Tipo IV/genética , Vacúolos/microbiologia , Animais , Autofagia , Proteínas de Bactérias/imunologia , Células CHO , Cricetulus , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/imunologia , Humanos , Legionella pneumophila/imunologia , Listeria monocytogenes/genética , Listeria monocytogenes/imunologia , Macrófagos/imunologia , Camundongos , Proteína Sequestossoma-1/genética , Proteína Sequestossoma-1/imunologia , Coloração e Rotulagem/métodos , Sistemas de Secreção Tipo IV/imunologia , Ubiquitina/genética , Ubiquitina/imunologia , Vacúolos/imunologia
8.
Methods Mol Biol ; 2136: 223-231, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32430824

RESUMO

Group A Streptococcus (GAS) is one of the major human pathogens that can invade nonphagocytic cells. GAS internalized through endocytosis secretes the pore-forming toxin Streptolysin O (SLO) to escape into the cytoplasm. The cytosolic GAS is selectively captured by autophagic membranes (GAS-containing autophagosome-like vacuoles, GcAVs) and delivered to lysosomes for degradation. Macroautophagy (referred to as autophagy hereafter) is a highly conserved lysosome-mediated catabolic process, which is critical for cellular homeostasis. Autophagy also acts as an intracellular immune system. In this section, we describe how to identify GcAVs in infected cells using fluorescent microscopy.


Assuntos
Autofagossomos/metabolismo , Streptococcus pyogenes/imunologia , Vacúolos/imunologia , Autofagossomos/imunologia , Autofagia/imunologia , Proteínas de Bactérias/metabolismo , Células HeLa , Humanos , Lisossomos/metabolismo , Fagossomos/metabolismo , Transporte Proteico , Infecções Estreptocócicas/imunologia , Estreptolisinas/metabolismo , Vacúolos/metabolismo
9.
Nature ; 582(7810): 104-108, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32427965

RESUMO

Malaria caused by Plasmodium falciparum remains the leading single-agent cause of mortality in children1, yet the promise of an effective vaccine has not been fulfilled. Here, using our previously described differential screening method to analyse the proteome of blood-stage P. falciparum parasites2, we identify P. falciparum glutamic-acid-rich protein (PfGARP) as a parasite antigen that is recognized by antibodies in the plasma of children who are relatively resistant-but not those who are susceptible-to malaria caused by P. falciparum. PfGARP is a parasite antigen of 80 kDa that is expressed on the exofacial surface of erythrocytes infected by early-to-late-trophozoite-stage parasites. We demonstrate that antibodies against PfGARP kill trophozoite-infected erythrocytes in culture by inducing programmed cell death in the parasites, and that vaccinating non-human primates with PfGARP partially protects against a challenge with P. falciparum. Furthermore, our longitudinal cohort studies showed that, compared to individuals who had naturally occurring anti-PfGARP antibodies, Tanzanian children without anti-PfGARP antibodies had a 2.5-fold-higher risk of severe malaria and Kenyan adolescents and adults without these antibodies had a twofold-higher parasite density. By killing trophozoite-infected erythrocytes, PfGARP could synergize with other vaccines that target parasite invasion of hepatocytes or the invasion of and egress from erythrocytes.


Assuntos
Apoptose/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Parasitos/imunologia , Plasmodium falciparum/citologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Adolescente , Adulto , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/química , Antígenos de Protozoários/imunologia , Aotidae/imunologia , Aotidae/parasitologia , Caspases/metabolismo , Criança , Estudos de Coortes , DNA de Protozoário/química , DNA de Protozoário/metabolismo , Ativação Enzimática , Eritrócitos/parasitologia , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/química , Quênia , Vacinas Antimaláricas/imunologia , Malária Falciparum/parasitologia , Masculino , Camundongos , Parasitos/citologia , Parasitos/crescimento & desenvolvimento , Plasmodium falciparum/crescimento & desenvolvimento , Proteínas de Protozoários/química , Tanzânia , Trofozoítos/citologia , Trofozoítos/crescimento & desenvolvimento , Trofozoítos/imunologia , Vacúolos/imunologia
10.
Infect Immun ; 88(5)2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32094251

RESUMO

Gamma interferon (IFN-γ)-induced innate immune responses play important roles in the inhibition of Toxoplasma gondii infection. It has been reported that IFN-γ stimulates non-acidification-dependent growth restriction of T. gondii in HeLa cells, but the mechanism remains unclear. Here, we found that γ-aminobutyric acid (GABA) receptor-associated protein-like 2 (GABARAPL2) plays a critical role in parasite restriction in IFN-γ-treated HeLa cells. GABARAPL2 is recruited to membrane structures surrounding parasitophorous vacuoles (PV). Autophagy adaptors are required for the proper localization and function of GABARAPL2 in the IFN-γ -induced immune response. These findings provide further understanding of a noncanonical autophagy pathway responsible for IFN-γ-dependent inhibition of T. gondii growth in human HeLa cells and demonstrate the critical role of GABARAPL2 in this response.


Assuntos
Família da Proteína 8 Relacionada à Autofagia/imunologia , Interferon gama/imunologia , Toxoplasma/imunologia , Toxoplasmose/imunologia , Autofagia/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Células HeLa , Humanos , Imunidade Inata/imunologia , Vacúolos/imunologia
11.
Science ; 367(6475): 301-305, 2020 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-31806695

RESUMO

Despite ongoing (macro)pinocytosis of extracellular fluid, the volume of the endocytic pathway remains unchanged. To investigate the underlying mechanism, we used high-resolution video imaging to analyze the fate of macropinosomes formed by macrophages in vitro and in situ. Na+, the primary cationic osmolyte internalized, exited endocytic vacuoles via two-pore channels, accompanied by parallel efflux of Cl- and osmotically coupled water. The resulting shrinkage caused crenation of the membrane, which fostered recruitment of curvature-sensing proteins. These proteins stabilized tubules and promoted their elongation, driving vacuolar remodeling, receptor recycling, and resolution of the organelles. Failure to resolve internalized fluid impairs the tissue surveillance activity of resident macrophages. Thus, osmotically driven increases in the surface-to-volume ratio of endomembranes promote traffic between compartments and help to ensure tissue homeostasis.


Assuntos
Vigilância Imunológica , Macrófagos/imunologia , Pinocitose/imunologia , Animais , Canais de Cálcio/genética , Canais de Cálcio/fisiologia , Endossomos/imunologia , Transporte de Íons , Lipídeos/imunologia , Camundongos , Camundongos Knockout , Organelas/imunologia , Osmose , Sódio/metabolismo , Canais de Potencial de Receptor Transitório/genética , Vacúolos/imunologia
12.
Exp Cell Res ; 386(2): 111727, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31759054

RESUMO

Following mating, leukocytes are recruited to the uterine epithelium where they phagocytose spermatozoa and mediate maternal immune tolerance as well as a mild inflammatory response. In this ultrastructural study we utilised array tomography, a high-resolution volume scanning electron microscopy approach to 3D reconstruct the cellular relationships formed by leukocytes recruited to the luminal uterine epithelium 12 h post-mating in the rat. We report that following mating, neutrophils and macrophages are internalised by the luminal uterine epithelium, with multiple leukocytes internalised via contortion through a small tunnel in the apical membrane into a large membrane-bound vacuole within the cytoplasm of luminal uterine epithelial cells (UECs). Once internalised within the UECs, recruited leukocytes appear to phagocytose material within the membrane-bound vacuole and most ultimately undergo a specialised cell death, including vacuolisation and loss of membrane integrity. As these observations involve ultrastructurally normal leukocytic cells internalised within non-phagocytic epithelial cells, these observations are consistent with the formation of cell-in-cell structures via entosis, rather than phagocytic engulfment by UECs. Although cell-in-cell structures have been reported in normal and pathological conditions elsewhere, the data collected herein represents the first evidence of the formation of cell-in-cell structures within the uterine epithelium as a novel component of the maternal inflammatory response to mating.


Assuntos
Copulação/fisiologia , Entose/imunologia , Células Epiteliais/ultraestrutura , Epitélio/ultraestrutura , Leucócitos/ultraestrutura , Útero/citologia , Animais , Morte Celular , Células Epiteliais/imunologia , Epitélio/imunologia , Feminino , Tolerância Imunológica , Leucócitos/imunologia , Masculino , Fagocitose , Gravidez , Ratos , Ratos Wistar , Espermatozoides/citologia , Espermatozoides/imunologia , Útero/imunologia , Vacúolos/imunologia , Vacúolos/ultraestrutura
13.
Methods Mol Biol ; 2071: 371-409, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31758463

RESUMO

Critical steps in resistance of mice against Toxoplasma gondii occur in the first 2 or 3 h after the pathogen has entered a cell that has been exposed to interferon γ (IFNγ). The newly formed parasitophorous vacuole is attacked by the IFNγ-inducible IRG proteins and disrupted, resulting in death of the parasite and necrotic death of the cell. Here we describe some techniques that we have used to describe and quantify these events in different combinations of the host and the parasite.


Assuntos
Imunidade Inata/fisiologia , Interferon gama/metabolismo , Toxoplasma/imunologia , Toxoplasma/patogenicidade , Animais , Linhagem Celular , Células Cultivadas , Citometria de Fluxo , Imunofluorescência , Proteína HMGB1/metabolismo , Imunidade Inata/genética , Camundongos , Plasmídeos/genética , Vacúolos/imunologia , Vacúolos/metabolismo
14.
Plant Mol Biol ; 101(4-5): 343-354, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31621005

RESUMO

KEY MESSAGE: Short review focussing on the role and targeting of vacuolar substructure in plant immunity and pathogenesis. Plants lack specialized immune cells, therefore each plant cell must defend itself against invading pathogens. A typical plant defense strategy is the hypersensitive response that results in host cell death at the site of infection, a process largely regulated by the vacuole. In plant cells, the vacuole is a vital organelle that plays a central role in numerous fundamental processes, such as development, reproduction, and cellular responses to biotic and abiotic stimuli. It shows divergent membranous structures that are continuously transforming. Recent technical advances in visualization and live-cell imaging have significantly altered our view of the vacuolar structures and their dynamics. Understanding the active nature of the vacuolar structures and the mechanisms of vacuole-mediated defense responses is of great importance in understanding plant-pathogen interactions. In this review, we present an overview of the current knowledge about the vacuole and its internal structures, as well as their role in plant-microbe interactions. There is so far limited information on the modulation of the vacuolar structures by pathogens, but recent research has identified the vacuole as a possible target of microbial interference.


Assuntos
Interações Hospedeiro-Patógeno , Imunidade Vegetal , Plantas/ultraestrutura , Vacúolos/ultraestrutura , Biomarcadores/metabolismo , Morte Celular , Membranas Intracelulares/imunologia , Membranas Intracelulares/microbiologia , Membranas Intracelulares/ultraestrutura , Proteínas de Plantas/metabolismo , Plantas/imunologia , Plantas/microbiologia , Vacúolos/imunologia , Vacúolos/microbiologia
15.
Front Immunol ; 10: 2104, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31555296

RESUMO

Toxoplasma gondii secretes rhoptry (ROP) and dense granule (GRA) effector proteins to evade host immune clearance mediated by interferon gamma (IFN-γ), immunity-related GTPase (IRG) effectors, and CD8+ T cells. Here, we investigated the role of parasite-secreted effectors in regulating host access to parasitophorous vacuole (PV) localized parasite antigens and their presentation to CD8+ T cells by the major histocompatibility class I (MHC-I) pathway. Antigen presentation of PV localized parasite antigens by MHC-I was significantly increased in macrophages and/or dendritic cells infected with mutant parasites that lacked expression of secreted GRA (GRA2, GRA3, GRA4, GRA5, GRA7, GRA12) or ROP (ROP5, ROP18) effectors. The ability of various secreted GRA or ROP effectors to suppress antigen presentation by MHC-I was dependent on cell type, expression of IFN-γ, or host IRG effectors. The suppression of antigen presentation by ROP5, ROP18, and GRA7 correlated with a role for these molecules in preventing PV disruption by IFN-γ-activated host IRG effectors. However, GRA2 mediated suppression of antigen presentation was not correlated with PV disruption. In addition, the GRA2 antigen presentation phenotypes were strictly co-dependent on the expression of the GRA6 protein. These results show that MHC-I antigen presentation of PV localized parasite antigens was controlled by mechanisms that were dependent or independent of IRG effector mediated PV disruption. Our findings suggest that the GRA6 protein underpins an important mechanism that enhances CD8+ T cell recognition of parasite-infected cells with damaged or ruptured PV membranes. However, in intact PVs, parasite secreted effector proteins that associate with the PV membrane or the intravacuolar network membranes play important roles to actively suppress antigen presentation by MHC-I to reduce CD8+ T cell recognition and clearance of Toxoplasma gondii infected host cells.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos de Protozoários/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Toxoplasmose Animal/imunologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Protozoários/imunologia , Toxoplasma/imunologia , Vacúolos/imunologia
16.
Transfus Clin Biol ; 26(4): 346-351, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30797678

RESUMO

Presentation of exogenous antigens loaded on major histocompatibility complex class I molecules by antigen presenting cells, termed cross-presentation, is essential for the induction of CD8+ T cells and is performed mainly by specialized dendritic cell subsets. Research into this field has described two main mechanisms of cross-presentation, the cytosolic pathway and the vacuolar pathway. As the first step in cross-presentation, surface receptors relating to cross-presentation are required in the recognition and uptake of Ags, which include C-type lectin receptors, immunoglobulin γ Fc region receptor, chemokine receptor, scavenger receptor etc. After uptake by the cells, there are also many molecules that enable Ags to participate in cross-presentation pathways. By this approach, exogenous Ags can induce CD8+ T cells into cytotoxic T lymphocytes, which is of great significance to induce antitumor and antiviral immune responses, and the molecular mechanism would facilitate the development of related adjuvants. However, the detailed mechanisms of cross-presentation still remain unknown. In this paper, some latest researches, including two major pathways, DC surface receptors and application prospects are summarized.


Assuntos
Apresentação de Antígeno , Antígenos Heterófilos/imunologia , Apresentação Cruzada , Isoantígenos/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Citosol/imunologia , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Lipoproteínas HDL/imunologia , Camundongos , Modelos Imunológicos , Receptores Fc/imunologia , Receptores Imunológicos/imunologia , Receptores de Lipoproteínas/imunologia , Vacúolos/imunologia
17.
Methods Mol Biol ; 1880: 679-690, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30610731

RESUMO

Bacteria that escape from membrane-enclosed vacuoles to the cytosol of cells are targeted by autophagy, which recognizes and captures bacteria into autophagosomes wherein their proliferation is restricted. Here we discuss two means by which antibacterial autophagy is assessed: (1) the visualization and enumeration of autophagy protein recruitment to the vicinity of cytosolic bacteria by means of immunofluorescence microscopy and (2) the measurement of autophagy-dependent restriction of bacterial proliferation by means of colony-forming unit assay.


Assuntos
Autofagossomos/imunologia , Autofagia/imunologia , Bioensaio/métodos , Interações Hospedeiro-Patógeno/imunologia , Autofagossomos/microbiologia , Bioensaio/instrumentação , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Contagem de Colônia Microbiana/instrumentação , Contagem de Colônia Microbiana/métodos , Células HeLa , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microscopia de Fluorescência/instrumentação , Microscopia de Fluorescência/métodos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Interferência de RNA , Salmonella typhimurium/genética , Salmonella typhimurium/imunologia , Salmonella typhimurium/isolamento & purificação , Transformação Bacteriana , Vacúolos/imunologia , Vacúolos/microbiologia
18.
Nat Commun ; 9(1): 2570, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29967419

RESUMO

Presentation of exogenous antigens on MHC-I molecules, termed cross-presentation, is essential for cytotoxic CD8+ T cell responses. In mice, dendritic cells (DCs) that arise from monocytes (mo-DCs) during inflammation have a key function in these responses by cross-presenting antigens locally in peripheral tissues. Whether human naturally-occurring mo-DCs can cross-present is unknown. Here, we use human mo-DCs and macrophages directly purified from ascites to address this question. Single-cell RNA-seq data show that ascites CD1c+ DCs contain exclusively monocyte-derived cells. Both ascites mo-DCs and monocyte-derived macrophages cross-present efficiently, but are inefficient for transferring exogenous proteins into their cytosol. Inhibition of cysteine proteases, but not of proteasome, abolishes cross-presentation in these cells. We conclude that human monocyte-derived cells cross-present exclusively using a vacuolar pathway. Finally, only ascites mo-DCs provide co-stimulatory signals to induce effector cytotoxic CD8+ T cells. Our findings thus provide important insights on how to harness cross-presentation for therapeutic purposes.


Assuntos
Apresentação de Antígeno , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Vacúolos/metabolismo , Antígenos CD1/metabolismo , Ascite/etiologia , Ascite/imunologia , Buffy Coat/citologia , Técnicas de Cultura de Células , Células Cultivadas , Citosol/metabolismo , Células Dendríticas/metabolismo , Feminino , Perfilação da Expressão Gênica , Glicoproteínas/metabolismo , Voluntários Saudáveis , Humanos , Macrófagos/metabolismo , Masculino , Monócitos/metabolismo , Neoplasias Ovarianas/complicações , Neoplasias Ovarianas/imunologia , Tonsila Palatina/citologia , Análise de Sequência de RNA , Análise de Célula Única , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Vacúolos/imunologia
19.
Sci Rep ; 7(1): 5209, 2017 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-28701773

RESUMO

Interferon gamma (IFNγ) is the major proinflammatory cytokine conferring resistance to the intracellular vacuolar pathogen Toxoplasma gondii by inducing the destruction of the parasitophorous vacuole (PV). We previously identified TRIM21 as an IFNγ-driven E3 ubiquitin ligase mediating the deposition of ubiquitin around pathogen inclusions. Here, we show that TRIM21 knockout mice were highly susceptible to Toxoplasma infection, exhibiting decreased levels of serum inflammatory cytokines and higher parasite burden in the peritoneum and brain. We demonstrate that IFNγ drives recruitment of TRIM21 to GBP1-positive Toxoplasma vacuoles, leading to Lys63-linked ubiquitination of the vacuole and restriction of parasite early replication without interfering with vacuolar disruption. As seen in vivo, TRIM21 impacted the secretion of inflammatory cytokines. This study identifies TRIM21 as a previously unknown modulator of Toxoplasma gondii resistance in vivo thereby extending host innate immune recognition of eukaryotic pathogens to include E3 ubiquitin ligases.


Assuntos
Fibroblastos/parasitologia , Proteínas de Ligação ao GTP/metabolismo , Interações Hospedeiro-Parasita/imunologia , Macrófagos/parasitologia , Ribonucleoproteínas/fisiologia , Toxoplasmose/parasitologia , Vacúolos/parasitologia , Animais , Autofagia , Citocinas/metabolismo , Feminino , Fibroblastos/imunologia , Fibroblastos/metabolismo , Interferon gama/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Toxoplasma/patogenicidade , Toxoplasmose/imunologia , Toxoplasmose/metabolismo , Ubiquitina/metabolismo , Ubiquitinação , Vacúolos/imunologia , Vacúolos/metabolismo , Fatores de Virulência/metabolismo
20.
J Immunol ; 199(3): 992-1002, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28659356

RESUMO

Leishmania donovani infects macrophages, disrupting immune homeostasis. The underlying mechanism that sustains infection remains unresolved. In view of the potential of Wnt5a signaling to support immune homeostasis, we evaluated the interrelationship of Wnt5a signaling and Leishmania donovani infection. Upon infecting macrophages separately with antimony drug-sensitive and -resistant L. donovani, we noted disruption in the steady-state level of Wnt5a. Moreover, inhibition of Wnt5a signaling by small interfering RNA transfection in vitro or by use of inhibitor of Wnt production in vivo led to an increase in cellular parasite load. In contrast, treatment of macrophages with recombinant Wnt5a caused a decrease in the load of antimony-sensitive and -resistant parasites, thus confirming that Wnt5a signaling antagonizes L. donovani infection. Using inhibitors of the Wnt5a signaling intermediates Rac1 and Rho kinase, we demonstrated that Wnt5a-mediated inhibition of parasite infection in macrophages is Rac1/Rho dependent. Furthermore, phalloidin staining and reactive oxygen species estimation of Wnt5a-treated macrophages suggested that a Wnt5a-Rac/Rho-mediated decrease in parasite load is associated with an increase in F- actin assembly and NADPH oxidase activity. Moreover, live microscopy of L. donovani-infected macrophages treated with Wnt5a demonstrated increased endosomal/lysosomal fusions with parasite-containing vacuoles (parasitophorous vacuoles [PV]). An increase in PV-endosomal/lysosomal fusion accompanied by augmented PV degradation in Wnt5a-treated macrophages was also apparent from transmission electron microscopy of infected cells. Our results suggest that, although L. donovani evades host immune response, at least in part through inhibition of Wnt5a signaling, revamping Wnt5a signaling can inhibit L. donovani infection, irrespective of drug sensitivity or resistance.


Assuntos
Leishmania donovani/imunologia , Macrófagos/imunologia , Macrófagos/parasitologia , Proteína Wnt-5a/metabolismo , Actinas/metabolismo , Animais , Antimônio/farmacologia , Antiprotozoários/farmacologia , Leishmania donovani/efeitos dos fármacos , Leishmania donovani/fisiologia , Leishmaniose Visceral/imunologia , Macrófagos/ultraestrutura , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Transmissão , NADPH Oxidases/metabolismo , Neuropeptídeos/metabolismo , Carga Parasitária , Faloidina/química , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Transfecção , Vacúolos/imunologia , Vacúolos/parasitologia , Proteína Wnt-5a/genética , Proteína Wnt-5a/farmacologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Quinases Associadas a rho/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...