Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 17(7): e1008911, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34320028

RESUMO

In order to sustain a persistent infection, Mycobacterium tuberculosis (Mtb) must adapt to a changing environment that is shaped by the developing immune response. This necessity to adapt is evident in the flexibility of many aspects of Mtb metabolism, including a respiratory chain that consists of two distinct terminal cytochrome oxidase complexes. Under the conditions tested thus far, the bc1/aa3 complex appears to play a dominant role, while the alternative bd oxidase is largely redundant. However, the presence of two terminal oxidases in this obligate pathogen implies that respiratory requirements might change during infection. We report that the cytochrome bd oxidase is specifically required for resisting the adaptive immune response. While the bd oxidase was dispensable for growth in resting macrophages and the establishment of infection in mice, this complex was necessary for optimal fitness after the initiation of adaptive immunity. This requirement was dependent on lymphocyte-derived interferon gamma (IFNγ), but did not involve nitrogen and oxygen radicals that are known to inhibit respiration in other contexts. Instead, we found that ΔcydA mutants were hypersusceptible to the low pH encountered in IFNγ-activated macrophages. Unlike wild type Mtb, cytochrome bd-deficient bacteria were unable to sustain a maximal oxygen consumption rate (OCR) at low pH, indicating that the remaining cytochrome bc1/aa3 complex is preferentially inhibited under acidic conditions. Consistent with this model, the potency of the cytochrome bc1/aa3 inhibitor, Q203, is dramatically enhanced at low pH. This work identifies a critical interaction between host immunity and pathogen respiration that influences both the progression of the infection and the efficacy of potential new TB drugs.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Evasão da Resposta Imune/fisiologia , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Adaptação Fisiológica/fisiologia , Animais , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Mycobacterium tuberculosis/enzimologia
2.
PLoS Pathog ; 17(7): e1009720, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34280245

RESUMO

Hepatitis C virus (HCV) chronically infects 70 million people worldwide with an estimated annual disease-related mortality of 400,000. A vaccine could prevent spread of this pervasive human pathogen, but has proven difficult to develop, partly due to neutralizing antibody evasion mechanisms that are inherent features of the virus envelope glycoproteins, E1 and E2. A central actor is the E2 motif, hypervariable region 1 (HVR1), which protects several non-overlapping neutralization epitopes through an incompletely understood mechanism. Here, we show that introducing different HVR1-isolate sequences into cell-culture infectious JFH1-based H77 (genotype 1a) and J4 (genotype 1b) Core-NS2 recombinants can lead to severe viral attenuation. Culture adaptation of attenuated HVR1-swapped recombinants permitted us to identify E1/E2 substitutions at conserved positions both within and outside HVR1 that increased the infectivity of attenuated HVR1-swapped recombinants but were not adaptive for original recombinants. H77 recombinants with HVR1 from multiple other isolates consistently acquired substitutions at position 348 in E1 and position 385 in HVR1 of E2. Interestingly, HVR1-swapped J4 recombinants primarily acquired other substitutions: F291I (E1), F438V (E2), F447L/V/I (E2) and V710L (E2), indicating a different adaptation pathway. For H77 recombinants, the adaptive E1/E2 substitutions increased sensitivity to the neutralizing monoclonal antibodies AR3A and AR4A, whereas for J4 recombinants, they increased sensitivity to AR3A, while having no effect on sensitivity to AR4A. To evaluate effects of the substitutions on AR3A and AR4A binding, we performed ELISAs on extracted E1/E2 protein and performed immunoprecipitation of relevant viruses. However, extracted E1/E2 protein and immunoprecipitation of HCV particles only reproduced the neutralization phenotypes of the J4 recombinants. Finally, we found that the HVR1-swap E1/E2 substitutions decrease virus entry dependency on co-receptor SR-BI. Our study identifies E1/E2 positions that could be critical for intra-complex HVR1 interactions while emphasizing the need for developing novel tools for molecular studies of E1/E2 interactions.


Assuntos
Adaptação Fisiológica/fisiologia , Hepacivirus/fisiologia , Evasão da Resposta Imune/fisiologia , Proteínas do Envelope Viral/fisiologia , Linhagem Celular , Quimera , Células HEK293 , Hepacivirus/patogenicidade , Humanos , Internalização do Vírus
3.
PLoS Pathog ; 17(7): e1009729, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34237115

RESUMO

Rabies virus phosphoprotein (P protein) is a multifunctional protein that plays key roles in replication as the polymerase cofactor that binds to the complex of viral genomic RNA and the nucleoprotein (N protein), and in evading the innate immune response by binding to STAT transcription factors. These interactions are mediated by the C-terminal domain of P (PCTD). The colocation of these binding sites in the small globular PCTD raises the question of how these interactions underlying replication and immune evasion, central to viral infection, are coordinated and, potentially, coregulated. While direct data on the binding interface of the PCTD for STAT1 is available, the lack of direct structural data on the sites that bind N protein limits our understanding of this interaction hub. The PCTD was proposed to bind via two sites to a flexible loop of N protein (Npep) that is not visible in crystal structures, but no direct analysis of this interaction has been reported. Here we use Nuclear Magnetic Resonance, and molecular modelling to show N protein residues, Leu381, Asp383, Asp384 and phosphor-Ser389, are likely to bind to a 'positive patch' of the PCTD formed by Lys211, Lys214 and Arg260. Furthermore, in contrast to previous predictions we identify a single site of interaction on the PCTD by this Npep. Intriguingly, this site is proximal to the defined STAT1 binding site that includes Ile201 to Phe209. However, cell-based assays indicate that STAT1 and N protein do not compete for P protein. Thus, it appears that interactions critical to replication and immune evasion can occur simultaneously with the same molecules of P protein so that the binding of P protein to activated STAT1 can potentially occur without interrupting interactions involved in replication. These data suggest that replication complexes might be directly involved in STAT1 antagonism.


Assuntos
Evasão da Resposta Imune/fisiologia , Chaperonas Moleculares/metabolismo , Vírus da Raiva/metabolismo , Raiva/virologia , Proteínas Estruturais Virais/metabolismo , Replicação Viral/fisiologia , Animais , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Proteínas do Nucleocapsídeo/metabolismo , Raiva/metabolismo , Fator de Transcrição STAT1/metabolismo
4.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34035171

RESUMO

Immunoevasins are viral proteins that prevent antigen presentation on major histocompatibility complex (MHC) class I, thus evading host immune recognition. Hepatitis C virus (HCV) evades immune surveillance to induce chronic infection; however, how HCV-infected hepatocytes affect immune cells and evade immune recognition remains unclear. Herein, we demonstrate that HCV core protein functions as an immunoevasin. Its expression interfered with the maturation of MHC class I molecules catalyzed by the signal peptide peptidase (SPP) and induced their degradation via HMG-CoA reductase degradation 1 homolog, thereby impairing antigen presentation to CD8+ T cells. The expression of MHC class I in the livers of HCV core transgenic mice and chronic hepatitis C patients was impaired but was restored in patients achieving sustained virological response. Finally, we show that the human cytomegalovirus US2 protein, possessing a transmembrane region structurally similar to the HCV core protein, targets SPP to impair MHC class I molecule expression. Thus, SPP represents a potential target for the impairment of MHC class I molecules by DNA and RNA viruses.


Assuntos
Ácido Aspártico Endopeptidases/metabolismo , Hepacivirus/fisiologia , Evasão da Resposta Imune/fisiologia , Animais , Apresentação de Antígeno/imunologia , Linhagem Celular , Regulação para Baixo , Hepacivirus/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Camundongos , Proteínas do Core Viral/fisiologia
5.
Sci Rep ; 11(1): 11220, 2021 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-34045609

RESUMO

NK/T cell lymphoma (NKTCL) represents an aggressive lymphoid malignancy characterized by dismal prognosis. Immune-checkpoint blockade has shown promising efficacy in NKTCL. However, the molecular mechanisms underlying immune evasion in NKTCL have never been explored. Here, proteomic analysis was used to identify the differentially expressed proteins between NKTCL patients and healthy individuals. We found that S100A9, an immunosuppressive molecule, was much higher in NKTCL patients both in serum and tumor stroma. Elevated level of S100A9 was associated with advanced stage, poor overall response and early recurrence. Moreover, percentage of myeloid-derived suppressor cells (MDSCs) in peripheral blood was positively correlated with levels of S100A9. Low concentration of S100A9 promoted proliferation of NKTCL cells, while did not affect cell apoptosis and cell cycles. Furthermore, programmed death ligand 1 (PD-L1) expression on NKTCL cells was up-regulated by S100A9 through activation of ERK1/2 signaling. Inhibition of ERK1/2 signaling significantly decreased tumor growth and PD-L1 expression induced by S100A9. In conclusion, our research firstly identified S100A9 as an immune suppressor in the tumorigenesis of NKTCL via accumulation of MDSCs and upregulation of PD-L1 expression. S100A9 may serve as a potential target to increase the efficacy of immunotherapy in NKTCL.


Assuntos
Biomarcadores Tumorais/metabolismo , Calgranulina B/metabolismo , Evasão da Resposta Imune/fisiologia , Linfoma Extranodal de Células T-NK/metabolismo , Apoptose/fisiologia , Biomarcadores Tumorais/sangue , Calgranulina B/sangue , Proliferação de Células/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Linfoma Extranodal de Células T-NK/sangue , Linfoma Extranodal de Células T-NK/patologia , Masculino , Pessoa de Meia-Idade , Prognóstico , Proteômica
6.
PLoS Pathog ; 17(3): e1009481, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33788895

RESUMO

TcpC is a virulence factor of uropathogenic E. coli (UPEC). It was found that TIR domain of TcpC impedes TLR signaling by direct association with MyD88. It has been a long-standing question whether bacterial pathogens have evolved a mechanism to manipulate MyD88 degradation by ubiquitin-proteasome pathway. Here, we show that TcpC is a MyD88-targeted E3 ubiquitin ligase. Kidney macrophages from mice with pyelonephritis induced by TcpC-secreting UPEC showed significantly decreased MyD88 protein levels. Recombinant TcpC (rTcpC) dose-dependently inhibited protein but not mRNA levels of MyD88 in macrophages. Moreover, rTcpC significantly promoted MyD88 ubiquitination and accumulation in proteasomes in macrophages. Cys12 and Trp106 in TcpC are crucial amino acids in maintaining its E3 activity. Therefore, TcpC blocks TLR signaling pathway by degradation of MyD88 through ubiquitin-proteasome system. Our findings provide not only a novel biochemical mechanism underlying TcpC-medicated immune evasion, but also the first example that bacterial pathogens inhibit MyD88-mediated signaling pathway by virulence factors that function as E3 ubiquitin ligase.


Assuntos
Proteínas de Escherichia coli/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais/fisiologia , Escherichia coli Uropatogênica/patogenicidade , Fatores de Virulência/metabolismo , Animais , Linhagem Celular , Feminino , Humanos , Evasão da Resposta Imune/fisiologia , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Pielonefrite/imunologia , Pielonefrite/microbiologia , Receptores Toll-Like/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Escherichia coli Uropatogênica/imunologia , Escherichia coli Uropatogênica/metabolismo , Virulência/fisiologia
7.
Nat Commun ; 12(1): 1482, 2021 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-33674596

RESUMO

Immune evasion is a hallmark of KRAS-driven cancers, but the underlying causes remain unresolved. Here, we use a mouse model of pancreatic ductal adenocarcinoma to inactivate KRAS by CRISPR-mediated genome editing. We demonstrate that at an advanced tumor stage, dependence on KRAS for tumor growth is reduced and is manifested in the suppression of antitumor immunity. KRAS-deficient cells retain the ability to form tumors in immunodeficient mice. However, they fail to evade the host immune system in syngeneic wild-type mice, triggering strong antitumor response. We uncover changes both in tumor cells and host immune cells attributable to oncogenic KRAS expression. We identify BRAF and MYC as key mediators of KRAS-driven tumor immune suppression and show that loss of BRAF effectively blocks tumor growth in mice. Applying our results to human PDAC we show that lowering KRAS activity is likewise associated with a more vigorous immune environment.


Assuntos
Evasão da Resposta Imune/fisiologia , Modelos Genéticos , Neoplasias Pancreáticas/imunologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Animais , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Edição de Genes , Perfilação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Ductos Pancreáticos/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas p21(ras)/imunologia , Transcriptoma , Neoplasias Pancreáticas
8.
J Parasitol ; 107(1): 115-124, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33592628

RESUMO

The immune response of animals, including insects, is overcome by some parasites. For example, dauer larvae (DL) of the obligate entomopathogenic nematodes (EPNs) Heterorhabditis and Steinernema can invade insects, evade their defenses, and cause death. Although DL were long assumed to be the only infective stage of nematodes, recent reports suggest that L2-L3 larvae of facultative EPNs are also capable of killing insects. There are no studies, to our knowledge, about the role of nonimmunological barriers (the exoskeleton and its openings) in avoiding infection by DL and L2-L3 larvae, or whether these larval stages evade the host immune system in the same way. The objective of this study was to examine these questions by infecting Galleria mellonella with the facultative parasitic nematode Rhabditis regina. DL or L2-L3 larvae were either deposited on or near the moths or injected into their hemocoel. Once nematodes reached the hemocoel, the following host immune response parameters were quantified: prophenoloxidase, phenoloxidase, lytic activity, and the number of granular hemocytes. DL showed a greater ability to penetrate the exoskeleton than L2-L3 larvae. Once inside, however, both went unnoticed by the immune system and killed the insect. A higher number of granular hemocytes was activated by L2-L3 larvae than DL. We show for the first time that L2-L3 larvae can penetrate and evade the insect immune system. Further research is needed to compare facultative and specialized EPNs to determine which is more likely, with both DL and L2-L3 larvae, to evade insect defense barriers and produce death. The results will contribute to understanding the evolution of virulence in entomopathogenic nematodes.


Assuntos
Evasão da Resposta Imune/fisiologia , Lepidópteros/parasitologia , Strongyloidea/imunologia , Análise de Variância , Animais , Catecol Oxidase/metabolismo , Proteínas Cardiotóxicas de Elapídeos/metabolismo , Precursores Enzimáticos/metabolismo , Larva/imunologia , Lepidópteros/enzimologia , Lepidópteros/imunologia , Monofenol Mono-Oxigenase/metabolismo , Strongyloidea/patogenicidade , Strongyloidea/ultraestrutura , Fatores de Tempo , Virulência
9.
Sci Rep ; 11(1): 946, 2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33441583

RESUMO

Macrophages and monocytes are important for clearance of Leishmania infections. However, immune evasion tactics employed by the parasite results in suppressed inflammatory responses, marked by deficient macrophage functions and increased accumulation of monocytes. This results in an ineffective ability to clear parasite loads. Allograft Inflammatory Factor-1 (AIF1) is expressed in myeloid cells and serves to promote immune responses. However, AIF1 involvement in monocyte and macrophage functions during parasitic infections has not been explored. This study now shows that Leishmania donovani inhibits AIF1 expression in macrophages to block pro-inflammatory responses. Mice challenged with the parasite had markedly reduced AIF1 expression in splenic macrophages. Follow-up studies using in vitro approaches confirmed that L. donovani infection in macrophages suppresses AIF1 expression, which correlated with reduction in pro-inflammatory cytokine production and increased parasite load. Ectopic overexpression of AIF1 in macrophages provided protection from infection, marked by robust pro-inflammatory cytokine production and efficient pathogen clearance. Further investigations found that inhibiting AIF1 expression in bone marrow cells or monocytes impaired differentiation into functional macrophages. Collectively, results show that AIF1 is a critical regulatory component governing monocyte and macrophage immune functions and that L. donovani infection can suppress the gene as an immune evasion tactic.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Inflamação/imunologia , Leishmania donovani/metabolismo , Proteínas dos Microfilamentos/metabolismo , Animais , Apoptose , Células da Medula Óssea/citologia , Proteínas de Ligação ao Cálcio/fisiologia , Diferenciação Celular , Feminino , Evasão da Resposta Imune/imunologia , Evasão da Resposta Imune/fisiologia , Inflamação/metabolismo , Leishmania donovani/patogenicidade , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/fisiologia , Monócitos/imunologia , Monócitos/metabolismo
10.
Elife ; 92020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-33300875

RESUMO

HIV-1 must replicate in cells that are equipped to defend themselves from infection through intracellular innate immune systems. HIV-1 evades innate immune sensing through encapsidated DNA synthesis and encodes accessory genes that antagonize specific antiviral effectors. Here, we show that both particle associated, and expressed HIV-1 Vpr, antagonize the stimulatory effect of a variety of pathogen associated molecular patterns by inhibiting IRF3 and NF-κB nuclear transport. Phosphorylation of IRF3 at S396, but not S386, was also inhibited. We propose that, rather than promoting HIV-1 nuclear import, Vpr interacts with karyopherins to disturb their import of IRF3 and NF-κB to promote replication in macrophages. Concordantly, we demonstrate Vpr-dependent rescue of HIV-1 replication in human macrophages from inhibition by cGAMP, the product of activated cGAS. We propose a model that unifies Vpr manipulation of nuclear import and inhibition of innate immune activation to promote HIV-1 replication and transmission.


Assuntos
Infecções por HIV/imunologia , Evasão da Resposta Imune/fisiologia , Imunidade Inata/imunologia , Replicação Viral/fisiologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia , Transporte Ativo do Núcleo Celular/fisiologia , Infecções por HIV/transmissão , Infecções por HIV/virologia , HIV-1/imunologia , HIV-1/metabolismo , HIV-1/patogenicidade , Humanos , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Carioferinas/imunologia , Carioferinas/metabolismo , Macrófagos/imunologia , Macrófagos/virologia , NF-kappa B/imunologia , NF-kappa B/metabolismo , Transdução de Sinais/fisiologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo
12.
Viruses ; 12(10)2020 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-32993049

RESUMO

Extracellular vesicles have recently emerged as a novel mode of viral transmission exploited by naked viruses to exit host cells through a nonlytic pathway. Extracellular vesicles can allow multiple viral particles to collectively traffic in and out of cells, thus enhancing the viral fitness and diversifying the transmission routes while evading the immune system. This has been shown for several RNA viruses that belong to the Picornaviridae, Hepeviridae, Reoviridae, and Caliciviridae families; however, recent studies also demonstrated that the BK and JC viruses, two DNA viruses that belong to the Polyomaviridae family, use a similar strategy. In this review, we provide an update on recent advances in understanding the mechanisms used by naked viruses to hijack extracellular vesicles, and we discuss the implications for the biology of polyomaviruses.


Assuntos
Vesículas Extracelulares/virologia , Evasão da Resposta Imune/fisiologia , Infecções por Polyomavirus/transmissão , Polyomavirus/metabolismo , Infecções Tumorais por Vírus/transmissão , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Endocitose/fisiologia , Humanos , Polyomavirus/imunologia , Infecções Tumorais por Vírus/virologia , Internalização do Vírus , Replicação Viral/fisiologia
13.
Nat Commun ; 11(1): 2688, 2020 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-32461612

RESUMO

Severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS) coronaviruses (CoVs) are zoonotic pathogens with high fatality rates and pandemic potential. Vaccine development focuses on the principal target of the neutralizing humoral immune response, the spike (S) glycoprotein. Coronavirus S proteins are extensively glycosylated, encoding around 66-87 N-linked glycosylation sites per trimeric spike. Here, we reveal a specific area of high glycan density on MERS S that results in the formation of oligomannose-type glycan clusters, which were absent on SARS and HKU1 CoVs. We provide a comparison of the global glycan density of coronavirus spikes with other viral proteins including HIV-1 envelope, Lassa virus glycoprotein complex, and influenza hemagglutinin, where glycosylation plays a known role in shielding immunogenic epitopes. Overall, our data reveal how organisation of glycosylation across class I viral fusion proteins influence not only individual glycan compositions but also the immunological pressure across the protein surface.


Assuntos
Glicoproteínas/imunologia , Coronavírus da Síndrome Respiratória do Oriente Médio , Polissacarídeos , Glicoproteína da Espícula de Coronavírus/imunologia , Proteínas Virais de Fusão/imunologia , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Microscopia Crioeletrônica , Epitopos/química , Epitopos/imunologia , Epitopos/metabolismo , Glicoproteínas/química , Glicoproteínas/ultraestrutura , Glicosilação , Células HEK293 , HIV-1/imunologia , HIV-1/metabolismo , Humanos , Evasão da Resposta Imune/fisiologia , Vírus Lassa/imunologia , Vírus Lassa/metabolismo , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Coronavírus da Síndrome Respiratória do Oriente Médio/metabolismo , Orthomyxoviridae/imunologia , Orthomyxoviridae/metabolismo , Polissacarídeos/química , Polissacarídeos/imunologia , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/ultraestrutura , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/ultraestrutura , Proteínas Virais/química , Proteínas Virais/imunologia , Proteínas Virais/ultraestrutura
14.
Parasite Immunol ; 42(7): e12712, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32187690

RESUMO

The ability of Toxoplasma gondii to cause clinical disease in immune-competent and immune-deficient hosts coupled with its ease of use in vitro and availability of murine models has led to its use as a model organism to study how the immune system controls an intracellular infection. This article reviews the studies that established the role of the cytokine IFN-γ in the activation of macrophages to control T gondii and the events that lead to the mobilization and expansion of macrophage populations and their ability to limit parasite replication. Macrophages also have pro-inflammatory functions that promote protective NK and T-cell activities as well as regulatory properties that facilitate the resolution of inflammation. Nevertheless, while macrophages are important in determining the outcome of infection, T gondii has evolved mechanisms to subvert macrophage activation and can utilize their migratory activities to promote dissemination and these two properties underlie the ability of this parasite to persist and cause disease.


Assuntos
Interferon gama/imunologia , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Toxoplasma/imunologia , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Interações Hospedeiro-Parasita/imunologia , Evasão da Resposta Imune/fisiologia , Interferon gama/metabolismo , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Camundongos , Linfócitos T/imunologia
15.
Parasit Vectors ; 13(1): 49, 2020 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-32029006

RESUMO

BACKGROUND: Cystic echinococcosis is a chronic disease caused by infection with the larvae of Echinococcus granulosus. The parasite's ability to establish persistent infection is partly due to its evolving immune evasion strategies. One strategy may involve the protective effect of arginase, which impedes the control of pathogens or tumors, whereas it remains largely unknown during E. granulosus infection. Here, we analyzed whether arginase was produced in peritoneal cells and assessed its role in immunosuppression in mice infected with protoscoleces of E. granulosus. METHODS: BALB/c mice injected with protoscoleces of E. granulosus were used to evaluate the expression of arginase (ARG) in mRNA and protein levels. The profiles of ARG-1 expression in peritoneal cells and CD3ζ expression in T cells from spleens were assessed at different time points (3, 6, 9 and 12 months post-infection) by flow cytometry. In vitro, peritoneal cells were co-cultured with purified T cells in a transwell system, and the levels of CD3ζ re-expression were compared by flow cytometry. Meanwhile, the changes of L-arginine and its related metabolites in serum were tested. RESULTS: Compared to the control group, the peritoneal cells from infected mice showed higher levels of ARG-1 mRNA and protein, unchanged ARG-2 and iNOS. Enhanced ARG-1 expression was present in SSClowCD11b+F4/80+, CD11b+CD11c+, CD11b+Gr-1+Ly-6C+Ly-6G-, CD11b+Gr-1+Ly-6C-Ly-6G+, CD11b+Gr-1+ and CD11b+Ly-6G+ cells. The proportion of cells and the proportion of ARG-1 expression in corresponding cells exhibited a rising trend along with the extension of infection time, except for fluctuations in SSClowCD11b+F4/80+ and CD11b+CD11c+ cells at 12 months post-infection, whereas the expression of CD3ζ chain in CD4+ and CD8+ T cells showed a descending trend. Purified T cells showed declined re-expression of CD3ζ when co-cultured with peritoneal cells from infected mice, and CD3ζ was regenerated by supplement of L-arginine or arginase inhibitor BEC, rather than NOS inhibitor L-NMMA or catalase. Meanwhile, the concentrations of L-arginine, L-citrulline and NO decreased, and those of L-ornithine and urea increased in serum post-infection. CONCLUSIONS: Our findings demonstrated that ARG-1 expression is enhanced in multiple myeloid cells from peritoneum and promotes immune evasion of E. granulosus in mice by inhibiting the expression of T cell receptor CD3ζ chain and antagonism against iNOS.


Assuntos
Arginase/imunologia , Echinococcus granulosus/imunologia , Evasão da Resposta Imune/fisiologia , Animais , Arginase/metabolismo , Equinococose/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Células Mieloides/imunologia , Óxido Nítrico Sintase Tipo II/imunologia , Óxido Nítrico Sintase Tipo II/metabolismo , Linfócitos T/imunologia
16.
Am J Respir Cell Mol Biol ; 62(3): 354-363, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31545652

RESUMO

Comparisons of infectivity among the clinically important nontuberculous mycobacteria (NTM) species have not been explored in great depth. Rapid-growing mycobacteria, including Mycobacterium abscessus and M. porcinum, can cause indolent but progressive lung disease. Slow-growing members of the M. avium complex are the most common group of NTM to cause lung disease, and molecular approaches can now distinguish between several distinct species of M. avium complex including M. intracellulare, M. avium, M. marseillense, and M. chimaera. Differential infectivity among these NTM species may, in part, account for differences in clinical outcomes and response to treatment; thus, knowing the relative infectivity of particular isolates could increase prognostication accuracy and enhance personalized treatment. Using human macrophages, we investigated the infectivity and virulence of nine NTM species, as well as multiple isolates of the same species. We also assessed their capacity to evade killing by the antibacterial peptide cathelicidin (LL-37). We discovered that the ability of different NTM species to infect macrophages varied among the species and among isolates of the same species. Our biochemical assays implicate modified phospholipids, which may include a phosphatidylinositol or cardiolipin backbone, as candidate antagonists of LL-37 antibacterial activity. The high variation in infectivity and virulence of NTM strains suggests that more detailed microbiological and biochemical characterizations are necessary to increase our knowledge of NTM pathogenesis.


Assuntos
Peptídeos Catiônicos Antimicrobianos/antagonistas & inibidores , Evasão da Resposta Imune/fisiologia , Lipídeos de Membrana/fisiologia , Micobactérias não Tuberculosas/patogenicidade , Fosfolipídeos/fisiologia , Peptídeos Catiônicos Antimicrobianos/metabolismo , Peptídeos Catiônicos Antimicrobianos/farmacologia , Membrana Celular/imunologia , Cromatografia em Camada Fina , Escherichia coli/efeitos dos fármacos , Humanos , Macrófagos/microbiologia , Macrófagos Alveolares/microbiologia , Lipídeos de Membrana/isolamento & purificação , Micobactérias não Tuberculosas/efeitos dos fármacos , Micobactérias não Tuberculosas/fisiologia , Fosfolipídeos/isolamento & purificação , Filogenia , Especificidade da Espécie , Células THP-1 , Virulência , Catelicidinas
17.
mBio ; 10(6)2019 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-31822584

RESUMO

Human cytomegalovirus (HCMV) encodes an endoplasmic reticulum (ER)-resident glycoprotein, UL148, which activates the unfolded protein response (UPR) but is fully dispensable for viral replication in cultured cells. Hence, its previously ascribed roles in immune evasion and modulation of viral cell tropism are hypothesized to cause ER stress. Here, we show that UL148 is necessary and sufficient to drive the formation of prominent ER-derived structures that on average occupy 5% of the infected cell cytoplasm. The structures are sites where UL148 coalesces with cellular proteins involved in ER quality control, such as HRD1 and EDEM1. Electron microscopy revealed that cells infected with wild-type but not UL148-null HCMV show prominent accumulations of densely packed ruffled ER membranes which connect to distended cisternae of smooth and partially rough ER. During ectopic expression of UL148-green fluorescent protein (GFP) fusion protein, punctate signals traffic to accumulate at conspicuous structures. The structures exhibit poor recovery of fluorescence after photobleaching, which suggests that their contents are poorly mobile and do not efficiently exchange with the rest of the ER. Small-molecule blockade of the integrated stress response (ISR) prevents the formation of puncta, leading to a uniform reticular fluorescent signal. Accordingly, ISR inhibition during HCMV infection abolishes the coalescence of UL148 and HRD1 into discrete structures, which argues that UL148 requires the ISR to cause ER reorganization. Given that UL148 stabilizes immature forms of a receptor binding subunit for a viral envelope glycoprotein complex important for HCMV infectivity, our results imply that stress-dependent ER remodeling contributes to viral cell tropism.IMPORTANCE Perturbations to endoplasmic reticulum (ER) morphology occur during infection with various intracellular pathogens and in certain genetic disorders. We identify that a human cytomegalovirus (HCMV) gene product, UL148, profoundly reorganizes the ER during infection and is sufficient to do so when expressed on its own. Our results reveal that UL148-dependent reorganization of the ER is a prominent feature of HCMV-infected cells. Moreover, we find that this example of virally induced organelle remodeling requires the integrated stress response (ISR), a stress adaptation pathway that contributes to a number of disease states. Since ER reorganization accompanies roles of UL148 in modulation of HCMV cell tropism and in evasion of antiviral immune responses, our results may have implications for understanding the mechanisms involved. Furthermore, our findings provide a basis to utilize UL148 as a tool to investigate organelle responses to stress and to identify novel drugs targeting the ISR.


Assuntos
Infecções por Citomegalovirus/metabolismo , Citomegalovirus/metabolismo , Retículo Endoplasmático/metabolismo , Glicoproteínas/metabolismo , Proteínas Virais de Fusão/metabolismo , Proteínas não Estruturais Virais/metabolismo , Linhagem Celular , Estresse do Retículo Endoplasmático/fisiologia , Humanos , Evasão da Resposta Imune/fisiologia , Proteínas de Membrana/metabolismo , Resposta a Proteínas não Dobradas/fisiologia , Proteínas do Envelope Viral/metabolismo , Tropismo Viral/fisiologia , Replicação Viral/fisiologia
18.
Int J Mol Sci ; 20(22)2019 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-31731575

RESUMO

Edwardsiella tarda is a Gram-negative bacterial pathogen with a broad host range, including fish, reptiles, and mammals. One prominent virulence feature of E. tarda is its ability to survive and replicate in host phagocytes, but the relevant molecular mechanism is largely unknown. In this study, we examined the transcriptome profiles of RAW264.7 cells, a murine macrophage cell line, infected with live E. tarda or stimulated with dead E. tarda for 4 h and 8 h. Eighteen libraries were constructed, and an average of 69 million clean reads per library were obtained, with ~81.63% of the reads being successfully mapped to the reference genome. In total, 208 and 232 differentially expressed genes (DEGs) were identified between live and dead E. tarda-treated cells at 4 h and 8 h post-infection, respectively. The DEGs were markedly enriched in the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways associated with immunity. Live E. tarda differed strikingly from dead E. tarda in the regulation of immune related genes. Compared with dead E. tarda-treated cells, live E. tarda-treated cells exhibited marked and significant suppression in the induction of a large amount of immune genes, including RIG-I-like receptors, cytokines, and interferon-related genes. Furthermore, some of the immune genes highly regulated by live E. tarda formed complicated interaction networks with each other. Together, the results of this study revealed a transcriptome profile specifically induced by the active virulence elements of live E. tarda during the infection process, thus adding new insights into the intracellular infection mechanism of E. tarda. This study also provided a valuable set of target genes for further study of the immune evasion strategy of E. tarda.


Assuntos
Edwardsiella tarda/imunologia , Edwardsiella tarda/patogenicidade , Evasão da Resposta Imune/fisiologia , Animais , Perfilação da Expressão Gênica , Macrófagos/metabolismo , Camundongos , Fosforilação , Células RAW 264.7 , Transcriptoma/genética , Virulência
19.
Nat Commun ; 10(1): 5315, 2019 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-31757950

RESUMO

To colonise their host, pathogens must counter local environmental and immunological challenges. Here, we reveal that the fungal pathogen Candida albicans exploits diverse host-associated signals to promote immune evasion by masking of a major pathogen-associated molecular pattern (PAMP), ß-glucan. Certain nutrients, stresses and antifungal drugs trigger ß-glucan masking, whereas other inputs, such as nitrogen sources and quorum sensing molecules, exert limited effects on this PAMP. In particular, iron limitation triggers substantial changes in the cell wall that reduce ß-glucan exposure. This correlates with reduced phagocytosis by macrophages and attenuated cytokine responses by peripheral blood mononuclear cells. Iron limitation-induced ß-glucan masking depends on parallel signalling via the iron transceptor Ftr1 and the iron-responsive transcription factor Sef1, and the protein kinase A pathway. Our data reveal that C. albicans exploits a diverse range of specific host signals to trigger protective anticipatory responses against impending phagocytic attack and promote host colonisation.


Assuntos
Candida albicans/metabolismo , Citocinas/imunologia , Evasão da Resposta Imune/fisiologia , Ferro/metabolismo , Macrófagos/imunologia , Fagocitose/imunologia , beta-Glucanas/metabolismo , Animais , Candida albicans/imunologia , Parede Celular/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Fúngicas/metabolismo , Humanos , Evasão da Resposta Imune/imunologia , Imunidade Inata/imunologia , Leucócitos Mononucleares/imunologia , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Moléculas com Motivos Associados a Patógenos/imunologia , Moléculas com Motivos Associados a Patógenos/metabolismo , Transdução de Sinais , beta-Glucanas/imunologia
20.
J Microbiol Biotechnol ; 29(12): 1873-1881, 2019 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-31650769

RESUMO

The innate immune response serves as a first-line-of-defense mechanism for a host against viral infection. Viruses must therefore subvert this anti-viral response in order to establish an efficient life cycle. In line with this fact, Kaposi's sarcoma-associated herpesvirus (KSHV) encodes numerous genes that function as immunomodulatory proteins to antagonize the host immune system. One such mechanism through which KSHV evades the host immunity is by encoding a viral homolog of cellular interferon (IFN) regulatory factors (IRFs), known as vIRFs. Herein, we summarize recent advances in the study of the immunomodulatory strategies of KSHV vIRFs and their effects on KSHV-associated pathogenesis.


Assuntos
Herpesvirus Humano 8/fisiologia , Evasão da Resposta Imune/fisiologia , Fatores Reguladores de Interferon/fisiologia , Proteínas Virais/fisiologia , Apoptose/fisiologia , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/imunologia , Interações Hospedeiro-Patógeno/imunologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Evasão da Resposta Imune/imunologia , Imunidade Inata , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/imunologia , Transdução de Sinais , Fatores de Transcrição , Proteínas Virais/genética , Proteínas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA