Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
2.
Nat Immunol ; 21(8): 868-879, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32690950

RESUMO

STING is essential for control of infections and for tumor immunosurveillance, but it can also drive pathological inflammation. STING resides on the endoplasmic reticulum (ER) and traffics following stimulation to the ERGIC/Golgi, where signaling occurs. Although STING ER exit is the rate-limiting step in STING signaling, the mechanism that drives this process is not understood. Here we identify STEEP as a positive regulator of STING signaling. STEEP was associated with STING and promoted trafficking from the ER. This was mediated through stimulation of phosphatidylinositol-3-phosphate (PtdIns(3)P) production and ER membrane curvature formation, thus inducing COPII-mediated ER-to-Golgi trafficking of STING. Depletion of STEEP impaired STING-driven gene expression in response to virus infection in brain tissue and in cells from patients with STING-associated diseases. Interestingly, STING gain-of-function mutants from patients interacted strongly with STEEP, leading to increased ER PtdIns(3)P levels and membrane curvature. Thus, STEEP enables STING signaling by promoting ER exit.


Assuntos
Retículo Endoplasmático/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais/fisiologia , Animais , Retículo Endoplasmático/imunologia , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Proteínas de Membrana/imunologia , Camundongos , Proteínas do Tecido Nervoso/imunologia , Proteínas Nucleares , Transporte Proteico/fisiologia
3.
Immunity ; 54(7): 1478-1493.e6, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34015257

RESUMO

Viral infections during pregnancy are a considerable cause of adverse outcomes and birth defects, and the underlying mechanisms are poorly understood. Among those, cytomegalovirus (CMV) infection stands out as the most common intrauterine infection in humans, putatively causing early pregnancy loss. We employed murine CMV as a model to study the consequences of viral infection on pregnancy outcome and fertility maintenance. Even though pregnant mice successfully controlled CMV infection, we observed highly selective, strong infection of corpus luteum (CL) cells in their ovaries. High infection densities indicated complete failure of immune control in CL cells, resulting in progesterone insufficiency and pregnancy loss. An abundance of gap junctions, absence of vasculature, strong type I interferon (IFN) responses, and interaction of innate immune cells fully protected the ovarian follicles from viral infection. Our work provides fundamental insights into the effect of CMV infection on pregnancy loss and mechanisms protecting fertility.


Assuntos
Corpo Lúteo/imunologia , Infecções por Citomegalovirus/imunologia , Fertilidade/imunologia , Imunidade Inata/imunologia , Animais , Corpo Lúteo/virologia , Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Feminino , Junções Comunicantes/imunologia , Interferon Tipo I/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Gravidez , Progesterona/imunologia
4.
Nature ; 2024 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-39048823

RESUMO

The brain is highly sensitive to damage caused by infection and inflammation1,2. Herpes simplex virus 1 (HSV-1) is a neurotropic virus and the cause of herpes simplex encephalitis3. It is unknown whether neuron-specific antiviral factors control virus replication to prevent infection and excessive inflammatory responses, hence protecting the brain. Here we identify TMEFF1 as an HSV-1 restriction factor using genome-wide CRISPR screening. TMEFF1 is expressed specifically in neurons of the central nervous system and is not regulated by type I interferon, the best-known innate antiviral system controlling virus infections. Depletion of TMEFF1 in stem-cell-derived human neurons led to elevated viral replication and neuronal death following HSV-1 infection. TMEFF1 blocked the HSV-1 replication cycle at the level of viral entry through interactions with nectin-1 and non-muscle myosin heavy chains IIA and IIB, which are core proteins in virus-cell binding and virus-cell fusion, respectively4-6. Notably, Tmeff1-/- mice exhibited increased susceptibility to HSV-1 infection in the brain but not in the periphery. Within the brain, elevated viral load was observed specifically in neurons. Our study identifies TMEFF1 as a neuron-specific restriction factor essential for prevention of HSV-1 replication in the central nervous system.

5.
Nat Immunol ; 17(2): 150-8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26595890

RESUMO

Mucosal surfaces are exposed to environmental substances and represent a major portal of entry for microorganisms. The innate immune system is responsible for early defense against infections and it is believed that the interferons (IFNs) constitute the first line of defense against viruses. Here we identify an innate antiviral pathway that works at epithelial surfaces before the IFNs. The pathway is activated independently of known innate sensors of viral infections through a mechanism dependent on viral O-linked glycans, which induce CXCR3 chemokines and stimulate antiviral activity in a manner dependent on neutrophils. This study therefore identifies a previously unknown layer of antiviral defense that exerts its action on epithelial surfaces before the classical IFN response is operative.


Assuntos
Imunidade Inata , Interferons/metabolismo , Mucosa/imunologia , Mucosa/metabolismo , Viroses/imunologia , Viroses/metabolismo , Animais , Linhagem Celular , Quimiocina CXCL10/biossíntese , Modelos Animais de Doenças , Feminino , Expressão Gênica , Glicosilação , Herpes Simples/genética , Herpes Simples/imunologia , Herpes Simples/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 2/imunologia , Humanos , Interferons/genética , Ligantes , Camundongos , Camundongos Knockout , Mucosa/virologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Polissacarídeos/imunologia , Receptores CXCR3/deficiência , Receptores CXCR3/metabolismo , Vagina/imunologia , Vagina/metabolismo , Vagina/virologia , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/metabolismo , Carga Viral , Viroses/virologia
6.
EMBO J ; 42(19): e113118, 2023 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-37646198

RESUMO

Neurotropic viruses, including herpes simplex virus (HSV) types 1 and 2, have the capacity to infect neurons and can cause severe diseases. This is associated with neuronal cell death, which may contribute to morbidity or even mortality if the infection is not controlled. However, the mechanistic details of HSV-induced neuronal cell death remain enigmatic. Here, we report that lytic HSV-2 infection of human neuron-like SH-SY5Y cells and primary human and murine brain cells leads to cell death mediated by gasdermin E (GSDME). HSV-2-induced GSDME-mediated cell death occurs downstream of replication-induced endoplasmic reticulum stress driven by inositol-requiring kinase 1α (IRE1α), leading to activation of caspase-2, cleavage of the pro-apoptotic protein BH3-interacting domain death agonist (BID), and mitochondria-dependent activation of caspase-3. Finally, necrotic neurons released alarmins, which activated inflammatory responses in human iPSC-derived microglia. In conclusion, lytic HSV infection in neurons activates an ER stress-driven pathway to execute GSDME-mediated cell death and promote inflammation.

7.
EMBO J ; 41(10): e109622, 2022 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-35178710

RESUMO

Understanding the molecular pathways driving the acute antiviral and inflammatory response to SARS-CoV-2 infection is critical for developing treatments for severe COVID-19. Here, we find decreasing number of circulating plasmacytoid dendritic cells (pDCs) in COVID-19 patients early after symptom onset, correlating with disease severity. pDC depletion is transient and coincides with decreased expression of antiviral type I IFNα and of systemic inflammatory cytokines CXCL10 and IL-6. Using an in vitro stem cell-based human pDC model, we further demonstrate that pDCs, while not supporting SARS-CoV-2 replication, directly sense the virus and in response produce multiple antiviral (interferons: IFNα and IFNλ1) and inflammatory (IL-6, IL-8, CXCL10) cytokines that protect epithelial cells from de novo SARS-CoV-2 infection. Via targeted deletion of virus-recognition innate immune pathways, we identify TLR7-MyD88 signaling as crucial for production of antiviral interferons (IFNs), whereas Toll-like receptor (TLR)2 is responsible for the inflammatory IL-6 response. We further show that SARS-CoV-2 engages the receptor neuropilin-1 on pDCs to selectively mitigate the antiviral interferon response, but not the IL-6 response, suggesting neuropilin-1 as potential therapeutic target for stimulation of TLR7-mediated antiviral protection.


Assuntos
COVID-19 , Células Dendríticas , Receptor 2 Toll-Like , Receptor 7 Toll-Like , COVID-19/imunologia , COVID-19/patologia , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/patologia , Humanos , Interferon Tipo I/imunologia , Interferon-alfa/imunologia , Interleucina-6/imunologia , Neuropilina-1/imunologia , SARS-CoV-2 , Receptor 2 Toll-Like/imunologia , Receptor 7 Toll-Like/imunologia
8.
EMBO Rep ; 2024 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-39060455

RESUMO

RNA vaccines elicit protective immunity against SARS-CoV-2, but the use of mRNA as an antiviral immunotherapeutic is unexplored. Here, we investigate the activity of lipidoid nanoparticle (LNP)-formulated mRNA encoding human IFNλ1 (ETH47), which is a critical driver of innate immunity at mucosal surfaces protecting from viral infections. IFNλ1 mRNA administration promotes dose-dependent protein translation, induction of interferon-stimulated genes without relevant signs of unspecific immune stimulation, and dose-dependent inhibition of SARS-CoV-2 replication in vitro. Pulmonary administration of IFNλ1 mRNA in mice results in a potent reduction of virus load, virus-induced body weight loss and significantly increased survival. These data support the development of inhaled administration of IFNλ1 mRNA as a potential prophylactic option for individuals exposed to SARS-CoV-2 or at risk suffering from COVID-19. Based on the broad antiviral activity of IFNλ1 regardless of virus or variant, this approach might also be utilized for other respiratory viral infections or pandemic preparedness.

9.
Proc Natl Acad Sci U S A ; 118(50)2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34876524

RESUMO

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has created an urgent need for new technologies to treat COVID-19. Here we report a 2'-fluoro protected RNA aptamer that binds with high affinity to the receptor binding domain (RBD) of SARS-CoV-2 spike protein, thereby preventing its interaction with the host receptor ACE2. A trimerized version of the RNA aptamer matching the three RBDs in each spike complex enhances binding affinity down to the low picomolar range. Binding mode and specificity for the aptamer-spike interaction is supported by biolayer interferometry, single-molecule fluorescence microscopy, and flow-induced dispersion analysis in vitro. Cell culture experiments using virus-like particles and live SARS-CoV-2 show that the aptamer and, to a larger extent, the trimeric aptamer can efficiently block viral infection at low concentration. Finally, the aptamer maintains its high binding affinity to spike from other circulating SARS-CoV-2 strains, suggesting that it could find widespread use for the detection and treatment of SARS-CoV-2 and emerging variants.


Assuntos
Aptâmeros de Nucleotídeos/farmacologia , SARS-CoV-2/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Enzima de Conversão de Angiotensina 2/metabolismo , Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/metabolismo , Humanos , Mutação , Testes de Neutralização , Conformação de Ácido Nucleico , Ligação Proteica/efeitos dos fármacos , Domínios e Motivos de Interação entre Proteínas , SARS-CoV-2/fisiologia , Técnica de Seleção de Aptâmeros , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/metabolismo
10.
J Virol ; 96(6): e0131121, 2022 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-35045263

RESUMO

Spread of herpes simplex virus 1 (HSV1) from the periphery to the central nervous system (CNS) can lead to extensive infection and pathological inflammation in the brain, causing herpes simplex encephalitis (HSE). It has been shown that microglia, the CNS-resident macrophages, are involved in early sensing of HSV1 and induction of antiviral responses. In addition, infiltration of peripheral immune cells may contribute to the control of viral infection. In this study, we tested the effect of microglia depletion in a mouse model of HSE. Increased viral titers and increased disease severity were observed in microglia-depleted mice. The effect of microglia depletion was more pronounced in wild-type than in cGas-/- mice, revealing that this immune sensor contributes to the antiviral activity of microglia. Importantly, microglia depletion led to reduced production of type I interferon (IFN), proinflammatory cytokines, and chemokines at early time points after viral entry into the CNS. In line with this, in vitro experiments on murine primary CNS cells demonstrated microglial presence to be essential for IFN RNA induction, and control of HSV1 replication. However, the effect of microglia depletion on the expression of IFNs, and inflammatory cytokines was restricted to the early time point of HSV1 entry into the CNS. There was no major alteration of infiltration of CD45-positive cells in microglia-depleted mice. Collectively, our data demonstrate a key role for microglia in controlling HSV1 replication early after viral entry into the CNS and highlight the importance of a prompt antiviral innate response to reduce the risk of HSE development. IMPORTANCE One of the most devastating and acute neurological conditions is encephalitis, i.e., inflammation of brain tissue. Herpes simplex virus 1 (HSV1) is a highly prevalent pathogen in humans, and the most frequent cause of viral sporadic encephalitis called herpes simplex encephalitis (HSE). HSV1 can infect peripheral neurons and reach the central nervous system (CNS) of humans, where it can be detected by brain resident cells and infiltrating immune cells, leading to protective and damaging immune responses. In this study, we investigated the effects of microglia depletion, the main brain-resident immune cell type. For this purpose, we used a mouse model of HSE. We found that viral levels increased, and disease symptoms worsened in microglia-depleted mice. In addition, mice lacking a major sensor of viral DNA, cGAS, manifested a more pronounced disease than wild-type mice, highlighting the importance of this immune sensor in the activity of microglia. Microglia depletion led to reduced production of many known antiviral factors, most notably type I interferon (IFN). The importance of microglia in the early control of HSV1 spread and the generation of antiviral responses is further demonstrated by experiments on murine mixed glial cell cultures. Interestingly, mice with microglia depletion exhibited an unaltered activation of antiviral responses and recruitment of immune cells from the periphery at later time points of infection, but this did not prevent the development of the disease. Overall, the data highlight the importance of rapid activation of the host defense, with microglia playing a critical role in controlling HSV1 infection, which eventually prevents damage to neurons and brain tissue.


Assuntos
Encefalite por Herpes Simples , Herpesvirus Humano 1 , Imunidade , Interferon Tipo I , Microglia , Internalização do Vírus , Animais , Encéfalo/imunologia , Encéfalo/virologia , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalite por Herpes Simples/imunologia , Encefalite por Herpes Simples/fisiopatologia , Herpesvirus Humano 1/metabolismo , Imunidade/imunologia , Inflamação/patologia , Interferon Tipo I/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/imunologia , Microglia/virologia , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo
11.
Mol Ther ; 30(9): 2998-3016, 2022 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-35526097

RESUMO

We established a split nanoluciferase complementation assay to rapidly screen for inhibitors that interfere with binding of the receptor binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike glycoprotein with its target receptor, angiotensin-converting enzyme 2 (ACE2). After a screen of 1,200 US Food and Drug Administration (FDA)-approved compounds, we identified bifonazole, an imidazole-based antifungal agent, as a competitive inhibitor of RBD-ACE2 binding. Mechanistically, bifonazole binds ACE2 around residue K353, which prevents association with the RBD, affecting entry and replication of spike-pseudotyped viruses as well as native SARS-CoV-2 and its variants of concern (VOCs). Intranasal administration of bifonazole reduces lethality in K18-hACE2 mice challenged with vesicular stomatitis virus (VSV)-spike by 40%, with a similar benefit after live SARS-CoV-2 challenge. Our screen identified an antiviral agent that is effective against SARS-CoV-2 and VOCs such as Omicron that employ the same receptor to infect cells and therefore has high potential to be repurposed to control, treat, or prevent coronavirus disease 2019 (COVID-19).


Assuntos
Antivirais , Tratamento Farmacológico da COVID-19 , Imidazóis , SARS-CoV-2 , Enzima de Conversão de Angiotensina 2/antagonistas & inibidores , Animais , Antivirais/farmacologia , Imidazóis/farmacologia , Camundongos , Ligação Proteica , SARS-CoV-2/efeitos dos fármacos , Glicoproteína da Espícula de Coronavírus/química , Estados Unidos , United States Food and Drug Administration
12.
EMBO J ; 37(8)2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29496741

RESUMO

Negative regulation of immune pathways is essential to achieve resolution of immune responses and to avoid excess inflammation. DNA stimulates type I IFN expression through the DNA sensor cGAS, the second messenger cGAMP, and the adaptor molecule STING Here, we report that STING degradation following activation of the pathway occurs through autophagy and is mediated by p62/SQSTM1, which is phosphorylated by TBK1 to direct ubiquitinated STING to autophagosomes. Degradation of STING was impaired in p62-deficient cells, which responded with elevated IFN production to foreign DNA and DNA pathogens. In the absence of p62, STING failed to traffic to autophagy-associated vesicles. Thus, DNA sensing induces the cGAS-STING pathway to activate TBK1, which phosphorylates IRF3 to induce IFN expression, but also phosphorylates p62 to stimulate STING degradation and attenuation of the response.


Assuntos
Nucleotidiltransferases/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteína Sequestossoma-1/fisiologia , Animais , Autofagia , Linhagem Celular , DNA/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais
13.
EMBO Rep ; 21(12): e51252, 2020 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-33112036

RESUMO

Respiratory infections, like the current COVID-19 pandemic, target epithelial cells in the respiratory tract. Alveolar macrophages (AMs) are tissue-resident macrophages located within the lung. They play a key role in the early phases of an immune response to respiratory viruses. AMs are likely the first immune cells to encounter SARS-CoV-2 during an infection, and their reaction to the virus will have a profound impact on the outcome of the infection. Interferons (IFNs) are antiviral cytokines and among the first cytokines produced upon viral infection. In this study, AMs from non-infectious donors are challenged with SARS-CoV-2. We demonstrate that challenged AMs are incapable of sensing SARS-CoV-2 and of producing an IFN response in contrast to other respiratory viruses, like influenza A virus and Sendai virus, which trigger a robust IFN response. The absence of IFN production in AMs upon challenge with SARS-CoV-2 could explain the initial asymptotic phase observed during COVID-19 and argues against AMs being the sources of pro-inflammatory cytokines later during infection.


Assuntos
COVID-19/imunologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/virologia , SARS-CoV-2/imunologia , Antivirais/imunologia , COVID-19/virologia , Células Cultivadas , Citocinas/imunologia , Células Epiteliais/imunologia , Células Epiteliais/virologia , Humanos , Evasão da Resposta Imune , Interferon Tipo I/imunologia , Pulmão/imunologia , Pulmão/virologia , Pandemias
14.
PLoS Pathog ; 15(12): e1008155, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31856218

RESUMO

Cellular response to environmental challenges requires immediate and precise regulation of transcriptional programs. During viral infections, this includes the expression of antiviral genes that are essential to combat the pathogen. Transcribed mRNAs are bound and escorted to the cytoplasm by the cap-binding complex (CBC). We recently identified a protein complex consisting of NCBP1 and NCBP3 that, under physiological conditions, has redundant function to the canonical CBC, consisting of NCBP1 and NCBP2. Here, we provide evidence that NCBP3 is essential to mount a precise and appropriate antiviral response. Ncbp3-deficient cells allow higher virus growth and elicit a reduced antiviral response, a defect happening on post-transcriptional level. Ncbp3-deficient mice suffered from severe lung pathology and increased morbidity after influenza A virus challenge. While NCBP3 appeared to be particularly important during viral infections, it may be more broadly involved to ensure proper protein expression.


Assuntos
Infecções por Orthomyxoviridae/imunologia , Proteínas de Ligação ao Cap de RNA/imunologia , Proteínas de Ligação ao Cap de RNA/metabolismo , Animais , Vírus da Influenza A/imunologia , Camundongos , Camundongos Knockout , Infecções por Orthomyxoviridae/metabolismo , Biossíntese de Proteínas/fisiologia
15.
EMBO J ; 35(13): 1385-99, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27234299

RESUMO

Herpes simplex virus (HSV) 1 stimulates type I IFN expression through the cGAS-STING-TBK1 signaling axis. Macrophages have recently been proposed to be an essential source of IFN during viral infection. However, it is not known how HSV-1 inhibits IFN expression in this cell type. Here, we show that HSV-1 inhibits type I IFN induction through the cGAS-STING-TBK1 pathway in human macrophages, in a manner dependent on the conserved herpesvirus protein ICP27. This viral protein was expressed de novo in macrophages with early nuclear localization followed by later translocation to the cytoplasm where ICP27 prevented activation of IRF3. ICP27 interacted with TBK1 and STING in a manner that was dependent on TBK1 activity and the RGG motif in ICP27. Thus, HSV-1 inhibits expression of type I IFN in human macrophages through ICP27-dependent targeting of the TBK1-activated STING signalsome.


Assuntos
Herpesvirus Humano 1/patogenicidade , Proteínas Imediatamente Precoces/metabolismo , Evasão da Resposta Imune , Interferon Tipo I/antagonistas & inibidores , Macrófagos/imunologia , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Células Cultivadas , Interações Hospedeiro-Patógeno , Humanos , Mapeamento de Interação de Proteínas
16.
PLoS Pathog ; 14(4): e1006976, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29608601

RESUMO

In recent years, there has been an increasing interest in immunomodulatory therapy as a means to treat various conditions, including infectious diseases. For instance, Toll-like receptor (TLR) agonists have been evaluated for treatment of genital herpes. However, although the TLR7 agonist imiquimod was shown to have antiviral activity in individual patients, no significant effects were observed in clinical trials, and the compound also exhibited significant side effects, including local inflammation. Cytosolic DNA is detected by the enzyme cyclic GMP-AMP (2'3'-cGAMP) synthase (cGAS) to stimulate antiviral pathways, mainly through induction of type I interferon (IFN)s. cGAS is activated upon DNA binding to produce the cyclic dinucleotide (CDN) 2'3'-cGAMP, which in turn binds and activates the adaptor protein Stimulator of interferon genes (STING), thus triggering type I IFN expression. In contrast to TLRs, STING is expressed broadly, including in epithelial cells. Here we report that natural and non-natural STING agonists strongly induce type I IFNs in human cells and in mice in vivo, without stimulating significant inflammatory gene expression. Systemic treatment with 2'3'-cGAMP reduced genital herpes simplex virus (HSV) 2 replication and improved the clinical outcome of infection. More importantly, local application of CDNs at the genital epithelial surface gave rise to local IFN activity, but only limited systemic responses, and this treatment conferred total protection against disease in both immunocompetent and immunocompromised mice. In direct comparison between CDNs and TLR agonists, only CDNs acted directly on epithelial cells, hence allowing a more rapid and IFN-focused immune response in the vaginal epithelium. Thus, specific activation of the STING pathway in the vagina evokes induction of the IFN system but limited inflammatory responses to allow control of HSV2 infections in vivo.


Assuntos
Antivirais/farmacologia , Herpes Genital/prevenção & controle , Herpesvirus Humano 2/efeitos dos fármacos , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Proteínas de Membrana/agonistas , Nucleotídeos Cíclicos/farmacologia , Animais , Células Cultivadas , Citosol/virologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Feminino , Herpes Genital/metabolismo , Herpes Genital/virologia , Herpesvirus Humano 2/patogenicidade , Humanos , Interferon Tipo I/metabolismo , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Queratinócitos/virologia , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Nucleotidiltransferases/fisiologia , Transdução de Sinais
17.
Hepatology ; 64(3): 746-59, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27312012

RESUMO

UNLABELLED: Hepatitis B virus (HBV) is a major human pathogen, and about one third of the global population will be exposed to the virus in their lifetime. HBV infects hepatocytes, where it replicates its DNA and infection can lead to acute and chronic hepatitis with a high risk of liver cirrhosis and hepatocellular carcinoma. Despite this, there is limited understanding of how HBV establishes chronic infections. In recent years it has emerged that foreign DNA potently stimulates the innate immune response, particularly type 1 interferon (IFN) production; and this occurs through a pathway dependent on the DNA sensor cyclic guanosine monophosphate-adenosine monophosphate synthase and the downstream adaptor protein stimulator of IFN genes (STING). In this work we describe that human and murine hepatocytes do not express STING. Consequently, hepatocytes do not produce type 1 IFN in response to foreign DNA or HBV infection and mice lacking STING or cyclic guanosine monophosphate-adenosine monophosphate synthase exhibit unaltered ability to control infection in an adenovirus-HBV model. Stimulation of IFN production in the murine liver by administration of synthetic RNA decreases virus infection, thus demonstrating that IFN possesses anti-HBV activity in the liver. Importantly, introduction of STING expression specifically in hepatocytes reconstitutes the DNA sensing pathway, which leads to improved control of HBV in vivo. CONCLUSION: The lack of a functional innate DNA-sensing pathway in hepatocytes hampers efficient innate control of HBV infection; this may explain why HBV has adapted to specifically replicate in hepatocytes and could contribute to the weak capacity of this cell type to clear HBV infection. (Hepatology 2016;64:746-759).


Assuntos
Hepatite B Crônica/imunologia , Hepatócitos/imunologia , Adenoviridae , Animais , Células Cultivadas , Feminino , Hepatócitos/metabolismo , Imunidade Inata , Interferons/fisiologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Nucleotidiltransferases/metabolismo
18.
Trends Immunol ; 35(2): 79-87, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24316012

RESUMO

The innate immune system mediates protection against neurotropic viruses capable of infecting the central nervous system (CNS). Neurotropic viruses include herpes simplex virus (HSV), West Nile virus (WNV), rabies virus, La Crosse virus, and poliovirus. Viral infection triggers activation of pattern recognition receptors (PRRs) such as Toll-like receptors (TLRs), retinoic acid-inducible gene 1 (RIG-I) like receptors (RLRs), nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs), and cytosolic DNA sensors. Although originally characterised in peripheral immune cells, emerging evidence points to important roles for these PRRs in cells of the CNS. Here, we review recent advances in our understanding of the mechanisms by which these PRRs provide protection against neurotropic viruses, and discuss instances in which these responses become detrimental and cause immunopathology in the CNS.


Assuntos
Sistema Nervoso Central/imunologia , Sistema Nervoso Central/virologia , Imunidade Inata , Animais , Viroses do Sistema Nervoso Central/imunologia , Humanos , Inflamassomos/imunologia , Modelos Imunológicos , Receptores de Reconhecimento de Padrão/imunologia , Transdução de Sinais/imunologia , Receptores Toll-Like/imunologia
19.
J Immunol ; 193(12): 6090-102, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25385819

RESUMO

Detection of microbes by TLRs on the plasma membrane leads to the induction of proinflammatory cytokines such as TNF-α, via activation of NF-κB. Alternatively, activation of endosomal TLRs leads to the induction of type I IFNs via IFN regulatory factors (IRFs). TLR4 signaling from the plasma membrane to NF-κB via the Toll/IL-1R (TIR) adaptor protein MyD88 requires the TIR sorting adaptor Mal, whereas endosomal TLR4 signaling to IRF3 via the TIR domain-containing adaptor-inducing IFN-ß (TRIF) requires the TRIF-related adaptor molecule (TRAM). Similar to TLR4 homodimers, TLR2 heterodimers can also induce both proinflammatory cytokines and type I IFNs. TLR2 plasma membrane signaling to NF-κB is known to require MyD88 and Mal, whereas endosomal IRF activation by TLR2 requires MyD88. However, it was unclear whether TLR2 requires a sorting adaptor for endosomal signaling, like TLR4 does. In this study, we show that TLR2-dependent IRF7 activation at the endosome is both Mal- and TRAM-dependent, and that TRAM is required for the TLR2-dependent movement of MyD88 to endosomes following ligand engagement. TRAM interacted with both TLR2 and MyD88, suggesting that TRAM can act as a bridging adapter between these two molecules. Furthermore, infection of macrophages lacking TRAM with herpes viruses or the bacterium Staphylococcus aureus led to impaired induction of type I IFN, indicating a role for TRAM in TLR2-dependent responses to human pathogens. Our work reveals that TRAM acts as a sorting adaptor not only for TLR4, but also for TLR2, to facilitate signaling to IRF7 at the endosome, which explains how TLR2 is capable of causing type I IFN induction.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Endossomos/metabolismo , Interferon Tipo I/biossíntese , Transdução de Sinais , Receptor 2 Toll-Like/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem Celular , Endocitose , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Fator Regulador 7 de Interferon/metabolismo , Interferon beta/biossíntese , Espaço Intracelular/metabolismo , Glicoproteínas de Membrana/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Peptídeos/farmacologia , Ligação Proteica , Transporte Proteico , Receptores de Interleucina-1/metabolismo , Receptor 2 Toll-Like/antagonistas & inibidores , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/química , Receptor 4 Toll-Like/metabolismo
20.
Cell Rep ; 43(2): 113792, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38363679

RESUMO

Pattern recognition receptors (PRRs) induce host defense but can also induce exacerbated inflammatory responses. This raises the question of whether other mechanisms are also involved in early host defense. Using transcriptome analysis of disrupted transcripts in herpes simplex virus (HSV)-infected cells, we find that HSV infection disrupts the hypoxia-inducible factor (HIF) transcription network in neurons and epithelial cells. Importantly, HIF activation leads to control of HSV replication. Mechanistically, HIF activation induces autophagy, which is essential for antiviral activity. HSV-2 infection in vivo leads to hypoxia in CNS neurons, and mice with neuron-specific HIF1/2α deficiency exhibit elevated viral load and augmented PRR signaling and inflammatory gene expression in the CNS after HSV-2 infection. Data from human stem cell-derived neuron and microglia cultures show that HIF also exerts antiviral and inflammation-restricting activity in human CNS cells. Collectively, the HIF transcription factor system senses virus-induced hypoxic stress to induce cell-intrinsic antiviral responses and limit inflammation.


Assuntos
Encefalite , Herpes Simples , Humanos , Animais , Camundongos , Inflamação , Neurônios , Hipóxia , Antivirais/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA