Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 54(6): 1102-1104, 2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34107267

RESUMO

The impact of inhibitory receptor NKG2A-mediated education on uterine NK (uNK) cell responsiveness to vascular remodeling on pregnancy outcomes has remained unclear. In this issue of Immunity, Shreeve et al. show that loss of NKG2A+ uNK cells results in deficient vascularization and restricted fetal growth.


Assuntos
Células Matadoras Naturais , Útero , Feminino , Humanos , Neovascularização Patológica , Gravidez , Remodelação Vascular
2.
Immunity ; 50(1): 1-3, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30650369

RESUMO

In this issue of Immunity, Daniels et al. (2019) demonstrate that RIPK3 signaling limits Zika virus (ZIKV) infection in the central nervous system independently of its function in necroptosis by promoting itaconate production in infected neurons, thereby revealing a neuron-specific mechanism of metabolite-mediated ZIKV control.


Assuntos
Antivirais , Infecção por Zika virus , Zika virus , Apoptose , Carboxiliases , Humanos , Necrose , Neurônios , Nucleotídeos , Proteínas , Proteína Serina-Treonina Quinases de Interação com Receptores
3.
J Cell Sci ; 134(5)2021 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-33277377

RESUMO

Autophagy is a degradative cellular pathway that targets cytoplasmic contents and organelles for turnover by the lysosome. Various autophagy pathways play key roles in the clearance of viral infections, and many families of viruses have developed unique methods for avoiding degradation. Some positive-stranded RNA viruses, such as enteroviruses and flaviviruses, usurp the autophagic pathway to promote their own replication. We previously identified the endoplasmic reticulum (ER)-localized protein BPIFB3 as an important negative regulator of non-canonical autophagy that uniquely impacts the replication of enteroviruses and flaviviruses. Here, we find that many components of the canonical autophagy machinery are not required for BPIFB3 depletion-induced autophagy and identify the host factors that facilitate its role in the replication of enteroviruses and flaviviruses. Using proximity-dependent biotinylation (BioID) followed by mass spectrometry, we identify ARFGAP1 and TMED9 as two cellular components that interact with BPIFB3 to regulate autophagy and viral replication. Importantly, our data demonstrate that non-canonical autophagy in mammalian cells can be controlled outside of the traditional pathway regulators and define the role of two proteins in BPIFB3 depletion mediated non-canonical autophagy.


Assuntos
Autofagia , Infecções por Vírus de RNA , Animais , Retículo Endoplasmático , Replicação Viral
4.
J Virol ; 96(13): e0033022, 2022 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-35699446

RESUMO

Echoviruses are among the most common worldwide causes of aseptic meningitis, which can cause long-term sequelae and death, particularly in neonates. However, the mechanisms by which these viruses induce meningeal inflammation are poorly understood, owing at least in part to the lack of in vivo models that recapitulate this aspect of echovirus pathogenesis. Here, we developed an in vivo neonatal mouse model that recapitulates key aspects of echovirus-induced meningitis. We show that expression of the human homologue of the primary echovirus receptor, the neonatal Fc receptor (FcRn), is not sufficient for infection of the brains of neonatal mice. However, ablation of type I, but not III, interferon (IFN) signaling in mice expressing human FcRn permitted high levels of echovirus replication in the brain, with corresponding clinical symptoms, including delayed motor skills and hind-limb weakness. Using this model, we defined the immunological response of the brain to echovirus infection and identified key cytokines, such as granulocyte colony-stimulating factor (G-CSF) and interleukin 6 (IL-6), that were induced by this infection. Lastly, we showed that echoviruses specifically replicate in the leptomeninges, where they induce profound inflammation and cell death. Together, this work establishes an in vivo model of aseptic meningitis associated with echovirus infections that delineates the differential roles of type I and type III IFNs in echovirus-associated neuronal disease and defines the specificity of echoviral infections within the meninges. IMPORTANCE Echoviruses are among the most common worldwide causes of aseptic meningitis, which can cause long-term sequelae or even death. The mechanisms by which echoviruses infect the brain are poorly understood, largely owing to the lack of robust in vivo models that recapitulate this aspect of echovirus pathogenesis. Here, we establish a neonatal mouse model of echovirus-induced aseptic meningitis and show that expression of the human homologue of the FcRn, the primary receptor for echoviruses, and ablation of type I IFN signaling are required to recapitulate echovirus-induced meningitis and clinical disease. These findings provide key insights into the host factors that control echovirus-induced meningitis and a model that could be used to test anti-echovirus therapeutics.


Assuntos
Infecções do Sistema Nervoso Central , Infecções por Echovirus , Meningite Asséptica , Animais , Infecções do Sistema Nervoso Central/fisiopatologia , Infecções do Sistema Nervoso Central/virologia , Infecções por Echovirus/complicações , Infecções por Echovirus/fisiopatologia , Infecções por Echovirus/virologia , Enterovirus Humano B/fisiologia , Humanos , Inflamação , Interferon Tipo I/metabolismo , Interferons , Meningite Asséptica/etiologia , Meningite Asséptica/fisiopatologia , Meningite Asséptica/virologia , Camundongos , Interferon lambda
5.
PLoS Pathog ; 17(1): e1009252, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33513208

RESUMO

Neonatal echovirus infections are characterized by severe hepatitis and neurological complications that can be fatal. Here, we show that expression of the human homologue of the neonatal Fc receptor (hFcRn), the primary receptor for echoviruses, and ablation of type I interferon (IFN) signaling are key host determinants involved in echovirus pathogenesis. We show that expression of hFcRn alone is insufficient to confer susceptibility to echovirus infections in mice. However, expression of hFcRn in mice deficient in type I interferon (IFN) signaling, hFcRn-IFNAR-/-, recapitulate the echovirus pathogenesis observed in humans. Luminex-based multianalyte profiling from E11 infected hFcRn-IFNAR-/- mice revealed a robust systemic immune response to infection, including the induction of type I IFNs. Furthermore, similar to the severe hepatitis observed in humans, E11 infection in hFcRn-IFNAR-/- mice caused profound liver damage. Our findings define the host factors involved in echovirus pathogenesis and establish in vivo models that recapitulate echovirus disease in humans.


Assuntos
Enterovirus Humano B/patogenicidade , Infecções por Enterovirus/virologia , Genoma Viral/genética , Hepatite/virologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Interferon Tipo I/metabolismo , Receptores Fc/metabolismo , Transdução de Sinais , Animais , Enterovirus Humano B/genética , Infecções por Enterovirus/imunologia , Feminino , Expressão Gênica , Hepatite/imunologia , Hepatócitos/imunologia , Hepatócitos/virologia , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Imunidade , Fígado/imunologia , Fígado/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Fc/genética
6.
Immunity ; 40(6): 936-48, 2014 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-24931123

RESUMO

Virus infection is sensed in the cytoplasm by retinoic acid-inducible gene I (RIG-I, also known as DDX58), which requires RNA and polyubiquitin binding to induce type I interferon (IFN) and activate cellular innate immunity. We show that the human IFN-inducible oligoadenylate synthetases-like (OASL) protein has antiviral activity and mediates RIG-I activation by mimicking polyubiquitin. Loss of OASL expression reduced RIG-I signaling and enhanced virus replication in human cells. Conversely, OASL expression suppressed replication of a number of viruses in a RIG-I-dependent manner and enhanced RIG-I-mediated IFN induction. OASL interacted and colocalized with RIG-I, and through its C-terminal ubiquitin-like domain specifically enhanced RIG-I signaling. Bone-marrow-derived macrophages from mice deficient for Oasl2 showed that among the two mouse orthologs of human OASL, Oasl2 is functionally similar to human OASL. Our findings show a mechanism by which human OASL contributes to host antiviral responses by enhancing RIG-I activation.


Assuntos
2',5'-Oligoadenilato Sintetase/imunologia , RNA Helicases DEAD-box/imunologia , Infecções por Vírus de DNA/imunologia , Interferon Tipo I/imunologia , Infecções por Vírus de RNA/imunologia , 2',5'-Oligoadenilato Sintetase/genética , Animais , Proteína DEAD-box 58 , Células HCT116 , Células HEK293 , Humanos , Imunidade Inata , Fator Regulador 7 de Interferon/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Poliubiquitina , Ligação Proteica/imunologia , Interferência de RNA , RNA Interferente Pequeno , Receptores Imunológicos , Transdução de Sinais/imunologia , Replicação Viral/imunologia
7.
PLoS Biol ; 18(8): e3000889, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32853196

RESUMO

The SARS-CoV-2 epidemic challenged universities and other academic institutions to rapidly adapt to urgent and life-threatening situations. It forced most institutions to shut down nearly every aspect of their research and educational enterprises. In doing so, university leaders were thrust into unchartered waters and forced them to make unprecedented decisions. Successes and failures along the way highlighted how the autonomous nature of the American academic research enterprise and skillsets normally required of university leaders were ill-suited to mounting an emergency response. Here, as faculty from medical centers in the United States, we draw lessons from these experiences and apply them as we plan for the next possible COVID-19-induced shutdown as well as other large-scale pandemics and emergencies at universities in the United States and throughout the world.


Assuntos
Betacoronavirus/patogenicidade , Pesquisa Biomédica/organização & administração , Defesa Civil/organização & administração , Infecções por Coronavirus/epidemiologia , Pandemias , Pneumonia Viral/epidemiologia , COVID-19 , Humanos , Guias de Prática Clínica como Assunto , Saúde Pública , SARS-CoV-2 , Estados Unidos/epidemiologia , Universidades
8.
J Cell Sci ; 134(5)2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-33093239

RESUMO

The function of microRNAs (miRNAs) can be cell autonomous or communicated to other cell types and has been implicated in diverse biological processes. We previously demonstrated that miR-517a-3p (miR-517a), a highly expressed member of the chromosome 19 miRNA cluster (C19MC) that is transcribed almost exclusively in human trophoblasts, attenuates viral replication via induction of autophagy in non-trophoblastic recipient cells. However, the molecular mechanisms underlying these effects remain unknown. Here, we identified unc-13 homolog D (UNC13D) as a direct, autophagy-related gene target of miR-517a, leading to repression of UNC13D. In line with the antiviral activity of miR-517a, silencing UNC13D suppressed replication of vesicular stomatitis virus (VSV), whereas overexpression of UNC13D increased VSV levels, suggesting a role for UNC13D silencing in the antiviral activity of miR-517a. We also found that miR-517a activated NF-κB signaling in HEK-293XL cells expressing TLR8, but the effect was not specific to C19MC miRNA. Taken together, our results define mechanistic pathways that link C19MC miRNA with inhibition of viral replication.


Assuntos
Cromossomos Humanos Par 19 , Proteínas de Membrana , MicroRNAs , Humanos , MicroRNAs/genética , NF-kappa B/genética , Trofoblastos
9.
Proc Natl Acad Sci U S A ; 116(9): 3758-3763, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30808762

RESUMO

Echoviruses are amongst the most common causative agents of aseptic meningitis worldwide and are particularly devastating in the neonatal population, where they are associated with severe hepatitis, neurological disease, including meningitis and encephalitis, and even death. Here, we identify the neonatal Fc receptor (FcRn) as a pan-echovirus receptor. We show that loss of expression of FcRn or its binding partner beta 2 microglobulin (ß2M) renders cells resistant to infection by a panel of echoviruses at the stage of virus attachment, and that a blocking antibody to ß2M inhibits echovirus infection in cell lines and in primary human intestinal epithelial cells. We also show that expression of human, but not mouse, FcRn renders nonpermissive human and mouse cells sensitive to echovirus infection and that the extracellular domain of human FcRn directly binds echovirus particles and neutralizes infection. Lastly, we show that neonatal mice expressing human FcRn are more susceptible to echovirus infection by the enteral route. Our findings thus identify FcRn as a pan-echovirus receptor, which may explain the enhanced susceptibility of neonates to echovirus infections.


Assuntos
Enterovirus Humano B/genética , Antígenos de Histocompatibilidade Classe I/genética , Receptores Fc/genética , Receptores Virais/genética , Microglobulina beta-2/genética , Animais , Infecções por Echovirus/genética , Infecções por Echovirus/imunologia , Infecções por Echovirus/virologia , Enterovirus Humano B/patogenicidade , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Camundongos , Ligação Proteica , Microglobulina beta-2/imunologia
10.
J Virol ; 94(9)2020 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-32102874

RESUMO

Flaviviruses, including dengue virus (DENV) and Zika virus (ZIKV), rely heavily on the availability of endoplasmic reticulum (ER) membranes throughout their life cycle, and degradation of ER membranes restricts flavivirus replication. Accordingly, DENV and ZIKV restrict ER turnover by protease-mediated cleavage of reticulophagy regulator 1 (RETREG1), also known as FAM134B, an autophagy receptor responsible for targeted ER sheet degradation. Given that the induction of autophagy may play an important role in flavivirus replication, the antiviral role of RETREG1 suggests that specialized autophagic pathways may have differential effects on the flavivirus life cycle. We previously identified BPI fold-containing family B member 3 (BPIFB3) as a regulator of autophagy that negatively controls enterovirus replication. Here, we show that in contrast to enteroviruses, BPIFB3 functions as a positive regulator of DENV and ZIKV infection and that its RNA interference-mediated silencing inhibits the formation of viral replication organelles. Mechanistically, we show that depletion of BPIFB3 enhances RETREG1-dependent reticulophagy, leading to enhanced ER turnover and the suppression of viral replication. Consistent with this, the antiviral effects of BPIFB3 depletion can be reversed by RETREG1 silencing, suggesting a specific role for BPIFB3 in regulating ER turnover. These studies define BPIFB3 as a required host factor for both DENV and ZIKV replication and further contribute to our understanding of the requirements for autophagy during flavivirus infection.IMPORTANCE Flaviviruses and other arthropod-transmitted viruses represent a widespread global health problem, with limited treatment options currently available. Thus, a better understanding of the cellular requirements for their infection is needed. Both DENV and ZIKV rely on expansion of the endoplasmic reticulum (ER) and the induction of autophagy to establish productive infections. However, little is known regarding the interplay between the requirements for autophagy initiation during infection and the mechanisms used by these viruses to avoid clearance through the autophagic pathway. Our study highlights the importance of the host factor BPIFB3 in regulating flavivirus replication and further confirms that the RETREG1-dependent reticulophagy pathway is antiviral to both DENV and ZIKV.


Assuntos
Proteínas de Transporte/metabolismo , Flavivirus/fisiologia , Replicação Viral/fisiologia , Autofagia , Proteínas de Transporte/fisiologia , Linhagem Celular , Vírus da Dengue/fisiologia , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/fisiologia , Retículo Endoplasmático/virologia , Flavivirus/metabolismo , Infecções por Flavivirus/virologia , Interações Hospedeiro-Patógeno/genética , Humanos , Interferência de RNA , Zika virus/fisiologia , Infecção por Zika virus/virologia
11.
J Virol ; 94(24)2020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-32999020

RESUMO

Dengue virus (DENV) is a mosquito-borne virus that infects upward of 300 million people annually and has the potential to cause fatal hemorrhagic fever and shock. While the parameters contributing to dengue immunopathogenesis remain unclear, the collapse of redox homeostasis and the damage induced by oxidative stress have been correlated with the development of inflammation and progression toward the more severe forms of disease. In the present study, we demonstrate that the accumulation of reactive oxygen species (ROS) late after DENV infection (>24 hpi) resulted from a disruption in the balance between oxidative stress and the nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent antioxidant response. The DENV NS2B3 protease complex strategically targeted Nrf2 for degradation in a proteolysis-independent manner; NS2B3 licensed Nrf2 for lysosomal degradation. Impairment of the Nrf2 regulator by the NS2B3 complex inhibited the antioxidant gene network and contributed to the progressive increase in ROS levels, along with increased virus replication and inflammatory or apoptotic gene expression. By 24 hpi, when increased levels of ROS and antiviral proteins were observed, it appeared that the proviral effect of ROS overcame the antiviral effects of the interferon (IFN) response. Overall, these studies demonstrate that DENV infection disrupts the regulatory interplay between DENV-induced stress responses, Nrf2 antioxidant signaling, and the host antiviral immune response, thus exacerbating oxidative stress and inflammation in DENV infection.IMPORTANCE Dengue virus (DENV) is a mosquito-borne pathogen that threatens 2.5 billion people in more than 100 countries annually. Dengue infection induces a spectrum of clinical symptoms, ranging from classical dengue fever to severe dengue hemorrhagic fever or dengue shock syndrome; however, the complexities of DENV immunopathogenesis remain controversial. Previous studies have reported the importance of the transcription factor Nrf2 in the control of redox homeostasis and antiviral/inflammatory or death responses to DENV. Importantly, the production of reactive oxygen species and the subsequent stress response have been linked to the development of inflammation and progression toward the more severe forms of the disease. Here, we demonstrate that DENV uses the NS2B3 protease complex to strategically target Nrf2 for degradation, leading to a progressive increase in oxidative stress, inflammation, and cell death in infected cells. This study underlines the pivotal role of the Nrf2 regulatory network in the context of DENV infection.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Células A549 , Linhagem Celular , Dengue/virologia , Vírus da Dengue/genética , Regulação Viral da Expressão Gênica , Técnicas de Inativação de Genes , Células HEK293 , Heme Oxigenase-1/genética , Humanos , Interferons , Fator 2 Relacionado a NF-E2/genética , Espécies Reativas de Oxigênio , Transdução de Sinais/efeitos dos fármacos
12.
Trends Immunol ; 39(10): 848-858, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30219309

RESUMO

Barrier surfaces such as the epithelium lining the respiratory and gastrointestinal (GI) tracts, the endothelium comprising the blood-brain barrier (BBB), and placental trophoblasts provide key physical and immunological protection against viruses. These barriers utilize nonredundant mechanisms to suppress viral infections including the production of interferons (IFNs), which induce a strong antiviral state following receptor binding. However, whereas type I IFNs control infection systemically, type III IFNs (IFN-λs) control infection locally at barrier surfaces and are often preferentially induced by these cells. In this review we focus on the role of IFN-λ at barrier surfaces, focusing on the respiratory and GI tracts, the BBB, and the placenta, and on how these IFNs act to suppress viral infections.


Assuntos
Trato Gastrointestinal/imunologia , Interferons/metabolismo , Placenta/imunologia , Sistema Respiratório/imunologia , Fenômenos Fisiológicos da Pele/imunologia , Viroses/imunologia , Animais , Barreira Hematoencefálica , Feminino , Humanos , Gravidez , Viroses/tratamento farmacológico , Interferon lambda
13.
Am J Obstet Gynecol ; 225(6): 593.e1-593.e9, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34364845

RESUMO

Pregnant individuals infected with SARS-CoV-2 have higher rates of intensive care unit admission, oxygen requirement, need for mechanical ventilation, and death than nonpregnant individuals. Increased COVID-19 disease severity may be associated with an increased risk of viremia and placental infection. Maternal SARS-CoV-2 infection is also associated with pregnancy complications such as preeclampsia and preterm birth, which can be either placentally mediated or reflected in the placenta. Maternal viremia followed by placental infection may lead to maternal-fetal transmission (vertical), which affects 1% to 3% of exposed newborns. However, there is no agreed-upon or standard definition of placental infection. The National Institutes of Health/Eunice Kennedy Shriver National Institute of Child Health and Human Development convened a group of experts to propose a working definition of placental infection to inform ongoing studies of SARS-CoV-2 during pregnancy. Experts recommended that placental infection be defined using techniques that allow virus detection and localization in placental tissue by one or more of the following methods: in situ hybridization with antisense probe (detects replication) or a sense probe (detects viral messenger RNA) or immunohistochemistry to detect viral nucleocapsid or spike proteins. If the abovementioned methods are not possible, reverse transcription polymerase chain reaction detection or quantification of viral RNA in placental homogenates, or electron microscopy are alternative approaches. A graded classification for the likelihood of placental infection as definitive, probable, possible, and unlikely was proposed. Manuscripts reporting placental infection should describe the sampling method (location and number of samples collected), method of preservation of tissue, and detection technique. Recommendations were made for the handling of the placenta, examination, and sampling and the use of validated reagents and sample protocols (included as appendices).


Assuntos
Teste para COVID-19/métodos , COVID-19/diagnóstico , Doenças Placentárias/diagnóstico , Doenças Placentárias/virologia , Complicações Infecciosas na Gravidez/diagnóstico , Complicações Infecciosas na Gravidez/virologia , SARS-CoV-2 , Teste de Ácido Nucleico para COVID-19 , Consenso , Feminino , Guias como Assunto , Humanos , Imuno-Histoquímica , Hibridização In Situ , Microscopia Eletrônica , National Institute of Child Health and Human Development (U.S.) , Gravidez , Estados Unidos/epidemiologia
14.
Proc Natl Acad Sci U S A ; 114(35): 9433-9438, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28784796

RESUMO

Protecting the fetus from the hematogenous spread of viruses requires multifaceted layers of protection and relies heavily on trophoblasts, the fetal-derived cells that comprise the placental barrier. We showed previously that trophoblasts isolated from full-term placentas resist infection by diverse viruses, including Zika virus (ZIKV), and transfer this resistance to nonplacental cells through the activity of paracrine effectors, including the constitutive release of type III interferons (IFNs). Here, we developed 3D cell-line-based models of human syncytiotrophoblasts, cells that lie in direct contact with maternal blood, and show that these cells recapitulate the antiviral properties of primary trophoblasts through the constitutive release of type III IFNs (IFNλ1 and IFNλ2) and become resistant to ZIKV infection. In addition, using organotypic human midgestation chorionic villous explants, we show that syncytiotrophoblasts isolated from the second trimester of pregnancy also constitutively release type III IFNs and use these IFNs in autocrine and paracrine manners to restrict ZIKV infection. Collectively, these data provide important insights into the defense mechanisms used by syncytiotrophoblasts at various stages of human gestation to resist ZIKV infection and new human models to study the role of type III IFNs in the vertical transmission of ZIKV and other viruses associated with congenital disease.


Assuntos
Fibroblastos/imunologia , Fibroblastos/virologia , Infecção por Zika virus/imunologia , Zika virus/fisiologia , Linhagem Celular , Humanos , Relações Materno-Fetais , Nitrilas , Pirazóis/farmacologia , Pirimidinas
15.
Proc Natl Acad Sci U S A ; 114(7): 1672-1677, 2017 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28137842

RESUMO

Enteroviruses are among the most common viral infectious agents of humans and are primarily transmitted by the fecal-oral route. However, the events associated with enterovirus infections of the human gastrointestinal tract remain largely unknown. Here, we used stem cell-derived enteroids from human small intestines to study enterovirus infections of the intestinal epithelium. We found that enteroids were susceptible to infection by diverse enteroviruses, including echovirus 11 (E11), coxsackievirus B (CVB), and enterovirus 71 (EV71), and that contrary to an immortalized intestinal cell line, enteroids induced antiviral and inflammatory signaling pathways in response to infection in a virus-specific manner. Furthermore, using the Notch inhibitor dibenzazepine (DBZ) to drive cellular differentiation into secretory cell lineages, we show that although goblet cells resist E11 infection, enteroendocrine cells are permissive, suggesting that enteroviruses infect specific cell populations in the human intestine. Taken together, our studies provide insights into enterovirus infections of the human intestine, which could lead to the identification of novel therapeutic targets and/or strategies to prevent or treat infections by these highly clinically relevant viruses.


Assuntos
Infecções por Enterovirus/virologia , Enterovirus/fisiologia , Intestino Delgado/virologia , Organoides/virologia , Células CACO-2 , Linhagem da Célula/efeitos dos fármacos , Linhagem da Célula/genética , Células Cultivadas , Dibenzazepinas/farmacologia , Resistência à Doença/genética , Infecções por Enterovirus/metabolismo , Inibidores Enzimáticos/farmacologia , Perfilação da Expressão Gênica/métodos , Interações Hospedeiro-Patógeno , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/virologia , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Organoides/citologia , Organoides/metabolismo , Transdução de Sinais/genética
16.
Pediatr Transplant ; 23(1): e13303, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30338634

RESUMO

Recent years have brought a rise in newly emergent viral infections, primarily in the form of previously known arthropod-transmitted viruses that have increased significantly in both incidence and geographical range. Of particular note are DENV, CHIKV, and ZIKV, which are transmitted mostly by Aedes species of mosquitoes that exhibit a wide and increasing global distribution. Being important pathogens for the general population, these viruses have the potential to be devastating in the international transplant community, with graft rejection and death as possible outcomes of infection. In this review, we discuss the current state of knowledge for these viruses as well as repercussions of infection in the solid organ and HSCT population, with a focus, when possible, on pediatric patients.


Assuntos
Infecções por Arbovirus , Doenças Transmissíveis Emergentes , Transplante de Órgãos , Complicações Pós-Operatórias , Infecções por Arbovirus/diagnóstico , Infecções por Arbovirus/epidemiologia , Infecções por Arbovirus/etiologia , Infecções por Arbovirus/terapia , Criança , Doenças Transmissíveis Emergentes/diagnóstico , Doenças Transmissíveis Emergentes/epidemiologia , Doenças Transmissíveis Emergentes/etiologia , Doenças Transmissíveis Emergentes/terapia , Saúde Global , Humanos , Pediatria , Complicações Pós-Operatórias/diagnóstico , Complicações Pós-Operatórias/epidemiologia , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/terapia , Fatores de Risco
17.
Proc Natl Acad Sci U S A ; 113(6): 1642-7, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26729873

RESUMO

Clinical observations link respiratory virus infection and Pseudomonas aeruginosa colonization in chronic lung disease, including cystic fibrosis (CF) and chronic obstructive pulmonary disease. The development of P. aeruginosa into highly antibiotic-resistant biofilm communities promotes airway colonization and accounts for disease progression in patients. Although clinical studies show a strong correlation between CF patients' acquisition of chronic P. aeruginosa infections and respiratory virus infection, little is known about the mechanism by which chronic P. aeruginosa infections are initiated in the host. Using a coculture model to study the formation of bacterial biofilm formation associated with the airway epithelium, we show that respiratory viral infections and the induction of antiviral interferons promote robust secondary P. aeruginosa biofilm formation. We report that the induction of antiviral IFN signaling in response to respiratory syncytial virus (RSV) infection induces bacterial biofilm formation through a mechanism of dysregulated iron homeostasis of the airway epithelium. Moreover, increased apical release of the host iron-binding protein transferrin during RSV infection promotes P. aeruginosa biofilm development in vitro and in vivo. Thus, nutritional immunity pathways that are disrupted during respiratory viral infection create an environment that favors secondary bacterial infection and may provide previously unidentified targets to combat bacterial biofilm formation.


Assuntos
Biofilmes/crescimento & desenvolvimento , Imunidade , Fenômenos Fisiológicos da Nutrição , Pseudomonas aeruginosa/fisiologia , Infecções por Vírus Respiratório Sincicial/patologia , Vírus Sinciciais Respiratórios/fisiologia , Animais , Antivirais/farmacologia , Brônquios/patologia , Líquido da Lavagem Broncoalveolar , Fibrose Cística/microbiologia , Fibrose Cística/patologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/microbiologia , Células Epiteliais/virologia , Homeostase/efeitos dos fármacos , Humanos , Interferon beta/farmacologia , Ferro/farmacologia , Camundongos , Interações Microbianas/efeitos dos fármacos , Modelos Biológicos , Pseudomonas aeruginosa/efeitos dos fármacos , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transferrina/metabolismo
18.
Infect Immun ; 86(11)2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30181350

RESUMO

Tissues and organs provide the structural and biochemical landscapes upon which microbial pathogens and commensals function to regulate health and disease. While flat two-dimensional (2-D) monolayers composed of a single cell type have provided important insight into understanding host-pathogen interactions and infectious disease mechanisms, these reductionist models lack many essential features present in the native host microenvironment that are known to regulate infection, including three-dimensional (3-D) architecture, multicellular complexity, commensal microbiota, gas exchange and nutrient gradients, and physiologically relevant biomechanical forces (e.g., fluid shear, stretch, compression). A major challenge in tissue engineering for infectious disease research is recreating this dynamic 3-D microenvironment (biological, chemical, and physical/mechanical) to more accurately model the initiation and progression of host-pathogen interactions in the laboratory. Here we review selected 3-D models of human intestinal mucosa, which represent a major portal of entry for infectious pathogens and an important niche for commensal microbiota. We highlight seminal studies that have used these models to interrogate host-pathogen interactions and infectious disease mechanisms, and we present this literature in the appropriate historical context. Models discussed include 3-D organotypic cultures engineered in the rotating wall vessel (RWV) bioreactor, extracellular matrix (ECM)-embedded/organoid models, and organ-on-a-chip (OAC) models. Collectively, these technologies provide a more physiologically relevant and predictive framework for investigating infectious disease mechanisms and antimicrobial therapies at the intersection of the host, microbe, and their local microenvironments.


Assuntos
Microambiente Celular , Interações Hospedeiro-Patógeno , Mucosa Intestinal/fisiologia , Técnicas de Cultura de Órgãos/métodos , Organoides , Engenharia Tecidual/métodos , História do Século XX , História do Século XXI , Humanos , Modelos Biológicos , Técnicas de Cultura de Órgãos/história , Engenharia Tecidual/história
19.
FASEB J ; 31(7): 2760-2770, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28289056

RESUMO

During pregnancy, placental trophoblasts at the feto-maternal interface produce a broad repertoire of microRNA (miRNA) species. These species include miRNA from the primate-specific chromosome 19 miRNA cluster (C19MC), which is expressed nearly exclusively in the placenta. Trafficking of these miRNAs among the maternal, placental, and fetal compartments is unknown. To determine miRNA expression and trafficking patterns during pregnancy, we sequenced miRNAs in triads of human placenta and of maternal and fetal blood and found large subject-to-subject variability, with C19MC exhibiting compartment-specific expression. We therefore created humanized mice that transgenically express the entire 160-kb human C19MC locus or lentivirally express C19MC miRNA members selectively in the placenta. C19MC transgenic mice expressed a low level of C19MC miRNAs in diverse organs. When pregnant, female C19MC mice exhibited a strikingly elevated (>40-fold) expression of C19MC miRNA in the placenta, compared with other organs, that resembled C19MC miRNAs patterns in humans. Our mouse models showed that placental miRNA traffic primarily to the maternal circulation and that maternal miRNA can traffic to the placenta and even into the fetal compartment. These findings define an extraordinary means of nonhormonal, miRNA-based communication between the placenta and feto-maternal compartments.-Chang, G., Mouillet, J.-F., Mishima, T., Chu, T., Sadovsky, E., Coyne, C. B., Parks, W. T., Surti, U., Sadovsky, Y. Expression and trafficking of placental microRNAs at the feto-maternal interface.


Assuntos
Cromossomos Humanos Par 19/genética , Regulação da Expressão Gênica/fisiologia , Troca Materno-Fetal , MicroRNAs/metabolismo , Placenta/fisiologia , Animais , Transporte Biológico , Feminino , Humanos , Camundongos , Camundongos Transgênicos , MicroRNAs/genética , Gravidez
20.
J Immunol ; 196(9): 3877-86, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-27016603

RESUMO

Moloney leukemia virus 10, homolog (MOV10) is an IFN-inducible RNA helicase, associated with small RNA-induced silencing. In this article, we report that MOV10 exhibits antiviral activity, independent of its helicase function, against a number of positive- and negative-strand RNA viruses by enhancing type I IFN induction. Using a number of genome-edited knockout human cells, we show that IFN regulatory factor 3-mediated IFN induction and downstream IFN signaling through IFN receptor was necessary to inhibit virus replication by MOV10. MOV10 enhanced IFN regulatory factor 3-mediated transcription of IFN. However, this IFN induction by MOV10 was unique and independent of the known retinoic acid-inducible gene I/mitochondrial antiviral-signaling protein-mediated RNA-sensing pathway. Upon virus infection, MOV10 specifically required inhibitor of κB kinase ε, not TANK-binding kinase 1, for its antiviral activity. The important role of MOV10 in mediating antiviral signaling was further supported by the finding that viral proteases from picornavirus family specifically targeted MOV10 as a possible innate immune evasion mechanism. These results establish MOV10, an evolutionary conserved protein involved in RNA silencing, as an antiviral gene against RNA viruses that uses an retinoic acid-inducible gene I-like receptor-independent pathway to enhance IFN response.


Assuntos
Infecções por Cardiovirus/imunologia , Vírus da Encefalomiocardite/imunologia , RNA Helicases/metabolismo , Infecções por Rhabdoviridae/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Evasão da Resposta Imune , Imunidade Inata , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/metabolismo , RNA Helicases/genética , Interferência de RNA , RNA Viral/genética , Receptores do Ácido Retinoico/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA