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.
J Med Virol ; 96(7): e29783, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38965890

RESUMO

Many COVID-19 patients suffer from gastrointestinal symptoms and impaired intestinal barrier function is thought to play a key role in Long COVID. Despite its importance, the impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on intestinal epithelia is poorly understood. To address this, we established an intestinal barrier model integrating epithelial Caco-2 cells, mucus-secreting HT29 cells and Raji cells. This gut epithelial model allows efficient differentiation of Caco-2 cells into microfold-like cells, faithfully mimics intestinal barrier function, and is highly permissive to SARS-CoV-2 infection. Early strains of SARS-CoV-2 and the Delta variant replicated with high efficiency, severely disrupted barrier function, and depleted tight junction proteins, such as claudin-1, occludin, and ZO-1. In comparison, Omicron subvariants also depleted ZO-1 from tight junctions but had fewer damaging effects on mucosal integrity and barrier function. Remdesivir, the fusion inhibitor EK1 and the transmembrane serine protease 2 inhibitor Camostat inhibited SARS-CoV-2 replication and thus epithelial barrier damage, while the Cathepsin inhibitor E64d was ineffective. Our results support that SARS-CoV-2 disrupts intestinal barrier function but further suggest that circulating Omicron variants are less damaging than earlier viral strains.


Assuntos
COVID-19 , Mucosa Intestinal , SARS-CoV-2 , Junções Íntimas , Replicação Viral , Humanos , SARS-CoV-2/patogenicidade , Células CACO-2 , COVID-19/virologia , COVID-19/patologia , Mucosa Intestinal/virologia , Mucosa Intestinal/patologia , Junções Íntimas/virologia , Alanina/análogos & derivados , Proteína da Zônula de Oclusão-1/metabolismo , Proteína da Zônula de Oclusão-1/genética , Antivirais/farmacologia , Células HT29 , Ocludina/metabolismo , Ocludina/genética , Monofosfato de Adenosina/análogos & derivados
2.
J Neurovirol ; 30(1): 22-38, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38189894

RESUMO

Neurotropic viruses can infiltrate the CNS by crossing the blood-brain barrier (BBB) through various mechanisms including paracellular, transcellular, and "Trojan horse" mechanisms during leukocyte diapedesis. These viruses belong to several families, including retroviruses; human immunodeficiency virus type 1 (HIV-1), flaviviruses; Japanese encephalitis (JEV); and herpesviruses; herpes simplex virus type 1 (HSV-1), Epstein-Barr virus (EBV), and mouse adenovirus 1 (MAV-1). For entering the brain, viral proteins act upon the tight junctions (TJs) between the brain microvascular endothelial cells (BMECs). For instance, HIV-1 proteins, such as glycoprotein 120, Nef, Vpr, and Tat, disrupt the BBB and generate a neurotoxic effect. Recombinant-Tat triggers amendments in the BBB by decreasing expression of the TJ proteins such as claudin-1, claudin-5, and zona occludens-1 (ZO-1). Thus, the breaching of BBB has been reported in myriad of neurological diseases including multiple sclerosis (MS). Neurotropic viruses also exhibit molecular mimicry with several myelin sheath proteins, i.e., antibodies against EBV nuclear antigen 1 (EBNA1) aa411-426 cross-react with MBP and EBNA1 aa385-420 was found to be associated with MS risk haplotype HLA-DRB1*150. Notably, myelin protein epitopes (PLP139-151, MOG35-55, and MBP87-99) are being used to generate model systems for MS such as experimental autoimmune encephalomyelitis (EAE) to understand the disease mechanism and therapeutics. Viruses like Theiler's murine encephalomyelitis virus (TMEV) are also commonly used to generate EAE. Altogether, this review provide insights into the viruses' association with BBB leakiness and MS along with possible mechanistic details which could potentially use for therapeutics.


Assuntos
Barreira Hematoencefálica , Esclerose Múltipla , Barreira Hematoencefálica/virologia , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Humanos , Animais , Esclerose Múltipla/virologia , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Camundongos , Junções Íntimas/virologia , Junções Íntimas/metabolismo , Permeabilidade Capilar , Células Endoteliais/virologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia
3.
Tissue Barriers ; 9(2): 1883965, 2021 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-33632074

RESUMO

The apical junctional complexes (AJCs) of airway epithelial cells are a key component of the innate immune system by creating barriers to pathogens, inhaled allergens, and environmental particles. AJCs form between adjacent cells and consist of tight junctions (TJs) and adherens junctions (AJs). Respiratory viruses have been shown to target various components of the AJCs, leading to airway epithelial barrier dysfunction by different mechanisms. Virus-induced epithelial permeability may allow for allergens and bacterial pathogens to subsequently invade. In this review, we discuss the pathophysiologic mechanisms leading to disruption of AJCs and the potential ensuing ramifications. We focus on the following viruses that affect the pulmonary system: respiratory syncytial virus, rhinovirus, influenza viruses, immunodeficiency virus, and other viruses such as coxsackievirus, adenovirus, coronaviruses, measles, parainfluenza virus, bocavirus, and vaccinia virus. Understanding the mechanisms by which viruses target the AJC and impair barrier function may help design therapeutic innovations to treat these infections.


Assuntos
Junções Íntimas/virologia , Viroses/fisiopatologia , Animais , Humanos , Camundongos
4.
Biochim Biophys Acta Biomembr ; 1862(7): 183296, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32268133

RESUMO

The hepatitis C virus (HCV) is a major cause of liver diseases ranging from liver inflammation to advanced liver diseases like cirrhosis and hepatocellular carcinoma (HCC). HCV infection is restricted to the liver, and more specifically to hepatocytes, which represent around 80% of liver cells. The mechanism of HCV entry in human hepatocytes has been extensively investigated since the discovery of the virus 30 years ago. The entry mechanism is a multi-step process relying on several host factors including heparan sulfate proteoglycan (HSPG), low density lipoprotein receptor (LDLR), tetraspanin CD81, Scavenger Receptor class B type I (SR-BI), Epidermal Growth Factor Receptor (EGFR) and Niemann-Pick C1-like 1 (NPC1L1). Moreover, in order to establish a persistent infection, HCV entry is dependent on the presence of tight junction (TJ) proteins Claudin-1 (CLDN1) and Occludin (OCLN). In the liver, tight junction proteins play a role in architecture and homeostasis including sealing the apical pole of adjacent cells to form bile canaliculi and separating the basolateral domain drained by sinusoidal blood flow. In this review, we will highlight the role of liver tight junction proteins in HCV infection, and we will discuss the potential targeted therapeutic approaches to improve virus eradication.


Assuntos
Hepatite C/genética , Interações Hospedeiro-Patógeno/genética , Junções Íntimas/genética , Internalização do Vírus , Hepacivirus/genética , Hepacivirus/patogenicidade , Hepatite C/virologia , Hepatócitos/virologia , Humanos , Fígado/patologia , Fígado/virologia , Proteínas de Membrana Transportadoras/genética , Ocludina/genética , Receptores de LDL/genética , Tetraspanina 28/genética , Junções Íntimas/virologia
5.
Virology ; 531: 93-99, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30856486

RESUMO

Tight junctions enable epithelial cells to form physical barriers that act as an innate immune defense against respiratory infection. However, the involvement of tight junction molecules in paramyxovirus infections, which include various respiratory pathogens, has not been examined in detail. Human parainfluenza virus type 2 (hPIV2) infects airway epithelial cells and causes respiratory illness. In the present study, we found that hPIV2 infection of cultured cells induces expression of claudin-1 (CLDN1), an essential component of tight junctions. This induction seemed to be intrinsically restricted by V, an accessory protein that modulates various host responses, to enable efficient virus propagation. By generating CLDN1 over-expressing and knockout cell lines, we showed that CLDN1 is involved in the restriction of hPIV2 spread via cell-to-cell contact. Taken together, we identified CLDN1 an inhibitory factor for hPIV2 dissemination, and that its V protein acts to counter this.


Assuntos
Claudina-1/metabolismo , Vírus da Parainfluenza 2 Humana/fisiologia , Infecções por Rubulavirus/metabolismo , Infecções por Rubulavirus/virologia , Claudina-1/genética , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Interações Hospedeiro-Patógeno , Humanos , Vírus da Parainfluenza 2 Humana/genética , Infecções por Rubulavirus/genética , Junções Íntimas/metabolismo , Junções Íntimas/virologia , Proteínas Virais/genética , Proteínas Virais/metabolismo
6.
Am J Physiol Lung Cell Mol Physiol ; 316(2): L358-L368, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30489157

RESUMO

Respiratory syncytial virus (RSV) is a major cause of hospitalization for infants and young children worldwide. RSV is known to infect epithelial cells and increase the permeability of model airway epithelial monolayers in vitro. We hypothesized that RSV infection also induces airway barrier dysfunction in vivo. C57BL/6 mice were intranasally inoculated with RSV, and on day 4 post-inoculation were examined for viral replication, lung inflammation, and barrier integrity as well as the structure and molecular composition of epithelial junctions. In parallel, primary mouse tracheal epithelial cells (mTEC) were cultured for in vitro studies. RSV-infected mice lost weight and showed significant peribronchial inflammation compared with noninfected controls and UV-inactivated RSV-inoculated animals. RSV infection increased the permeability of the airway epithelial barrier and altered the molecular composition of epithelial tight junctions. The observed RSV-induced barrier disruption was accompanied by decreased expression of several tight-junction proteins and accumulation of cleaved extracellular fragments of E-cadherin in bronchoalveolar lavage and mTEC supernatants. Similarly, in vitro RSV infection of mTEC monolayers resulted in enhanced permeability and disruption of tight-junction structure. Furthermore, incubation of mTEC monolayers with a recombinant fragment of E-cadherin caused tight-junction disassembly. Taken together, these data indicate that RSV infection leads to airway barrier dysfunction in vivo, mediated by either decreased expression or cleavage of junctional proteins. Our observations provide further insights into the pathophysiology of RSV infection and provide a rationale for development of barrier-protecting agents to alleviate the pathogenic effects of RSV infection.


Assuntos
Células Epiteliais/virologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/patogenicidade , Junções Íntimas/virologia , Animais , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Camundongos Endogâmicos C57BL , Sistema Respiratório/metabolismo , Junções Íntimas/metabolismo
7.
J Virol ; 93(4)2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30463963

RESUMO

The genus Sapovirus belongs to the family Caliciviridae, and its members are common causative agents of severe acute gastroenteritis in both humans and animals. Some caliciviruses are known to use either terminal sialic acids or histo-blood group antigens as attachment factors and/or cell surface proteins, such as CD300lf, CD300ld, and junctional adhesion molecule 1 of tight junctions (TJs), as receptors. However, the roles of TJs and their proteins in sapovirus entry have not been examined. In this study, we found that porcine sapovirus (PSaV) significantly decreased transepithelial electrical resistance and increased paracellular permeability early in infection of LLC-PK cells, suggesting that PSaV dissociates TJs of cells. This led to the interaction between PSaV particles and occludin, which traveled in a complex into late endosomes via Rab5- and Rab7-dependent trafficking. Inhibition of occludin using small interfering RNA (siRNA), a specific antibody, or a dominant-negative mutant significantly blocked the entry of PSaV. Transient expression of occludin in nonpermissive Chinese hamster ovary (CHO) cells conferred susceptibility to PSaV, but only for a limited time. Although claudin-1, another TJ protein, neither directly interacted nor was internalized with PSaV particles, it facilitated PSaV entry and replication in the LLC-PK cells. We conclude that PSaV particles enter LLC-PK cells by binding to occludin as a coreceptor in PSaV-dissociated TJs. PSaV and occludin then form a complex that moves to late endosomes via Rab5- and Rab7-dependent trafficking. In addition, claudin-1 in the TJs opened by PSaV infection facilitates PSaV entry and infection as an entry factor.IMPORTANCE Sapoviruses (SaVs) cause severe acute gastroenteritis in humans and animals. Although they replicate in intestinal epithelial cells, which are tightly sealed by apical-junctional complexes, such as tight junctions (TJs), the mechanisms by which SaVs hijack TJs and their proteins for successful entry and infection remain largely unknown. Here, we demonstrate that porcine SaVs (PSaVs) induce early dissociation of TJs, allowing them to bind to the TJ protein occludin as a functional coreceptor. PSaVs then travel in a complex with occludin into late endosomes through Rab5- and Rab7-dependent trafficking. Claudin-1, another TJ protein, does not directly interact with PSaV but facilitates the entry of PSaV into cells as an entry factor. This work contributes to our understanding of the entry of SaV and other caliciviruses into cells and may aid in the development of efficient and affordable drugs to treat SaV infections.


Assuntos
Ocludina/metabolismo , Sapovirus/fisiologia , Junções Íntimas/virologia , Animais , Células CHO , Cricetulus , Endossomos/metabolismo , Células Epiteliais/virologia , Gastroenterite/virologia , Células LLC-PK1 , Ocludina/fisiologia , Sapovirus/metabolismo , Sapovirus/patogenicidade , Suínos/virologia , Junções Íntimas/metabolismo , Viroses/metabolismo
8.
Laryngoscope ; 128(12): E393-E401, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30325507

RESUMO

OBJECTIVE: Rhinoviruses (RV), which are responsible for the majority of common colds, induce mucus overproduction, increased vascular permeability, and secondary bacterial infection. These symptoms are primarily caused by barrier function disruption, which is controlled by intercellular junctions. In this study, we investigated whether reactive oxygen species (ROS) are closely involved in tight junction disruption of primary human nasal epithelial (HNE) cells induced by infection of RV . METHODS AND RESULTS: Incubation with RV resulted in disruption of tight junction proteins (ZO-1, E-cadherin, claudin-1, and occludin) in HNE cells. Pretreatment with diphenylene iodonium (DPI) decreased RV-induced disruption of tight junction in HNE cells. RV-induced generation of ROS was diminished by DPI. However, rotenone was not inhibited in HNE cells following incubation with RV. Rhinoviruses resulted in a marked decrease in protein phosphatases activity and an increase in protein tyrosine phosphorylation levels in HNE cells. Diphenylene iodonium inhibited the RV-induced inactivation of phosphatases and phosphorylation of protein tyrosine. In addition, inhibition of protein tyrosine phosphatases with phenylarsine oxide resulted in a marked decrease in protein phosphatase activity and disruption of tight junction proteins in HNE cells. CONCLUSION: Our results suggest that ROS-mediated inhibition of phosphatases plays a crucial role in disruption of tight junctions in HNE cells by RV. The data suggest that RV infection may damage nasal epithelial barrier function. LEVEL OF EVIDENCE: NA Laryngoscope, 128:E393-E401, 2018.


Assuntos
Resfriado Comum/virologia , Células Epiteliais/virologia , Espécies Reativas de Oxigênio/metabolismo , Rhinovirus/metabolismo , Junções Íntimas/virologia , Células Epiteliais/efeitos dos fármacos , Humanos , Mucosa Nasal/citologia , Mucosa Nasal/virologia , Oniocompostos/farmacologia , Rhinovirus/efeitos dos fármacos
9.
Exp Cell Res ; 371(2): 322-329, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30142324

RESUMO

Tight junctions (TJs) alteration is commonly seen in airway inflammatory diseases. Oncostatin M (OSM) is an inflammatory mediator associated with chronic rhinosinusitis with nasal polyps (CRSwNP). We have previously shown that human nasal epithelial cells (hNECs) are highly permissive cells for influenza A virus (IAV). However, its role in TJs alteration and the effects of IAV on inducing OSM expression in nasal epithelium remains to be further investigated. In this study, OSM and TJs expression was measured and compared between inferior turbinate from healthy controls and nasal polyps from CRSwNP. Additionally, hNECs cultured at air-liquid interface (ALI) were infected with H3N2 influenza virus to study the role of influenza virus in inducing epithelial OSM expression as a possible means of exacerbation. The expression of ZO-1, claudin-1, and occludin was markedly decreased and correlated negatively with that of OSM in CRSwNP. By using the in vitro hNEC model, H3N2 infection resulted in significantly increased OSM expression (2.2-, 4.7- and 3.9-fold higher at 8, 24, and 48 h post-infection vs. mock infection). Furthermore, OSM is found to co-localize with ciliated and goblet cells in hNECs infected with H3N2 influenza virus. Our findings demonstrated that increased OSM expression is implicated in CRSwNP as a possible mechanism of TJs' impairment, which can be further augmented following influenza infection via epithelial OSM expression, possibly contributing to exacerbations.


Assuntos
Vírus da Influenza A Subtipo H3N2/genética , Influenza Humana/genética , Mucosa Nasal/metabolismo , Pólipos Nasais/genética , Oncostatina M/genética , Rinite/genética , Sinusite/genética , Adulto , Estudos de Casos e Controles , Diferenciação Celular , Doença Crônica , Claudina-1/genética , Claudina-1/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Células Epiteliais/virologia , Feminino , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Humanos , Vírus da Influenza A Subtipo H3N2/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H3N2/metabolismo , Influenza Humana/metabolismo , Influenza Humana/patologia , Influenza Humana/virologia , Masculino , Pessoa de Meia-Idade , Mucosa Nasal/patologia , Mucosa Nasal/virologia , Pólipos Nasais/metabolismo , Pólipos Nasais/patologia , Pólipos Nasais/virologia , Ocludina/genética , Ocludina/metabolismo , Oncostatina M/metabolismo , Cultura Primária de Células , Rinite/metabolismo , Rinite/patologia , Rinite/virologia , Transdução de Sinais , Sinusite/metabolismo , Sinusite/patologia , Sinusite/virologia , Junções Íntimas/metabolismo , Junções Íntimas/patologia , Junções Íntimas/virologia , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo
10.
Clin Exp Immunol ; 190(3): 351-359, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28856667

RESUMO

Respiratory syncytial virus (RSV) is an important risk factor of asthma development and is responsible for severe respiratory tract infections. However, the influence of RSV infection on barrier function of bronchial epithelial cells in vitro and in vivo is still unclear. The aim of this study was to analyse the role of RSV in tight junction (TJ) regulation and to compare epithelial integrity between asthmatic and healthy individuals upon RSV infection. Healthy and asthmatic human bronchial epithelial cells (HBECs) were differentiated at air-liquid interface (ALI) and infected with RSV and ultraviolet (UV)-irradiated RSV. TJ expression and their integrity were analysed by quantitative polymerase chain reaction (qPCR), transepithelial resistance (TER) and paracellular flux. To determine the effect in vivo, BALB/c mice were infected intranasally with RSV or UV-irradiated RSV A2. Bronchoalveolar lavage and TJ integrity were analysed on days 1, 2, 4 and 6 post-infection by qPCR, bioplex and confocal microscopy. RSV increased barrier integrity in ALI cultures of HBEC from healthy subjects, but no effect was found in HBECs from asthmatics. This was not associated with an increase in TJ mRNA expression. In vivo, RSV induced lung inflammation in mice and down-regulated claudin-1 and occludin mRNA expression in whole lungs. Surprisingly, RSV infection was not observed in bronchial epithelial cells, but was found in the lung parenchyma. Decreased expression of occludin upon RSV infection was visible in mouse bronchial epithelial cells in confocal microscopy. However, there was no regulation of claudin-1 and claudin-7 at protein level.


Assuntos
Brônquios/imunologia , Células Epiteliais/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Junções Íntimas/imunologia , Animais , Asma/etiologia , Asma/imunologia , Asma/patologia , Asma/virologia , Brônquios/patologia , Brônquios/virologia , Lavagem Broncoalveolar , Células Cultivadas , Claudina-1/imunologia , Claudinas/imunologia , Células Epiteliais/patologia , Células Epiteliais/virologia , Regulação da Expressão Gênica/imunologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Vírus Respiratório Sincicial/patologia , Fatores de Risco , Junções Íntimas/patologia , Junções Íntimas/virologia
11.
J Virol ; 91(10)2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28275187

RESUMO

Porcine epidemic diarrhea virus (PEDV), the causative agent of porcine epidemic diarrhea, has caused huge economic losses in pig-producing countries. Although PEDV was long believed to replicate in the intestinal epithelium by using aminopeptidase N as a receptor, the mechanisms of PEDV infection are not fully characterized. In this study, we found that PEDV infection of epithelial cells results in disruption of the tight junctional distribution of occludin to its intracellular location. Overexpression of occludin in target cells makes them more susceptible to PEDV infection, whereas ablation of occludin expression by use of small interfering RNA (siRNA) in target cells significantly reduces their susceptibility to virus infection. However, the results observed with occludin siRNA indicate that occludin is not required for virus attachment. We conclude that occludin plays an essential role in PEDV infection at the postbinding stages. Furthermore, we observed that macropinocytosis inhibitors blocked occludin internalization and virus entry, indicating that virus entry and occludin internalization are closely coupled. However, the macropinocytosis inhibitors could not impede virus replication once the virus had entered host cells. This suggests that occludin internalization by macropinocytosis or a macropinocytosis-like process is involved in the virus entry events. Immunofluorescence confocal microscopy showed that PEDV was trapped at cellular junctional regions upon macropinocytosis inhibitor treatment, indicating that occludin may serve as a scaffold in the vicinity of virus entry. Collectively, these data show that occludin plays an essential role in PEDV infection during late entry events. Our observation may provide novel insights into PEDV infection and related pathogenesis.IMPORTANCE Tight junctions are highly specialized membrane domains whose main function is to attach adjacent cells to each other, thereby forming intercellular seals. Here we investigate, for the first time, the role of the tight junction protein occludin in PEDV infection. We observed that PEDV infection induced the internalization of occludin. By using genetic modification methods, we demonstrate that occludin plays an essential role in PEDV infection. Moreover, PEDV entry and occludin internalization seem to be closely coupled. Our findings reveal a new mechanism of PEDV infection.


Assuntos
Ocludina/metabolismo , Vírus da Diarreia Epidêmica Suína/fisiologia , Junções Íntimas/química , Ligação Viral , Internalização do Vírus , Animais , Linhagem Celular , Chlorocebus aethiops , Citoplasma/metabolismo , Células Epiteliais/virologia , Ocludina/deficiência , Ocludina/genética , Vírus da Diarreia Epidêmica Suína/efeitos dos fármacos , Vírus da Diarreia Epidêmica Suína/patogenicidade , RNA Interferente Pequeno , Suínos , Junções Íntimas/patologia , Junções Íntimas/virologia , Células Vero , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
12.
Mediators Inflamm ; 2017: 3164375, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29515292

RESUMO

Recent clinical reports indicate that infection with dengue virus (DENV) commonly has ocular manifestations. The most serious threat to vision is dengue retinopathy, including retinal vasculopathy and macular edema. Mechanisms of retinopathy are unstudied, but observations in patients implicate retinal pigment epithelial cells and retinal endothelial cells. Human retinal cells were inoculated with DENV-2 and monitored for up to 72 hours. Epithelial and endothelial cells supported DENV replication and release, but epithelial cells alone demonstrated clear cytopathic effect, and infection was more productive in those cells. Infection induced type I interferon responses from both cells, but this was stronger in epithelial cells. Endothelial cells increased expression of adhesion molecules, with sustained overexpression of vascular adhesion molecule-1. Transcellular impedance decreased for epithelial monolayers, but not endothelial monolayers, coinciding with cytopathic effect. This reduction was accompanied by disorganization of intracellular filamentous-actin and decreased expression of junctional molecules, zonula occludens 1, and catenin-ß1. Changes in endothelial expression of adhesion molecules are consistent with the retinal vasculopathy seen in patients infected with DENV; decreases in epithelial junctional protein expression, paralleling loss of integrity of the epithelium, provide a molecular basis for DENV-associated macular edema. These molecular processes present potential therapeutic targets for vision-threatening dengue retinopathy.


Assuntos
Vírus da Dengue/patogenicidade , Células Endoteliais/virologia , Retina/citologia , Linhagem Celular , Células Cultivadas , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Humanos , Imuno-Histoquímica , Junções Íntimas/imunologia , Junções Íntimas/virologia , Replicação Viral/fisiologia
13.
J Gen Virol ; 98(3): 405-412, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27983476

RESUMO

Preventing virally induced liver disease begins with an understanding of the host factors that define susceptibility to infection. Hepatitis C virus (HCV) is a global health issue, with an estimated 170 million infected individuals at risk of developing liver disease including fibrosis and hepatocellular carcinoma. The liver is the major reservoir supporting HCV replication and this hepatocellular tropism is defined by HCV engagement of cellular entry receptors. Hepatocytes are polarized in vivo and this barrier function limits HCV entry. We previously reported that activated macrophages promote HCV entry into polarized hepatocytes via a TNF-α-dependent process; however, the underlying mechanism was not defined. In this study, we show that several TNF superfamily members, including TNF-α, TNF-ß, TWEAK and LIGHT, promote HCV entry via NF-κB-mediated activation of myosin light chain kinase (MLCK) and disruption of tight junctions. These observations support a model where HCV hijacks an inflammatory immune response to stimulate infection and uncovers a role for NF-κB-MLCK signalling in maintaining hepatocellular tight junctions.


Assuntos
Hepacivirus/fisiologia , Hepatite C/virologia , Fígado/virologia , Quinase de Cadeia Leve de Miosina/metabolismo , NF-kappa B/metabolismo , Fatores de Necrose Tumoral/metabolismo , Internalização do Vírus , Carcinoma Hepatocelular/virologia , Ativação Enzimática , Hepatite C/metabolismo , Hepatócitos/virologia , Humanos , Fígado/metabolismo , Cirrose Hepática/virologia , Neoplasias Hepáticas/virologia , Transdução de Sinais , Junções Íntimas/metabolismo , Junções Íntimas/virologia , Fator de Transcrição RelA/metabolismo
14.
J Neurochem ; 140(2): 216-230, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27718541

RESUMO

Developing therapies for central nervous system (CNS) diseases is exceedingly difficult because of the blood-brain barrier (BBB). Notably, emerging technologies may provide promising new options for the treatment of CNS disorders. Adeno-associated virus serotype 9 (AAV9) has been shown to transduce cells in the CNS following intravascular administration in rodents, cats, pigs, and non-human primates. These results suggest that AAV9 is capable of crossing the BBB. However, mechanisms that govern AAV9 transendothelial trafficking at the BBB remain unknown. Furthermore, possibilities that AAV9 may transduce brain endothelial cells or affect BBB integrity still require investigation. Using primary human brain microvascular endothelial cells as a model of the human BBB, we performed transduction and transendothelial trafficking assays comparing AAV9 to AAV2, a serotype that does not cross the BBB or transduce endothelial cells effectively in vivo. Results of our in vitro studies indicate that AAV9 penetrates brain microvascular endothelial cells barriers more effectively than AAV2, but has reduced transduction efficiency. In addition, our data suggest that (i) AAV9 penetrates endothelial barriers through an active, cell-mediated process, and (ii) AAV9 fails to disrupt indicators of BBB integrity such as transendothelial electrical resistance, tight junction protein expression/localization, and inflammatory activation status. Overall, this report shows how human brain endothelial cells configured in BBB models can be utilized for evaluating transendothelial movement and transduction kinetics of various AAV capsids. Importantly, the use of a human in vitro BBB model can provide import insight into the possible effects that candidate AVV gene therapy vectors may have on the status of BBB integrity. Read the Editorial Highlight for this article on page 192.


Assuntos
Barreira Hematoencefálica/virologia , Encéfalo/virologia , Movimento Celular/fisiologia , Dependovirus , Células Endoteliais/virologia , Junções Íntimas/virologia , Barreira Hematoencefálica/citologia , Encéfalo/metabolismo , Células Cultivadas , Humanos , Transcitose/fisiologia
15.
FASEB J ; 30(3): 1234-46, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26601824

RESUMO

HIV invades the brain early after infection; however, its interactions with the cells of the blood-brain barrier (BBB) remain poorly understood. Our goal was to evaluate the role of occludin, one of the tight junction proteins that regulate BBB functions in HIV infection of BBB pericytes. We provide evidence that occludin levels largely control the metabolic responses of human pericytes to HIV. Occludin in BBB pericytes decreased by 10% during the first 48 h after HIV infection, correlating with increased nuclear translocation of the gene repressor C-terminal-binding protein (CtBP)-1 and NFκB-p65 activation. These changes were associated with decreased expression and activation of the class III histone deacetylase sirtuin (SIRT)-1. Occludin levels recovered 96 h after infection, restoring SIRT-1 and reducing HIV transcription to 20% of its highest values. We characterized occludin biochemically as a novel NADH oxidase that controls the expression and activation of SIRT-1. The inverse correlation between occludin and HIV transcription was then replicated in human primary macrophages and differentiated monocytic U937 cells, in which occludin silencing resulted in 75 and 250% increased viral transcription, respectively. Our work shows that occludin has previously unsuspected metabolic properties and is a target of HIV infection, opening the possibility of designing novel pharmacological approaches to control HIV transcription.


Assuntos
Barreira Hematoencefálica/virologia , Infecções por HIV/virologia , HIV/genética , Ocludina/metabolismo , Oxirredutases do Álcool/metabolismo , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Encéfalo/virologia , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , HIV/metabolismo , Infecções por HIV/metabolismo , Humanos , NF-kappa B/metabolismo , Pericitos/metabolismo , Pericitos/virologia , Sirtuína 1/metabolismo , Junções Íntimas/metabolismo , Junções Íntimas/virologia , Transcrição Gênica/genética
16.
J Infect Dis ; 211(3): 383-93, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25149763

RESUMO

Viruses that cause systemic disease often spread through the bloodstream to infect target tissues. Although viremia is an important step in the pathogenesis of many viruses, how viremia is established is not well understood. Reovirus has been used to dissect mechanisms of viral pathogenesis and is being evaluated in clinical trials as an oncolytic agent. After peroral entry into mice, reovirus replicates within the gastrointestinal tract and disseminates systemically via hematogenous or neural routes. Junctional adhesion molecule-A (JAM-A) is a tight junction protein that serves as a receptor for reovirus. JAM-A is required for establishment of viremia and viral spread to sites of secondary replication. JAM-A also is expressed on the surface of circulating hematopoietic cells. To determine contributions of endothelial and hematopoietic JAM-A to reovirus dissemination and pathogenesis, we generated strains of mice with altered JAM-A expression in these cell types and assessed bloodstream spread of reovirus strain type 1 Lang (T1L), which disseminates solely by hematogenous routes. We found that endothelial JAM-A but not hematopoietic JAM-A facilitates reovirus T1L bloodstream entry and egress. Understanding how viruses establish viremia may aid in development of inhibitors of this critical step in viral pathogenesis and foster engineering of improved oncolytic viral vectors.


Assuntos
Moléculas de Adesão Celular/metabolismo , Células Endoteliais/metabolismo , Receptores de Superfície Celular/metabolismo , Reoviridae/metabolismo , Junções Íntimas/metabolismo , Viremia/metabolismo , Animais , Células Cultivadas , Células Endoteliais/virologia , Fibroblastos/metabolismo , Fibroblastos/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Virais/metabolismo , Junções Íntimas/virologia , Viremia/virologia
17.
J Cell Sci ; 127(Pt 16): 3401-13, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25125573

RESUMO

Tight junctions are a component of the epithelial junctional complex, and they form the paracellular diffusion barrier that enables epithelial cells to create cellular sheets that separate compartments with different compositions. The assembly and function of tight junctions are intimately linked to the actomyosin cytoskeleton and, hence, are under the control of signalling mechanisms that regulate cytoskeletal dynamics. Tight junctions not only receive signals that guide their assembly and function, but transmit information to the cell interior to regulate cell proliferation, migration and survival. As a crucial component of the epithelial barrier, they are often targeted by pathogenic viruses and bacteria, aiding infection and the development of disease. In this Commentary, we review recent progress in the understanding of the molecular signalling mechanisms that drive junction assembly and function, and the signalling processes by which tight junctions regulate cell behaviour and survival. We also discuss the way in which junctional components are exploited by pathogenic viruses and bacteria, and how this might affect junctional signalling mechanisms.


Assuntos
Fenômenos Fisiológicos Bacterianos , Diferenciação Celular , Células Epiteliais/metabolismo , Transdução de Sinais , Junções Íntimas/metabolismo , Fenômenos Fisiológicos Virais , Animais , Bactérias/patogenicidade , Células Epiteliais/citologia , Células Epiteliais/microbiologia , Células Epiteliais/virologia , Humanos , Junções Íntimas/microbiologia , Junções Íntimas/virologia , Vírus/patogenicidade
18.
PLoS Pathog ; 10(6): e1004198, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24968145

RESUMO

Effective antiretroviral therapy (ART) dramatically reduces AIDS-related complications, yet the life expectancy of long-term ART-treated HIV-infected patients remains shortened compared to that of uninfected controls, due to increased risk of non-AIDS related morbidities. Many propose that these complications result from translocated microbial products from the gut that stimulate systemic inflammation--a consequence of increased intestinal paracellular permeability that persists in this population. Concurrent intestinal immunodeficiency and structural barrier deterioration are postulated to drive microbial translocation, and direct evidence of intestinal epithelial breakdown has been reported in untreated pathogenic SIV infection of rhesus macaques. To assess and characterize the extent of epithelial cell damage in virally-suppressed HIV-infected patients, we analyzed intestinal biopsy tissues for changes in the epithelium at the cellular and molecular level. The intestinal epithelium in the HIV gut is grossly intact, exhibiting no decreases in the relative abundance and packing of intestinal epithelial cells. We found no evidence for structural and subcellular localization changes in intestinal epithelial tight junctions (TJ), but observed significant decreases in the colonic, but not terminal ileal, transcript levels of TJ components in the HIV+ cohort. This result is confirmed by a reduction in TJ proteins in the descending colon of HIV+ patients. In the HIV+ cohort, colonic TJ transcript levels progressively decreased along the proximal-to-distal axis. In contrast, expression levels of the same TJ transcripts stayed unchanged, or progressively increased, from the proximal-to-distal gut in the healthy controls. Non-TJ intestinal epithelial cell-specific mRNAs reveal differing patterns of HIV-associated transcriptional alteration, arguing for an overall change in intestinal epithelial transcriptional regulation in the HIV colon. These findings suggest that persistent intestinal epithelial dysregulation involving a reduction in TJ expression is a mechanism driving increases in colonic permeability and microbial translocation in the ART-treated HIV-infected patient, and a possible immunopathogenic factor for non-AIDS related complications.


Assuntos
Fármacos Anti-HIV/efeitos adversos , Colo/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Infecções por HIV/tratamento farmacológico , Mucosa Intestinal/efeitos dos fármacos , Proteínas de Junções Íntimas/antagonistas & inibidores , Junções Íntimas/efeitos dos fármacos , Centros Médicos Acadêmicos , Fármacos Anti-HIV/uso terapêutico , Estudos de Coortes , Colo/metabolismo , Colo/patologia , Colo/virologia , Colo Ascendente/efeitos dos fármacos , Colo Ascendente/metabolismo , Colo Ascendente/patologia , Colo Ascendente/virologia , Colo Descendente/efeitos dos fármacos , Colo Descendente/metabolismo , Colo Descendente/patologia , Colo Descendente/virologia , Colo Transverso/efeitos dos fármacos , Colo Transverso/metabolismo , Colo Transverso/patologia , Colo Transverso/virologia , Feminino , Infecções por HIV/metabolismo , Infecções por HIV/patologia , Infecções por HIV/virologia , Humanos , Íleo/efeitos dos fármacos , Íleo/metabolismo , Íleo/patologia , Íleo/virologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Mucosa Intestinal/virologia , Masculino , Pessoa de Meia-Idade , Ohio , Especificidade de Órgãos , Permeabilidade/efeitos dos fármacos , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo , Junções Íntimas/metabolismo , Junções Íntimas/patologia , Junções Íntimas/virologia
19.
PLoS One ; 9(2): e88803, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586397

RESUMO

Herpes simplex virus (HSV) types 1 and 2 are the most common opportunistic infections in HIV/AIDS. In these immunocompromised individuals, HSV-1 reactivates and replicates in oral epithelium, leading to oral disorders such as ulcers, gingivitis, and necrotic lesions. Although the increased risk of HSV infection may be mediated in part by HIV-induced immune dysfunction, direct or indirect interactions of HIV and HSV at the molecular level may also play a role. In this report we show that prolonged interaction of the HIV proteins tat and gp120 and cell-free HIV virions with polarized oral epithelial cells leads to disruption of tight and adherens junctions of epithelial cells through the mitogen-activated protein kinase signaling pathway. HIV-induced disruption of oral epithelial junctions facilitates HSV-1 paracellular spread between the epithelial cells. Furthermore, HIV-associated disruption of adherens junctions exposes sequestered nectin-1, an adhesion protein and critical receptor for HSV envelope glycoprotein D (gD). Exposure of nectin-1 facilitates binding of HSV-1 gD, which substantially increases HSV-1 infection of epithelial cells with disrupted junctions over that of cells with intact junctions. Exposed nectin-1 from disrupted adherens junctions also increases the cell-to-cell spread of HSV-1 from infected to uninfected oral epithelial cells. Antibodies to nectin-1 and HSV-1 gD substantially reduce HSV-1 infection and cell-to-cell spread, indicating that HIV-promoted HSV infection and spread are mediated by the interaction of HSV gD with HIV-exposed nectin-1. Our data suggest that HIV-associated disruption of oral epithelial junctions may potentiate HSV-1 infection and its paracellular and cell-to-cell spread within the oral mucosal epithelium. This could be one of the possible mechanisms of rapid development of HSV-associated oral lesions in HIV-infected individuals.


Assuntos
Células Epiteliais/patologia , Infecções por HIV/complicações , Herpes Simples/transmissão , Sistema de Sinalização das MAP Quinases/fisiologia , Boca/citologia , Junções Íntimas/patologia , Western Blotting , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Células Epiteliais/virologia , Imunofluorescência , Produtos do Gene tat/metabolismo , Proteína gp120 do Envelope de HIV/metabolismo , Herpes Simples/etiologia , Humanos , Nectinas , Junções Íntimas/metabolismo , Junções Íntimas/virologia , Proteínas do Envelope Viral/metabolismo
20.
Mol Oncol ; 8(3): 533-43, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24462519

RESUMO

High-risk human papillomavirus (HPV) infection is the principal risk factor for the development of cervical cancer. The HPV E6 oncoprotein has the ability to target and interfere with several PSD-95/DLG/ZO-1 (PDZ) domain-containing proteins that are involved in the control of cell polarity. This function can be significant for E6 oncogenic activity because a deficiency in cell polarisation is a marker of tumour progression. The establishment and control of polarity in epithelial cells depend on the correct asymmetrical distribution of proteins and lipids at the cell borders and on specialised cell junctions. In this report, we have investigated the effects of HPV E6 protein on the polarity machinery, with a focus on the PDZ partitioning defective 3 (Par3) protein, which is a key component of tight junctions (TJ) and the polarity network. We demonstrate that E6 is able to bind and induce the mislocalisation of Par3 protein in a PDZ-dependent manner without significant reduction in Par3 protein levels. In addition, the high-risk HPV-18 E6 protein promotes a delay in TJ formation when analysed by calcium switch assays. Taken together, the data presented in this study contribute to our understanding of the molecular mechanism by which HPVs induce the loss of cell polarity, with potential implications for the development and progression of HPV-associated tumours.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Polaridade Celular , Proteínas de Ligação a DNA/metabolismo , Células Epiteliais/virologia , Interações Hospedeiro-Patógeno , Papillomavirus Humano 18/fisiologia , Proteínas de Membrana/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Infecções por Papillomavirus/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Ciclo Celular/análise , Linhagem Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Proteínas de Membrana/análise , Infecções por Papillomavirus/patologia , Infecções por Papillomavirus/virologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Junções Íntimas/metabolismo , Junções Íntimas/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA