Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Med Virol ; 96(5): e29671, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38747003

RESUMEN

The coronavirus disease of 2019 (COVID-19) pandemic has led to more than 700 million confirmed cases and nearly 7 million deaths. Although severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) virus mainly infects the respiratory system, neurological complications are widely reported in both acute infection and long-COVID cases. Despite the success of vaccines and antiviral treatments, neuroinvasiveness of SARS-CoV-2 remains an important question, which is also centered on the mystery of whether the virus is capable of breaching the barriers into the central nervous system. By studying the K18-hACE2 infection model, we observed clear evidence of microvascular damage and breakdown of the blood-brain barrier (BBB). Mechanistically, SARS-CoV-2 infection caused pericyte damage, tight junction loss, endothelial activation and vascular inflammation, which together drive microvascular injury and BBB impairment. In addition, the blood-cerebrospinal fluid barrier at the choroid plexus was also impaired after infection. Therefore, cerebrovascular and choroid plexus dysfunctions are important aspects of COVID-19 and may contribute to neurological complications both acutely and in long COVID.


Asunto(s)
Barrera Hematoencefálica , COVID-19 , Plexo Coroideo , SARS-CoV-2 , Barrera Hematoencefálica/virología , Animales , Plexo Coroideo/virología , Plexo Coroideo/patología , COVID-19/virología , COVID-19/patología , COVID-19/complicaciones , COVID-19/fisiopatología , Ratones , Uniones Estrechas/virología , Modelos Animales de Enfermedad , Enzima Convertidora de Angiotensina 2/metabolismo , Inflamación/virología , Humanos , Pericitos/virología , Pericitos/patología
2.
J Neurovirol ; 30(1): 22-38, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38189894

RESUMEN

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.


Asunto(s)
Barrera Hematoencefálica , Esclerosis Múltiple , Barrera Hematoencefálica/virología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Humanos , Animales , Esclerosis Múltiple/virología , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Ratones , Uniones Estrechas/virología , Uniones Estrechas/metabolismo , Permeabilidad Capilar , Células Endoteliales/virología , Células Endoteliales/metabolismo , Células Endoteliales/patología
3.
Signal Transduct Target Ther ; 6(1): 337, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34489403

RESUMEN

SARS-CoV-2 has been reported to show a capacity for invading the brains of humans and model animals. However, it remains unclear whether and how SARS-CoV-2 crosses the blood-brain barrier (BBB). Herein, SARS-CoV-2 RNA was occasionally detected in the vascular wall and perivascular space, as well as in brain microvascular endothelial cells (BMECs) in the infected K18-hACE2 transgenic mice. Moreover, the permeability of the infected vessel was increased. Furthermore, disintegrity of BBB was discovered in the infected hamsters by administration of Evans blue. Interestingly, the expression of claudin5, ZO-1, occludin and the ultrastructure of tight junctions (TJs) showed unchanged, whereas, the basement membrane was disrupted in the infected animals. Using an in vitro BBB model that comprises primary BMECs with astrocytes, SARS-CoV-2 was found to infect and cross through the BMECs. Consistent with in vivo experiments, the expression of MMP9 was increased and collagen IV was decreased while the markers for TJs were not altered in the SARS-CoV-2-infected BMECs. Besides, inflammatory responses including vasculitis, glial activation, and upregulated inflammatory factors occurred after SARS-CoV-2 infection. Overall, our results provide evidence supporting that SARS-CoV-2 can cross the BBB in a transcellular pathway accompanied with basement membrane disrupted without obvious alteration of TJs.


Asunto(s)
Membrana Basal/metabolismo , Barrera Hematoencefálica/metabolismo , COVID-19/metabolismo , SARS-CoV-2/metabolismo , Uniones Estrechas/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Membrana Basal/patología , Membrana Basal/virología , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/virología , COVID-19/genética , COVID-19/patología , Chlorocebus aethiops , Modelos Animales de Enfermedad , Humanos , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Transgénicos , SARS-CoV-2/genética , Uniones Estrechas/genética , Uniones Estrechas/patología , Uniones Estrechas/virología , Células Vero
4.
Front Immunol ; 12: 636966, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34557180

RESUMEN

Since 2003, the world has been confronted with three new betacoronaviruses that cause human respiratory infections: SARS-CoV, which causes severe acute respiratory syndrome (SARS), MERS-CoV, which causes Middle East respiratory syndrome (MERS), and SARS-CoV-2, which causes Coronavirus Disease 2019 (COVID-19). The mechanisms of coronavirus transmission and dissemination in the human body determine the diagnostic and therapeutic strategies. An important problem is the possibility that viral particles overcome tissue barriers such as the intestine, respiratory tract, blood-brain barrier, and placenta. In this work, we will 1) consider the issue of endocytosis and the possibility of transcytosis and paracellular trafficking of coronaviruses across tissue barriers with an emphasis on the intestinal epithelium; 2) discuss the possibility of antibody-mediated transcytosis of opsonized viruses due to complexes of immunoglobulins with their receptors; 3) assess the possibility of the virus transfer into extracellular vesicles during intracellular transport; and 4) describe the clinical significance of these processes. Models of the intestinal epithelium and other barrier tissues for in vitro transcytosis studies will also be briefly characterized.


Asunto(s)
Endocitosis , Mucosa Intestinal/virología , SARS-CoV-2/metabolismo , Anticuerpos Antivirales/metabolismo , Antivirales/farmacología , Antivirales/uso terapéutico , COVID-19/transmisión , COVID-19/virología , Ensayos Clínicos como Asunto , Endocitosis/efectos de los fármacos , Humanos , Mucosa Intestinal/metabolismo , Modelos Biológicos , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/inmunología , Uniones Estrechas/metabolismo , Uniones Estrechas/virología , Transcitosis/efectos de los fármacos , Acoplamiento Viral , Tratamiento Farmacológico de COVID-19
5.
Tissue Barriers ; 9(4): 1943274, 2021 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-34241579

RESUMEN

Oropharyngeal, airway, intestinal, and genital mucosal epithelia are the main portals of entry for the majority of human pathogenic viruses. To initiate systemic infection, viruses must first be transmitted across the mucosal epithelium and then spread across the body. However, mucosal epithelia have well-developed tight junctions, which have a strong barrier function that plays a critical role in preventing the spread and dissemination of viral pathogens. Viruses can overcome these barriers by disrupting the tight junctions of mucosal epithelia, which facilitate paracellular viral penetration and initiate systemic disease. Disruption of tight and adherens junctions may also release the sequestered viral receptors within the junctional areas, and liberation of hidden receptors may facilitate viral infection of mucosal epithelia. This review focuses on possible molecular mechanisms of virus-associated disruption of mucosal epithelial junctions and its role in transmucosal viral transmission and spread.


Asunto(s)
Uniones Estrechas , Virosis , Epitelio/virología , Humanos , Membrana Mucosa/virología , Uniones Estrechas/virología , Virosis/transmisión
6.
Tissue Barriers ; 9(2): 1883965, 2021 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-33632074

RESUMEN

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.


Asunto(s)
Uniones Estrechas/virología , Virosis/fisiopatología , Animales , Humanos , Ratones
7.
PLoS Negl Trop Dis ; 14(6): e0008335, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32511241

RESUMEN

Zika virus (ZIKV), a neglected tropical disease until its re-emergence in 2007, causes microcephaly in infants and Guillain-Barré syndrome in adults. Its re-emergence and spread to more than 80 countries led the World Health Organization in 2016 to declare a Public Health Emergency. ZIKV is mainly transmitted by mosquitos, but can persist in infected human male semen for prolonged periods and may be sexually transmitted. Testicular Sertoli cells support ZIKV replication and may be a reservoir for persistent ZIKV infection. Electrical impedance analyses indicated ZIKV infection rapidly disrupted Vero cell monolayers but had little effect upon human Sertoli cells (HSerC). We determined ZIKV-induced proteomic changes in HSerC using an aptamer-based multiplexed technique (SOMAscan) targeting >1300 human proteins. ZIKV infection caused differential expression of 299 proteins during three different time points, including 5 days after infection. Dysregulated proteins are involved in different bio-functions, including cell death and survival, cell cycle, maintenance of cellular function, cell signaling, cellular assembly, morphology, movement, molecular transport, and immune response. Many signaling pathways important for maintenance of HSerC function and spermatogenesis were highly dysregulated. These included IL-6, IGF1, EGF, NF-κB, PPAR, ERK/MAPK, and growth hormone signaling. Down-regulation of the PPAR signaling pathway might impact cellular energy supplies. Upstream molecule analysis also indicated microRNAs involved in germ cell development were downregulated by infection. Overall, this study leads to a better understanding of Sertoli cellular mechanisms used by ZIKV during persistent infection and possible ZIKV impacts on spermatogenesis.


Asunto(s)
Células de Sertoli/inmunología , Espermatogénesis , Uniones Estrechas/inmunología , Infección por el Virus Zika/inmunología , Animales , Chlorocebus aethiops , Humanos , Masculino , Proteómica , Semen/virología , Células de Sertoli/virología , Transducción de Señal , Uniones Estrechas/virología , Células Vero , Replicación Viral , Virus Zika
8.
Biochim Biophys Acta Biomembr ; 1862(7): 183296, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32268133

RESUMEN

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.


Asunto(s)
Hepatitis C/genética , Interacciones Huésped-Patógeno/genética , Uniones Estrechas/genética , Internalización del Virus , Hepacivirus/genética , Hepacivirus/patogenicidad , Hepatitis C/virología , Hepatocitos/virología , Humanos , Hígado/patología , Hígado/virología , Proteínas de Transporte de Membrana/genética , Ocludina/genética , Receptores de LDL/genética , Tetraspanina 28/genética , Uniones Estrechas/virología
9.
Virology ; 531: 93-99, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30856486

RESUMEN

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.


Asunto(s)
Claudina-1/metabolismo , Virus de la Parainfluenza 2 Humana/fisiología , Infecciones por Rubulavirus/metabolismo , Infecciones por Rubulavirus/virología , Claudina-1/genética , Células Epiteliales/metabolismo , Células Epiteliales/virología , Interacciones Huésped-Patógeno , Humanos , Virus de la Parainfluenza 2 Humana/genética , Infecciones por Rubulavirus/genética , Uniones Estrechas/metabolismo , Uniones Estrechas/virología , Proteínas Virales/genética , Proteínas Virales/metabolismo
10.
J Virol ; 93(4)2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30463963

RESUMEN

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.


Asunto(s)
Ocludina/metabolismo , Sapovirus/fisiología , Uniones Estrechas/virología , Animales , Células CHO , Cricetulus , Endosomas/metabolismo , Células Epiteliales/virología , Gastroenteritis/virología , Células LLC-PK1 , Ocludina/fisiología , Sapovirus/metabolismo , Sapovirus/patogenicidad , Porcinos/virología , Uniones Estrechas/metabolismo , Virosis/metabolismo
11.
Am J Physiol Lung Cell Mol Physiol ; 316(2): L358-L368, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30489157

RESUMEN

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.


Asunto(s)
Células Epiteliales/virología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/patogenicidad , Uniones Estrechas/virología , Animales , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Ratones Endogámicos C57BL , Sistema Respiratorio/metabolismo , Uniones Estrechas/metabolismo
12.
Laryngoscope ; 128(12): E393-E401, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30325507

RESUMEN

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.


Asunto(s)
Resfriado Común/virología , Células Epiteliales/virología , Especies Reactivas de Oxígeno/metabolismo , Rhinovirus/metabolismo , Uniones Estrechas/virología , Células Epiteliales/efectos de los fármacos , Humanos , Mucosa Nasal/citología , Mucosa Nasal/virología , Compuestos Onio/farmacología , Rhinovirus/efectos de los fármacos
13.
Virus Genes ; 54(5): 684-693, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30173363

RESUMEN

Porcine bocavirus (PBoV) has a high prevalence in both healthy and diseased swine around the world. It was recently reported that PBoV and porcine circovirus type 2 (PCV2)-which contribute to porcine diarrheal disease-have a high rate of co-infection. To clarify the pathogenesis of PBoV, we examined the co-infection rate and effects of these two pathogens in IPEC-J2 porcine intestinal enterocytes. Both single and co-infection had cytopathic effects in IPEC-J2 cells. The apoptosis and proliferation rates of cells infected with both viruses did not differ significantly from those of cells infected with either one alone. PBoV and PCV2 induced the upregulation of inflammatory cytokines and the downregulation of the tight junction proteins occludin and claudin 1 in the early stage of infection, leading to destruction of epithelial barrier integrity and enhanced cytotoxicity. These findings provide insight into the pathogenic mechanisms of PBoV and PCV2 and a basis for developing effective strategies to prevent the spread of gastrointestinal diseases in pigs and other livestock.


Asunto(s)
Bocavirus/patogenicidad , Circovirus/patogenicidad , Enfermedades de los Porcinos/virología , Uniones Estrechas/virología , Animales , Apoptosis , Línea Celular , Infecciones por Circoviridae/prevención & control , Infecciones por Circoviridae/virología , Coinfección , Citocinas/biosíntesis , Efecto Citopatogénico Viral , Infecciones por Parvoviridae/virología , Porcinos , Enfermedades de los Porcinos/patología , Enfermedades de los Porcinos/prevención & control , Replicación Viral
14.
Cell Host Microbe ; 24(3): 405-416.e3, 2018 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-30173956

RESUMEN

Sexual transmission of filoviruses was first reported in 1968 after an outbreak of Marburg virus (MARV) disease and recently caused flare-ups of Ebola virus disease in the 2013-2016 outbreak. How filoviruses establish testicular persistence and are shed in semen remain unknown. We discovered that persistent MARV infection of seminiferous tubules, an immune-privileged site that harbors sperm production, is a relatively common event in crab-eating macaques that survived infection after antiviral treatment. Persistence triggers severe testicular damage, including spermatogenic cell depletion and inflammatory cell invasion. MARV mainly persists in Sertoli cells, leading to breakdown of the blood-testis barrier formed by inter-Sertoli cell tight junctions. This disruption is accompanied by local infiltration of immunosuppressive CD4+Foxp3+ regulatory T cells. Our study elucidates cellular events associated with testicular persistence that may promote sexual transmission of filoviruses and suggests that targeting immunosuppression may be warranted to clear filovirus persistence in damaged immune-privileged sites.


Asunto(s)
Enfermedad del Virus de Marburg/virología , Marburgvirus/fisiología , Enfermedades de los Primates/virología , Testículo/virología , Animales , Macaca , Masculino , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/metabolismo , Enfermedades de los Primates/inmunología , Enfermedades de los Primates/metabolismo , Células de Sertoli/metabolismo , Células de Sertoli/virología , Sobrevivientes , Linfocitos T Reguladores/inmunología , Uniones Estrechas/metabolismo , Uniones Estrechas/virología
15.
Exp Cell Res ; 371(2): 322-329, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30142324

RESUMEN

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.


Asunto(s)
Subtipo H3N2 del Virus de la Influenza A/genética , Gripe Humana/genética , Mucosa Nasal/metabolismo , Pólipos Nasales/genética , Oncostatina M/genética , Rinitis/genética , Sinusitis/genética , Adulto , Estudios de Casos y Controles , Diferenciación Celular , Enfermedad Crónica , Claudina-1/genética , Claudina-1/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Epiteliales/virología , Femenino , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Humanos , Subtipo H3N2 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H3N2 del Virus de la Influenza A/metabolismo , Gripe Humana/metabolismo , Gripe Humana/patología , Gripe Humana/virología , Masculino , Persona de Mediana Edad , Mucosa Nasal/patología , Mucosa Nasal/virología , Pólipos Nasales/metabolismo , Pólipos Nasales/patología , Pólipos Nasales/virología , Ocludina/genética , Ocludina/metabolismo , Oncostatina M/metabolismo , Cultivo Primario de Células , Rinitis/metabolismo , Rinitis/patología , Rinitis/virología , Transducción de Señal , Sinusitis/metabolismo , Sinusitis/patología , Sinusitis/virología , Uniones Estrechas/metabolismo , Uniones Estrechas/patología , Uniones Estrechas/virología , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo
16.
Clin Exp Allergy ; 48(5): 513-524, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29350877

RESUMEN

BACKGROUND: Bronchial epithelial tight junctions (TJ) have been extensively assessed in healthy airway epithelium. However, no studies have yet assessed the effect of human rhinovirus (HRV) infection on the expression and resultant barrier function in epithelial tight junctions (TJ) in childhood asthma. OBJECTIVES: To investigate the impact of HRV infection on airway epithelial TJ expression and barrier function in airway epithelial cells (AECs) of children with and without asthma. Furthermore, to test the hypothesis that barrier integrity and function is compromised to a greater extent by HRV in AECs from asthmatic children. METHODS: Primary AECs were obtained from children with and without asthma, differentiated into air-liquid interface (ALI) cultures and infected with rhinovirus. Expression of claudin-1, occludin and zonula occluden-1 (ZO-1) was assessed via qPCR, immunocytochemistry (ICC), in-cell western (ICW) and confocal microscopy. Barrier function was assessed by transepithelial electrical resistance (TER; RT ) and permeability to fluorescent dextran. RESULTS: Basal TJ gene expression of claudin-1 and occludin was significantly upregulated in asthmatic children compared to non-asthmatics; however, no difference was seen with ZO-1. Interestingly, claudin-1, occludin and ZO-1 protein expression was significantly reduced in AEC of asthmatic children compared to non-asthmatic controls suggesting possible post-transcriptional inherent differences. HRV infection resulted in a transient dissociation of TJ and airway barrier integrity in non-asthmatic children. Although similar dissociation of TJ was observed in asthmatic children, a significant and sustained reduction in TJ expression concurrent with both a significant decrease in TER and an increase in permeability in asthmatic children was observed. CONCLUSION: This study demonstrates novel intrinsic differences in TJ gene and protein expression between AEC of children with and without asthma. Furthermore, it correlates directly the relationship between HRV infection and the resultant dissociation of epithelial TJ that causes a continued altered barrier function in children with asthma.


Asunto(s)
Asma/patología , Asma/virología , Infecciones por Picornaviridae/patología , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología , Adolescente , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Rhinovirus , Uniones Estrechas/patología , Uniones Estrechas/virología
17.
Infect Immun ; 86(1)2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29084896

RESUMEN

Staphylococcus aureus (S. aureus) infections are among the most common and severe infections, garnering notoriety in an era of increasing resistance to antibiotics. It is therefore important to define molecular mechanisms by which this pathogen attacks host cells. Here, we demonstrate that alpha-toxin, one of the major toxins of S. aureus, induces activation of acid sphingomyelinase and concomitant release of ceramide in endothelial cells treated with the toxin. Activation of acid sphingomyelinase by alpha-toxin is mediated via ADAM10. Infection experiments employing alpha-toxin-deficient S. aureus and the corresponding wild-type strain reveal that activation of acid sphingomyelinase in endothelial cells requires alpha-toxin expression by the pathogen. Activation of acid sphingomyelinase is linked to degradation of tight junctions in endothelial cells in vitro, which is blocked by pharmacological inhibition of acid sphingomyelinase. Most importantly, alpha-toxin induces severe degradation of tight junctions in the lung and causes lung edema in vivo, which is prevented by genetic deficiency of acid sphingomyelinase. These data indicate a novel and important role of the acid sphingomyelinase/ceramide system for the endothelial response to toxins and provide a molecular link between alpha-toxin and the degradation of tight junctions. The data also suggest that inhibition of acid sphingomyelinase may provide a novel treatment option to prevent lung edema caused by S. aureus alpha-toxin.


Asunto(s)
Toxinas Bacterianas/metabolismo , Ceramidas/metabolismo , Células Endoteliales/metabolismo , Proteínas Hemolisinas/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Staphylococcus aureus/metabolismo , Uniones Estrechas/metabolismo , Proteína ADAM10/metabolismo , Animales , Células Cultivadas , Células Endoteliales/virología , Pulmón/metabolismo , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Edema Pulmonar/metabolismo , Edema Pulmonar/virología , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/virología , Uniones Estrechas/virología
18.
Clin Exp Immunol ; 190(3): 351-359, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28856667

RESUMEN

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.


Asunto(s)
Bronquios/inmunología , Células Epiteliales/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Uniones Estrechas/inmunología , Animales , Asma/etiología , Asma/inmunología , Asma/patología , Asma/virología , Bronquios/patología , Bronquios/virología , Lavado Broncoalveolar , Células Cultivadas , Claudina-1/inmunología , Claudinas/inmunología , Células Epiteliales/patología , Células Epiteliales/virología , Regulación de la Expresión Génica/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Infecciones por Virus Sincitial Respiratorio/patología , Factores de Riesgo , Uniones Estrechas/patología , Uniones Estrechas/virología
19.
J Virol ; 91(10)2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28275187

RESUMEN

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.


Asunto(s)
Ocludina/metabolismo , Virus de la Diarrea Epidémica Porcina/fisiología , Uniones Estrechas/química , Acoplamiento Viral , Internalización del Virus , Animales , Línea Celular , Chlorocebus aethiops , Citoplasma/metabolismo , Células Epiteliales/virología , Ocludina/deficiencia , Ocludina/genética , Virus de la Diarrea Epidémica Porcina/efectos de los fármacos , Virus de la Diarrea Epidémica Porcina/patogenicidad , ARN Interferente Pequeño , Porcinos , Uniones Estrechas/patología , Uniones Estrechas/virología , Células Vero , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...