Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 16(6)2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38932195

RESUMO

Antiretroviral treatments have notably extended the lives of individuals with HIV and reduced the occurrence of comorbidities, including ocular manifestations. The involvement of endoplasmic reticulum (ER) stress in HIV-1 pathogenesis raises questions about its correlation with cellular senescence or its role in initiating senescent traits. This study investigated how ER stress and dysregulated autophagy impact cellular senescence triggered by HIV-1 Tat in the MIO-M1 cell line (human Müller glial cells). Cells exposed to HIV-1 Tat exhibited increased vimentin expression combined with markers of ER stress (BiP, p-eIF2α), autophagy (LC3, Beclin-1, p62), and the senescence marker p21 compared to control cells. Western blotting and staining techniques like SA-ß-gal were employed to examine these markers. Additionally, treatments with ER stress inhibitor 4-PBA before HIV-1 Tat exposure led to a decreased expression of ER stress, senescence, and autophagy markers. Conversely, pre-treatment with the autophagy inhibitor 3-MA resulted in reduced autophagy and senescence markers but did not alter ER stress markers compared to control cells. The findings suggest a link between ER stress, dysregulated autophagy, and the initiation of a senescence phenotype in MIO-M1 cells induced by HIV-1 Tat exposure.


Assuntos
Autofagia , Senescência Celular , Estresse do Retículo Endoplasmático , HIV-1 , Produtos do Gene tat do Vírus da Imunodeficiência Humana , Humanos , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética , HIV-1/fisiologia , Linhagem Celular , Células Ependimogliais/metabolismo , Células Ependimogliais/virologia , Infecções por HIV/virologia
2.
Invest Ophthalmol Vis Sci ; 58(10): 4355­4365, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28810265

RESUMO

Purpose: Emerging evidence has shown that both congenital and adult Zika virus (ZIKV) infection can cause eye diseases. The goals of the current study were to explore mechanisms and pathophysiology of ZIKV-induced eye defects. Methods: Wild-type or A129 interferon type I receptor-deficient mice were infected by either FSS13025 or Mex1-7 strain of ZIKV. Retinal histopathology was measured at different time points after infection. The presence of viral RNA and protein in the retina was determined by in situ hybridization and immunofluorescence staining, respectively. Growth curves of ZIKV in permissive retinal cells were assessed in cultured retinal pigment epithelial (RPE) and Müller glial cells. Results: ZIKV-infected mice developed a spectrum of ocular pathologies that affected multiple layers of the retina. A primary target of ZIKV in the eye was Müller glial cells, which displayed decreased neurotrophic function and increased expression of proinflammatory cytokines after infection. ZIKV also infected RPE; and both the RPE and Müller cells expressed viral entry receptors TYRO3 and AXL. Retinitis, focal retinal degeneration, and ganglion cell loss were observed after the clearance of viral particles. Conclusions: Our data suggest that ZIKV can infect infant eyes with immature blood-retinal barrier and cause structural damages to the retina. The ocular findings in microcephalic infants may not be solely caused by ZIKV-induced impairment of neurodevelopment.


Assuntos
Modelos Animais de Doenças , Células Ependimogliais/virologia , Infecções Oculares Virais/virologia , Doenças Retinianas/virologia , Epitélio Pigmentado da Retina/virologia , Infecção por Zika virus/virologia , Animais , Barreira Hematorretiniana , Células Cultivadas , Células Ependimogliais/patologia , Infecções Oculares Virais/patologia , Citometria de Fluxo , Hibridização in Situ Fluorescente , Camundongos , Camundongos Endogâmicos C57BL , RNA Viral/genética , Reação em Cadeia da Polimerase em Tempo Real , Doenças Retinianas/patologia , Epitélio Pigmentado da Retina/patologia , Uveíte Posterior/patologia , Uveíte Posterior/virologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/fisiologia , Zika virus/fisiologia , Infecção por Zika virus/patologia
3.
Antiviral Res ; 145: 70-81, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28739278

RESUMO

Zika virus (ZIKV) infection has been associated with ocular abnormalities such as chorioretinal atrophy, optic nerve abnormalities, posterior uveitis and idiopathic maculopathy. Yet our knowledge about ZIKV infection in retinal cells and its potential contribution to retinal pathology is still very limited. Here we found that primary Müller cells, the principal glial cells in the retina, expressed a high level of ZIKV entry cofactor AXL gene and were highly permissive to ZIKV infection. In addition, ZIKV-infected Müller cells exhibited a pro-inflammatory phenotype and produced many inflammatory and growth factors. While a number of inflammatory signaling pathways such as ERK, p38MAPK, NF-κB, JAK/STAT3 and endoplasmic reticulum stress were activated after ZIKV infection, inhibition of p38MAPK after ZIKV infection most effectively blocked ZIKV-induced inflammatory and growth molecules. In comparison to ZIKV, Dengue virus (DENV), another Flavivirus infected Müller cells more efficiently but induced much lower pro-inflammatory responses. These data suggest that Müller cells play an important role in ZIKV-induced ocular pathology by induction of inflammatory and growth factors in which the p38MAPK pathway has a central role. Blocking p38MAPK may provide a novel approach to control ZIKV-induced ocular inflammation.


Assuntos
Células Ependimogliais/imunologia , Células Ependimogliais/virologia , Zika virus/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Células Cultivadas , Vírus da Dengue/fisiologia , Estresse do Retículo Endoplasmático , Inflamação , Camundongos , Fenótipo , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Internalização do Vírus , Zika virus/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética
4.
J Neuroinflammation ; 14(1): 43, 2017 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-28253931

RESUMO

BACKGROUND: Ocular abnormalities present in microcephalic infants with presumed Zika virus (ZIKV) congenital disease includes focal pigment mottling of the retina, chorioretinal atrophy, optic nerve abnormalities, and lens dislocation. Target cells in the ocular compartment for ZIKV infectivity are unknown. The cellular response of ocular cells to ZIKV infection has not been described. Mechanisms for viral dissemination in the ocular compartment of ZIKV-infected infants and adults have not been reported. Here, we identify target cells for ZIKV infectivity in both the inner and outer blood-retinal barriers (IBRB and OBRB), describe the cytokine expression profile in the IBRB after ZIKV exposure, and propose a mechanism for viral dissemination in the retina. METHODS: We expose primary cellular components of the IBRB including human retinal microvascular endothelial cells, retinal pericytes, and Müller cells as well as retinal pigmented epithelial cells of the OBRB to the PRVABC56 strain of ZIKV. Viral infectivity was analyzed by microscopy, immunofluorescence, and reverse transcription polymerase chain reaction (RT-PCR and qRT-PCR). Angiogenic and proinflammatory cytokines were measured by Luminex assays. RESULTS: We find by immunofluorescent staining using the Flavivirus 4G2 monoclonal antibody that retinal endothelial cells and pericytes of the IBRB and retinal pigmented epithelial cells of the OBRB are fully permissive for ZIKV infection but not Müller cells when compared to mock-infected controls. We confirmed ZIKV infectivity in retinal endothelial cells, retinal pericytes, and retinal pigmented epithelial cells by RT-PCR and qRT-PCR using ZIKV-specific oligonucleotide primers. Expression profiles by Luminex assays in retinal endothelial cells infected with ZIKV revealed a marginal increase in levels of beta-2 microglobulin (ß2-m), granulocyte macrophage colony-stimulating factor (GMCSF), intercellular adhesion molecule 1 (ICAM-1), interleukin-6 (IL-6), monocyte chemotactic protein-1 (MCP1), and vascular cell adhesion molecule 1 (VCAM-1) and higher levels of regulated upon activation, normal T cell expressed and presumably secreted (RANTES) but lower levels of interleukin-4 (IL-4) compared to controls. CONCLUSIONS: Retinal endothelial cells, retinal pericytes, and retinal pigmented epithelial cells are fully permissive for ZIKV lytic replication and are primary target cells in the retinal barriers for infection. ZIKV infection of retinal endothelial cells and retinal pericytes induces significantly higher levels of RANTES that likely contributes to ocular inflammation.


Assuntos
Barreira Hematorretiniana/patologia , Células Ependimogliais/patologia , Oftalmopatias/patologia , Infecção por Zika virus/patologia , Zika virus , Adulto , Animais , Barreira Hematorretiniana/imunologia , Barreira Hematorretiniana/virologia , Células Cultivadas , Chlorocebus aethiops , Células Ependimogliais/imunologia , Células Ependimogliais/virologia , Oftalmopatias/imunologia , Oftalmopatias/virologia , Humanos , Células Vero , Zika virus/imunologia , Zika virus/metabolismo , Infecção por Zika virus/imunologia
5.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795408

RESUMO

Schmallenberg virus (SBV) was discovered in Germany in late 2011 and then spread rapidly to many European countries. SBV is an orthobunyavirus that causes abortion and congenital abnormalities in ruminants. A virus-encoded nonstructural protein, termed NSs, is a major virulence factor of SBV, and it is known to promote the degradation of Rpb1, a subunit of the RNA polymerase II (Pol II) complex, and therefore hampers global cellular transcription. In this study, we found that NSs is mainly localized in the nucleus of infected cells and specifically appears to target the nucleolus through a nucleolar localization signal (NoLS) localized between residues 33 and 51 of the protein. NSs colocalizes with nucleolar markers such as B23 (nucleophosmin) and fibrillarin. We observed that in SBV-infected cells, B23 undergoes a nucleolus-to-nucleoplasm redistribution, evocative of virus-induced nucleolar disruption. In contrast, the nucleolar pattern of B23 was unchanged upon infection with an SBV recombinant mutant with NSs lacking the NoLS motif (SBVΔNoLS). Interestingly, unlike wild-type SBV, the inhibitory activity of SBVΔNoLS toward RNA Pol II transcription is impaired. Overall, our results suggest that a putative link exists between NSs-induced nucleolar disruption and its inhibitory function on cellular transcription, which consequently precludes the cellular antiviral response and/or induces cell death. IMPORTANCE: Schmallenberg virus (SBV) is an emerging arbovirus of ruminants that spread in Europe between 2011 and 2013. SBV induces fetal abnormalities during gestation, with the central nervous system being one of the most affected organs. The virus-encoded NSs protein acts as a virulence factor by impairing host cell transcription. Here, we show that NSs contains a nucleolar localization signal (NoLS) and induces disorganization of the nucleolus. The NoLS motif in the SBV NSs is absolutely necessary for virus-induced inhibition of cellular transcription. To our knowledge, this is the first report of nucleolar functions for NSs within the Bunyaviridae family.


Assuntos
Nucléolo Celular/virologia , Células Ependimogliais/virologia , Interações Hospedeiro-Patógeno , Orthobunyavirus/patogenicidade , RNA Polimerase II/química , Proteínas não Estruturais Virais/química , Animais , Linhagem Celular Transformada , Nucléolo Celular/metabolismo , Nucléolo Celular/ultraestrutura , Plexo Corióideo/citologia , Plexo Corióideo/metabolismo , Plexo Corióideo/virologia , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Células Ependimogliais/metabolismo , Células Ependimogliais/ultraestrutura , Regulação da Expressão Gênica , Células HeLa , Humanos , Mutação , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Orthobunyavirus/genética , Orthobunyavirus/metabolismo , Sinais Direcionadores de Proteínas , Transporte Proteico , Proteólise , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Ovinos , Transdução de Sinais , Transcrição Gênica , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
6.
Cell Res ; 26(6): 645-54, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27174054

RESUMO

The recent Zika virus (ZIKV) epidemic in Latin America coincided with a marked increase in microcephaly in newborns. However, the causal link between maternal ZIKV infection and malformation of the fetal brain has not been firmly established. Here we show a vertical transmission of ZIKV in mice and a marked effect on fetal brain development. We found that intraperitoneal (i.p.) injection of a contemporary ZIKV strain in pregnant mice led to the infection of radial glia cells (RGs) of dorsal ventricular zone of the fetuses, the primary neural progenitors responsible for cortex development, and caused a marked reduction of these cortex founder cells in the fetuses. Interestingly, the infected fetal mice exhibited a reduced cavity of lateral ventricles and a discernable decrease in surface areas of the cortex. This study thus supports the conclusion that vertically transmitted ZIKV affects fetal brain development and provides a valuable animal model for the evaluation of potential therapeutic or preventative strategies.


Assuntos
Córtex Cerebral/embriologia , Córtex Cerebral/virologia , Células Ependimogliais/patologia , Células Ependimogliais/virologia , Infecção por Zika virus/transmissão , Zika virus/fisiologia , Animais , Ciclo Celular/genética , Proliferação de Células , Córtex Cerebral/anormalidades , Córtex Cerebral/patologia , Feto/patologia , Feto/virologia , Regulação da Expressão Gênica , Camundongos Endogâmicos C57BL , Microcefalia/genética , Microcefalia/patologia , Microcefalia/virologia , Células-Tronco Neurais/patologia , Infecção por Zika virus/patologia , Infecção por Zika virus/virologia
7.
J Vis Exp ; (98)2015 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-25938717

RESUMO

Müller cells are the principal glial cells of the retina. Their end-feet form the limits of the retina at the outer and inner limiting membranes (ILM), and in conjunction with astrocytes, pericytes and endothelial cells they establish the blood-retinal barrier (BRB). BRB limits material transport between the bloodstream and the retina while the ILM acts as a basement membrane that defines histologically the border between the retina and the vitreous cavity. Labeling Müller cells is particularly relevant to study the physical state of the retinal barriers, as these cells are an integral part of the BRB and ILM. Both BRB and ILM are frequently altered in retinal disease and are responsible for disease symptoms. There are several well-established methods to study the integrity of the BRB, such as the Evans blue assay or fluorescein angiography. However these methods do not provide information on the extent of BRB permeability to larger molecules, in nanometer range. Furthermore, they do not provide information on the state of other retinal barriers such as the ILM. To study BRB permeability alongside retinal ILM, we used an AAV based method that provides information on permeability of BRB to larger molecules while indicating the state of the ILM and extracellular matrix proteins in disease states. Two AAV variants are useful for such study: AAV5 and ShH10. AAV5 has a natural tropism for photoreceptors but it cannot get across to the outer retina when administered into the vitreous when the ILM is intact (i.e., in wild-type retinas). ShH10 has a strong tropism towards glial cells and will selectively label Müller glia in both healthy and diseased retinas. ShH10 provides more efficient gene delivery in retinas where ILM is compromised. These viral tools coupled with immunohistochemistry and blood-DNA analysis shed light onto the state of retinal barriers in disease.


Assuntos
Barreira Hematorretiniana/fisiopatologia , Dependovirus/fisiologia , Doenças Retinianas/fisiopatologia , Animais , Barreira Hematorretiniana/patologia , Barreira Hematorretiniana/virologia , Permeabilidade Capilar , Dependovirus/genética , Células Ependimogliais/química , Células Ependimogliais/patologia , Células Ependimogliais/virologia , Técnicas de Transferência de Genes , Genes Reporter , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Pericitos/química , Pericitos/patologia , Pericitos/virologia , Doenças Retinianas/patologia , Doenças Retinianas/virologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...