Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 214
Filtrar
1.
PLoS Pathog ; 20(4): e1012156, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38598560

RESUMEN

SARS-CoV-2 has been shown to cause wide-ranging ocular abnormalities and vision impairment in COVID-19 patients. However, there is limited understanding of SARS-CoV-2 in ocular transmission, tropism, and associated pathologies. The presence of viral RNA in corneal/conjunctival tissue and tears, along with the evidence of viral entry receptors on the ocular surface, has led to speculation that the eye may serve as a potential route of SARS-CoV-2 transmission. Here, we investigated the interaction of SARS-CoV-2 with cells lining the blood-retinal barrier (BRB) and the role of the eye in its transmission and tropism. The results from our study suggest that SARS-CoV-2 ocular exposure does not cause lung infection and moribund illness in K18-hACE2 mice despite the extended presence of viral remnants in various ocular tissues. In contrast, intranasal exposure not only resulted in SARS-CoV-2 spike (S) protein presence in different ocular tissues but also induces a hyperinflammatory immune response in the retina. Additionally, the long-term exposure to viral S-protein caused microaneurysm, retinal pigmented epithelium (RPE) mottling, retinal atrophy, and vein occlusion in mouse eyes. Notably, cells lining the BRB, the outer barrier, RPE, and the inner barrier, retinal vascular endothelium, were highly permissive to SARS-CoV-2 replication. Unexpectedly, primary human corneal epithelial cells were comparatively resistant to SARS-CoV-2 infection. The cells lining the BRB showed induced expression of viral entry receptors and increased susceptibility towards SARS-CoV-2-induced cell death. Furthermore, hyperglycemic conditions enhanced the viral entry receptor expression, infectivity, and susceptibility of SARS-CoV-2-induced cell death in the BRB cells, confirming the reported heightened pathological manifestations in comorbid populations. Collectively, our study provides the first evidence of SARS-CoV-2 ocular tropism via cells lining the BRB and that the virus can infect the retina via systemic permeation and induce retinal inflammation.


Asunto(s)
Barrera Hematorretinal , COVID-19 , Retina , SARS-CoV-2 , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Animales , Barrera Hematorretinal/virología , COVID-19/inmunología , COVID-19/virología , Ratones , Humanos , Retina/virología , Retina/inmunología , Retina/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Internalización del Virus , Glicoproteína de la Espiga del Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/inmunología , Inflamación/inmunología , Inflamación/virología , Betacoronavirus/fisiología , Tropismo Viral , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/patología
2.
Am J Pathol ; 191(10): 1787-1804, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34197777

RESUMEN

Although pathologies associated with acute virus infections have been extensively studied, the effects of long-term latent virus infections are less well understood. Human cytomegalovirus, which infects 50% to 80% of humans, is usually acquired during early life and persists in a latent state for the lifetime. The purpose of this study was to determine whether systemic murine cytomegalovirus (MCMV) infection acquired early in life disseminates to and becomes latent in the eye and if ocular MCMV can trigger in situ inflammation and occurrence of ocular pathology. This study found that neonatal infection of BALB/c mice with MCMV resulted in dissemination of virus to the eye, where it localized principally to choroidal endothelia and pericytes and less frequently to the retinal pigment epithelium (RPE) cells. MCMV underwent ocular latency, which was associated with expression of multiple virus genes and from which MCMV could be reactivated by immunosuppression. Latent ocular infection was associated with significant up-regulation of several inflammatory/angiogenic factors. Retinal and choroidal pathologies developed in a progressive manner, with deposits appearing at both basal and apical aspects of the RPE, RPE/choroidal atrophy, photoreceptor degeneration, and neovascularization. The pathologies induced by long-term ocular MCMV latency share features of previously described human ocular diseases, such as age-related macular degeneration.


Asunto(s)
Envejecimiento/patología , Coroides/patología , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Muromegalovirus/fisiología , Retina/patología , Inductores de la Angiogénesis/metabolismo , Animales , Animales Recién Nacidos , Antígenos Virales/metabolismo , Coroides/diagnóstico por imagen , Coroides/ultraestructura , Coroides/virología , ADN Viral/metabolismo , Regulación Viral de la Expresión Génica , Infecciones por Herpesviridae/diagnóstico por imagen , Interacciones Huésped-Patógeno , Terapia de Inmunosupresión , Inflamación/patología , Ratones Endogámicos BALB C , Muromegalovirus/genética , Fagocitos/patología , Retina/diagnóstico por imagen , Retina/ultraestructura , Retina/virología , Epitelio Pigmentado de la Retina/diagnóstico por imagen , Epitelio Pigmentado de la Retina/patología , Tomografía de Coherencia Óptica , Activación Viral
3.
J Pathol ; 251(2): 200-212, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32243583

RESUMEN

Age-related macular degeneration (AMD) is a complex, multifactorial, progressive disease which represents a leading cause of irreversible visual impairment and blindness in older individuals. Human cytomegalovirus (HCMV), which infects 50-80% of humans, is usually acquired during early life and persists in a latent state for the life of the individual. In view of its previously described pro-angiogenic properties, we hypothesized that cytomegalovirus might be a novel risk factor for progression to an advanced form, neovascular AMD, which is characterized by choroidal neovascularization (CNV). The purpose of this study was to investigate if latent ocular murine cytomegalovirus (MCMV) infection exacerbated the development of CNV in vascular endothelial growth factor (VEGF)-overexpressing VEGF-Ahyper mice. Here we show that neonatal infection with MCMV resulted in dissemination of virus to various organs throughout the body including the eye, where it localized principally to the choroid in both VEGF-overexpressingVEGF-Ahyper and wild-type(WT) 129 mice. By 6 months post-infection, no replicating virus was detected in eyes and extraocular tissues, although virus DNA was still present in all eyes and extraocular tissues of both VEGF-Ahyper and WT mice. Expression of MCMV immediate early (IE) 1 mRNA was detected only in latently infected eyes of VEGF-Ahyper mice, but not in eyes of WT mice. Significantly increased CNV was observed in eyes of MCMV-infected VEGF-Ahyper mice compared to eyes of uninfected VEGF-Ahyper mice, while no CNV lesions were observed in eyes of either infected or uninfected WT mice. Protein levels of several inflammatory/angiogenic factors, particularly VEGF and IL-6, were significantly higher in eyes of MCMV-infected VEGF-Ahyper mice, compared to uninfected controls. Initial studies of ocular tissue from human cadavers revealed that HCMV DNA was present in four choroid/retinal pigment epithelium samples from 24 cadavers. Taken together, our data suggest that ocular HCMV latency could be a significant risk factor for the development of AMD. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Neovascularización Coroidal/virología , Retinitis por Citomegalovirus/virología , Degeneración Macular/virología , Muromegalovirus/patogenicidad , Retina/virología , Latencia del Virus , Anciano , Anciano de 80 o más Años , Animales , Neovascularización Coroidal/genética , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Retinitis por Citomegalovirus/genética , Retinitis por Citomegalovirus/metabolismo , Retinitis por Citomegalovirus/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , Degeneración Macular/genética , Degeneración Macular/metabolismo , Degeneración Macular/patología , Masculino , Ratones de la Cepa 129 , Ratones Transgénicos , Persona de Mediana Edad , Retina/metabolismo , Retina/ultraestructura , Factores de Riesgo , Transducción de Señal , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Int J Mol Sci ; 22(15)2021 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-34360899

RESUMEN

(1) Background: caspase-12 is activated during cytomegalovirus retinitis, although its role is presently unclear. (2) Methods: caspase-12-/- (KO) or caspase-12+/+ (WT) mice were immunosup eyes were analyzed by plaque assay, TUNEL assay, immunohistochemical staining, western blotting, and real-time PCR. (3) Results: increased retinitis and a more extensive virus spread were detected in the retina of infected eyes of KO mice compared to WT mice at day 14 p.i. Compared to MCMV injected WT eyes, mRNA levels of interferons α, ß and γ were significantly reduced in the neural retina of MCMV-infected KO eyes at day 14 p.i. Although similar numbers of MCMV infected cells, similar virus titers and similar numbers of TUNEL-staining cells were detected in injected eyes of both KO and WT mice at days 7 and 10 p.i., significantly lower amounts of cleaved caspase-3 and p53 protein were detected in infected eyes of KO mice at both time points. (4) Conclusions: caspase-12 contributes to caspase-3-dependent and independent retinal bystander cell death during MCMV retinitis and may also play an important role in innate immunity against virus infection of the retina.


Asunto(s)
Apoptosis/genética , Caspasa 12/deficiencia , Retinitis por Citomegalovirus/enzimología , Inmunidad Innata/genética , Muromegalovirus/fisiología , Retina/enzimología , Neuronas Retinianas/enzimología , Animales , Caspasa 12/genética , Retinitis por Citomegalovirus/genética , Retinitis por Citomegalovirus/virología , Femenino , Etiquetado Corte-Fin in Situ/métodos , Interferones/biosíntesis , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Retina/virología , Neuronas Retinianas/virología , Transducción de Señal/genética , Proteína p53 Supresora de Tumor/metabolismo , Replicación Viral/genética
5.
J Med Virol ; 92(3): 394-398, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31670405

RESUMEN

The mechanisms that contribute to retinal tissue destruction during the onset and progression of AIDS-related human cytomegalovirus (HCMV) retinitis remain unclear. Evidence for the stimulation of multiple cell death pathways including apoptosis, necroptosis, and pyroptosis during the pathogenesis of experimental murine cytomegalovirus (MCMV) retinitis in mice with retrovirus-induced immunosuppression (MAIDS) has been reported. Parthanatos is a caspase-independent cell death pathway mediated by rapid overactivation of poly (ADP-ribose) polymerase-1 (PARP-1) and distinct from other cell death pathways. Using the MAIDS model of MCMV retinitis, studies were performed to test the hypothesis that intraocular MCMV infection of mice with MAIDS stimulates parthanatos-associated messenger RNAs (mRNAs) and proteins within the eye during the development of retinal necrosis that takes place by 10 days after MCMV infection. MCMV-infected eyes of MAIDS mice exhibited significant stimulation of PARP-1 mRNA and proteins at 3 days after infection but declined thereafter at 6 and 10 days after infection. Additional studies showed the intraocular stimulation of mRNAs or proteins before MCMV retinitis development for two additional participants in parthanatos, polymer of ADP-ribose and poly (ADP-ribose) glycohydrolase. These results provide new evidence for a role for parthanatos during MAIDS-related MCMV retinitis that may also extend to AIDS-related HCMV retinitis.


Asunto(s)
Retinitis por Citomegalovirus/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Murino/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Murino/virología , Parthanatos , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Animales , Muerte Celular , Retinitis por Citomegalovirus/complicaciones , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Síndrome de Inmunodeficiencia Adquirida del Murino/complicaciones , Muromegalovirus , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli Adenosina Difosfato Ribosa/genética , Poli Adenosina Difosfato Ribosa/metabolismo , ARN Mensajero/metabolismo , Retina/patología , Retina/virología , Retroviridae/inmunología
6.
Vet Pathol ; 57(3): 409-417, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32202218

RESUMEN

Investigations describing the ocular and lacrimal gland lesions associated with rabies are sparse. Here we characterize the pathological changes and distribution of rabies viral antigen in the eye, optic nerve, and lacrimal gland of 18 rabies cases from different mammalian species. Histology and immunohistochemistry for rabies virus, CD3, CD20, and Iba1 were performed on tissue sections of eye, optic nerve, and lacrimal gland. Polymerase chain reaction (PCR) for rabies was performed on all cases, including 7 formalin-fixed, paraffin-embedded (FFPE) and 11 frozen tissue samples of eye and lacrimal gland. Pathological changes in the eye consisted of retinal necrosis (12/18 cases) with occasional viral inclusions within ganglion cells (8/12 cases). Immunohistochemically, viral antigen was detected within the nerve fiber layer, ganglion cells, and inner plexiform layer in all 12 cases with retinal lesions and in 2 cases with no retinal lesions, as well as optic nerve (6/18 cases) and lacrimal gland epithelium (3/18 cases). CD3+ T lymphocytes were present in the retina (11/18 cases), optic nerve (2/18 cases), and lacrimal gland (11/18 cases). No CD20+ B lymphocytes or Iba1+ macrophages were detected. PCR for rabies virus was positive in 9 of 11 frozen samples but in only 2 of 7 FFPE samples. Five samples that were negative for rabies by PCR were positive by immunohistochemistry, and 2 samples were negative by both tests. These results provide evidence that rabies virus infection extends to the eye, likely via the ocular nerve, and that the lacrimal gland might be a source of viral infection.


Asunto(s)
Ojo/virología , Mamíferos/virología , Virus de la Rabia , Rabia , Animales , Antígenos CD20/metabolismo , Linfocitos B/metabolismo , Complejo CD3/metabolismo , Ojo/patología , Inmunohistoquímica/veterinaria , Aparato Lagrimal/patología , Aparato Lagrimal/virología , Nervio Óptico/patología , Nervio Óptico/virología , Reacción en Cadena de la Polimerasa/veterinaria , Rabia/patología , Rabia/transmisión , Rabia/veterinaria , Virus de la Rabia/inmunología , Virus de la Rabia/aislamiento & purificación , Retina/patología , Retina/virología , Linfocitos T/metabolismo , Lágrimas/virología
7.
Gene Ther ; 26(12): 479-490, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31562387

RESUMEN

Adeno-associated virus vectors (rAAV) are currently the most common vehicle used in clinical trials of retinal gene therapy, usually delivered through subretinal injections to target cells of the outer retina. However, targeting the inner retina requires intravitreal injections, a simple and safe procedure, which is effective for transducing the rodent retina, but still of low efficiency in the eyes of primates. We investigated whether adjuvant pharmacological agents may enhance rAAV transduction of the retinas of mouse and rat after intravitreal delivery. Tyrosine kinase inhibitors were highly efficient in mice, especially imatinib and genistein, and promoted transduction even of the outer retina. In rats, however, we report that they were not effective. Even with direct proteasomal inhibition in rats, the effects upon transduction were only minimal and restricted to the inner retina. Even tyrosine capsid mutant rAAVs in rats had a transduction profile similar to wtAAV. Thus, the differences between mouse and rat, in both eye size and the inner limiting membrane, compromise the efficiency of AAV vectors penetration from the vitreous into the retina, and impact the efficacy of strategies developed to enhance intravitreal retinal rAAV transduction. Further improvement of strategies, then are required.


Asunto(s)
Adyuvantes Farmacéuticos/administración & dosificación , Dependovirus/genética , Vectores Genéticos/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Retina/virología , Animales , Electrorretinografía , Terapia Genética , Genisteína/administración & dosificación , Mesilato de Imatinib/administración & dosificación , Inyecciones Intravítreas , Ratones , Mutación , Ratas , Transducción Genética
8.
Gene Ther ; 26(9): 386-398, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31308478

RESUMEN

Recombinant adeno-associated virus (rAAV) has been widely used for gene delivery in animal models and successfully applied in clinical trials for treating inherited retinal disease. Although subretinal delivery of AAVs can effectively transduce photoreceptors and/or retinal pigmental epithelium (RPE), cells most affected by inherited retinal diseases, the procedure is invasive and complicated, and only delivers the gene to a limited retinal area. AAVs can also be delivered intravitreally to the retina, a much less invasive nonsurgical procedure. However, intravitreal administration of non-modified AAV serotypes tends to transduce only ganglion cells and inner nuclear layer cells. To date, most non-modified AAV serotypes that have been identified are incapable of efficiently transducing photoreceptors and/or RPE when delivered intravitreally. In this study, we investigate the retinal tropism of AAVrh10 vector administered by intravitreal injection to mouse, rat, and rabbit eyes. Our results demonstrate that AAVrh10 is capable of transducing not only inner retinal cells, but also outer retinal cells in all three species, though the transduction efficiency in rabbit was low. In addition, AAVrh10 preferentially transduced outer retinal cells in mouse models of retinal disease. Therefore, AAVrh10 vector could be a useful candidate to intravitreally deliver genes to photoreceptor and RPE cells.


Asunto(s)
Dependovirus/genética , Retina , Transducción Genética/métodos , Animales , Citomegalovirus/genética , Dependovirus/fisiología , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Inyecciones Intravítreas , Masculino , Ratones , Células Fotorreceptoras/virología , Conejos , Ratas , Ratas Sprague-Dawley , Retina/efectos de los fármacos , Retina/virología , Enfermedades de la Retina/terapia , Tropismo Viral
9.
Biochem Biophys Res Commun ; 515(1): 222-227, 2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31146917

RESUMEN

Adeno-associated virus (AAV) has been studied as a safe delivery tool for gene therapy of retinal blinding diseases such as Leber's congenital amaurosis (LCA). The tropism of recombinant AAV (rAAV) including its specificity and efficiency in targeting retinal cell types has been studied with native or engineered capsids, along with specific promoters. However, one of the rAAV serotypes, rAAV2/6, has not been well-studied based on a report of low infection efficiency in the retina. We investigated the tropism of several rAAVs by subretinal injection in the adult mouse and found that rAAV2/6 predominantly infected cone photoreceptors including the main spectral type. Our data suggest that subretinal injection with rAAV2/6 may provide both an efficacious and specific means of gene delivery to cone photoreceptors in murine retinas.


Asunto(s)
Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Enfermedades de la Retina/terapia , Animales , Vectores Genéticos/administración & dosificación , Inyecciones , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/terapia , Ratones de la Cepa 129 , Opsinas/genética , Opsinas/metabolismo , Retina/virología , Células Fotorreceptoras Retinianas Conos/virología , Enfermedades de la Retina/genética , Resultado del Tratamiento
10.
J Virol ; 92(14)2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29743365

RESUMEN

Severe dengue virus (DENV) infection is associated with overactivity of the complement alternative pathway (AP) in patient studies. Here, the molecular changes in components of the AP during DENV infection in vitro were investigated. mRNA for factor H (FH), a major negative regulator of the AP, was significantly increased in DENV-infected endothelial cells (EC) and macrophages, but, in contrast, production of extracellular FH protein was not. This discord was not seen for the AP activator factor B (FB), with DENV induction of both FB mRNA and protein, nor was it seen with Toll-like receptor 3 or 4 stimulation of EC and macrophages, which induces both FH and FB mRNA and protein. Surface-bound and intracellular FH protein was, however, induced by DENV, but only in DENV antigen-positive cells, while in two other DENV-susceptible immortalized cell lines (ARPE-19 and human retinal endothelial cells), FH protein was induced both intracellularly and extracellularly by DENV infection. Regardless of the cell type, there was an imbalance in AP components and an increase in markers of complement AP activity associated with DENV-infected cells, with lower FH relative to FB protein, an increased ability to promote AP-mediated lytic activity, and increased deposition of complement component C3b on the surface of DENV-infected cells. For EC in particular, these changes are predicted to result in higher complement activity in the local cellular microenvironment, with the potential to induce functional changes that may result in increased vascular permeability, a hallmark of dengue disease.IMPORTANCE Dengue virus (DENV) is a significant human viral pathogen with a global medical and economic impact. DENV may cause serious and life-threatening disease, with increased vascular permeability and plasma leakage. The pathogenic mechanisms underlying these features remain unclear; however, overactivity of the complement alternative pathway has been suggested to play a role. In this study, we investigate the molecular events that may be responsible for this observed alternative pathway overactivity and provide novel findings of changes in the complement system in response to DENV infection in primary cell types that are a major target for DENV infection (macrophages) and pathogenesis (endothelial cells) in vivo Our results suggest a new dimension of cellular events that may influence endothelial cell barrier function during DENV infection that could expand strategies for developing therapeutics to prevent or control DENV-mediated vascular disease.


Asunto(s)
Factor B del Complemento/inmunología , Factor H de Complemento/inmunología , Vía Alternativa del Complemento , Proteínas del Sistema Complemento/inmunología , Virus del Dengue/inmunología , Dengue/inmunología , Células Cultivadas , Factor B del Complemento/metabolismo , Factor H de Complemento/metabolismo , Proteínas del Sistema Complemento/metabolismo , Dengue/metabolismo , Dengue/virología , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Endotelio Vascular/virología , Humanos , Retina/inmunología , Retina/patología , Retina/virología
11.
Int J Mol Sci ; 20(6)2019 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-30909422

RESUMEN

Pericytes are multipotent cells of the vascular system with cytoplasmic extensions proximal to endothelial cells that occur along the abluminal surface of the endothelium. The interactions between endothelial cells and pericytes are essential for proper microvascular formation, development, stabilization, and maintenance. Pericytes are essential for the regulation of paracellular flow between cells, transendothelial fluid transport, angiogenesis, and vascular immunosurveillance. They also influence the chemical composition of the surrounding microenvironment to protect endothelial cells from potential harm. Dysregulation or loss of pericyte function can result in microvascular instability and pathological consequences. Human pericytes have been shown to be targets for human cytomegalovirus (HCMV) infection and lytic replication that likely contribute to vascular inflammation. This review focuses on human vascular pericytes and their permissiveness for HCMV infection. It also discusses their implication in pathogenesis in the blood⁻brain barrier (BBB), the inner blood⁻retinal barrier (IBRB), the placenta⁻blood barrier, and the renal glomerulus as well as their potential role in subclinical vascular disease.


Asunto(s)
Infecciones por Citomegalovirus/virología , Citomegalovirus/fisiología , Interacciones Huésped-Patógeno , Pericitos/metabolismo , Pericitos/virología , Animales , Susceptibilidad a Enfermedades , Endotelio Vascular/metabolismo , Endotelio Vascular/virología , Femenino , Humanos , Células Mesangiales/metabolismo , Células Mesangiales/virología , Placenta/metabolismo , Placenta/virología , Embarazo , Retina/metabolismo , Retina/virología , Medición de Riesgo , Factores de Riesgo
12.
Mol Vis ; 24: 379-394, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29853772

RESUMEN

Purpose: The purpose of this study was to determine whether the blood-retina barrier is compromised by choroidal murine cytomegalovirus (MCMV) infection, using electron microscopy. Methods: BALB/c mice were immunosuppressed with methylprednisolone and monoclonal antibodies to CD4 and CD8. At several time points post-MCMV intraperitoneal inoculation, the eyes were removed and analyzed with western blotting and immunoelectron microscopy for the presence of MCMV early antigen (EA) and the host protein RIP3. Posterior eyecups from RIP3-/- and RIP3+/+ mice were cultured and inoculated with MCMV. At days 4, 7, and 11 post-infection, cultures were collected and analyzed with plaque assay, immunohistochemical staining, and real-time PCR (RT-PCR). Results: MCMV EA was observed in the nuclei of vascular endothelial cells and pericytes in the choriocapillaris. Disruption of Bruch's membrane was observed, especially at sites adjacent to activated platelets, and a few RPE cells containing some enlarged vesicles were found directly beneath disrupted Bruch's membrane. Some virus particles were also observed in the enlarged vesicles of RPE cells. Levels of the RIP3 protein, which was observed mainly in the RPE cells and the basement membrane of the choriocapillaris, were greatly increased following MCMV infection, while depletion of RIP3 resulted in greatly decreased inflammasome formation, as well as expression of downstream inflammation factors. Conclusions: The results suggest that systemic MCMV spreads to the choroid and replicates in vascular endothelia and pericytes of the choriocapillaris during immunosuppression. Choroidal MCMV infection is associated with in situ inflammation and subsequent disruption of Bruch's membrane and the outer blood-retina barrier.


Asunto(s)
Coroides/inmunología , Infecciones por Citomegalovirus/inmunología , Infecciones Virales del Ojo/inmunología , Huésped Inmunocomprometido , Retina/inmunología , Retinitis/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Antígenos Virales/genética , Plaquetas/inmunología , Plaquetas/patología , Plaquetas/virología , Barrera Hematorretinal/inmunología , Barrera Hematorretinal/patología , Barrera Hematorretinal/virología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Coroides/irrigación sanguínea , Coroides/patología , Coroides/virología , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/virología , Células Endoteliales , Infecciones Virales del Ojo/patología , Infecciones Virales del Ojo/virología , Femenino , Proteínas Inmediatas-Precoces/genética , Inflamasomas/inmunología , Metilprednisolona/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Muromegalovirus/crecimiento & desarrollo , Muromegalovirus/patogenicidad , Pericitos/inmunología , Pericitos/patología , Pericitos/virología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Retina/patología , Retina/virología , Epitelio Pigmentado de la Retina/inmunología , Epitelio Pigmentado de la Retina/patología , Epitelio Pigmentado de la Retina/virología , Retinitis/patología , Retinitis/virología
13.
Fish Shellfish Immunol ; 74: 627-636, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29414318

RESUMEN

Viral diseases are responsible for high rates of mortality and subsequent economic losses in modern aquaculture. The nervous necrosis virus (NNV) produces viral encephalopathy and retinopathy (VER), which affects the central nervous system, is considered one of the most serious viral diseases in marine aquaculture. Although some studies have localized NNV in the retina cells, none has dealt with immunity in the retina. Thus, for the first time, we intravitreally infected healthy specimens of European sea bass (Dicentrarchus labrax) with NNV with the aim of characterizing the immune response in the retina. Ultrastructural analysis detected important retinal injuries and structure degradation, including pycnosis, hydropic degeneration and vacuolization in some cell layers as well as myelin sheaths in the optic nerve fibres. Immunohistochemistry demonstrated that NNV replicated in the eyes. Regarding retinal immunity, NNV infection elicited the transcription of genes encoding proteins involved in the interferon (IFN) and cell-mediated cytotoxicity (CMC) responses as well as B and T cell markers, demonstrating that viral replication influences innate and adaptive responses. Further studies are needed to understand the retina immunity and whether the main retinal function, vision, is affected by nodavirus.


Asunto(s)
Lubina/genética , Lubina/inmunología , Enfermedades de los Peces/inmunología , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Retina/inmunología , Animales , Enfermedades de los Peces/virología , Nodaviridae/fisiología , Infecciones por Virus ARN/inmunología , Infecciones por Virus ARN/virología , Retina/virología , Enfermedades de la Retina/inmunología , Enfermedades de la Retina/virología
14.
Gene Ther ; 24(12): 787-800, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28872643

RESUMEN

There is much debate on the adeno-associated virus (AAV) serotype that best targets specific retinal cell types and the route of surgical delivery-intravitreal or subretinal. This study compared three of the most efficacious AAV vectors known to date in a mouse model of retinal degeneration (rd1 mouse) and macaque and human retinal explants. Green fluorescent protein (GFP) driven by a ubiquitous promoter was packaged into three AAV capsids: AAV2/8(Y733F), AAV2/2(quad Y-F) and AAV2/2(7m8). Overall, AAV2/2(7m8) transduced the largest area of retina and resulted in the highest level of GFP expression, followed by AAV2/2(quad Y-F) and AAV2/8(Y733F). AAV2/2(7m8) and AAV2/2(quad Y-F) both resulted in similar patterns of transduction whether they were injected intravitreally or subretinally. AAV2/8(Y733F) transduced a significantly smaller area of retina when injected intravitreally compared with subretinally. Retinal ganglion cells, horizontal cells and retinal pigment epithelium expressed relatively high levels of GFP in the mouse retina, whereas amacrine cells expressed low levels of GFP and bipolar cells were infrequently transduced. Cone cells were the most frequently transduced cell type in macaque retina explants, whereas Müller cells were the predominant transduced cell type in human retinal explants. Of the AAV serotypes tested, AAV2/2(7m8) was the most effective at transducing a range of cell types in degenerate mouse retina and macaque and human retinal explants.


Asunto(s)
Dependovirus/genética , Recombinación Genética , Retina/metabolismo , Tropismo Viral/genética , Animales , Modelos Animales de Enfermedad , Vectores Genéticos , Humanos , Inyecciones Intravítreas , Macaca , Ratones , Regiones Promotoras Genéticas , Retina/citología , Retina/virología , Degeneración Retiniana/genética , Células Ganglionares de la Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Ensamble de Virus
15.
J Gen Virol ; 98(12): 2993-3007, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29182510

RESUMEN

Plasma leakage is the main pathophysiological feature in severe dengue, resulting from altered vascular barrier function associated with an inappropriate immune response triggered upon infection. The present study investigated functional changes using an electric cell-substrate impedance sensing system in four (brain, dermal, pulmonary and retinal) human microvascular endothelial cell (MEC) lines infected with purified dengue virus, followed by assessment of cytokine profiles and the expression of inter-endothelial junctional proteins. Modelling of changes in electrical impedance suggests that vascular leakage in dengue-infected MECs is mostly due to the modulation of cell-to-cell interactions, while this loss of vascular barrier function observed in the infected MECs varied between cell lines and DENV serotypes. High levels of inflammatory cytokines (IL-6 and TNF-α), chemokines (CXCL1, CXCL5, CXCL11, CX3CL1, CCL2 and CCL20) and adhesion molecules (VCAM-1) were differentially produced in the four infected MECs. Further, the tight junctional protein, ZO-1, was down-regulated in both the DENV-1-infected brain and pulmonary MECs, while claudin-1, PECAM-1 and VE-cadherin were differentially expressed in these two MECs after infection. Non-purified virus stock was also studied to investigate the impact of virus stock purity on dengue-specific immune responses, and the results suggest that virus stock propagated through cell culture may include factors that mask or alter the DENV-specific immune responses of the MECs. The findings of the present study show that high DENV load differentially modulates human microvascular endothelial barrier function and disrupts the function of inter-endothelial junctional proteins during early infection with organ-specific cytokine production.


Asunto(s)
Células Endoteliales/virología , Endotelio Vascular/virología , Interacciones Huésped-Patógeno , Carga Viral/inmunología , Antígenos CD/genética , Antígenos CD/inmunología , Encéfalo/citología , Encéfalo/inmunología , Encéfalo/virología , Cadherinas/genética , Cadherinas/inmunología , Línea Celular , Quimiocina CCL2/genética , Quimiocina CCL2/inmunología , Quimiocina CCL20/genética , Quimiocina CCL20/inmunología , Quimiocina CX3CL1/genética , Quimiocina CX3CL1/inmunología , Quimiocinas CXC/genética , Quimiocinas CXC/inmunología , Claudina-1/genética , Claudina-1/inmunología , Virus del Dengue/genética , Virus del Dengue/crecimiento & desarrollo , Virus del Dengue/inmunología , Dermis/citología , Dermis/inmunología , Dermis/virología , Impedancia Eléctrica , Células Endoteliales/citología , Células Endoteliales/inmunología , Endotelio Vascular/citología , Endotelio Vascular/inmunología , Regulación de la Expresión Génica , Humanos , Interleucina-6/genética , Interleucina-6/inmunología , Pulmón/citología , Pulmón/inmunología , Pulmón/virología , Especificidad de Órganos , Permeabilidad , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Retina/citología , Retina/inmunología , Retina/virología , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/inmunología , Internalización del Virus , Proteína de la Zonula Occludens-1/genética
16.
J Virol ; 90(21): 9878-9888, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27558418

RESUMEN

Many adeno-associated virus (AAV) serotypes efficiently transduce the retina when delivered to the subretinal space but show limited success when delivered to the vitreous due to the inner limiting membrane (ILM). Subretinal delivery of AAV serotype 2 (AAV2) and its heparan sulfate (HS)-binding-deficient capsid led to similar expression, indicating transduction of the outer retina occurred by HS-independent mechanisms. However, intravitreal delivery of HS-ablated recombinant AAV2 (rAAV2) led to a 300-fold decrease in transduction compared to AAV2. Fluorescence in situ hybridization of AAV transgenes was used to identify differences in retinal trafficking and revealed that HS binding was responsible for AAV2 accumulation at the ILM. This mechanism was tested on human ex vivo retinas and showed similar accumulation with HS-binding AAV2 capsid only. To evaluate if HS binding could be applied to other AAV serotypes to enhance their transduction, AAV1 and AAV8 were modified to bind HS with a single-amino-acid mutation and tested in mice. Both HS-binding mutants of AAV1 and AAV8 had higher intravitreal transduction than their non-HS-binding parent capsid due to increased retinal accumulation. To understand the influence that HS binding has on tropism, chimeric AAV2 capsids with dual-glycan usage were tested intravitreally in mice. Compared to HS binding alone, these chimeric capsids displayed enhanced transduction that was correlated with a change in tropism. Taken together, these data indicate that HS binding serves to sequester AAV capsids from the vitreous to the ILM but does not influence retinal tropism. The enhanced retinal transduction of HS-binding capsids provides a rational design strategy for engineering capsids for intravitreal delivery. IMPORTANCE: Adeno-associated virus (AAV) has become the vector of choice for viral gene transfer and has shown great promise in clinical trials. The need for development of an easy, less invasive injection route for ocular gene therapy is met by intravitreal delivery, but delivery of AAV by this route results in poor transduction outcomes. The inner limiting membrane (ILM) creates a barrier separating the vitreous and the retina. Binding of AAV to heparan sulfate proteoglycan (HSPG) at the ILM may allow the virus to traverse this barrier for better retinal transduction. We show that HSPG binding is correlated with greater accumulation and penetration of AAV in the retina. We demonstrated that this accumulation is conserved across mouse and human retinas and that the addition of HSPG binding to other AAV capsids can increase the number of vectors accumulating at the ILM without dictating tropism.


Asunto(s)
Dependovirus/metabolismo , Dependovirus/fisiología , Vectores Genéticos/metabolismo , Heparitina Sulfato/metabolismo , Retina/virología , Tropismo/fisiología , Animales , Cápside/metabolismo , Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/genética , Humanos , Ratones , Transducción Genética/métodos , Transgenes/genética , Tropismo/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA