Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Retina ; 42(2): 236-243, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35050927

ABSTRACT

PURPOSE: Retinal manifestations have been described in COVID-19 patients, but it is unknown whether SARS-CoV-2, the causal agent in COVID-19, can directly infect posterior ocular tissues. Here, we investigate SARS-CoV-2 host factor gene expression levels and their distribution across retinal and choroidal cell types. METHODS: Query of single-cell RNA sequencing data from human retina and choroid. RESULTS: We find no relevant expression of two key genes involved in SARS-CoV-2 entry, ACE2 and TMPRSS2, in retinal cell types. By contrast, scarce expression levels could be detected in choroidal vascular cells. CONCLUSION: Given the current understanding of viral host cell entry, these findings suggest a low vulnerability of the posterior eye segment to SARS-CoV-2 with a potential weak spot in the vasculature, which could play a putative causative role in ocular lesions in COVID-19 patients. This may qualify the vasculature of the human posterior eye segment as an in vivo biomarker for life-threatening vascular occlusions in COVID-19 patients.


Subject(s)
COVID-19/epidemiology , Eye Infections, Viral/virology , Gene Expression Regulation, Viral , Posterior Eye Segment/virology , SARS-CoV-2 , Serine Endopeptidases/genetics , Virus Internalization , COVID-19/virology , Eye Infections, Viral/epidemiology , Eye Infections, Viral/pathology , Humans , Posterior Eye Segment/pathology , RNA, Viral/genetics , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/virology , Serine Endopeptidases/biosynthesis
3.
JCI Insight ; 5(24)2020 12 17.
Article in English | MEDLINE | ID: mdl-33180748

ABSTRACT

Congenital Zika syndrome (CZS) is associated with microcephaly and various neurological, musculoskeletal, and ocular abnormalities, but the long-term pathogenesis and postnatal progression of ocular defects in infants are not well characterized. Rhesus macaques are superior to rodents as models of CZS because they are natural hosts of the virus and share similar immune and ocular characteristics, including blood-retinal barrier characteristics and the unique presence of a macula. Using a previously described model of CZS, we infected pregnant rhesus macaques with Zika virus (ZIKV) during the late first trimester and characterized postnatal ocular development and evolution of ocular defects in 2 infant macaques over 2 years. We found that one of them exhibited colobomatous chorioretinal atrophic lesions with macular and vascular dragging as well as retinal thinning caused by loss of retinal ganglion neuron and photoreceptor layers. Despite these congenital ocular malformations, axial elongation and retinal development in these infants progressed at normal rates compared with healthy animals. The ZIKV-exposed infants displayed a rapid loss of ZIKV-specific antibodies, suggesting the absence of viral replication after birth, and did not show any behavioral or neurological defects postnatally. Our findings suggest that ZIKV infection during early pregnancy can impact fetal retinal development and cause congenital ocular anomalies but does not appear to affect postnatal ocular growth.


Subject(s)
Prenatal Exposure Delayed Effects/virology , Retina/embryology , Zika Virus Infection/metabolism , Animals , Blood-Retinal Barrier/virology , Female , Macaca/virology , Macaca mulatta , Pregnancy , Pregnancy Complications, Infectious/virology , Retina/virology , Retinal Degeneration/virology , Retinal Ganglion Cells/physiology , Retinal Ganglion Cells/virology , Virus Replication , Zika Virus/immunology , Zika Virus/pathogenicity , Zika Virus Infection/complications , Zika Virus Infection/physiopathology
4.
Curr Gene Ther ; 19(6): 404-412, 2020.
Article in English | MEDLINE | ID: mdl-32072884

ABSTRACT

BACKGROUND: Adeno-associated Virus (AAV) vectors are the most promising vehicles for therapeutic gene delivery to the retina. To develop a practical gene delivery tool, achieving high AAV transduction efficiency in specific cell types is often required. AAV-mediated targeted expression in retinal bipolar cells is needed in certain applications such as optogenetic therapy, however, the transduction efficiency driven by endogenous cell-specific promoters is usually low. Methods that can improve AAV transduction efficiency in bipolar cells need to be developed. OBJECTIVE: The study aimed to examine the effect of proteasome inhibitors on AAV-mediated transduction efficiency in retinal bipolar cells. METHODS: Quantitative analysis of fluorescent reporter protein expression was performed to assess the effect of two proteasome inhibitors, doxorubicin and MG132, on AAV-mediated transduction efficiency in retinal bipolar cells in mice. RESULTS: Our results showed that doxorubicin can increase the AAV transduction efficiency in retinal bipolar cells in a dose-dependent manner. We also observed doxorubicin-mediated cytotoxicity in retinal neurons, but the cytotoxicity could be mitigated by the coapplication of dexrazoxane. Three months after the coapplication of doxorubicin (300 µM) and dexrazoxane, the AAV transduction efficiency in retinal bipolar cells increased by 33.8% and no cytotoxicity was observed in all the layers of the retina. CONCLUSION: Doxorubicin could enhance the AAV transduction efficiency in retinal bipolar cells in vivo. The potential long-term cytotoxicity caused by doxorubicin to retinal neurons could be partially mitigated by dexrazoxane. The coapplication of doxorubicin and dexrazoxane may serve as a potential adjuvant regimen for improving AAV transduction efficiency in retinal bipolar cells.


Subject(s)
Gene Expression/drug effects , Proteasome Inhibitors/pharmacology , Retinal Bipolar Cells/drug effects , Retinal Bipolar Cells/metabolism , Animals , Dependovirus/genetics , Dexrazoxane/pharmacology , Doxorubicin/pharmacology , Genetic Vectors , Leupeptins/pharmacology , Mice , Mice, Inbred C57BL , Models, Animal , Retina/metabolism , Retina/virology , Retinal Bipolar Cells/virology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/virology , Transduction, Genetic/methods
5.
Methods Mol Biol ; 2060: 419-428, 2020.
Article in English | MEDLINE | ID: mdl-31617195

ABSTRACT

Mammalian nervous tissues are heterogeneous. The retina, brain, spinal cord, and peripheral sensory and autonomic ganglia are each composed of neuronal and glial cell partners embedded in a connective tissue bed and supplied by vascular and immune cells. This complicated structure presents many challenges to neuroscientists and cell biologists (e.g., how to carry out a quantitative study of neurons surrounded by the hormonal and immune stimuli of supporting cells). A reductionist view has led investigators to study tissue slices and cultures of isolated neurons in vitro, subtracting the immune and vascular components to simplify the problem. Recently, investigators have extended the approach and produced organoids which are derived from embryonic neurons from induced pluripotent stem cells (Muffat et al., Proc Natl Acad Sci U S A 115:7117-7122, 2018).Using this approach advances have been made in the study of viral infections of the nervous system. For example, by using a genetically modified carrier virus, they can compare the effect of different viral envelope proteins on viral tropism and viral response pathways. However, the timed delivery of hormonal stimuli and interactions with immune cells remain problematic.We present an alternative method for studying these issues using the axonal transport of Herpes simplex virus in mature retinal neurons in vivo. Using genetically identical mice and carefully controlling the delivery of virus, an investigator can obtain insight into the transport of virus to and from the neuron cell body within the cellular environment of an intact, mature animal. This allows confirmation and extension of results seen in vitro.


Subject(s)
DNA, Viral/metabolism , Herpes Simplex , Herpesvirus 1, Human/metabolism , Retinal Ganglion Cells , Animals , Biological Transport, Active , Disease Models, Animal , Herpes Simplex/metabolism , Herpes Simplex/pathology , Male , Mice , Mice, Inbred BALB C , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/virology
6.
mSphere ; 4(3)2019 05 08.
Article in English | MEDLINE | ID: mdl-31068433

ABSTRACT

Zika virus (ZIKV) infection during pregnancy leads to devastating fetal outcomes, including neurological (microcephaly) and ocular pathologies such as retinal lesions, optic nerve abnormalities, chorioretinal atrophy, and congenital glaucoma. Only clinical case reports have linked ZIKV infection to causing glaucoma, a major blinding eye disease. In the present study, we have investigated the role of ZIKV in glaucoma pathophysiology using in vitro and in vivo experimental models. We showed that human primary trabecular meshwork (Pr. TM) cells, as well as a human GTM3 cell line, were permissive to ZIKV infection. ZIKV induced the transcription of various genes expressing pattern recognition receptors (TLR2, TLR3, and RIG-I), cytokines/chemokines (TNF-α, IL-1ß, CCL5, and CXCL10), interferons (IFN-α2, IFN-ß1, and IFN-γ), and interferon-stimulated genes (ISG15 and OAS2) in Pr. TM cells. ZIKV infection in IFNAR1-/- and wild-type (WT) mouse eyes resulted in increased intraocular pressure (IOP) and the development of chorioretinal atrophy. Anterior chamber (AC) inoculation of ZIKV caused infectivity in iridocorneal angle and TM, leading to the death of TM cells in the mouse eyes. Moreover, anterior segment tissue of infected eyes exhibited increased expression of inflammatory mediators and interferons. Furthermore, ZIKV infection in IFNAR1-/- mice resulted in retinal ganglion cell (RGC) death and loss, coinciding with optic nerve infectivity and disruption of anterograde axonal transport. Because of similarity in glaucomatous pathologies in our study and other experimental glaucoma models, ZIKV infection can be used to study infectious triggers of glaucoma, currently an understudied area of investigation.IMPORTANCE Ocular complications due to ZIKV infection remains a major public health concern because of their ability to cause visual impairment or blindness. Most of the previous studies have shown ZIKV-induced ocular pathology in the posterior segment (i.e., retina) of the eye. However, some recent clinical reports from affected countries highlighted the importance of ZIKV in affecting the anterior segment of the eye and causing congenital glaucoma. Because glaucoma is the second leading cause of blindness worldwide, it is imperative to study ZIKV infection in causing glaucoma to identify potential targets for therapeutic intervention. In this study, we discovered that ZIKV permissively infects human TM cells and evokes inflammatory responses causing trabeculitis. Using a mouse model, we demonstrated that ZIKV infection resulted in higher IOP, increased RGC loss, and optic nerve abnormalities, the classical hallmarks of glaucoma. Collectively, our study provides new insights into ocular ZIKV infection resulting in glaucomatous pathology.


Subject(s)
Eye/pathology , Eye/virology , Glaucoma/virology , Trabecular Meshwork/virology , Zika Virus Infection/complications , Zika Virus/pathogenicity , Animals , Cell Death , Cell Line , Chemokines/genetics , Cytokines/genetics , Disease Models, Animal , Female , Glaucoma/physiopathology , Humans , Interferons/genetics , Intraocular Pressure , Male , Mice , Mice, Inbred C57BL , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/virology , Trabecular Meshwork/pathology , Transcriptome , Zika Virus Infection/pathology
7.
Indian J Med Res ; 150(5): 498-503, 2019 11.
Article in English | MEDLINE | ID: mdl-31939394

ABSTRACT

Background & objectives: Mouse is a preferred animal model for studying pathogenesis of Japanese encephalitis virus (JEV) infections, and different routes of inoculation have been tried. Some neurotropic viruses can reach the brain following infection through ocular route. This study was undertaken to establish JEV-induced clinical disease in mouse model through conjunctival route and document the neuropathological effects. Methods: Ten two-week old Swiss albino mice were inoculated with 5 µl Vero cell cultured virus containing 104.7 TCID50 JEV through conjunctival route. Clinical signs of mice were observed twice daily. After necropsy examination, different organs including eyes and olfactory bulbs were collected for histopathological examination, quantification of viral copy number and antigen by real-time TaqMan assay and immunohistochemistry, respectively. Results: Infected mice showed characteristic clinical signs of JE by 4 days post-infection (dpi). Histopathological lesions in brain included perivascular cuffing by mononuclear cells, focal gliosis, necrosis of neurons and neuronophagia and astrocytosis in the cerebrum, cerebellum and the brainstem. JEV viral load was highest in the brain followed by intestine, heart, liver, spleen, lung and kidney. JEV antigen was detected in the bipolar and ganglion cells of the retina and in the mitral cells and periglomerular cells of olfactory bulb and other parts of the brain. Interpretation & conclusions: JEV infection in mice through conjunctival route produced characteristic clinical signs of the disease and neuropathological lesions. Demonstration of JEV antigen in association with neuropathological lesions in the central nervous system and neuronal cells of the eye showed that conjunctival route could be an effective alternate route for virus invasion into the brain. These findings have biosafety implications for researchers, veterinary practitioners and pig farmers.


Subject(s)
Conjunctiva/virology , Encephalitis Virus, Japanese/pathogenicity , Encephalitis, Japanese/transmission , Encephalitis, Japanese/virology , Animals , Central Nervous System/pathology , Central Nervous System/virology , Chlorocebus aethiops , Conjunctiva/pathology , Disease Models, Animal , Encephalitis, Japanese/pathology , Humans , Mice , Neuropathology , Retinal Ganglion Cells/virology , Vero Cells
8.
Emerg Microbes Infect ; 7(1): 96, 2018 May 25.
Article in English | MEDLINE | ID: mdl-29802245

ABSTRACT

Zika-infected patients can have eye involvement ranging from mild conjunctivitis to severe chorioretinal lesions, however the possible long-term sequelae of infection and timeline to recovery remain unknown. Here we describe the partial recovery of chorioretinal lesions in an immunocompetent patient diagnosed with bilateral posterior uveitis associated with Zika infection and show that some lesions resolved with focal atrophy evident as pigmentary changes on funduscopy. To better understand the progression of the lesions and correlate the changes in fundus imaging with local viral load, immune responses, and retinal damage, we developed a symptomatic mouse model of ocular Zika virus infection. Imaging of the fundus revealed multiple hypopigmentary patches indicative of chorioretinal degeneration as well as thinning of the retina that mirror the lesions in patients. Microscopically, the virus primarily infected the optic nerve, retinal ganglion cells, and inner nuclear layer cells, showing thinning of the outer plexiform layer. During acute infection, the eyes showed retinal layer disorganization, retinitis, vitritis, and focal choroiditis, with mild cellular infiltration and increased expression of tumor necrosis factor, interferon-γ, granzyme B, and perforin. Focal areas of gliosis and retinal degeneration persisted 60 dpi. The model recapitulates features of ZIKA infections in patients and should help elucidate the mechanisms underlying the damage to the eyes and aid in the development of effective therapeutics.


Subject(s)
Chorioretinitis/virology , Retina/virology , Uveitis, Posterior/virology , Zika Virus Infection/pathology , Zika Virus/isolation & purification , Adult , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Conjunctivitis, Viral/virology , Humans , Killer Cells, Natural/immunology , Male , Mice , Mice, Inbred C57BL , Optic Nerve/virology , Retinal Ganglion Cells/virology
9.
JAMA Ophthalmol ; 135(10): 1069-1076, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28880978

ABSTRACT

Importance: A better pathophysiologic understanding of the neurodevelopmental abnormalities observed in neonates exposed in utero to Zika virus (ZIKV) is needed to develop treatments. The retina as an extension of the diencephalon accessible to in vivo microcopy with spectral-domain optical coherence tomography (SD-OCT) can provide an insight into the pathophysiology of congenital Zika syndrome (CZS). Objective: To quantify the microstructural changes of the retina in CZS and compare these changes with those of cobalamin C (cblC) deficiency, a disease with potential retinal maldevelopment. Design, Setting, and Participants: This case series included 8 infants with CZS and 8 individuals with cblC deficiency. All patients underwent ophthalmologic evaluation at 2 university teaching hospitals and SD-OCT imaging in at least 1 eye. Patients with cblC deficiency were homozygous or compound heterozygotes for mutations in the methylmalonic aciduria and homocystinuria type C (MMACHC) gene. Data were collected from January 1 to March 17, 2016, for patients with CZS and from May 4, 2015, to April 23, 2016, for patients with cblC deficiency. Main Outcomes and Measures: The SD-OCT cross-sections were segmented using automatic segmentation algorithms embedded in the SD-OCT systems. Each retinal layer thickness was measured at critical eccentricities using the position of the signal peaks and troughs on longitudinal reflectivity profiles. Results: Eight infants with CZS (5 girls and 3 boys; age range, 3-5 months) and 8 patients with cblC deficiency (3 girls and 5 boys; age range, 4 months to 15 years) were included in the analysis. All 8 patients with CZS had foveal abnormalities in the analyzed eyes (8 eyes), including discontinuities of the ellipsoid zone, thinning of the central retina with increased backscatter, and severe structural disorganization, with 3 eyes showing macular pseudocolobomas. Pericentral retina with normal lamination showed a thinned (<30% of normal thickness) ganglion cell layer (GCL) that colocalized in 7 of 8 eyes with a normal photoreceptor layer. The inner nuclear layer was normal or had borderline thinning. The central retinal degeneration was similar to that of cblC deficiency. Conclusions and Relevance: Congenital Zika syndrome showed a central retinal degeneration with severe GCL loss, borderline inner nuclear layer thinning, and less prominent photoreceptor loss. The findings provide the first, to date, in vivo evidence in humans for possible retinal maldevelopment with a predilection for retinal GCL loss in CZS, consistent with a murine model of the disease and suggestive of in utero depletion of this neuronal population as a consequence of Zika virus infection.


Subject(s)
Eye Infections, Viral/diagnosis , Pregnancy Complications, Infectious , Retinal Degeneration/diagnosis , Retinal Ganglion Cells/pathology , Zika Virus Infection/diagnosis , Adolescent , Antibodies, Viral/blood , Child , Child, Preschool , Eye Infections, Viral/congenital , Eye Infections, Viral/virology , Female , Humans , Infant , Male , Photoreceptor Cells, Vertebrate/pathology , Pregnancy , Retinal Degeneration/congenital , Retinal Degeneration/virology , Retinal Ganglion Cells/virology , Retrospective Studies , Tomography, Optical Coherence , Visual Acuity , Vitamin B 12 Deficiency/diagnosis , Zika Virus/immunology , Zika Virus Infection/congenital , Zika Virus Infection/virology
10.
PLoS One ; 10(4): e0124646, 2015.
Article in English | MEDLINE | ID: mdl-25894397

ABSTRACT

Herpes simplex virus-1 (HSV-1) infection causes severe conditions, with serious complications, including corneal blindness from uncontrolled ocular infections. An important cellular defense mechanism against HSV-1 infection is autophagy. The autophagic response of the host cell was suggested to be regulated by HSV-1. In this study, we performed a detailed analysis of autophagy in multiple HSV-1-targeted cell types, and under various infection conditions that recapitulate a productive infection model. We found that autophagy was slightly inhibited in one cell type, while in other cell types autophagy maintained its basal levels mostly unchanged during productive infection. This study refines the concept of HSV-1-mediated autophagy regulation to imply either inhibition, or prevention of activation, of the innate immune pathway.


Subject(s)
Autophagy , Herpes Simplex/pathology , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Host-Pathogen Interactions , Models, Biological , Animals , Epithelial Cells/pathology , Epithelial Cells/virology , Epithelium, Corneal/pathology , Epithelium, Corneal/virology , Fibroblasts/pathology , Fibroblasts/virology , HeLa Cells , Humans , Mice , Rats , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/virology
11.
Invest Ophthalmol Vis Sci ; 55(11): 7137-46, 2014 Oct 08.
Article in English | MEDLINE | ID: mdl-25298417

ABSTRACT

PURPOSE: Extensive death of uninfected bystander neuronal cells is an important component of the pathogenesis of cytomegalovirus retinitis. Our previous results have shown that caspase 3-dependent and -independent pathways are involved in death of uninfected bystander cells during murine cytomegalovirus (MCMV) retinitis and also that Bcl-2, an important inhibitor of apoptosis via the Bax-mediated mitochondrial pathway, is downregulated during this process. The purpose of this study was to determine whether Bax-mediated mitochondrial damage has a significant role in the death of uninfected retinal cells. METHODS: BALB/c mice, Bax(-/-) mice, or Bax(+/+) mice were immunosuppressed with methylprednisolone and infected with 5 × 10(3) plaque-forming units (PFU) of the K181 strain of MCMV via the supraciliary route. Injected eyes were analyzed by plaque assay, electron microscopy, hematoxylin and eosin (H&E) staining, TUNEL assay, Western blot (for caspase 3, caspase 12, Bax, receptor interacting protein-1 [RIP1] and receptor interacting protein-3 [RIP3]), as well as immunohistochemical staining for MCMV early antigen and cleaved caspase 3. RESULTS: Significantly more Bax was detected in mitochondrial fractions of MCMV-infected eyes than in mitochondrial fractions of mock-infected control eyes. Furthermore, the level of cleaved caspase 3 was significantly lower in MCMV-infected Bax(-/-) eyes than in MCMV-infected Bax(+/+) eyes. However, more caspase 3-independent cell death of uninfected bystander retinal cells and more cleaved RIP1 were observed in Bax(-/-) than in Bax(+/+) eyes. CONCLUSIONS: During MCMV retinitis, Bax is activated and has an important role in death of uninfected bystander retinal cells by caspase 3-dependent apoptosis. Although the exact mechanism remains to be deciphered, active Bax might also prevent death of some types of uninfected retinal cells by a caspase 3-independent pathway.


Subject(s)
Cell Death , Cytomegalovirus Retinitis/pathology , Retinal Ganglion Cells/ultrastructure , bcl-2-Associated X Protein/physiology , Animals , Blotting, Western , Caspase 3/metabolism , Cytomegalovirus Retinitis/metabolism , Cytomegalovirus Retinitis/virology , Disease Models, Animal , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Inbred BALB C , Microscopy, Electron , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/virology
12.
Methods Mol Biol ; 1144: 283-92, 2014.
Article in English | MEDLINE | ID: mdl-24671691

ABSTRACT

The mammalian retina, brain, spinal cord, and peripheral ganglia are all heterogeneous tissues. Each is composed of neuronal and glial cell partners embedded in a connective tissue bed and supplied by vascular and immune cells. This complicated structure presents many challenges to neuroscientists and cell biologists, e.g., how to carry out a quantitative study of neurons in a mature animal surrounded by the hormonal and immune stimuli. A reductionist view leads investigators to study single neurons in vitro, subtracting the immune and vascular components and simplifying the problem. While this has advantages, it limits relevance of the study. We present a method for studying the axonal transport of Herpes simplex virus in mature neurons in situ. Using genetically identical mice and carefully controlling the delivery of virus, an investigator can obtain insight into the transport of virus to and from the neuron cell body within the cellular environment of an intact animal.


Subject(s)
Axonal Transport/genetics , DNA/genetics , Herpesvirus 1, Human/metabolism , Molecular Biology/methods , Animals , DNA/metabolism , Herpesvirus 1, Human/genetics , Humans , Mice , Neurons/cytology , Neurons/virology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/virology , Virus Replication/genetics
13.
Invest Ophthalmol Vis Sci ; 54(2): 962-7, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23322573

ABSTRACT

PURPOSE: How herpes simplex virus (HSV) is transported from the infected neuron cell body to the axon terminal is poorly understood. Several viral proteins are candidates for regulating the process, but the evidence is controversial. We compared the results of Us9 deletions in two HSV strains (F and NS) using a novel quantitative assay to test the hypothesis that the viral protein Us9 regulates the delivery of viral DNA to the distal axon of retinal ganglion cells in vivo. We also deleted a nine-amino acid motif in the Us9 protein of F strain (Us9-30) to define the role of this domain in DNA delivery. METHODS: The vitreous chambers of murine eyes were infected with equivalent amounts of F or NS strains of HSV. At 3, 4, or 5 days post infection (dpi), both optic tracts (OT) were dissected and viral genome was quantified by qPCR. RESULTS: At 3 dpi, the F strain Us9- and Us9-30 mutants delivered less than 10% and 1%, respectively, of the viral DNA delivered after infection with the Us9R (control) strain. By 4 and 5 dpi, delivery of viral DNA had only partially recovered. Deletion of Us9 in NS-infected mice has a less obvious effect on delivery of new viral DNA to the distal OT. By 3 dpi the NS Us9-strain delivered 22% of the DNA that was delivered by the NS wt, and by 4 and 5 dpi the amount of Us9-viral DNA was 96% and 81%, respectively. CONCLUSIONS: A highly conserved acidic cluster within the Us9 protein plays a critical role for genome transport to the distal axon. The transport is less dependent on Us9 expression in the NS than in the F strain virus. This assay can be used to compare transport efficiency in other neurotropic viral strains.


Subject(s)
Axons/virology , DNA, Viral/genetics , Gene Expression Regulation, Viral , Retinal Ganglion Cells/virology , Simplexvirus/genetics , Viral Envelope Proteins/genetics , Viral Proteins/genetics , Animals , Axons/metabolism , Axons/pathology , Cell Line , Disease Models, Animal , Eye Infections, Viral/genetics , Eye Infections, Viral/pathology , Eye Infections, Viral/virology , Genome, Viral , Male , Mice , Mice, Inbred BALB C , Polymerase Chain Reaction , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Viral Envelope Proteins/biosynthesis , Viral Proteins/metabolism
14.
J Vis Exp ; (68)2012 Oct 17.
Article in English | MEDLINE | ID: mdl-23117695

ABSTRACT

Classical methods for studying neuronal circuits are fairly low throughput. Transsynaptic viruses, particularly the pseudorabies (PRV) and rabies virus (RABV), and more recently vesicular stomatitis virus (VSV), for studying circuitry, is becoming increasingly popular. These higher throughput methods use viruses that transmit between neurons in either the anterograde or retrograde direction. Recently, a modified RABV for monosynaptic retrograde tracing was developed. (Figure 1A). In this method, the glycoprotein (G) gene is deleted from the viral genome, and resupplied only in targeted neurons. Infection specificity is achieved by substituting a chimeric G, composed of the extracellular domain of the ASLV-A glycoprotein and the cytoplasmic domain of the RABV-G (A/RG), for the normal RABV-G(1). This chimeric G specifically infects cells expressing the TVA receptor(1). The gene encoding TVA can been delivered by various methods(2-8). Following RABV-G infection of a TVA-expressing neuron, the RABV can transmit to other, synaptically connected neurons in a retrograde direction by nature of its own G which was co-delivered with the TVA receptor. This technique labels a relatively large number of inputs (5-10%)(2) onto a defined cell type, providing a sampling of all of the inputs onto a defined starter cell type. We recently modified this technique to use VSV as a transsynaptic tracer(9). VSV has several advantages, including the rapidity of gene expression. Here we detail a new viral tracing system using VSV useful for probing microcircuitry with increased resolution. While the original published strategies by Wickersham et al.(4) and Beier et al.(9) permit labeling of any neurons that project onto initially-infected TVA-expressing-cells, here VSV was engineered to transmit only to TVA-expressing cells (Figure 1B). The virus is first pseudotyped with RABV-G to permit infection of neurons downstream of TVA-expressing neurons. After infecting this first population of cells, the virus released can only infect TVA-expressing cells. Because the transsynaptic viral spread is limited to TVA-expressing cells, presence of absence of connectivity from defined cell types can be explored with high resolution. An experimental flow chart of these experiments is shown in Figure 2. Here we show a model circuit, that of direction-selectivity in the mouse retina. We examine the connectivity of starburst amacrine cells (SACs) to retinal ganglion cells (RGCs).


Subject(s)
Neural Pathways/physiology , Neural Pathways/virology , Vesiculovirus/physiology , Amacrine Cells/physiology , Amacrine Cells/virology , Animals , Mice , Neurons/physiology , Neurons/virology , Retinal Ganglion Cells/physiology , Retinal Ganglion Cells/virology , Synapses/physiology , Synapses/virology , Vesiculovirus/genetics
15.
Acad Radiol ; 17(10): 1242-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20619698

ABSTRACT

RATIONALE AND OBJECTIVES: The aim of this study was to investigate whether ultrasound-mediated microbubble destruction enhances the transduction efficiency of recombinant adeno-associated virus (rAAV)-mediated enhanced green fluorescent protein (EGFP) gene into retinal ganglion cells (RGCs) of rats and whether it causes relevant adverse effects. MATERIALS AND METHODS: Thirty-two adult Sprague-Dawley rats were divided into four groups with different ultrasound powers, and retinal flat mounts and hematoxylin and eosin staining sections were made for optimization of parameters. A further 70 adult Sprague-Dawley rats were divided into four groups randomly. The first group (group A) was used as a normal control with 10 rats, and the remaining rats were evenly divided into groups B, C, and D. Each group included 20 rats. Groups B and C received rAAV-encoding EGFP gene (rAAV(2)-EGFP) in phosphate-buffered saline without and with ultrasound to the retina, respectively. Group D received microbubbles and rAAV(2)-EGFP mixture and ultrasound to the retina. The injection approach was intravitreal injection for all eyes. After 21 days, RGCs were labeled retrogradely with Fluoro-Gold. After 28 days, retinal flat mounts, frozen sections, and pathologic sections were assessed in each group. Expression of EGFP reporter gene was observed on laser confocal microscopy and evaluated according to average optical density and transfected RGC rate. To evaluate adverse effects with retinal flat mounts, labeled RGCs were counted, and retinal pathologic sections were examined. RESULTS: When ultrasound parameters (frequency, 0.3 MHz; power, 0.5 W/cm(2); total time, 60 seconds [irradiation time, 5 seconds; interval time, 10 seconds; four times]) were selected, EGFP expression was stronger, and retinas were not damaged. In the second part of the experiment, RGCs were labeled with Fluoro-Gold successfully. Green fluorescence can be observed in labeled RGCs in groups B to D. While average optical density and transfected RGC rate in group D were the highest compared to the other groups, no significant reduction in RGC number was detected with retrograde labeling. No obvious damage was observed with pathologic sections. CONCLUSIONS: Ultrasound-mediated microbubble destruction can effectively and safely enhance rAAV delivery to RGCs in rats, and it may represent a novel gene delivery method in gene therapy for glaucomatous optic neuroprotection.


Subject(s)
Adenoviridae/physiology , Genetic Vectors/genetics , Microbubbles , Retinal Ganglion Cells/radiation effects , Retinal Ganglion Cells/virology , Sonication/methods , Transfection/methods , Animals , Cells, Cultured , Female , In Vitro Techniques , Male , Rats , Rats, Sprague-Dawley
16.
Curr Biol ; 17(11): 981-8, 2007 Jun 05.
Article in English | MEDLINE | ID: mdl-17524644

ABSTRACT

Intrinsically photosensitive melanopsin-containing retinal ganglion cells (ipRGCs) control important physiological processes, including the circadian rhythm, the pupillary reflex, and the suppression of locomotor behavior (reviewed in [1]). ipRGCs are also activated by classical photoreceptors, the rods and cones, through local retinal circuits [2, 3]. ipRGCs can be transsynaptically labeled through the pupillary-reflex circuit with the derivatives of the Bartha strain of the alphaherpesvirus pseudorabies virus(PRV) [4, 5] that express GFP [6-12]. Bartha-strain derivatives spread only in the retrograde direction [13]. There is evidence that infected cells function normally for a while during GFP expression [7]. Here we combine transsynaptic PRV labeling, two-photon laser microscopy, and electrophysiological techniques to trace the local circuit of different ipRGC subtypes in the mouse retina and record light-evoked activity from the transsynaptically labeled ganglion cells. First, we show that ipRGCs are connected by monostratified amacrine cells that provide strong inhibition from classical-photoreceptor-driven circuits. Second, we show evidence that dopaminergic interplexiform cells are synaptically connected to ipRGCs. The latter finding provides a circuitry link between light-dark adaptation and ipRGC function.


Subject(s)
Retinal Ganglion Cells/physiology , Rod Opsins/metabolism , Visual Pathways/physiology , Amacrine Cells/physiology , Amacrine Cells/virology , Animals , Green Fluorescent Proteins/analysis , Herpesvirus 1, Suid/genetics , Herpesvirus 1, Suid/metabolism , Mice , Retinal Ganglion Cells/radiation effects , Retinal Ganglion Cells/virology , Synaptic Transmission
17.
Neuroscience ; 146(3): 974-85, 2007 May 25.
Article in English | MEDLINE | ID: mdl-17382478

ABSTRACT

Many membranous organelles and protein complexes are normally transported anterograde within axons to the presynaptic terminal, and details of the motors, adaptors and cargoes have received significant attention. Much less is known about the transport in neurons of non-membrane bound particles, such as mRNAs and their associated proteins. We propose that herpes simplex virus type 1 (HSV) can be used to study the detailed mechanisms regulating long distance transport of particles in axons. A critical step in the transmission of HSV from one infected neuron to the next is the polarized anterograde axonal transport of viral DNA from the host infected nerve cell body to the axon terminal. Using the in vivo mouse retinal ganglion cell model infected with wild type virus or a mutant strain that lacks the protein Us9, we found that Us9 protein was necessary for long distance anterograde axonal transport of viral nucleocapsid (DNA surrounded by capsid proteins), but unnecessary for transport of virus envelope. Thus, we conclude that nucleocapsid can be transported independently down axons via a Us9-dependent mechanism.


Subject(s)
Axonal Transport/physiology , Capsid Proteins/metabolism , Herpesvirus 1, Human/metabolism , Herpesvirus 1, Human/physiology , Animals , Blotting, Western , Cell Proliferation/drug effects , DNA/biosynthesis , DNA/genetics , Electrophoresis, Polyacrylamide Gel , Eye , Glycoproteins/metabolism , Herpesvirus 1, Human/genetics , Immunohistochemistry , Injections , Intracellular Signaling Peptides and Proteins , Lipoproteins/genetics , Mice , Mice, Knockout , Phosphoproteins/genetics , Presynaptic Terminals/metabolism , Retinal Ganglion Cells/virology , Reverse Transcriptase Polymerase Chain Reaction , Viral Proteins/genetics , Viral Proteins/metabolism
18.
Gene Ther ; 13(18): 1328-41, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16708079

ABSTRACT

We compared the effects of intravitreal injection of bi-cistronic adeno-associated viral (AAV-2) vectors encoding enhanced green fluorescent protein (GFP) and either ciliary neurotrophic factor (CNTF), brain-derived neurotrophic factor (BDNF) or growth-associated protein-43 (GAP43) on adult retinal ganglion cell (RGC) survival and regeneration following (i) optic nerve (ON) crush or (ii) after ON cut and attachment of a peripheral nerve (PN). At 7 weeks after ON crush, quantification of betaIII-tubulin immunostaining revealed that, compared to AAV-GFP controls, RGC survival was not enhanced by AAV-GAP43-GFP but was increased in AAV-CNTF-GFP (mean RGCs/retina: 17 450+/-358 s.e.m.) and AAV-BDNF-GFP injected eyes (10 200+/-4064 RGCs/retina). Consistent with increased RGC viability in AAV-CNTF-GFP and AAV-BDNF-GFP injected eyes, these animals possessed many betaIII-tubulin- and GFP-positive fibres proximal to the ON crush. However, only in the AAV-CNTF-GFP group were regenerating RGC axons seen in distal ON (1135+/-367 axons/nerve, 0.5 mm post-crush), some reaching the optic chiasm. RGCs were immunoreactive for CNTF and quantitative RT-PCR revealed a substantial increase in CNTF mRNA expression in retinas transduced with AAV-CNTF-GFP. The combination of AAV-CNTF-GFP transduction of RGCs with autologous PN-ON transplantation resulted in even greater RGC survival and regeneration. At 7 weeks after PN transplantation there were 27 954 (+/-2833) surviving RGCs/retina, about 25% of the adult RGC population. Of these, 13 352 (+/-1868) RGCs/retina were retrogradely labelled after fluorogold injections into PN grafts. In summary, AAV-mediated expression of CNTF promotes long-term survival and regeneration of injured adult RGCs, effects that are substantially enhanced by combining gene and cell-based therapies/interventions.


Subject(s)
Ciliary Neurotrophic Factor/genetics , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Optic Nerve Injuries/therapy , Transduction, Genetic/methods , Animals , Axotomy , Cell Survival , Ciliary Neurotrophic Factor/analysis , Ciliary Neurotrophic Factor/metabolism , Female , Gene Expression , Genetic Vectors/genetics , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Immunohistochemistry , Injections , Nerve Regeneration , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Rats , Rats, Wistar , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/virology , Reverse Transcriptase Polymerase Chain Reaction , Vitreous Body
19.
J Virol ; 79(17): 11142-50, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16103165

ABSTRACT

Herpes simplex virus (HSV) infects both epithelial cells and neuronal cells of the human host. Although HSV assembly has been studied extensively for cultured epithelial and neuronal cells, cultured neurons are biochemically, physiologically, and anatomically significantly different than mature neurons in vivo. Therefore, it is imperative that viral maturation and assembly be studied in vivo. To study viral assembly in vivo, we inoculated wild-type and replication-defective viruses into the posterior chamber of mouse eyes and followed infection in retinal ganglion cell bodies and axons. We used PCR techniques to detect viral DNA and RNA and electron microscopy immunohistochemistry and Western blotting to detect viral proteins in specific portions of the optic tract. This approach has shown that viral DNA replication is necessary for viral DNA movement into axons. Movement of viral DNA along ganglion cell axons occurs within capsid-like structures at the speed of fast axonal transport. These studies show that the combined use of intravitreal injections of replication-defective viruses and molecular probes allows the genetic analysis of essential viral replication and maturation processes in neurons in vivo. The studies also provide novel direct evidence for the axonal transport of viral DNA and support for the subassembly hypothesis of viral maturation in situ.


Subject(s)
Retinal Ganglion Cells/virology , Simplexvirus/physiology , Animals , Axons/virology , Biological Transport , DNA, Viral/metabolism , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Simplexvirus/metabolism , Virus Assembly
20.
Microbes Infect ; 7(9-10): 1087-96, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15893497

ABSTRACT

The study of neurons in culture would benefit from the development of a gene transduction system capable of delivering foreign genes at high efficiency, as transduction of primary neurons with existing systems is inefficient. The efficacy of lytic vaccinia virus (VV) infection of primary retinal cultures and PC12 cells (a model of neuronal differentiation) was examined in order to determine the efficiency of gene transduction using VV in neuronal primary culture. VV was able to infect retinal cells and PC12 cells and express transgenes of Escherichia coli beta-galactosidase (lacZ) and epithelial fatty acid binding protein (E-FABP) in a virus dose-dependent manner. Most (50-100%) of the retinal cells were positive for transgene protein at multiplicities of infection (MOI) between 10 and 100 plaque-forming units (PFU), while over 50% of VV-infected PC12 cells expressed the virus encoded gene at an MOI = 10. The production of foreign mRNA and protein by VV following infection was verified by PCR and Western blot. Because VV is a lytic virus, cytopathic effects were examined. Retinal cultures maintained for 0.5 days in vitro showed greater than 90% survival at 24 h post-infection, while 14-day cultures were equally viable for 48 h. Retinal ganglion cells and differentiated PC12 cells appear to be more protected against lytic VV infection than proliferating glial and undifferentiated PC12 cells. These data suggest that VV may be a useful vector for delivering foreign genes to neuronal cells with an efficient transient transgene expression.


Subject(s)
Neuroglia/virology , Neurons/virology , Retinal Ganglion Cells/virology , Transduction, Genetic/methods , Vaccinia virus/genetics , Vaccinia virus/physiology , Animals , Blotting, Western , Cell Survival , Cells, Cultured , Escherichia coli Proteins/analysis , Escherichia coli Proteins/genetics , Gene Expression , Genes, Reporter , Immunohistochemistry , Microscopy, Confocal , Neuroglia/chemistry , PC12 Cells , RNA, Bacterial/analysis , RNA, Messenger/analysis , Rats , Retinal Ganglion Cells/chemistry , beta-Galactosidase/analysis , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...