Next Article in Journal
Topical Diacerein Decreases Skin and Splenic CD11c+ Dendritic Cells in Psoriasis
Next Article in Special Issue
Long-Term Evaluation of Retinal Morphology and Function in Rosa26-Cas9 Knock-In Mice
Previous Article in Journal
The miR-20a/miR-92b Profile Is Associated with Circulating γδ T-Cell Perturbations in Mild Psoriasis
Previous Article in Special Issue
Analysis of Electric Field Stimulation in Blue Light Stressed 661W Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Transcriptome Analysis of Retinal and Choroidal Pathologies in Aged BALB/c Mice Following Systemic Neonatal Murine Cytomegalovirus Infection

1
Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
2
James and Jean Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
3
Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
4
Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
5
Department of Ophthamology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(5), 4322; https://doi.org/10.3390/ijms24054322
Submission received: 25 January 2023 / Revised: 16 February 2023 / Accepted: 17 February 2023 / Published: 21 February 2023
(This article belongs to the Special Issue Retinal Degeneration—From Genetics to Therapy)

Abstract

:
Our previous studies have shown that systemic neonatal murine cytomegalovirus (MCMV) infection of BALB/c mice spread to the eye with subsequent establishment of latency in choroid/RPE. In this study, RNA sequencing (RNA-Seq) analysis was used to determine the molecular genetic changes and pathways affected by ocular MCMV latency. MCMV (50 pfu per mouse) or medium as control were injected intra-peritoneally (i.p.) into BALB/c mice at <3 days after birth. At 18 months post injection, the mice were euthanized, and the eyes were collected and prepared for RNA-Seq. Compared to three uninfected control eyes, we identified 321 differentially expressed genes (DEGs) in six infected eyes. Using the QIAGEN Ingenuity Pathway Analysis (QIAGEN IPA), we identified 17 affected canonical pathways, 10 of which function in neuroretinal signaling, with the majority of DEGs being downregulated, while 7 pathways function in upregulated immune/inflammatory responses. Retinal and epithelial cell death pathways involving both apoptosis and necroptosis were also activated. MCMV ocular latency is associated with upregulation of immune and inflammatory responses and downregulation of multiple neuroretinal signaling pathways. Cell death signaling pathways are also activated and contribute to the degeneration of photoreceptors, RPE, and choroidal capillaries.

1. Introduction

Human cytomegalovirus (HCMV) is a common virus which infects 40 to 80% of individuals in the human population [1]. The virus is usually acquired during early life when the innate and adaptive immune systems are not fully mature [2], with the eye being one of the major target organs. Thus, the incidence of HCMV chorioretinitis is reported to be 25% in infants with symptomatic congenital HCMV infection [3,4,5,6]. Furthermore, the choroid/RPE may be a site of HCMV latency, since recent studies in our laboratory of ocular tissue from human cadavers have revealed that HCMV DNA was present in 17% (4 of 24) of choroid/RPE samples [7].
In order to investigate the effects of lifelong infection with cytomegalovirus, we established an in vivo mouse model in which systemic neonatal murine cytomegalovirus (MCMV) infection of BALB/c mice spreads to the eye, with subsequent establishment of latency in the choroid/RPE and the development of retinal and choroidal pathologies that appear in aged, infected mice, including deposits at both basal and apical aspects of the RPE (basal lamina deposits and subretinal deposits), together with degeneration of the choriocapillaris, RPE, and photoreceptors [8]. These pathologies exhibit some features of human age-related macular degeneration (AMD), which is characterized in its early stages by lipoprotein deposits at the basal and apical aspects of the RPE and in its advanced forms by CNV [9,10] or geographic atrophy (GA) of the outer retinal tissue, RPE, and choriocapillaris [11,12,13]. AMD is a complex, multifactorial, progressive disease and a leading cause of severe, permanent vision loss in older individuals [9,10,14]. Although the exact events which contribute to the development of AMD remain uncertain, studies have implicated various immunological and inflammatory mechanisms [15,16,17]. Many studies support correlations between oxidative stress, persistent low-grade inflammation, and AMD pathogenesis [18,19,20,21,22,23]. Inflammation and oxidative stress are correlated physiological processes [18,24,25,26,27]. Superoxide radicals are formed by NADPH oxidase from activated immune cells during inflammatory reactions, while oxidants are activators of the NF-κB pathway triggering inflammation/immune response [28].
Our studies have indicated that MCMV most likely remains latent in eyes of aged mice, since replicating virus was never recovered and expression of virus genes m83 and m54, which, respectively, encode a virion protein expressed late in the viral replication cycle [29] and a virus DNA polymerase, [30] was not detected in any of the eyes of the infected mice. However, expression of several other virus genes was detected in the eyes of aged mice [8]. The majority of these genes function in either immune-modulation, such as m04 and m138 [31], or inhibition of cell death, including m36, m38.5, m41, and m45 [32], which could facilitate the establishment of ocular virus latency. Furthermore, other genes with the potential to stimulate immune responses, such as IE1, m80, and m18, were also expressed in the eyes of latently infected, aged mice [8]. IE1 can trigger a proinflammatory host transcriptional response via a STAT1-dependent mechanism [33,34], and m80 encodes a MCMV assembly protein protease [35], while m18 encodes a protein which is an antigenic peptide recognized by CD8 T cells [36] and also drives expression of the RAE-1 family of NKG2D ligands leading to subsequent activation of NK cells [37]. Therefore, activation of these genes could stimulate an inflammatory/immune response, which could, in turn, induce retinal and choroidal pathologies observed in infected aged BALB/c mice. In this study, we analyzed eye samples from aged, infected mice and age-matched uninfected controls by performing RNA sequencing (RNA-Seq) to identify molecular pathways involved in immune/inflammatory responses as well as others affected by ocular MCMV latency.

2. Results

2.1. SD-OCT (Spectral-Domain Optical Coherence Tomography) Analysis

Six eyes from the MCMV latently infected mice at 18 months p.i. and three eyes from age-matched uninfected controls were used for sequencing analysis. Prior to removal of the eyes, SD-OCT examinations were performed, and retinal thickness was calculated using a Leica Envisu R2210 system (Bioptigen, Leica, Morrisville, NC, USA). Compared to age-matched uninfected control eyes, a significantly reduced retinal thickness was observed in all six eyes of the infected mice (Figure 1B) as previously reported [8], while severe photoreceptor (PRC) degeneration, including disappearance of the entire outer nuclear layer (ONL) in some areas, was observed in three of six eyes (Figure 1A). In addition, one choroidal neovascularization (CNV)-like lesion was observed in one of these three eyes with severe PRC degeneration (not shown).

2.2. Identification of Differentially Expressed Genes

Each sample generated 80 to 107 million paired reads with 100% passing filter (PF) clusters, >96% perfect barcode, >95% bases with Q30, and >39 mean quality scores.
The number of genes (≥1 normalized count) expressed in each sample ranged from 12,875 to 14,230 with a cutoff of |fold change| ≥ 2 and q ≤ 0.05 used to identify differentially expressed genes (DEGs). Compared to the uninfected control eyes (samples c1, c2, c3), 321 DEGs (208 downregulated and 113 upregulated, Supplementary Table S1) were identified in the six virus latently infected eyes. The top 15 upregulated and downregulated genes are listed in Table 1, together with the fold changes and FDR values. Among virus-infected eyes, 48 DEGs (38 downregulated and 10 upregulated, Supplementary Table S2) were identified in three eyes with severe retinal degeneration (samples h1, h2, h3), compared to three infected eyes with milder retinal degeneration (samples m1, m2, m3). A heat map for all nine samples is shown in Figure 1D.

2.3. Validation of Differentially Expressed Genes

RNA-seq transcriptome data were validated by qRT-PCR analysis and expression levels of eight genes were analyzed in three control and six virus-infected samples. As shown in Figure 1C, similar expression trends were observed by both qRT-PCR analysis and RNA-Seq for all eight genes. A correlation (R2) of 0.8904 was observed between the log2FC of ΔCT values derived by qRT-PCR analysis and the log2FC value derived by RNA-seq analysis.

2.4. Comparison of Infected and Uninfected Eyes

2.4.1. Analysis of Canonical Pathways

QIAGEN IPA analysis was used to identify involvement of canonical pathways with a p-value of less than 10−2. These pathways, together with the differentially expressed genes in each canonical pathway, are listed in Table 2. Among a total of 17 canonical pathways identified, 10 function in neuroretinal signaling, with the majority of differentiated regulated genes involved in these pathways being downregulated in infected eyes. As shown in Figure 2, for example, all 22 differentially expressed genes linked to the Phototransduction Pathway of rod and cone cells, were downregulated. These include genes involved in cAMP and PKA-mediated signaling, which participate in the modulation of presynaptic GABA release [38] and are themselves regulated by Relaxin signaling in a biphasic manner [39]. cAMP signaling also plays a role in regulating glutamatergic transmission via phosphorylation of certain ionotropic glutamate receptors [40] through dopamine-DARPP32 feedback. Since DARPP-32 is localized in horizontal cells, amacrine cells, and Müller glial cells, this may result in the disfunction of these important retinal cell types. Expression of genes involved in RNA splicing was also downregulated in infected eyes, consistent with previous results which have shown that RNA processing defects are associated with many diseases of the neuron, including retinal degeneration [41].
Other pathways affected include the Endothelin-1 (ET-1) pathway, which was also downregulated, and G beta gamma signaling. The neuropeptide ET-1 is expressed in multiple retinal cell types, including RPE, photoreceptors, the inner plexiform layer, and ganglion cell layer [42] and functions in neuromodulation and neurotransmission [42,43]. However, ET-1 could also activate ET receptors in the retinal and choroidal vasculature, suggesting an important role in regulating in situ blood flow [42]. G beta gamma signaling plays a critical role in rod cell function in low light conditions [44], as well as neuronal CREB signaling, which is one of the major regulators of neurotrophin responses [45,46], and G-protein coupled receptor signaling.
In contrast, the transcription of genes involved in several immune response pathways was upregulated. These included pathways involved in NFAT-regulated dendritic cell maturation, phagosome formation, and communication between innate and adaptive immune cells. Upregulated gene transcription was also observed in the GP6 signaling pathway, which functions in platelet activation and thrombus formation [47]. The Tec kinase signaling pathway, which regulates lymphocyte development, activation, and differentiation [48], as well as in the opioid signaling pathway, which is critically involved in many physiological processes including neuroprotection and immune response [49,50,51].

2.4.2. Analysis of Diseases and Functions

The QIAGEN IPA analysis also permitted the categorization of differentially expressed genes according to known disease associations and function as listed below.
  • Ophthalmic Disease, Organismal Injury, and Visual System Function. Many differentially expressed genes were noted in sections of this category. As shown in Table 3, differential expression of genes implicated in retinal degeneration, including degeneration of the photoreceptor, rod, and outer segments of the cone cell, was detected with z scores greater than 2. Seven differentially expressed genes were noted with a z score of 1.568. In addition, differential expression of genes required for maintenance of retinal cell and photoreceptor function and quantity were noted with z scores less than −2.
2.
Cell Death and Survival. As shown in Table 4, many differentially expressed genes implicated in the death of retinal cells and epithelial tissues, either by apoptosis or necrosis, were detected. TUNEL assays and Western blots were used to confirm this observation. As shown in Figure 3A, many TUNEL-positive cells were observed in the choroid (indicated by white arrows), RPE layer (indicated by arrow heads), and outer nuclear layer (indicated by red arrows) in virus-infected eyes, while in contrast, only a few TUNEL-positive cells were observed in the outer nuclear layer of eyes from age-matched uninfected mice. Eyes of MCMV neonatally infected mice, as well as the eyes of the age-matched, uninfected controls, were also analyzed by Western blotting. As shown in Figure 3B, MCMV infection was associated with increased production of cleaved caspase 3, RIP3, MLKL, and decreased production of rhodopsin, indicating that both apoptosis and necroptosis may contribute to cell death.
3.
Cellular Movement and Immune Cell Trafficking. As shown in Table 5, significant changes in the transcription of genes involved in migration, infiltration, and activation of multiple immune cell types were observed in infected eyes. Immunostaining was used to confirm these observations. As shown in Figure 4, accumulation of Iba1 positive macrophage/microglia and GFAP positive Müller cells/glia was observed in the subretinal space and outer nuclear layer of the eyes of aged, infected mice, but not in age-matched uninfected controls.

2.4.3. Analysis of Upstream Regulators

Pathway analysis identified 36 upstream regulators of these differentially expressed genes with activation Z scores of greater than 1.5 and p-values of overlap less than 0.05 (Table 6). The majority of these upstream regulators are cytokines (IL1, IFN, IL17A, TNF, etc.) or transcription factors (STAT1, STAT3) that are involved in innate immunity/inflammation. Several growth factors (VEGF, TGF) were also identified as activated upstream regulators.
As shown in the summary graph (Figure 5), upstream regulators, such as IL1, OSM, IL17A, and STAT1, stimulate activation and migration of immune cells, tissue degeneration, and antigen presentation.

2.5. Comparison of Infected Eyes with and without Severe Retinal Degeneration

Compared to infected eyes without severe retinal degeneration, we identified 48 DEGs in three infected eyes with severe retinal degeneration (Supplementary Table S2). As shown in Table 7, the majority of differentially regulated genes were involved in pathways related to neuroretinal signaling, cell death, and retinal degeneration. In contrast, no significant differences were detected in immune response pathways, such as those involved in migration, infiltration, or activation of immune cells, between infected eyes with and without severe retinal degeneration.

3. Discussion

The data presented here complements our earlier studies on lifelong MCMV latency in the retina. These studies demonstrated that systemic MCMV infection of newborn mice resulted in ocular pathology later in life which exhibited some similarities to AMD [8]. Although no infectious virus could be isolated from the retina during latent ocular infection, many gross pathological changes in the retinal architecture were observed. The transcriptional data presented here provides a theoretical framework for understanding retinal and choroidal pathologies in latently infected eyes and demonstrates widespread alterations in the expression of genes critical for maintaining retinal form and function. Thus, systemic virus infection of neonates results in disruption of normal ocular gene expression patterns even in advanced age.
The studies presented herein support the idea that cytomegalovirus ocular latency could be associated with in situ inflammation. Transcription of many inflammatory molecules was elevated, and pathway analysis indicated that several inflammatory pathways, including IL1, STAT1, IL17A, and OSM, were activated with activation Z scores greater than 2 and p-values of overlap less than 0.05. These inflammatory molecules may activate immune cells including macrophages/microglia and could also induce degeneration of photoreceptor, RPE, or choriocapillaris, either directly or indirectly, via activation of immune cells. Our results also demonstrate that multiple cell death pathways, including caspase-3 dependent apoptosis, necrosis, and necroptosis are activated and contribute to degeneration of photoreceptors, RPE, and choroidal capillaries, although future studies are still needed to determine which pathways contribute to which aspects of retinal degeneration.
Our previous studies have shown that during systemic neonatal MCMV infection of 129S1/SvImJ (129S1) mice [7], MCMV spreads to the eye with subsequent establishment of latency in the choroid/RPE. Unlike the neonatal infection of BALB/c mice, only a few MCMV genes are expressed, and no remarkable retinal or choroidal pathology, such as deposits, degeneration of the choriocapillaris, RPE, or photoreceptors, was observed [7]. BALB/c mice, which are susceptible to light damage due to a lack of melanin [52,53], exhibited photoreceptor degeneration (infiltrating cells, loss of outer segments, and decreased retinal thickness) in aged, uninfected control mice in both our own previous studies [8], as well as those of Bell and colleagues [54]. Thus, melanin in the RPE may prevent light-induced retinal damage by absorbing most of the light passing through the pupil and helping to scavenge free radicals [53,55,56], thereby protecting photoreceptors from oxidative stress [57,58]. Melanin might also prevent toxicity through its anti-oxidative function [59] and could play a role in protecting choroidal blood vessels from light damage [60]. Therefore, the susceptibility of BALB/c mice to light damage [61,62,63,64] may be due to an accumulation of reactive oxygen species (ROS) and subsequent oxidative stress and inflammation [65,66]. Previous studies have suggested that oxidative stress mediates the initial activation of viral gene expression during cytomegalovirus latency [67,68]. Following light damage, oxidative stress in MCMV latently infected BALB/c mice could activate expression of ocular virus genes, which in turn might promote production of inflammatory/angiogenic factors, thereby facilitating development of retinal and choroidal pathologies.
The choroid/RPE may be a site of HCMV latency since we have shown that HCMV DNA is present in some human choroid/RPE samples [7]. However, whether HCMV ocular latency contributes to human AMD remains to be determined. AMD is a complex multifactorial disease, and the majority of risk factors, including genetics, environmental insults, and age-related issues, are linked to the induction of oxidative stress, which could activate expression of ocular HCMV genes, resulting in the production of inflammatory/angiogenic factors and thereby facilitating development of an AMD-like pathology.

4. Materials and Methods

4.1. Cells and Virus

MCMV strain K181 was originally provided by Dr. Edward Morcarski, Emory University, Atlanta, GA. The virus was prepared from the salivary glands of MCMV-infected immunosuppressed BALB/c mice, and virus stocks were titered on monolayers of mouse embryo fibroblast (MEF) cells, as described previously [69]. A fresh aliquot of virus stocks was thawed and diluted to the appropriate concentration for each experiment.

4.2. Mice

We purchased breeding pairs of BALB/c mice from Jackson Laboratory (Bar Harbor, ME). All mice were given unrestricted access to food and water and were maintained on a 12 h light cycle alternating with a 12 h dark cycle. The breeding and treatment of animals in this study adhered to the Association for Research in Vision and Ophthalmology (ARVO) Statement for the Use of Animals in Ophthalmic and Vision Research and was approved by the Institutional Animal Care and Use Committee of Augusta University. The rd8 mutation was excluded by genotyping.

4.3. Antibodies

Anti-RPE65 (ab231782), anti-RIP3 (phosphor s232, ab195117), and anti-Rodopsin (ab5417) were purchased from abcam (Boston, MA, USA). Other antibodies used in this study were obtained from the following sources: anti-cleaved caspase 3 (#9664), anti-RIP1 (#3493), and anti-mouse β-actin (#3700) were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA). Mouse anti-GFAP (glial fibrillary acidic protein, specific for glia/Müller cells) was obtained from BD Biosciences (San Jose, CA, USA). Anti-mouse Iba1 was purchased from FUJIFILM WAKO Chemicals, U.S.A. Corporation (019-19741, Richmond, VA, USA). Goat anti-rabbit IgG HRP (sc2004) and rabbit anti-goat IgG HRP (sc2768) were sourced from Santa Cruz (Santa Cruz Biotechnology, CA, USA). Anti-Mouse IgG HRP Conjugate (W402B) was obtained from Promega (Madison WI, USA). Anti-mouse Alexa 488 and anti-rabbit Alexa 594 were obtained from Vector Laboratories, Inc. (Burlingame, CA, USA).

4.4. Experimental Design

A total amount of 50 pfu of MCMV or culture medium as control were injected into BALB/c mice within 3 days after birth via the intraperitoneal (i.p.) route. At 18 months post infection (p.i.), the mice were anesthetized, and spectral-domain (SD) optical coherence tomography (SD-OCT) was performed using the Bioptigen Spectral-Domain Ophthalmic Imaging System (En-visu R2200; Bioptigen, Morrisville, NC, USA). The OCT imaging protocol included averaged single B scan and volume intensity scans with images centered on the optic nerve head (1.4 mm × 1.4 mm, @0.0, 1000X100X4X1). Total retinal thickness was measured by manual assessment of retinal layers using InVivoVue™ Diver 2.4 software (Bioptigen) following the software introduction, as described previously [8]. The mice were euthanized, and the eyes were collected and prepared for RNA-Seq, immunofluorescence staining, Western blot, and real time RT-PCR (qRT-PCR) as described below.

4.5. RNA-Seq

4.5.1. RNA Extraction and Quality Control

Six eyes from infected mice at eighteen months p.i., and three eyes from age-matched, uninfected controls were used for RNA sequencing. Following removal of the lens, total RNA was extracted from whole eyes using Trizol (Invitrogen, CA, USA) according to the manufacturer’s instructions, and all nine RNA samples were then treated with DNase to exclude genomic DNA contamination. RNA concentrations were obtained using a Nanodrop 2000c spectrophotometer (Thermo Scientific Inc., Waltham, MA, USA), while RNA integrity was assessed using an Agilent 2200 Tape station instrument (Agilent Technologies, Santa Clara, CA, USA. RNA Integrity Number (RIN) scores for the nine samples were 7.6, 7.7, 7.7, 7.8, 8.0, 8.2, 8.2, 8.8, and 8.9, respectively. One microgram of total RNA from each sample was used to prepare Ribo-Zero RNA-Seq libraries.

4.5.2. Total RNA-Seq with Ribo-Zero Depletion

RNA-Seq libraries were prepared using the Illumina TruSeq Stranded Total RNA kit (Illumina, Inc., San Diego, CA, USA) according to the manufacturer’s protocol. Briefly, ribosomal RNA (rRNA) was removed using biotinylated, target-specific oligos combined with Ribo-Zero rRNA removal beads according to the Illumina Reference Guide (Illumina, San Diego, CA, USA). Following purification, the RNA was fragmented into small pieces using divalent cations at an elevated temperature. First strand cDNA synthesis was performed at 25 °C for 10 min, 42 °C for 15 min, and 70 °C for 15 min, using random hexamers and ProtoScript II Reverse Transcriptase (NEW ENGLAND BioLabs Inc.). For second strand cDNA synthesis, RNA templates were removed, and a second replacement strand was generated through the incorporation of dUTP (in place of dTTP, to maintain strand identity) and double-strand cDNA was generated. Blunt-ended cDNA was isolated from the second strand reaction mix using beads. The three ends of the cDNA were then adenylated, and the cDNA was ligated to indexing adaptors. PCR (15 cycles of 98 °C for 10 s, 60 °C for 30 s, and 72 °C for 30 s) was used to selectively enrich for DNA fragments with adapter molecules on both ends, and to amplify the amount of DNA in the library. Libraries were quantified and qualified using the D1000 Screen Tape on an Agilent 2200 Tape Station instrument and were normalized, pooled, and subjected to cluster and pair read sequencing for 150 cycles on a HiSeqX10 instrument (Illumina, Inc. San Diego, CA, USA), according to the manufacturer’s instructions.

4.5.3. Data Analysis Methods

Coding RNA data were analyzed by Rosalind (https://rosalind.onramp.bio/) (accessed on 21 September 2020), with a HyperScale architecture developed by OnRamp BioInformatics, Inc. (San Diego, CA, USA) [70]. Reads were trimmed using cutadapt [71]. Quality scores were assessed using FastQC [72], and reads were aligned to the Mus musculus genome build mm10 using STAR [73]. Individual sample reads were quantified using HTseq [74] and normalized via Relative Log Expression (RLE) using the DESeq2 R library [75]. Read distribution percentages, violin plots, identity heatmaps, and sample MDS plots were generated as part of the QC step using RSeQC [76]. DEseq2 was also used to calculate fold changes (FDs) and p-values. The significant gene set was selected with a q-value (p-value that has been adjusted for False Discovery Rate, FDR) of <0.05 threshold.
Using QIAGEN Ingenuity Pathway Analysis (QIAGEN IPA) (Ingenuity® Systems, www.ingenuity.com) (accessed on 23 March 2021), we illustrated clustering of genes for the final heatmap of differentially expressed genes and performed functional analyses to identify relevant gene pathways and networks.

4.6. qRT-PCR

RNA-seq transcriptome data were validated by qRT-PCR analysis. All primer sequences used for qRT-PCR are shown in Supplementary Table S3. Genes were amplified in 20 µL reaction consisting of 10 µL 2×SYBR Mix (Bio-Rad), 0.2 µL of 20 pmol/µL primer mixture, and 1 µL cDNA, using CFX96TM Real Time PCR System (Bio-Rad). PCR conditions were as follows: 3 min at 94°C, followed by 40 cycles of 94 °C for 10 s, 60 °C for 20 s, and 72 °C for 30 s. All CT values were analyzed and normalized to β-actin using the method of 2–ΔΔCT.

4.7. Immunofluorescence Staining

Eyes (four eyes in each group) were embedded in OCT compound, frozen, and sectioned in a cryostat. Sections were then fixed with 4% paraformaldehyde for 15 min and stained by TUNEL assay (in Situ Cell Death Detection Kit, Fluorescein; Roche Diagnostics, Indianapolis, IN, USA) and/or for RPE-65, GFAP, Iba-1 as described previously [77,78].

4.8. Western Blot

Lenses were removed from harvested eyes and the remaining eye tissues homogenized in a lysis buffer containing protease inhibitors (Complete™ Lysis-M, Roche, Germany). Proteins were extracted as previously described [79,80], and equal amounts of protein were separated by 10% or 12% SDS-PAGE, followed by electroblotting onto polyvinylidene difluoride membranes (Amersham Biosciences, Amersham, UK). Following blocking with 5% nonfat dry milk for 1 h at room temperature, membranes were incubated overnight at 4 °C with primary antibody. Binding of HRP–conjugated secondary antibody was performed for 1 h at room temperature, and bands were visualized using chemiluminescence (ECL; GE Healthcare, Chicago, IL, USA).

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms24054322/s1.

Author Contributions

Conceptualization, M.Z., X.Z., J.X.; methodology, J.X., X.Z.; formal analysis, X.Z., J.X.; validation, M.Z., X.Z., J.X.; investigation, X.Z., J.X.; resources, Z.D., D.G.E.-H., Y.L.; data curation, M.Z., X.Z., J.X.; writing—original draft preparation, M.Z.; writing—review and editing, M.Z., B.M.; visualization, X.Z.; supervision, M.Z.; funding acquisition, M.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by NIH grant RO1 EY026642, Retinal Research Foundation grant, Brightfocus Foundation grant, and NIH grant P30 EY031631.

Institutional Review Board Statement

All experiments and breeding followed the guidelines of the National Institutes of Health for the care and maintenance of mice and adhered to the ARVO Statement for the Use of Animals in Ophthalmic and Vision Research. The protocols were also approved by the Institutional Animal Care and Use Committee of Augusta University (#2009-0252, Date: 24 September 2020).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are included in this published article. All the data can be shared upon request by email.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Demmler, G.J.; Infectious Diseases Society of America and Centers for Disease Control. Summary of a workshop on surveillance for congenital cytomegalovirus disease. Rev. Infect. Dis. 1991, 13, 315–329. [Google Scholar] [CrossRef] [PubMed]
  2. Lanzieri, T.M.; Kruszon-Moran, D.; Amin, M.M.; Bialek, S.R.; Cannon, M.J.; Carroll, M.D.; Dollard, S.C. Seroprevalence of cytomegalovirus among children 1 to 5 years of age in the United States from the National Health and Nutrition Examination Survey of 2011 to 2012. Clin. Vaccine Immunol. 2015, 22, 245–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Boppana, S.; Amos, C.; Britt, W.; Stagno, S.; Alford, C.; Pass, R. Late onset and reactivation of chorioretinitis in children with congenital cytomegalovirus infection. Pediatr. Infect. Dis. J. 1994, 13, 1139–1142. [Google Scholar] [CrossRef]
  4. Pass, R.F.; Stagno, S.; Myers, G.J.; Alford, C.A. Outcome of symptomatic congenital cytomegalovirus infection: Results of long-term longitudinal follow-up. Pediatrics 1980, 66, 758–762. [Google Scholar] [CrossRef] [PubMed]
  5. Istas, A.S.; Demmler, G.J.; Dobbins, J.G.; Stewart, J.A. Surveillance for congenital cytomegalovirus disease: A report from the National Congenital Cytomegalovirus Disease Registry. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 1995, 20, 665–670. [Google Scholar] [CrossRef]
  6. Stagno, S.; Pass, R.F.; Dworsky, M.E.; Alford, C.A. Congenital and perinatal cytomegalovirus infections. Semin. Perinatol. 1983, 7, 31–42. [Google Scholar]
  7. Xu, J.; Liu, X.; Zhang, X.; Marshall, B.; Dong, Z.; Liu, Y.; Espinosa-Heidmann, D.G.; Zhang, M. Ocular cytomegalovirus latency exacerbates the development of choroidal neovascularization. J. Pathol. 2020, 251, 200–212. [Google Scholar] [CrossRef]
  8. Xu, J.; Liu, X.; Zhang, X.; Marshall, B.; Dong, Z.; Smith, S.B.; Espinosa-Heidmann, D.G.; Zhang, M. Retinal and choroidal pathologies in aged BALB/c mice following 1 systemic neonatal murine cytomegalovirus infection. Am. J. Pathol. 2021, 191, 1787–1804. [Google Scholar]
  9. Bowes Rickman, C.; Farsiu, S.; Toth, C.A.; Klingeborn, M. Dry age-related macular degeneration: Mechanisms, therapeutic targets, and imaging. Investig. Ophthalmol. Vis. Sci. 2013, 54, ORSF68–ORSF80. [Google Scholar] [CrossRef] [Green Version]
  10. Bird, A.C. Therapeutic targets in age-related macular disease. J. Clin. Investig. 2010, 120, 3033–3041. [Google Scholar] [CrossRef] [Green Version]
  11. Boyer, D.S.; Schmidt-Erfurth, U.; van Lookeren Campagne, M.; Henry, E.C.; Brittain, C. The Pathophysiology of Geographic Atrophy Secondary to Age-Related Macular Degeneration and the Complement Pathway as a Therapeutic Target. Retina 2017, 37, 819–835. [Google Scholar] [CrossRef]
  12. Danis, R.P.; Lavine, J.A.; Domalpally, A. Geographic atrophy in patients with advanced dry age-related macular degeneration: Current challenges and future prospects. Clin. Ophthalmol. 2015, 9, 2159–2174. [Google Scholar] [CrossRef] [Green Version]
  13. Kaszubski, P.; Ben Ami, T.; Saade, C.; Smith, R.T. Geographic Atrophy and Choroidal Neovascularization in the Same Eye: A Review. Ophthalmic. Res. 2016, 55, 185–193. [Google Scholar] [CrossRef] [Green Version]
  14. Gorin, M.B. Genetic insights into age-related macular degeneration: Controversies addressing risk, causality, and therapeutics. Mol. Asp. Med. 2012, 33, 467–486. [Google Scholar] [CrossRef] [Green Version]
  15. Pennesi, M.E.; Neuringer, M.; Courtney, R.J. Animal models of age related macular degeneration. Mol. Asp. Med. 2012, 33, 487–509. [Google Scholar] [CrossRef] [Green Version]
  16. Fletcher, E.L.; Jobling, A.I.; Greferath, U.; Mills, S.A.; Waugh, M.; Ho, T.; de Iongh, R.U.; Phipps, J.A.; Vessey, K.A. Studying age-related macular degeneration using animal models. Optom. Vis. Sci. 2014, 91, 878–886. [Google Scholar] [CrossRef] [Green Version]
  17. Ding, J.D.; Kelly, U.; Groelle, M.; Christenbury, J.G.; Zhang, W.; Bowes Rickman, C. The role of complement dysregulation in AMD mouse models. Adv. Exp. Med. Biol. 2014, 801, 213–219. [Google Scholar]
  18. Donato, L.; Scimone, C.; Alibrandi, S.; Scalinci, S.Z.; Rinaldi, C.; D'Angelo, R.; Sidoti, A. Epitranscriptome Analysis of Oxidative Stressed Retinal Epithelial Cells Depicted a Possible RNA Editing Landscape of Retinal Degeneration. Antioxidants 2022, 11, 1967. [Google Scholar] [CrossRef]
  19. Copland, D.A.; Theodoropoulou, S.; Liu, J.; Dick, A.D. A Perspective of AMD Through the Eyes of Immunology. Investig. Ophthalmol. Vis. Sci. 2018, 59, AMD83–AMD92. [Google Scholar] [CrossRef] [Green Version]
  20. Buschini, E.; Piras, A.; Nuzzi, R.; Vercelli, A. Age related macular degeneration and drusen: Neuroinflammation in the retina. Prog. Neurobiol. 2011, 95, 14–25. [Google Scholar] [CrossRef]
  21. Hadziahmetovic, M.; Malek, G. Age-Related Macular Degeneration Revisited: From Pathology and Cellular Stress to Potential Therapies. Front. Cell Dev. Biol. 2020, 8, 612812. [Google Scholar] [CrossRef] [PubMed]
  22. Hadziahmetovic, M.; Song, Y.; Wolkow, N.; Iacovelli, J.; Kautz, L.; Roth, M.P.; Dunaief, J.L. Bmp6 regulates retinal iron homeostasis and has altered expression in age-related macular degeneration. Am. J. Pathol. 2011, 179, 335–348. [Google Scholar] [CrossRef] [PubMed]
  23. Song, D.; Wilson, B.; Zhao, L.; Bhuyan, R.; Bandyopadhyay, M.; Lyubarsky, A.; Yu, C.; Li, Y.; Kanu, L.; Miwa, T.; et al. Retinal Pre-Conditioning by CD59a Knockout Protects against Light-Induced Photoreceptor Degeneration. PLoS ONE 2016, 11, e0166348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Louka, A.M.; Sagris, D.; Ntaios, G. Iotammunity, Vascular Aging, and Stroke. Curr. Med. Chem. 2022, 29, 5510–5521. [Google Scholar] [CrossRef]
  25. Scimone, C.; Donato, L.; Marino, S.; Alafaci, C.; D’Angelo, R.; Sidoti, A. Vis-a-vis: A focus on genetic features of cerebral cavernous malformations and brain arteriovenous malformations pathogenesis. Neurol. Sci. 2019, 40, 243–251. [Google Scholar] [CrossRef]
  26. Scimone, C.; Donato, L.; Alibrandi, S.; Esposito, T.; Alafaci, C.; D’Angelo, R.; Sidoti, A. Transcriptome analysis provides new molecular signatures in sporadic Cerebral Cavernous Malformation endothelial cells. Biochim. Biophys. Acta Mol. Basis. Dis. 2020, 1866, 165956. [Google Scholar] [CrossRef]
  27. Donato, L.; Alibrandi, S.; Scimone, C.; Rinaldi, C.; Dascola, A.; Calamuneri, A.; D’Angelo, R.; Sidoti, A. The impact of modifier genes on cone-rod dystrophy heterogeneity: An explorative familial pilot study and a hypothesis on neurotransmission impairment. PLoS ONE 2022, 17, e0278857. [Google Scholar] [CrossRef]
  28. Pantano, C.; Reynaert, N.L.; van der Vliet, A.; Janssen-Heininger, Y.M. Redox-sensitive kinases of the nuclear factor-kappaB signaling pathway. Antioxid Redox Signal 2006, 8, 1791–1806. [Google Scholar] [CrossRef]
  29. Morello, C.S.; Cranmer, L.D.; Spector, D.H. In vivo replication, latency, and immunogenicity of murine cytomegalovirus mutants with deletions in the M83 and M84 genes, the putative homologs of human cytomegalovirus pp65 (UL83). J. Virol. 1999, 73, 7678–7693. [Google Scholar] [CrossRef] [Green Version]
  30. Morello, C.S.; Kelley, L.A.; Munks, M.W.; Hill, A.B.; Spector, D.H. DNA immunization using highly conserved murine cytomegalovirus genes encoding homologs of human cytomegalovirus UL54 (DNA polymerase) and UL105 (helicase) elicits strong CD8 T-cell responses and is protective against systemic challenge. J. Virol. 2007, 81, 7766–7775. [Google Scholar] [CrossRef] [Green Version]
  31. Lacaze, P.; Forster, T.; Ross, A.; Kerr, L.E.; Salvo-Chirnside, E.; Lisnic, V.J.; Lopez-Campos, G.H.; Garcia-Ramirez, J.J.; Messerle, M.; Trgovcich, J.; et al. Temporal profiling of the coding and noncoding murine cytomegalovirus transcriptomes. J. Virol. 2011, 85, 6065–6076. [Google Scholar] [CrossRef] [Green Version]
  32. Handke, W.; Krause, E.; Brune, W. Live or let die: Manipulation of cellular suicide programs by murine cytomegalovirus. Med. Microbiol. Immunol. 2012, 201, 475–486. [Google Scholar] [CrossRef]
  33. Knoblach, T.; Grandel, B.; Seiler, J.; Nevels, M.; Paulus, C. Human cytomegalovirus IE1 protein elicits a type II interferon-like host cell response that depends on activated STAT1 but not interferon-gamma. PLoS Pathog. 2011, 7, e1002016. [Google Scholar] [CrossRef] [Green Version]
  34. Reitsma, J.M.; Sato, H.; Nevels, M.; Terhune, S.S.; Paulus, C. Human cytomegalovirus IE1 protein disrupts interleukin-6 signaling by sequestering STAT3 in the nucleus. J. Virol. 2013, 87, 10763–10776. [Google Scholar] [CrossRef] [Green Version]
  35. Sloan, J.H.; Loutsch, J.M.; Boyce, S.Y.; Holwerda, B.C. Expression and characterization of recombinant murine cytomegalovirus protease. J. Virol. 1997, 71, 7114–7118. [Google Scholar] [CrossRef] [Green Version]
  36. Holtappels, R.; Grzimek, N.K.A.; Thomas, D.; Reddehase, M.J. Early gene m18, a novel player in the immune response to murine cytomegalovirus. J. Gen. Virol. 2002, 83 Pt 2, 311–316. [Google Scholar] [CrossRef] [Green Version]
  37. Greene, T.T.; Tokuyama, M.; Knudsen, G.M.; Kunz, M.; Lin, J.; Greninger, A.L.; DeFilippis, V.R.; DeRisi, J.L.; Raulet, D.H.; Coscoy, L. A Herpesviral induction of RAE-1 NKG2D ligand expression occurs through release of HDAC mediated repression. Elife 2016, 5, e14749. [Google Scholar] [CrossRef]
  38. Zhou, X.; Zhang, R.; Zhang, S.; Wu, J.; Sun, X. Activation of 5-HT1A Receptors Promotes Retinal Ganglion Cell Function by Inhibiting the cAMP-PKA Pathway to Modulate Presynaptic GABA Release in Chronic Glaucoma. J. Neurosci. Off. J. Soc. Neurosci. 2019, 39, 1484–1504. [Google Scholar] [CrossRef] [Green Version]
  39. Dessauer, C.W.; Nguyen, B.T. Relaxin stimulates multiple signaling pathways: Activation of cAMP, PI3K, and PKCzeta in THP-1 cells. Ann. N. Y. Acad. Sci. 2005, 1041, 272–279. [Google Scholar] [CrossRef] [Green Version]
  40. Witkovsky, P.; Svenningsson, P.; Yan, L.; Bateup, H.; Silver, R. Cellular localization and function of DARPP-32 in the rodent retina. Eur. J. Neurosci. 2007, 25, 3233–3242. [Google Scholar] [CrossRef] [Green Version]
  41. Zelinger, L.; Swaroop, A. RNA Biology in Retinal Development and Disease. Trends Genet. 2018, 34, 341–351. [Google Scholar] [CrossRef] [PubMed]
  42. Salvatore, S.; Vingolo, E.M. Endothelin-1 role in human eye: A review. J. Ophthalmol. 2010, 2010, 354645. [Google Scholar] [CrossRef] [PubMed]
  43. Dashwood, M.R.; Loesch, A. Endothelin-1 as a neuropeptide: Neurotransmitter or neurovascular effects? J. Cell. Commun. Signal 2010, 4, 51–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Kolesnikov, A.V.; Rikimaru, L.; Hennig, A.K.; Lukasiewicz, P.D.; Fliesler, S.J.; Govardovskii, V.I.; Kefalov, V.J.; Kisselev, O.G. G-protein betagamma-complex is crucial for efficient signal amplification in vision. J. Neurosci. Off. J. Soc. Neurosci. 2011, 31, 8067–8077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Guido, W. Refinement of the retinogeniculate pathway. J. Physiol. 2008, 586, 4357–4362. [Google Scholar] [CrossRef]
  46. Wang, H.; Xu, J.; Lazarovici, P.; Quirion, R.; Zheng, W. cAMP Response Element-Binding Protein (CREB): A Possible Signaling Molecule Link in the Pathophysiology of Schizophrenia. Front. Mol. Neurosci. 2018, 11, 255. [Google Scholar] [CrossRef] [Green Version]
  47. Nieswandt, B.; Watson, S.P. Platelet-collagen interaction: Is GPVI the central receptor? Blood 2003, 102, 449–461. [Google Scholar] [CrossRef]
  48. Andreotti, A.H.; Schwartzberg, P.L.; Joseph, R.E.; Berg, L.J. T-cell signaling regulated by the Tec family kinase, Itk. Cold Spring Harb. Perspect. Biol. 2010, 2, a002287. [Google Scholar] [CrossRef] [Green Version]
  49. Husain, S.; Abdul, Y.; Crosson, C.E. Preservation of retina ganglion cell function by morphine in a chronic ocular-hypertensive rat model. Invest. Ophthalmol. Vis. Sci. 2012, 53, 4289–4298. [Google Scholar] [CrossRef]
  50. Sobczak, M.; Salaga, M.; Storr, M.A.; Fichna, J. Physiology, signaling, and pharmacology of opioid receptors and their ligands in the gastrointestinal tract: Current concepts and future perspectives. J. Gastroenterol. 2014, 49, 24–45. [Google Scholar] [CrossRef] [Green Version]
  51. Husain, S.; Liou, G.I.; Crosson, C.E. Opioid receptor activation: Suppression of ischemia/reperfusion-induced production of TNF-alpha in the retina. Invest. Ophthalmol. Vis. Sci. 2011, 52, 2577–2583. [Google Scholar] [CrossRef] [Green Version]
  52. Sanyal, S.; Zeilmaker, G.H. Retinal damage by constant light in chimaeric mice: Implications for the protective role of melanin. Exp. Eye Res. 1988, 46, 731–743. [Google Scholar] [CrossRef]
  53. van Norren, D.; Vos, J.J. Light damage to the retina: An historical approach. Eye 2016, 30, 169–172. [Google Scholar] [CrossRef] [Green Version]
  54. Bell, B.A.; Kaul, C.; Bonilha, V.L.; Rayborn, M.E.; Shadrach, K.; Hollyfield, J.G. The BALB/c mouse: Effect of standard vivarium lighting on retinal pathology during aging. Exp. Eye Res. 2015, 135, 192–205. [Google Scholar] [CrossRef] [Green Version]
  55. Bressler, N.M.; Bressler, S.B.; Fine, S.L. Age-related macular degeneration. Surv. Ophthalmol. 1988, 32, 375–413. [Google Scholar] [CrossRef]
  56. Weiter, J.J.; Delori, F.C.; Wing, G.L.; Fitch, K.A. Retinal pigment epithelial lipofuscin and melanin and choroidal melanin in human eyes. Invest. Ophthalmol. Vis. Sci. 1986, 27, 145–152. [Google Scholar]
  57. Zadlo, A.; Rozanowska, M.B.; Burke, J.M.; Sarna, T.J. Photobleaching of retinal pigment epithelium melanosomes reduces their ability to inhibit iron-induced peroxidation of lipids. Pigment. Cell. Res. 2007, 20, 52–60. [Google Scholar] [CrossRef]
  58. Sarna, T.; Burke, J.M.; Korytowski, W.; Rozanowska, M.; Skumatz, C.M.; Zareba, A.; Zareba, M. Loss of melanin from human RPE with aging: Possible role of melanin photooxidation. Exp. Eye Res. 2003, 76, 89–98. [Google Scholar] [CrossRef]
  59. Taubitz, T.; Tschulakow, A.V.; Tikhonovich, M.; Illing, B.; Fang, Y.; Biesemeier, A.; Julien-Schraermeyer, S.; Schraermeyer, U. Ultrastructural alterations in the retinal pigment epithelium and photoreceptors of a Stargardt patient and three Stargardt mouse models: Indication for the central role of RPE melanin in oxidative stress. PeerJ 2018, 6, e5215. [Google Scholar] [CrossRef] [Green Version]
  60. Peters, S.; Lamah, T.; Kokkinou, D.; Bartz-Schmidt, K.U.; Schraermeyer, U. Melanin protects choroidal blood vessels against light toxicity. Z. Nat. C. J. Biosci. 2006, 61, 427–433. [Google Scholar] [CrossRef]
  61. Nusinowitz, S.; Nguyen, L.; Radu, R.; Kashani, Z.; Farber, D.; Danciger, M. Electroretinographic evidence for altered phototransduction gain and slowed recovery from photobleaches in albino mice with a MET450 variant in RPE65. Exp. Eye Res. 2003, 77, 627–638. [Google Scholar] [CrossRef] [PubMed]
  62. Wenzel, A.; Reme, C.E.; Williams, T.P.; Hafezi, F.; Grimm, C. The Rpe65 Leu450Met variation increases retinal resistance against light-induced degeneration by slowing rhodopsin regeneration. J. Neurosci. 2001, 21, 53–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Danciger, M.; Matthes, M.T.; Yasamura, D.; Akhmedov, N.B.; Rickabaugh, T.; Gentleman, S.; Redmond, T.M.; La Vail, M.M.; Farber, D.B. A QTL on distal chromosome 3 that influences the severity of light-induced damage to mouse photoreceptors. Mamm. Genome 2000, 11, 422–427. [Google Scholar] [CrossRef] [PubMed]
  64. Song, D.; Song, Y.; Hadziahmetovic, M.; Zhong, Y.; Dunaief, J.L. Systemic administration of the iron chelator deferiprone protects against light-induced photoreceptor degeneration in the mouse retina. Free Radic. Biol. Med. 2012, 53, 64–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Sahin, K.; Gencoglu, H.; Akdemir, F.; Orhan, C.; Tuzcu, M.; Sahin, N.; Yilmaz, I.; Juturu, V. Lutein and zeaxanthin isomers may attenuate photo-oxidative retinal damage via modulation of G protein-coupled receptors and growth factors in rats. Biochem. Biophys. Res. Commun. 2019, 516, 163–170. [Google Scholar] [CrossRef]
  66. Song, D.; Song, J.; Wang, C.; Li, Y.; Dunaief, J.L. Berberine protects against light-induced photoreceptor degeneration in the mouse retina. Exp. Eye Res. 2016, 145, 1–9. [Google Scholar] [CrossRef] [Green Version]
  67. Forte, E.; Zhang, Z.; Thorp, E.B.; Hummel, M. Cytomegalovirus Latency and Reactivation: An Intricate Interplay With the Host Immune Response. Front. Cell Infect. Microbiol. 2020, 10, 130. [Google Scholar] [CrossRef]
  68. Kim, S.J.; Varghese, T.K.; Zhang, Z.; Zhao, L.C.; Thomas, G.; Hummel, M.; Abecassis, M. Renal ischemia/reperfusion injury activates the enhancer domain of the human cytomegalovirus major immediate early promoter. Am. J. Transpl. 2005, 5, 1606–1613. [Google Scholar] [CrossRef]
  69. Atherton, S.S.; Newell, C.K.; Kanter, M.Y.; Cousins, S.W. T cell depletion increases susceptibility to murine cytomegalovirus retinitis. Investig. Ophthalmol. Vis. Sci. 1992, 33, 3353–3360. [Google Scholar]
  70. Shoskes, D.A.; Keslar, K.S.; Gotwald, P.; Berglund, R.; Vij, S. Neuroinflammatory gene expression in chronic prostatitis/chronic pelvic pain syndrome patients: Insights into etiology and phenotype biology. Transl. Urol. 2021, 10, 3340–3347. [Google Scholar] [CrossRef]
  71. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet. J. 2011, 17, 3. [Google Scholar] [CrossRef]
  72. Andrews, S. FastQC: A Quality Control Tool for High Throughput Sequence Data. 2010. Available online: http://www.bioinformatics.babraham.ac.uk/projects/fastqc/ (accessed on 25 August 2020).
  73. Dobin, A.; Davis, C.A.; Schlesinger, F.; Drenkow, J.; Zaleski, C.; Jha, S.; Batut, P.; Chaisson, M.; Gingeras, T.R. STAR: Ultrafast universal RNA-seq aligner. Bioinformatics 2013, 29, 15–21. [Google Scholar] [CrossRef]
  74. Anders, S.; Pyl, P.T.; Huber, W. HTSeq—A Python framework to work with high-throughput sequencing data. Bioinformatics 2015, 31, 166–169. [Google Scholar] [CrossRef] [Green Version]
  75. Love, M.I.; Huber, W.; Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014, 15, 550. [Google Scholar] [CrossRef] [Green Version]
  76. Wang, L.; Wang, S.; Li, W. RSeQC: Quality control of RNA-seq experiments. Bioinformatics 2012, 28, 2184–2185. [Google Scholar] [CrossRef] [Green Version]
  77. Zhang, M.; Xin, H.; Duan, Y.; Atherton, S.S. Ocular reactivation of MCMV after immunosuppression of latently infected BALB/c mice. Investig. Ophthalmol. Vis. Sci. 2005, 46, 252–258. [Google Scholar] [CrossRef] [Green Version]
  78. Zhang, M.; Xin, H.; Roon, P.; Atherton, S.S. Infection of retinal neurons during murine cytomegalovirus retinitis. Investig. Ophthalmol. Vis. Sci. 2005, 46, 2047–2055. [Google Scholar] [CrossRef]
  79. Zhang, M.; Covar, J.; Marshall, B.; Dong, Z.; Atherton, S.S. Lack of TNF-alpha promotes caspase-3-independent apoptosis during murine cytomegalovirus retinitis. Investig. Ophthalmol. Vis. Sci. 2011, 52, 1800–1808. [Google Scholar] [CrossRef] [Green Version]
  80. Zhang, M.; Marshall, B.; Atherton, S.S. Murine cytomegalovirus infection and apoptosis in organotypic retinal cultures. Investig. Ophthalmol. Vis. Sci. 2008, 49, 295–303. [Google Scholar] [CrossRef] [Green Version]
Figure 1. (A) Representative images of SD-OCT in eyes of latently infected mice with medium retinal degeneration (m) and severe retinal degeneration (h) at 18 months p.i., together with age-matched control eyes (c). (B) Retinal thickness by SD-OCT. **** p < 0.0001 using the Mann-Whitney test. (C) Validation of RNA-seq transcriptome data by qRT-PCR analysis of eight genes in three control and six virus-infected samples. (D) A Heat map for three control samples (c1, c2, c3) and six virus-infected samples (m1, m2, m3, h1, h2, h3).
Figure 1. (A) Representative images of SD-OCT in eyes of latently infected mice with medium retinal degeneration (m) and severe retinal degeneration (h) at 18 months p.i., together with age-matched control eyes (c). (B) Retinal thickness by SD-OCT. **** p < 0.0001 using the Mann-Whitney test. (C) Validation of RNA-seq transcriptome data by qRT-PCR analysis of eight genes in three control and six virus-infected samples. (D) A Heat map for three control samples (c1, c2, c3) and six virus-infected samples (m1, m2, m3, h1, h2, h3).
Ijms 24 04322 g001
Figure 2. Downregulation of 22 differentially expressed genes linked to the phototransduction pathway of rod and cone cells.
Figure 2. Downregulation of 22 differentially expressed genes linked to the phototransduction pathway of rod and cone cells.
Ijms 24 04322 g002
Figure 3. (A) Representative merged photomicrograph of staining for RPE-65 (red), TUNEL (green), and DAPI (blue) in eyes of latently infected mice at 18 months p.i. (MCMV) together with age-matched control eyes (control). (B) Western blot stained with antibodies against cleaved caspase 3, pRIP3, MLKL, RIP1, Rodopsin, and β-actin in eyes of latently infected mice at 18 months p.i. (MCMV) together with age-matched control eyes (control).
Figure 3. (A) Representative merged photomicrograph of staining for RPE-65 (red), TUNEL (green), and DAPI (blue) in eyes of latently infected mice at 18 months p.i. (MCMV) together with age-matched control eyes (control). (B) Western blot stained with antibodies against cleaved caspase 3, pRIP3, MLKL, RIP1, Rodopsin, and β-actin in eyes of latently infected mice at 18 months p.i. (MCMV) together with age-matched control eyes (control).
Ijms 24 04322 g003
Figure 4. (A) Representative merged photomicrographs of stainings for (A) GFAP (red), TUNEL (green), and DAPI (blue) or (B) Iba-1 (red) and DAPI (blue) in eyes of latently infected mice at 18 months p.i. (MCMV) together with age-matched control eyes (control).
Figure 4. (A) Representative merged photomicrographs of stainings for (A) GFAP (red), TUNEL (green), and DAPI (blue) or (B) Iba-1 (red) and DAPI (blue) in eyes of latently infected mice at 18 months p.i. (MCMV) together with age-matched control eyes (control).
Ijms 24 04322 g004
Figure 5. The summary graph of upstream regulators.
Figure 5. The summary graph of upstream regulators.
Ijms 24 04322 g005
Table 1. Top mRNAs differentially expressed.
Table 1. Top mRNAs differentially expressed.
Gene SymbolGene NameFold Changeq Value
Gm14308predicted gene 1430811,223,3503.56 × 10−9
Kdm5dlysine (K)-specific demethylase 5D2,210.251.17 ×10−2
Utyubiquitously transcribed tetratricopeptide repeat containing, Y-linked968.763.60 × 10−4
Ddx3yDEAD-box helicase 3 Y-linked5125.80 × 10−5
Eif2s3yeukaryotic translation initiation factor 2, subunit 3, structural gene Y-linked382.682.10 × 10−5
Krt16keratin 1638.854.24 × 10−3
S100a9S100 calcium binding protein A928.441.62 × 10−3
Stfa2l1stefin A2 like 125.633.49 × 10−2
Mmp13matrix metallopeptidase 1320.531.09 × 10−2
Sbsnsuprabasin17.022.00 × 10−2
Ccr1C-C motif chemokine receptor 111.081.96 × 10−2
Gm4951predicted gene 49519.123.49 × 10−7
Gm4841predicted gene 48418.514.24 × 10−4
Msr1macrophage scavenger receptor 18.163.83 × 10−3
Nlrc5NLR family CARD domain containing 55.811.83 × 10−2
Ccdc24coiled-coil domain containing 24−6.105.79 × 10−4
Pdzph1PDZ and pleckstrin homology domains 1−5.654.31 × 10−4
Apobapolipoprotein B−5.201.40 × 10−5
Sntg2syntrophin gamma 2−5.131.70 × 10−5
Glb1l3galactosidase beta 1 like 3−4.639.80 × 10−5
Albalbumin−4.569.31 × 10−9
Nr2e3nuclear receptor subfamily 2 group E member 3−4.145.00 × 10−6
Nrlneural retina leucine zipper−4.021.94 × 10−3
Pdcphosducin−48.00 × 10−6
Cabp4calcium binding protein 4−3.861.00 × 10−6
Wdr31WD repeat domain 31−3.862.71 × 10−4
Kcnv2potassium voltage-gated channel modifier subfamily V member 2−3.831.34 × 10−4
Pde6bphosphodiesterase 6B−3.836.23 × 10−4
Gnat1G protein subunit alpha transducin 1−3.836.81 × 10−4
Kcnj14potassium inwardly rectifying channel subfamily J member 14−3.758.52 × 10−4
Table 2. Ingenuity canonical pathways.
Table 2. Ingenuity canonical pathways.
Ingenuity Canonical Pathwaysp-ValueRatio
Phototransduction Pathway2.51 × 10−280.415
Relaxin Signaling6.31 × 10−70.08
cAMP-mediated signaling4.57 ×10−50.0526
tRNA Splicing2.34 × 10−40.116
Protein Kinase A Signaling2.45 × 10−40.0376
Dendritic Cell Maturation6.61 × 10−40.0492
Phagosome Formation0.00120.056
Dopamine-DARPP32 Feedback in cAMP Signaling0.00130.0491
Role of NFAT in Regulation of the Immune Response0.00250.0442
Endothelin-1 Signaling0.00320.0426
GP6 Signaling Pathway0.00460.0504
Opioid Signaling Pathway0.0050.0364
G Beta Gamma Signaling0.00510.0492
Tec Kinase Signaling0.00560.0427
CREB Signaling in Neurons0.00560.0386
Communication between Innate and Adaptive Immune Cells0.00810.0521
G-Protein Coupled Receptor Signaling0.00930.0331
Table 3. Ophthalmic disease and visual system function.
Table 3. Ophthalmic disease and visual system function.
Diseases or Functions Annotationp-ValueActivation
z-Score
Number of Molecules
Affected
Degeneration of photoreceptors1.58 × 10−274.89738
Degeneration of retinal cone cells2.29 × 10−92.95610
Degeneration of outer nuclear layer6.12 × 10−93.13810
Degeneration of photoreceptor outer segments4.49 × 10−82.1218
Degeneration of retinal rods3.31 × 10−72.4338
Function of retina8.69 × 10−31−2.94734
Function of retinal cells9.64 × 10−31−2.78633
Quantity of photoreceptors1.18 × 10−18−3.90824
Quantity of retinal cells6.24 × 10−17−3.52125
Function of photoreceptors2.05 × 10−14−2.78613
Photoresponse2.o × 10−12−2.32814
Quantity of retinal cone cells6.12 × 10−09−2.34310
Function of retinal rods4.73 × 10−08−2.4337
Table 4. Cell death and survival.
Table 4. Cell death and survival.
Diseases or Functions Annotationp-ValueActivation z-ScoreNumber of Molecules
Affected
Cell death of retinal cells6.42 × 10−90.94311
Cell death of photoreceptors1.40 × 10−71.38
Apoptosis of photoreceptors2.20 × 10−50.1525
Apoptosis of retinal cells6.68 × 10−50.1526
Apoptosis4.98 × 10−40.93681
Necrosis3.0 × 10−40.25880
Cell survival9.14 × 10−40.02949
Necrosis of epithelial tissue0.0031.65122
Table 5. Immune cells and glia.
Table 5. Immune cells and glia.
Diseases or Functions Annotationp-ValueActivation z-ScoreNumber of Molecules
Affected
Activation of Müller glia9.3 × 10−7 4
Activation of neuroglia1.65 × 10−51.07911
Cellular infiltration by leukocytes2.21 × 10−52.72922
Leukocyte migration8.61 × 10−52.47238
Cell movement of leukocytes2.25 × 10−42.51832
Activation of phagocytes4.93 × 10−41.67317
Cellular infiltration by lymphocytes6.84 × 10−40.95710
Activation of macrophages0.0011.37512
Activation of antigen presenting cells0.001931.63814
Cell movement of phagocytes0.00311.59722
Table 6. Upstream regulators.
Table 6. Upstream regulators.
Upstream RegulatorMolecule TypeActivation z-Scorep-Value of OverlapNumber of Molecules
Affected
TGFB1growth factor3.0053.99 × 10−648
APPother2.3758.44 × 10−630
IFNGcytokine4.0338.72 × 10−640
OSMcytokine2.4395.58 × 10−525
CKAP2Lother1.6336.31 × 10−56
IFNB1cytokine1.9751.4 × 10−413
ELAVL1other2.9613.84 × 10−410
CHUKkinase1.9765.0 × 10−410
CSF2cytokine2.5976.47 × 10−417
IL17RAtransmembrane receptor28.51 × 10−44
SMAD3transcription regulator1.7690.0011611
IL1Bcytokine2.6150.0012526
AGTgrowth factor2.9370.0013923
Vegfgroup2.350.0014816
ARNT2transcription regulator1.6670.001619
IKBKGkinase2.4010.002147
SIM1transcription regulator1.6670.002169
PTGER2G-protein coupled receptor2.4330.00246
MRTFAtranscription regulator1.6640.002898
STAT3transcription regulator1.6890.0041617
ERKgroup2.420.005229
TNFcytokine1.9460.0054838
RARAligand-dependent nuclear receptor1.7060.0084411
EGFgrowth factor2.2960.0093215
IL17Acytokine2.7680.009339
FOXO3transcription regulator1.9530.015310
PI3K (complex)complex1.8120.01689
IL1Acytokine2.1460.0178
IFN-α/βgroup1.9410.02224
TGFAgrowth factor1.980.02224
STAT1transcription regulator2.6840.022510
BMP4growth factor1.9020.02717
PDGF BBcomplex2.0860.03149
PRLcytokine1.6430.03358
PTGS2enzyme2.2160.03596
CD3complex1.9780.040814
Table 7. Infected eyes with severe retinal degeneration vs. infected eyes without severe retinal degeneration.
Table 7. Infected eyes with severe retinal degeneration vs. infected eyes without severe retinal degeneration.
Ingenuity Canonical Pathways and Diseases or Functionsp-ValueActivation z-ScoreNumber of Molecules Affected
Phototransduction pathway1.26 × 10−20N/A11
Relaxin signaling2.69 × 10−3N/A3
tRNA Splicing2.95 × 10−3N/A2
Wnt/Ca+ pathway5.89 × 10−3N/A2
Protein Kinase A signaling6.17 × 10−3N/A4
cAMP-mediated signaling8.51 × 10−3N/A3
Cell death of retinal cells1.22 × 10−100.6557
Cell death of photoreceptors2.39 × 10−81.2135
Retinal degeneration3.6 × 10−202.76618
Degeneration of photoreceptors1.72 × 10−112.28
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhang, X.; Xu, J.; Marshall, B.; Dong, Z.; Liu, Y.; Espinosa-Heidmann, D.G.; Zhang, M. Transcriptome Analysis of Retinal and Choroidal Pathologies in Aged BALB/c Mice Following Systemic Neonatal Murine Cytomegalovirus Infection. Int. J. Mol. Sci. 2023, 24, 4322. https://doi.org/10.3390/ijms24054322

AMA Style

Zhang X, Xu J, Marshall B, Dong Z, Liu Y, Espinosa-Heidmann DG, Zhang M. Transcriptome Analysis of Retinal and Choroidal Pathologies in Aged BALB/c Mice Following Systemic Neonatal Murine Cytomegalovirus Infection. International Journal of Molecular Sciences. 2023; 24(5):4322. https://doi.org/10.3390/ijms24054322

Chicago/Turabian Style

Zhang, Xinyan, Jinxian Xu, Brendan Marshall, Zheng Dong, Yutao Liu, Diego G. Espinosa-Heidmann, and Ming Zhang. 2023. "Transcriptome Analysis of Retinal and Choroidal Pathologies in Aged BALB/c Mice Following Systemic Neonatal Murine Cytomegalovirus Infection" International Journal of Molecular Sciences 24, no. 5: 4322. https://doi.org/10.3390/ijms24054322

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop