Investigative Ophthalmology & Visual Science Cover Image for Volume 66, Issue 1
January 2025
Volume 66, Issue 1
Open Access
Retina  |   January 2025
CAMK2D and Complement Factor I–Involved Calcium/Calmodulin Signaling Modulates Sodium Iodate-Induced Mouse Retinal Degeneration
Author Affiliations & Notes
  • Weixing Xu
    Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou City, China
    School of Graduate, Dalian Medical University, Dalian City, China
    Key Laboratory of Age-related Macular Degeneration of Liaoning Province, Jinzhou Medical University, Jinzhou City, China
  • Liu Cao
    Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang City, China
  • Hua Liu
    School of Graduate, Dalian Medical University, Dalian City, China
    Key Laboratory of Age-related Macular Degeneration of Liaoning Province, Jinzhou Medical University, Jinzhou City, China
  • Correspondence: Weixing Xu, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou City, Henan Province 450000, China; [email protected]
Investigative Ophthalmology & Visual Science January 2025, Vol.66, 63. doi:https://doi.org/10.1167/iovs.66.1.63
  • Views
  • PDF
  • Share
  • Tools
    • Alerts
      ×
      This feature is available to authenticated users only.
      Sign In or Create an Account ×
    • Get Citation

      Weixing Xu, Liu Cao, Hua Liu; CAMK2D and Complement Factor I–Involved Calcium/Calmodulin Signaling Modulates Sodium Iodate-Induced Mouse Retinal Degeneration. Invest. Ophthalmol. Vis. Sci. 2025;66(1):63. https://doi.org/10.1167/iovs.66.1.63.

      Download citation file:


      © ARVO (1962-2015); The Authors (2016-present)

      ×
  • Supplements
Abstract

Purpose: To investigate the effect of Ca2+/calmodulin-dependent protein kinase II (CAMKII) δ subtypes (CAMK2D) on sodium iodate (NaIO3)-induced retinal degeneration in mice.

Methods: Bioinformatics analysis and Western blot experiments were used to screen the significantly differentially expressed genes in age-related macular degeneration (AMD) disease. CAMK2D knockdown and overexpression models were constructed by lentivirus (LV) infection of adult retinal pigment epithelial cell line-19 (ARPE-19) cells in vitro. Flow cytometry was used to detect ARPE-19 cell apoptosis induced by NaIO3. In vivo, CAMK2D knockdown and overexpression mouse models were generated by infecting mouse retinal pigment epithelium (RPE) with adeno-associated virus (AAV). Retinography, optical coherence tomography (OCT), and histological analysis (hematoxylin and eosin staining) were used to detect NaIO3-induced retinal structural changes in mice. Electroretinography (ERG) was used to detect NaIO3-induced retinal function changes in mice. TdT-mediated dUTP nick-end labeling (TUNEL) staining was used to detect the apoptosis of retinal cells induced by NaIO3. RNA sequencing (RNA-Seq) and bioinformatics analysis were used to screen for target genes affected by CAMK2D in CAMK2D-overexpressing ARPE-19 cells. And flow cytometry, OCT, and ERG were used to evaluate the regulatory effect of CAMK2D on target genes.

Results: Bioinformatics analysis found the expression of genes related to Ca2+ signal was significantly reduced in AMD patients. Western blot showed that in a mouse model of dry AMD induced by NaIO3, CAMK2D expression in RPE-Choroid tissue significantly lower than normal mice. In vitro, our results showed that overexpression of CAMK2D in ARPE-19 cells decreased apoptosis induced by NaIO3 and knockdown increased apoptosis. In vivo, CAMK2D overexpression in RPE cells can attenuate the retina degeneration induced by NaIO3 and CAMK2D knockdown aggravated degeneration. The bioinformatics analysis indicated that CAMK2D might affect AMD pathology through complement factor I (CFI). In vitro, knockdown of CFI in ARPE-19 cells increased apoptosis induced by NaIO3. In knockdown CFI ARPE-19 cells, overexpression of CAMK2D reduced the above apoptosis. In mice retina, CFI knockdown can aggravate the retina degeneration induced by NaIO3. In knockdown CFI mice, overexpression of CAMK2D in RPE can attenuate the above retina degeneration. Western blot confirmed that CAMK2D regulated the expression of CFI in mice.

Conclusions: CAMK2D can attenuate the retinal degeneration induced by NaIO3, which was achieved by regulating the CFI.

Age-related macular degeneration (AMD) is a leading cause of visual dysfunction worldwide, as a progressive retinal degenerative disease influenced by both environmental and genetic factors.1 Genetic factors play an important role in AMD, and it is estimated that the genetic factors can account for 45% to 70% of the disease.2 Retinal pigment epithelium (RPE) dysfunction is considered to be the key cause of AMD pathogenesis. For now, no methods could cure AMD.1,3 To explore the abnormal gene expression of AMD patients, we analyzed the gene expression profile GSE125564 from RPE cells of AMD patients in Gene Expression Omnibus database. The data indicated that the genes expression related to Calcium ion binding and Voltage gated calcium channel activity were significantly reduced in AMD patients compared with non-AMD patients. Calcium (Ca2+) regulates a range of cellular processes as a highly versatile intracellular signal.4,5 Ca2+/calmodulin-dependent protein kinase II (CAMKII) is a multifunctional kinase as a central coordinator and executor of Ca2+ signal transduction.6,7 Studies have shown that CAMKII was involved in the regulation of multiple biological processes such as cell apoptosis8,9and angiogenesis.1013 The occurrence and development of AMD are closely related to RPE cell apoptosis and angiogenesis.1416 However, the effect of CAMKII on RPE has not yet been studied. Our Western blot experiments show that in a mouse model of dry AMD induced by NaIO3, CAMK2D expression in RPE-choroid tissue significantly lower than normal mice. Therefore we evaluated the effect of CAMK2D on dry AMD in vitro and in vivo, and explored its mechanism of action. 
Material and Methods
Bioinformatics Analysis
Data sets were downloaded from Gene Expression Omnibus database (http://www.ncbi.nlm.nih.gov/geo). The microarray data of GSE125564 is based on the GPL23159 platform (Affymetrix Clariom S Assay, Human), which contains a dataset of systems-level transcriptome analysis of RPE samples differentiated from induced pluripotent stem cells (iPScs) of four advanced AMD and age-matched three non-AMD donors.17 The data sets had been normalized through log transformation. The DEGs were identified by the “limma” package through NetworkAnalyst (V.3.0) online database (https://www.networkanalyst.ca/). The volcano plot was graphed through NetworkAnalyst online database, and the GO functional enrichment analysis was graphed through “ggplot” package in R 4.3.0 software.18 
Cell Culture
At 37°C temperature incubator with 5% CO2, ARPE-19 cells were cultured in complete medium (89% DME/F12, 10% FBS, and 1% penicillin-streptomycin).19 
Lentiviral Infection of ARPE-19 Cells
The lentivirus-based vectors were used to construct overexpression of CAMK2D and shRNA-mediated lentivirus vectors were used to construct knockdown of CAMK2D (Gene-Chem, Shanghai, China). Lentiviral infection effects were displayed in Supplementary Figure S1. Puromycin 2 µg/mL was used to selected ARPE-19 cells and then amplified (Beyotime Biotechnology, Shanghai, China). ShRNA-mediated lentivirus vectors were used to construct knockdown of complement factor I (CFI) (Gene-Chem). 200 µg/ml geneticin was used to selected ARPE-19 cells and then amplified (Beyotime Biotechnology). All the sequences are displayed in Supplementary Tables S1 through S3
Quantitative Real-Time Reverse Transcription Polymerase Chain Reaction (qRT-PCR)
Total RNA was extracted from the ARPE-19 cells using an PrimeScript RT-PCR kit (TaKaRa, Shiga, Japan). The polymerase chain reaction (PCR) reaction conditions were as follows: pre-degeneration at 95°C for 30 seconds, Reps: 1; degeneration at 95°C for five seconds and annealing/stretching at 60°C for 30 seconds, Reps: 40. The primers of CAMK2D: forward primer: 5′-CAGTGACACCTGAAGCCAAAGA-3′; reverse primer: 5′-GTGCATCATGGAGGCAACAGTA-3′. The endogenous control was β-Actin. The relative mRNA level of CAMK2D was calculated by the 2−ΔΔCt method.20 
Western Blot Analysis
ARPE-19 cells were lysed using a mixture of RIPA, PMSF, and phosphatase inhibitors, and then total protein was extracted. Mice were anesthetized by intraperitoneal injection of excess pentobarbital sodium. Carefully separate the tissue around the eye, cut the optic nerve and quickly remove the eye, and then place it on ice. Carefully remove the conjunctiva, cornea, lens, and the rest to retain the complete eye cup. The retinal layer was carefully removed and the RPE-chorioscleral tissue was preserved. The pigmented tissue on the inner surface was scraped with a 1 mL syringe needle and collected into a centrifuge tube. The sclera and residual tissue were discarded. A sufficient amount of pigmented tissue was collected and spun in a centrifuge at 4°C. The dark brown precipitate was RPE-choroidal tissue. Total proteins were extracted from RPE-choroidal tissue using a Minute Total Protein Extraction Kit (Invent Biotechnologies, Eden Prairie, MN, USA) according to the manufacturer's instructions.21 The protein was configured into samples of concentration. Equal amounts of samples were subjected to electrophoresis and then transferred to the PVDF membrane.22 The endogenous control was β-actin. The intensities of the bands were measured by ImageJ software.22 
Flow Cytometry
PE Annexin V Apoptosis Detection Kit with 7-AAD was purchased from Becton, Dickinson and Company.23 FITC Annexin V Apoptosis Detection Kit with PI was purchased from Beyotime Biotechnology. According to the manufacturer's instructions, the ARPE-19 cells were labeled for apoptosis.22 The data was analyzed by FlowJo Software.22,24 
Animals and Model of NaIO3-Induced Retinal Degeneration
The adult Male MC57BL/6J mice aged 6–8 weeks were purchased from Beijing Vital River Laboratory Animal Technology Co. Ltd. (Beijing, China). Mice were maintained in 23° to 25°C, 12-hour light/dark cycle and free access of water and food.25,26 All animal experiments were reviewed and approved by the Experimental Animal Ethics Committee of Jinzhou Medical University and conformed to the “Guide for the Care and Use of Laboratory Animals” published by the National Institutes of Health (Publication, 8th Edition, 2011). The study is reported in accordance with ARRIVE guidelines (https://arriveguidelines.org).22 Mice were intraperitoneally injected with 25 mg/kg NaIO3 once. One week after injection, fundus photography and optical coherence tomography (OCT) of Micron IV retinal imaging camera system were performed. The model was successful when the retina showed deposits like drusen. 
Construction of AAV9 Vectors
AAV9 vectors were constructed by the Shanghai Genechem Co. Ltd. (Shanghai, China). AAV9-rpe65p-MCS-EGFP-3Flag-SV40 PolyA was the vector element sequence. Mouse CAMK2D(NM_001025438) was constructed into the AAV9 vector using the restriction enzymes NcoI/NcoI. Mouse CAMK2D (mir155[CAMK2D]) was constructed into the AAV9 vector using the restriction enzymes BsrGI/NheI. The specific RPE65 (retinoid isomerohydrolase RPE65) promoter was proved to be an RPE tissue-specific promoter. After AAV9 infects the retina, expression of the foreign gene under the regulation of this promoter normally occurs only in the RPE.27 
Intravitreal Injection of Viral Vector
Mice were anesthetized by the intraperitoneal injection of pentobarbital sodium (80 mg/kg). After mydriasis with compound tropicamide eye drops, a sterile disposable injection needle (33-gauge) was used to pierce the sclera gently about 1 mm behind the limbus under an operating microscope. Subsequently, a 33-gauge microsyringe was used to enter through the scleral puncture port. Under the microscope, the tip of the needle was located behind the lens and in front of the retina, and about 1 µL of virus was slowly injected. After the injection was completed, the eyeballs were smeared with gatifloxacin eye gel, and the mice were promptly rewarmed and revived. Gatifloxacin eye gel was applied daily for three days after surgery to prevent infection.26 
Retinography and OCT Imaging
Mice were anesthetized through the intraperitoneal injection of pentobarbital sodium (80 mg/kg). Their pupils were dilated with tropicamide and corneas were protected by gatifloxacin eye gel. Retinography and OCT were taken by Micron IV retinal imaging camera system (Phoenix Research Laboratories, Pleasanton, CA, USA).22 Based on OCT results, the peripheral nerve thickness of the optic nerve head was measured by Image-Pro Plus 6.0 software. 
Histological Analysis
The eyes were enucleated and embedded in paraffin. Retinal section tissues were sliced for hematoxylin and eosin (H&E) staining. Images were taken by a microscope.22,28 
ERG
Mice were dark adapted overnight and then anesthetized, and their pupils were dilated.29 The full-field ERGs were recorded by Micron IV retinal imaging camera system. The parameters were as follows: flash strength: 6.2 log (cd/m2), duration: 10 ms, aiming light: 6.2 log(cd/m2), 50 Hz clean filter, delay between sweeps: 5.0 sec. The amplitude of a-wave and b-wave were statistically analyzed separately.22 
TUNEL Staining
Mice were anesthetized by intraperitoneal injection of excess pentobarbital sodium. Cardiac perfusion with normal saline solution was performed first and continued until the liver turned white. Then, normal saline solution was replaced by 4% paraformaldehyde to continue perfusion until the body stiffness of the mice. Carefully separate the tissue around the eye, cut the optic nerve and quickly remove the eye, and then place it in eye-specific fixative solution at 4 °C. After 72 hours, carefully remove the conjunctiva, cornea, lens, etc. to retain the complete eye cup. The eye cup was gradually dehydrated and embedded in optimal cutting temperature compound embedding agent (O.C.T. Sakura; Sakura Finetek USA, Inc., Torrance, CA, USA) and then frozen overnight. The next day, the eye cup was made into a cryosection with a thickness of 10 µm for TUNEL staining. Cryosections of eyeball were stained by the In Situ Cell Death Detection Kit (Roche, Basel, Switzerland) according to the instructions. Nuclei were stained with DAPI, and photographs were taken by fluorescence microscope (Leica, Wetzlar, Germany). The apoptosis cells were counted and averaged in five visual fields selected randomly.30,31 
RNA Seq and Bioinformatics Analysis
RNA library sequencing was performed on the Illumina Hiseq 2500/4000 by Gene Denovo Biotechnology Co., Ltd (Guangzhou, China). Bioinformatic analysis was performed using Omicsmart, a real-time interactive online platform for data analysis (http://www.omicsmart.com).32 
Statistical Analysis
Each experiment was repeated a minimum of three times. GraphPad Prism 8 (GraphPad, San Diego, CA, USA) was used for statistical analysis and graph. T-test was used for two groups statistical analyses. For three or more groups, statistical analyses were calculated with one-Way analysis of variance, followed by Tukey tests for between-group analysis. P < 0.05 was considered as a significant difference.22 
Results
Bioinformatics analysis found that the Ca2+-related molecular functions (MF) were downregulated in AMD and Western Blot indicated that CAMK2D expression was downregulated in a mouse model of dry AMD. Eight hundred twenty-five significant differentially expressed genes (DEGs) of GSE125564 were identified, include 475 upregulated genes and 350 downregulated genes in AMD samples, as shown in the Volcano plot (P < 0.05, |log fold change (FC)|>1, Fig. 1A). The downregulated genes were used to perform gene ontology (GO) through the PANTHER classification system. Sorted by descending order of P value for the GO enrichment terms, the term of calcium ion binding ranked the first one and hit the largest number of genes in molecular functions. The term of voltage gated calcium channel activity ranked third in molecular functions (Fig. 1B). NaIO3-induced retinal degeneration is an animal model for AMD and frequently used to study cellular and molecular mechanisms underlying AMD. We constructed the model, and our Western blot showed that the CAMK2D expression in the NaIO3 group was significantly lower than the normal group (P < 0.05, Figs. 1C, 1D). This suggests that CAMK2D expression was downregulated in the dry AMD model induced by NaIO3
Figure 1.
 
CAMK2D expression in the dry AMD. (A) Volcano plot showing 825 significant DEGs of GSE125564 (Adjusted P value < 0.05, │log FC│ > 1). (B) The molecular functions terms in GO of downregulated genes by PANTHER. (C) CAMK2D protein level in RPE-choroid tissue. (D) Quantification based on (C) (n = 3, **P < 0.01).
Figure 1.
 
CAMK2D expression in the dry AMD. (A) Volcano plot showing 825 significant DEGs of GSE125564 (Adjusted P value < 0.05, │log FC│ > 1). (B) The molecular functions terms in GO of downregulated genes by PANTHER. (C) CAMK2D protein level in RPE-choroid tissue. (D) Quantification based on (C) (n = 3, **P < 0.01).
Knockdown or Overexpression of CAMK2D Increased or Decreased ARPE-19 Cell Apoptosis Induced by NaIO3 In Vitro
As assayed by the PE Annexin V Apoptosis Detection Kit with 7-AAD, treatment with 1250 µg/mL NaIO3 for 24 hours, total apoptosis rate in LV-CON313 + NaIO3 group was approximately 10%. With NaIO3 treatment, the total apoptosis rate in RNAi (119303) + NaIO3 group was significantly increased compared with the LV-CON313 + NaIO3 group. Without NaIO3 treatment, the knockdown of CAMK2D did not affect ARPE-19 cells apoptosis (Figs. 2A, 2B). With NaIO3 treatment or not, the total apoptosis rate in LV-CAMK2D was lower than LV-CON335 (Figs. 2C, 2D). The data demonstrated that knockdown of CAMK2D in the ARPE-19 cells (RNAi [119303]) increased cells apoptosis and overexpression (LV-CAMK2D) decreased cells apoptosis induced by NaIO3
Figure 2.
 
Flow cytometry analysis of apoptosis of ARPE‐19 cells. (A) About knockdown of CAMK2D, the percentage of cells in each quadrant is presented. (B) The total apoptosis rate in four groups were statistically analyzed based on (A). (C) About overexpression of CAMK2D, the percentage of cells in each quadrant is presented. (D) The total apoptosis rate in four groups were statistically analyzed based on (C). (Q2 represents late apoptosis, Q3 represents early apoptosis, Q2 + Q3 represents total apoptosis. n = 3 per group, ns: P > 0.05, **P < 0.01, ****P < 0.0001).
Figure 2.
 
Flow cytometry analysis of apoptosis of ARPE‐19 cells. (A) About knockdown of CAMK2D, the percentage of cells in each quadrant is presented. (B) The total apoptosis rate in four groups were statistically analyzed based on (A). (C) About overexpression of CAMK2D, the percentage of cells in each quadrant is presented. (D) The total apoptosis rate in four groups were statistically analyzed based on (C). (Q2 represents late apoptosis, Q3 represents early apoptosis, Q2 + Q3 represents total apoptosis. n = 3 per group, ns: P > 0.05, **P < 0.01, ****P < 0.0001).
Overexpression or Knockdown of CAMK2D Did Not Alter the Retinal Structure in Normal Mice
The above results showed that CAMK2D overexpression in ARPE-19 cells decreased cells apoptosis and knockdown increased cells apoptosis from oxidative damage. We further performed adeno-associated virus 9 (AAV9)-CAMK2D overexpression vector (AAV9-CAMK2D) and AAV9-CAMK2D knockdown vector (AAV9-shRNA-CAMK2D) in RPE cells of C57BL/6J mice to clarify the role of CAMK2D. Three weeks after the intravitreal injection of AAV9 vector, infection efficiency and CAMK2D expression levels were measured. We selected mouse retinas successfully expressing green fluorescent protein for Western blotting through Micron IV retinal imaging camera system (Fig. 3A). Western blotting indicated that CAMK2D was increased in RPE after AAV9-CAMK2D infection and downregulated in AAV9-shRNA-CAMK2D (Figs. 3B–E). To determine whether CAMK2D can change retina, fundus camera, OCT, and H&E staining were used to visualize retinal structures (Figs. 3F–H), and ERG was used to examine retinal function (Figs. 3I–K). AAV9-CAMK2D and AAV9-shRNA-CAMK2D showed no significant differences compared with the normal and control. These results indicate that neither overexpression nor knockdown of CAMK2D in RPE affects the retinal structure and function in normal mice. 
Figure 3.
 
Overexpression or knockdown of CAMK2D did not alter the retinal structure and function in normal mice. (A) Retinas expressing green fluorescent protein (GFP) were measured through Micron IV retinal imaging camera system. (B) Western blotting of CAMK2D after the injection of AAV9-CAMK2D vector. (C) Quantification based on (B). (D) Western blotting of CAMK2D after the injection of AAV9-shRNA-CAMK2D vector. (E) Quantification based on (D). (n = 3; *P < 0.05, **P < 0.01). (F) Fundus camera showed no significant changes in retinal structure after the AAV9 injection. (G) OCT showed no significant changes in retinal structure after the AAV9 injection. (H) H&E staining showed no significant changes in retinal structure after the AAV9 injection. (IK) ERG showed no significant differences of a-wave and b-wave after AAV9 vector injection. (n = 5; ns: P > 0.05).
Figure 3.
 
Overexpression or knockdown of CAMK2D did not alter the retinal structure and function in normal mice. (A) Retinas expressing green fluorescent protein (GFP) were measured through Micron IV retinal imaging camera system. (B) Western blotting of CAMK2D after the injection of AAV9-CAMK2D vector. (C) Quantification based on (B). (D) Western blotting of CAMK2D after the injection of AAV9-shRNA-CAMK2D vector. (E) Quantification based on (D). (n = 3; *P < 0.05, **P < 0.01). (F) Fundus camera showed no significant changes in retinal structure after the AAV9 injection. (G) OCT showed no significant changes in retinal structure after the AAV9 injection. (H) H&E staining showed no significant changes in retinal structure after the AAV9 injection. (IK) ERG showed no significant differences of a-wave and b-wave after AAV9 vector injection. (n = 5; ns: P > 0.05).
CAMK2D Overexpression in RPE Cells Attenuated the Retinal Degeneration and Knockdown Exacerbated Retinal Degeneration Induced by Oxidative Stress
In order to assess whether CAMK2D changes could affect the retina in oxidative damage, the mice were intraperitoneally injected with 25 mg/kg NaIO3, and one week later the retina was examined through Micron IV retinal imaging camera system. Compared to the AAV9-CON427 + NaIO3 group, less yellow-white exudation, thicker retina, and lighter structural damage in AAV9-CAMK2D+NaIO3 (Figs. 4A–E). And ERG indicated that a-wave and b-wave amplitudes were higher in AAV9-CAMK2D+NaIO3 than AAV9-CON427 + NaIO3 (Figs. 4F–H). TUNEL-positive signals were lower in AAV9-CAMK2D + NaIO3 group than AAV9-CON427 + NaIO3 (Figs. 4I–J). On the contrary, thinner retina and more severe structural damage were in AAV9-shRNA-CAMK2D + NaIO3 than AAV9-shRNA-NC + NaIO3 (Figs. 4A–E). Moreover, lower a/b-wave amplitude (Figs. 4F–H) and higher TUNEL-positive signals (Figs. 4I–J) was in AAV9-shRNA-CAMK2D + NaIO3 than AAV9-shRNA-NC + NaIO3
Figure 4.
 
Overexpression of CAMK2D in RPE attenuated NaIO3-induced retinal degeneration and knockdown exacerbated NaIO3-induced retinal degeneration. (A) Fundus camera (white arrows indicate foci of yellow-white exudation). (B) OCT (black arrow indicates the OCT imaging location; red arrows indicate bulging deposits like drusen. Scale bar: 100 µm.). (C) H&E staining (green arrows indicate bulging deposits. Scale bar: 50 µm.). (D) OCT detecting retinal thickness (black circular arrow indicates the OCT imaging location. Scale bar: 100 µm.). (E) The retinal thickness quantitative analysis based on (D). (n = 5; ****P < 0.0001). (F) ERG results. (G) The a-wave quantitative analysis based on (F). (H) The b-wave quantitative analysis based on (F) (n = 5; **P < 0.01, ***P < 0.001, ****P < 0.0001). (I) TUNEL results indicated retina apoptotic cells (white arrows indicate apoptotic cells. Scale bar: 50 µm. Below is the organizational structure of partial enlargement of the green rectangle in above.). (J) Quantitative analysis the number of apoptotic cells based on of (I) (n = 5; **P < 0.01, ****P < 0.0001).
Figure 4.
 
Overexpression of CAMK2D in RPE attenuated NaIO3-induced retinal degeneration and knockdown exacerbated NaIO3-induced retinal degeneration. (A) Fundus camera (white arrows indicate foci of yellow-white exudation). (B) OCT (black arrow indicates the OCT imaging location; red arrows indicate bulging deposits like drusen. Scale bar: 100 µm.). (C) H&E staining (green arrows indicate bulging deposits. Scale bar: 50 µm.). (D) OCT detecting retinal thickness (black circular arrow indicates the OCT imaging location. Scale bar: 100 µm.). (E) The retinal thickness quantitative analysis based on (D). (n = 5; ****P < 0.0001). (F) ERG results. (G) The a-wave quantitative analysis based on (F). (H) The b-wave quantitative analysis based on (F) (n = 5; **P < 0.01, ***P < 0.001, ****P < 0.0001). (I) TUNEL results indicated retina apoptotic cells (white arrows indicate apoptotic cells. Scale bar: 50 µm. Below is the organizational structure of partial enlargement of the green rectangle in above.). (J) Quantitative analysis the number of apoptotic cells based on of (I) (n = 5; **P < 0.01, ****P < 0.0001).
CAMK2D Regulates the Expression of CFI in ARPE-19 Cells Induced by NaIO3
To explore the mechanism of CAMK2D, the ARPE-19 cells after up-regulation of CAMK2D expression was detected by RNA-seq. RNA-seq identified 163 DEGs, which include 62 upregulated transcripts and 101 downregulated transcripts after CAMK2D overexpression (Fig. 5A). Seventy-seven disease genes of AMD were found in online mendelian inheritance in man. Venn diagrams show that CFI as the only overlapping gene was found between 163 DEGs and 77 AMD disease genes (Fig. 5B). CFI has two transcripts, encoding CFI and novel protein, respectively. In RNA-seq, CFI was upregulated, and novel protein was downregulated. Western blot results showed that CFI in overexpression CAMK2D was upregulated and knockdown was downregulated after ARPE-19 Cells induced by NaIO3 (Figs. 5C–F). 
Figure 5.
 
(A) DEGs of overexpression CAMK2D in ARPE-19 cells were shown in a volcano plot (P < 0.05, |log2FC| > log2(1.8)). (B) Venn diagrams show overlapping genes between disease genes of AMD and the DEGs. (C) Western blotting of CFI in ARPE-19 cells of overexpression CAMK2D. (D) Quantification based on (C). (E) Western blotting of CFI in ARPE-19 cells of knockdown CAMK2D. (F) Quantification based on (E) (n = 3 per group. ns: P > 0.05, *P < 0.05, **P < 0.01, ***P < 0.001).
Figure 5.
 
(A) DEGs of overexpression CAMK2D in ARPE-19 cells were shown in a volcano plot (P < 0.05, |log2FC| > log2(1.8)). (B) Venn diagrams show overlapping genes between disease genes of AMD and the DEGs. (C) Western blotting of CFI in ARPE-19 cells of overexpression CAMK2D. (D) Quantification based on (C). (E) Western blotting of CFI in ARPE-19 cells of knockdown CAMK2D. (F) Quantification based on (E) (n = 3 per group. ns: P > 0.05, *P < 0.05, **P < 0.01, ***P < 0.001).
CAMK2D Regulates CFI to Affect Retinal Degeneration Induced by NaIO3
Western blotting indicated that CFI was downregulated in ARPE-19 cells after LV-shRNA-CFI (CFI[RNAi]) infection (Figs. 6A, 6B). As assayed by the Annexin V-FITC Apoptosis Detection Kit with PI, without NaIO3 treatment, the knockdown of CFI did not affect ARPE-19 cell apoptosis. With NaIO3 treatment, the total apoptosis rate in CFI(RNAi) + NaIO3 group was significantly increased compared with the control (Normal + NaIO3 and LV-CON389 + NaIO3) group (Figs. 6C, 6E). The above data demonstrated that knockdown of CFI in the ARPE-19 cells increased apoptosis induced by NaIO3. As assayed by the PE Annexin V Apoptosis Detection Kit with 7AAD, with NaIO3 treatment, the total apoptosis rate in CFI(RNAi) + LV-CAMK2D + NaIO3 group was significantly decreased compared with the control (CFI(RNAi) + LV-CON335 + NaIO3 group) group (Figs. 6D, 6F). The results indicated overexpression of CAMK2D reduced the apoptosis induced by NaIO3 in ARPE-19 cells of CFI knockdown. Next, we further investigated the effect of overexpression of CAMK2D in mice retina of CFI knockdown on NaIO3-induced retinal degeneration. Western blotting indicated that CFI was downregulated in RPE-choroidal tissue after LV-shRNA-CFI (CFI[RNAi]) infection. CFI expression was higher than the Normal group after AAV9-CAMK2D infection. And CFI expression in the CFI(RNAi) + AAV-CAMK2D group was higher than the CFI(RNAi) group (Figs. 6G, 6H). The results indicated that overexpression of CAMK2D in RPE-Choroidal tissue can increase CFI expression. OCT examination of the mice retina revealed that, compared to the Normal + NaIO3 group, the retina was thinner in CFI(RNAi) + NaIO3 group. And the retina was thicker in CFI(RNAi) + AAV9-CAMK2D + NaIO3 group than the CFI(RNAi) + NaIO3 group (Figs. 6I, 6L). ERG indicated that a-wave and b-wave amplitudes were lower in CFI(RNAi) + NaIO3 group than the Normal + NaIO3 group. And the a-wave and b-wave amplitudes were higher in CFI(RNAi) + AAV9-CAMK2D + NaIO3 group than the CFI(RNAi) + NaIO3 group (Figs. 6J, 6K, 6M). The above results demonstrate that overexpression of CAMK2D can attenuate retinal degeneration induced by NaIO3 in mice retina of CFI knockdown. 
Figure 6.
 
(A) Western blotting of CFI in ARPE-19 cells of CFI knockdown. (B). Quantification based on (A) (n = 3. ns: P > 0.05, **P < 0.01). (C) The total apoptosis rates were statistically analyzed based on (E) the Annexin V-FITC Apoptosis Detection Kit with PI. (D) The total apoptosis rates were statistically analyzed based on (F) the Annexin V-PE Apoptosis Detection Kit with 7AAD. (E) The percentage of cells in each quadrant is presented on the Annexin V-FITC Apoptosis Detection Kit with PI. (F) The percentage of cells in each quadrant is presented on the Annexin V-PE Apoptosis Detection Kit with 7AAD (n = 3. ns: P > 0.05, **P < 0.01, ****P < 0.0001). (G) Western blotting of CFI in RPE-Choroidal tissue of mice. (H) Quantification based on (G) (n = 3. ns: P > 0.05, *P < 0.05, **P < 0.01, ***P < 0.001). (I) The retinal thickness quantitative analysis based on (L). (n = 5; **P < 0.01, ****P < 0.0001). (J) The a-wave quantitative analysis based on (M). (K) The b-wave quantitative analysis based on (M) (n = 5; *P < 0.05, **P < 0.01, ****P < 0.0001). (L) OCT detecting retinal thickness (Scale bar: 100 µm.). (M) ERG results.
Figure 6.
 
(A) Western blotting of CFI in ARPE-19 cells of CFI knockdown. (B). Quantification based on (A) (n = 3. ns: P > 0.05, **P < 0.01). (C) The total apoptosis rates were statistically analyzed based on (E) the Annexin V-FITC Apoptosis Detection Kit with PI. (D) The total apoptosis rates were statistically analyzed based on (F) the Annexin V-PE Apoptosis Detection Kit with 7AAD. (E) The percentage of cells in each quadrant is presented on the Annexin V-FITC Apoptosis Detection Kit with PI. (F) The percentage of cells in each quadrant is presented on the Annexin V-PE Apoptosis Detection Kit with 7AAD (n = 3. ns: P > 0.05, **P < 0.01, ****P < 0.0001). (G) Western blotting of CFI in RPE-Choroidal tissue of mice. (H) Quantification based on (G) (n = 3. ns: P > 0.05, *P < 0.05, **P < 0.01, ***P < 0.001). (I) The retinal thickness quantitative analysis based on (L). (n = 5; **P < 0.01, ****P < 0.0001). (J) The a-wave quantitative analysis based on (M). (K) The b-wave quantitative analysis based on (M) (n = 5; *P < 0.05, **P < 0.01, ****P < 0.0001). (L) OCT detecting retinal thickness (Scale bar: 100 µm.). (M) ERG results.
Discussion
AMD as a neurodegenerative disease is closely related to aging and genetics.1 The RPE plays a key role on maintaining retinal homeostasis through multiple biological functions.27 We found that the expression of genes related to Ca2+ signal was significantly reduced in AMD patients through bioinformatics analysis. CAMKII is a multifunctional kinase as a central coordinator and executor of Ca2+ signal transduction,6,7 which was involved in cell apoptosis8,9 and angiogenesis.1013 These biological processes are closely related to AMD.1416 However, there was a lack of direct evidence to elucidate the role of CAMKII in AMD. Our Western Blot experiments show that in a mouse model of dry AMD induced by NaIO3, CAMKII δ subtypes (CAMK2D) expression in RPE-Choroid tissue significantly lower than normal mice. Accordingly, we hypothesized that CAMK2D may be involved in regulating the pathological process of AMD. And then this hypothesis was verified by experiments. 
In vitro, we found that CAMK2D was involved in apoptosis induced by NaIO3 in ARPE-19 cells. Knockdown increased ARPE-19 cells apoptosis and overexpression decreased apoptosis, which was consistent with the conclusions of most studies about CAMKII in other cells4,3335 For retinal ganglion cells (RGCs) in stressful environment, basal levels of CAMKII can constantly afford them protection.33,34 Overexpression of CAMKIIαB by pIRES-hyg3/αB enhanced RGC-5 cells survival against glutamate treatment.33 The Paeoniflorin upregulated the level of p-CAMKII and then triggered AMPK activation against apoptosis induced by oxidative stress in ARPE-19 cells.35 Correspondingly, in RGCs with knocked down levels of CAMKIIalphaB by specific siRNA, cell apoptosis increase induced by glutamate.33 To further confirm the role of CAMK2D on apoptosis in vivo, gene transfer experiments were performed. The results showed that CAMK2D overexpression in RPE cells attenuated the retinal degeneration and knockdown exacerbated retinal degeneration induced by NaIO3. Such results further demonstrate the important role of CAMK2D in retina, which may affect AMD developing. 
To find the mechanism through which CAMK2D affects AMD, RNA-seq experiments were conducted. We overlapped 163 DEGs and 77 disease genes of AMD to find the key genes which CAMK2D may regulate. CFI as a major factor in the complement system was the only overlapping gene. CFI, a complement inhibitor, regulates the complement system activation by degrading complement component 3b (C3b).36 A growing body of research has shown that the complement pathway was engaged in the pathogenesis of AMD.37 In aging, complement may act through affecting inflammation, metabolism, apoptosis and other biological processes.38,39 Hyperactivation of the complement replacement pathway is one of the major causes of AMD.38,40 Complement component 3 (C3) as the central point of the three-cascade activation pathway of the complement system can lead to apoptosis by forming the membrane attack complex via the complement-activated pathway.38 CFI can reduce complement activity through restraining C3,19 which may reduce cells apoptosis. Moreover, haploinsufficiency of CFI is associated with decreased retinal thickness and is a strong risk factor for AMD development.41,42 Therefore we considered CFI as a major signaling gene for validation. In RNA-seq, CFI was upregulated. Western blotting analysis confirmed that CFI in overexpression CAMK2D was upregulated and knockdown was downregulated after ARPE-19 Cells induced by NaIO3. These suggest that CAMK2D might affect RPE cells apoptosis and AMD pathology through CFI. 
To further investigate whether CAMK2D could affect NaIO3-induced retinal degeneration by regulating CFI, CFI knockdown and CAMK2D overexpression models were constructed In vitro and vivo. As assayed by the Flow cytometry, knockdown of CFI in the ARPE-19 cells increased apoptosis induced by NaIO3 and overexpression of CAMK2D reduced the apoptosis induced by NaIO3 in ARPE-19 cells of CFI knockdown. Western blotting indicated that CAMK2D can regulate CFI expression. Overexpression CAMK2D in RPE-Choroid tissue can increase CFI expression. OCT examination revealed that CFI knockdown make retinal thickness thinning induced by NaIO3 and overexpression of CAMK2D can increased retinal thickness in NaIO3-induced mice of CFI knockdown. ERG indicated that CFI knockdown make retinal function more severe damage induced by NaIO3 and overexpression of CAMK2D can alleviate this retinal dysfunction in NaIO3-induced mice of CFI knockdown. The above results demonstrated that overexpression of CAMK2D can attenuate retinal degeneration induced by NaIO3 in mice retina of CFI knockdown. 
In conclusion, our study indicates that CAMK2D can attenuate the retinal degeneration induced by NaIO3, which may be achieved by regulating the CFI. But there still needs a lot of follow-up experiments to verify. Our results provide new insights into potential therapies for AMD. 
Acknowledgments
The authors thank Jianhua Huang for providing experimental technical guidance. 
Disclosure: W. Xu, None; L. Cao, None; H. Liu, None 
References
Ding JD, Johnson LV, Herrmann R, et al. Anti-amyloid therapy protects against retinal pigmented epithelium damage and vision loss in a model of age-related macular degeneration. Proc Natl Acad Sci USA. 2011; 108(28): E279–E287. [CrossRef] [PubMed]
Guymer RH, Chen FK, Hodgson LAB, et al. Subthreshold nanosecond laser in age-related macular degeneration: observational extension study of the LEAD Clinical Trial. Ophthalmol Retina. 2021; 5: 1196–1203. [CrossRef] [PubMed]
Gehrs KM, Anderson DH, Johnson LV, et al. Age-related macular degeneration–emerging pathogenetic and therapeutic concepts. Ann Med. 2006; 38: 450–471. [CrossRef] [PubMed]
Guo X, Zhou J, Starr C, et al. Preservation of vision after CaMKII-mediated protection of retinal ganglion cells. Cell. 2021; 184: 4299–4314.e12. [CrossRef] [PubMed]
Berridge MJ, Lipp P, Bootman MD. The versatility and universality of calcium signalling. Nat Rev Mol Cell Biol. 2000; 1: 11–21. [CrossRef] [PubMed]
Chi M, Evans H, Gilchrist J, et al. Phosphorylation of calcium/calmodulin-stimulated protein kinase II at T286 enhances invasion and migration of human breast cancer cells. Sci Rep. 2016; 6: 33132. [CrossRef] [PubMed]
Hudmon A, Schulman H. Neuronal CA2+/calmodulin-dependent protein kinase II: the role of structure and autoregulation in cellular function. Ann Rev Biochem. 2002; 71: 473–510. [CrossRef] [PubMed]
Salas MA, Valverde CA, Sánchez G, et al. The signalling pathway of CaMKII-mediated apoptosis and necrosis in the ischemia/reperfusion injury. J Mol Cell Cardiol. 2010; 48: 1298–1306. [CrossRef] [PubMed]
Li J, Wang P, Yu S, et al. Calcium entry mediates hyperglycemia-induced apoptosis through Ca(2+)/calmodulin-dependent kinase II in retinal capillary endothelial cells. Mol Vis. 2012; 18: 2371–2379. [PubMed]
Ashraf S, Bell S, O'Leary C, et al. CAMKII as a therapeutic target for growth factor-induced retinal and choroidal neovascularization. JCI Insight. 2019; 4(6): e122442. [PubMed]
Cuello F, Lorenz K. Inhibition of cardiac CaMKII to cure heart failure: step by step towards translation? Basic Res Cardiol. 2016; 111(6): 66. [CrossRef] [PubMed]
Mollova MY, Katus HA, Backs J. Regulation of CaMKII signaling in cardiovascular disease. Front Pharmacol. 2015; 6: 178. [CrossRef] [PubMed]
Toussaint F, Charbel C, Allen BG, et al. Vascular CaMKII: heart and brain in your arteries. Am J Physiol Cell Physiol. 2016; 311(3): C462–C478. [CrossRef] [PubMed]
Luo X, Gu S, Zhang Y, et al. Kinsenoside ameliorates oxidative stress-induced RPE cell apoptosis and inhibits angiogenesis via Erk/p38/NF-κB/VEGF signaling. Front Pharmacol. 2018; 9: 240. [CrossRef] [PubMed]
Tawfik A, Elsherbiny NM, Zaidi Y, et al. Homocysteine and age-related central nervous system diseases: role of inflammation. Int J Mol Sci. 2021; 22: 6259. [CrossRef] [PubMed]
Samelska K, Kupis M, Izdebska J, et al. Novel approach to antiangiogenic factors in age-related macular degeneration therapy. Cent Eur J Immunol. 2022; 47: 117–123. [CrossRef] [PubMed]
Vetreno RP, Qin L, Coleman LG, Jr. et al. Increased toll-like receptor-MyD88-NFκB-Proinflammatory neuroimmune signaling in the orbitofrontal cortex of humans with alcohol use disorder. Alcohol Clin Exp Res. 2021; 45: 1747–1761. [CrossRef] [PubMed]
Liu C, Liu Y, Yu Y, et al. Identification of up-regulated ANXA3 resulting in fracture non-union in patients with T2DM. Front Endocrinol. 2022; 13: 890941. [CrossRef]
Zhang Y, Goodfellow RX, Ghiringhelli Borsa N, et al. Complement factor I variants in complement-mediated renal diseases. Front Immunol. 2022; 13: 866330. [CrossRef] [PubMed]
Cai C, Zhou K, Jing J, et al. Confirmation of the predictive function of cuproptosis-related gene FDX1 in clear cell renal carcinoma using qRT-PCR and western blotting. Aging. 2023; 15: 6117–6134. [CrossRef] [PubMed]
Yi X, Wu L, Liu J, et al. Low-intensity pulsed ultrasound protects subchondral bone in rabbit temporomandibular joint osteoarthritis by suppressing TGF-β1/Smad3 pathway. J Orthop Res. 2020; 38: 2505–2512. [CrossRef] [PubMed]
Aung KH, Liu H, Ke Z, et al. Glabridin attenuates the retinal degeneration induced by sodium iodate in vitro and in vivo. Front Pharmacol. 2020; 11: 566699. [CrossRef] [PubMed]
Su F, Yang H, Guo A, et al. Mitochondrial BK(Ca) mediates the protective effect of low-dose ethanol preconditioning on oxygen-glucose deprivation and reperfusion-induced neuronal apoptosis. Front Physiol. 2021; 12: 719753. [CrossRef] [PubMed]
Wang P, Liang T, Zhan H, et al. Unique metabolism and protein expression signature in human decidual NK cells. Front Immunol. 2023; 14: 1136652. [CrossRef] [PubMed]
Qiu Q, Lei X, Wang Y, et al. Naringin protects against Tau hyperphosphorylation in Aβ (25–35)-injured PC12 cells through modulation of ER, PI3K/AKT, and GSK-3β signaling pathways. Behav Neurol. 2023; 2023: 1857330. [CrossRef] [PubMed]
Yao J, Wu XY, Yu Q, et al. The requirement of phosphoenolpyruvate carboxykinase 1 for angiogenesis in vitro and in vivo. Sci Adv. 2022; 8(21): eabn6928. [CrossRef] [PubMed]
Han S, Chen J, Hua J, et al. MITF protects against oxidative damage-induced retinal degeneration by regulating the NRF2 pathway in the retinal pigment epithelium. Redox Biol. 2020; 34: 101537. [CrossRef] [PubMed]
Tsai HY, Lai H, Chen ZY, et al. Inhibition of DUSP6 activates autophagy and rescues the retinal pigment epithelium in sodium iodate-induced retinal degeneration models in vivo and in vitro. Biomedicines. 2022; 10(1): 159. [CrossRef] [PubMed]
Zhan Z, Wu Y, Liu Z, et al. Reduced dendritic spines in the visual cortex contralateral to the optic nerve crush eye in adult mice. Invest Ophthalmol Vis Sci. 2020; 61(10): 55. [CrossRef] [PubMed]
Zhang H, Luo Q, Lu X, et al. Effects of hPMSCs on granulosa cell apoptosis and AMH expression and their role in the restoration of ovary function in premature ovarian failure mice. Stem Cell Res Ther. 2018; 9(1): 20. [CrossRef] [PubMed]
Wu S, Chang G, Gao L, et al. Trimetazidine protects against myocardial ischemia/reperfusion injury by inhibiting excessive autophagy. J Mol Med. 2018; 96: 791–806. [CrossRef] [PubMed]
Liu W, Lu J-Y, Wang Y-J, et al. Vaccinia virus induces EMT-like transformation and RhoA-mediated mesenchymal migration. J Med Virol. 2023; 95(8): e29041. [CrossRef] [PubMed]
Fan W, Li X, Cooper NG. CaMKIIalphaB mediates a survival response in retinal ganglion cells subjected to a glutamate stimulus. Invest Ophthalmol Vis Sci. 2007; 48: 3854–3863. [CrossRef] [PubMed]
Laabich A, Li G, Cooper NG. Calcium/calmodulin-dependent protein kinase II containing a nuclear localizing signal is altered in retinal neurons exposed to N-methyl-D-aspartate. Brain Res Mol Brain Res. 2000; 76: 253–265. [CrossRef] [PubMed]
Zhu X, Wang K, Zhou F, et al. Paeoniflorin attenuates atRAL-induced oxidative stress, mitochondrial dysfunction and endoplasmic reticulum stress in retinal pigment epithelial cells via triggering Ca/CaMKII-dependent activation of AMPK. Arch Pharm Res. 2018; 41: 1009–1018. [CrossRef] [PubMed]
Liu Y, Lv Z, Xiao T, et al. Functional and expressional analyses reveal the distinct role of complement factor I in regulating complement system activation during GCRV infection in Ctenopharyngodon idella. Int J Mol Sci. 2022; 23(19): 11369. [CrossRef] [PubMed]
Frederick PA, Kleinman ME. The immune system and AMD. Curr Ophthalmol Rep. 2014; 2: 14–19. [CrossRef] [PubMed]
Zheng R, Zhang Y, Zhang K, et al. The complement system, aging, and aging-related diseases. Int J Mol Sci. 2022; 23: 8689. [CrossRef] [PubMed]
Fishelson Z, Attali G, Mevorach D. Complement and apoptosis. Mol Immunol. 2001, 38(2–3): 207–219. [PubMed]
Armento A, Ueffing M, Clark SJ. The complement system in age-related macular degeneration. Cell Mol Life Sci. 2021; 78: 4487–4505. [CrossRef] [PubMed]
Hallam TM, Sharp SJ, Andreadi A, et al. Complement factor I: regulatory nexus, driver of immunopathology, and therapeutic. Immunobiology. 2023; 228(5): 152410. [CrossRef] [PubMed]
Hallam TM, Marchbank KJ, Harris CL, et al. Rare genetic variants in complement factor I lead to low FI plasma levels resulting in increased risk of age-related macular degeneration. Invest Ophthalmol Vis Sci. 2020; 61(6): 18. [CrossRef] [PubMed]
Figure 1.
 
CAMK2D expression in the dry AMD. (A) Volcano plot showing 825 significant DEGs of GSE125564 (Adjusted P value < 0.05, │log FC│ > 1). (B) The molecular functions terms in GO of downregulated genes by PANTHER. (C) CAMK2D protein level in RPE-choroid tissue. (D) Quantification based on (C) (n = 3, **P < 0.01).
Figure 1.
 
CAMK2D expression in the dry AMD. (A) Volcano plot showing 825 significant DEGs of GSE125564 (Adjusted P value < 0.05, │log FC│ > 1). (B) The molecular functions terms in GO of downregulated genes by PANTHER. (C) CAMK2D protein level in RPE-choroid tissue. (D) Quantification based on (C) (n = 3, **P < 0.01).
Figure 2.
 
Flow cytometry analysis of apoptosis of ARPE‐19 cells. (A) About knockdown of CAMK2D, the percentage of cells in each quadrant is presented. (B) The total apoptosis rate in four groups were statistically analyzed based on (A). (C) About overexpression of CAMK2D, the percentage of cells in each quadrant is presented. (D) The total apoptosis rate in four groups were statistically analyzed based on (C). (Q2 represents late apoptosis, Q3 represents early apoptosis, Q2 + Q3 represents total apoptosis. n = 3 per group, ns: P > 0.05, **P < 0.01, ****P < 0.0001).
Figure 2.
 
Flow cytometry analysis of apoptosis of ARPE‐19 cells. (A) About knockdown of CAMK2D, the percentage of cells in each quadrant is presented. (B) The total apoptosis rate in four groups were statistically analyzed based on (A). (C) About overexpression of CAMK2D, the percentage of cells in each quadrant is presented. (D) The total apoptosis rate in four groups were statistically analyzed based on (C). (Q2 represents late apoptosis, Q3 represents early apoptosis, Q2 + Q3 represents total apoptosis. n = 3 per group, ns: P > 0.05, **P < 0.01, ****P < 0.0001).
Figure 3.
 
Overexpression or knockdown of CAMK2D did not alter the retinal structure and function in normal mice. (A) Retinas expressing green fluorescent protein (GFP) were measured through Micron IV retinal imaging camera system. (B) Western blotting of CAMK2D after the injection of AAV9-CAMK2D vector. (C) Quantification based on (B). (D) Western blotting of CAMK2D after the injection of AAV9-shRNA-CAMK2D vector. (E) Quantification based on (D). (n = 3; *P < 0.05, **P < 0.01). (F) Fundus camera showed no significant changes in retinal structure after the AAV9 injection. (G) OCT showed no significant changes in retinal structure after the AAV9 injection. (H) H&E staining showed no significant changes in retinal structure after the AAV9 injection. (IK) ERG showed no significant differences of a-wave and b-wave after AAV9 vector injection. (n = 5; ns: P > 0.05).
Figure 3.
 
Overexpression or knockdown of CAMK2D did not alter the retinal structure and function in normal mice. (A) Retinas expressing green fluorescent protein (GFP) were measured through Micron IV retinal imaging camera system. (B) Western blotting of CAMK2D after the injection of AAV9-CAMK2D vector. (C) Quantification based on (B). (D) Western blotting of CAMK2D after the injection of AAV9-shRNA-CAMK2D vector. (E) Quantification based on (D). (n = 3; *P < 0.05, **P < 0.01). (F) Fundus camera showed no significant changes in retinal structure after the AAV9 injection. (G) OCT showed no significant changes in retinal structure after the AAV9 injection. (H) H&E staining showed no significant changes in retinal structure after the AAV9 injection. (IK) ERG showed no significant differences of a-wave and b-wave after AAV9 vector injection. (n = 5; ns: P > 0.05).
Figure 4.
 
Overexpression of CAMK2D in RPE attenuated NaIO3-induced retinal degeneration and knockdown exacerbated NaIO3-induced retinal degeneration. (A) Fundus camera (white arrows indicate foci of yellow-white exudation). (B) OCT (black arrow indicates the OCT imaging location; red arrows indicate bulging deposits like drusen. Scale bar: 100 µm.). (C) H&E staining (green arrows indicate bulging deposits. Scale bar: 50 µm.). (D) OCT detecting retinal thickness (black circular arrow indicates the OCT imaging location. Scale bar: 100 µm.). (E) The retinal thickness quantitative analysis based on (D). (n = 5; ****P < 0.0001). (F) ERG results. (G) The a-wave quantitative analysis based on (F). (H) The b-wave quantitative analysis based on (F) (n = 5; **P < 0.01, ***P < 0.001, ****P < 0.0001). (I) TUNEL results indicated retina apoptotic cells (white arrows indicate apoptotic cells. Scale bar: 50 µm. Below is the organizational structure of partial enlargement of the green rectangle in above.). (J) Quantitative analysis the number of apoptotic cells based on of (I) (n = 5; **P < 0.01, ****P < 0.0001).
Figure 4.
 
Overexpression of CAMK2D in RPE attenuated NaIO3-induced retinal degeneration and knockdown exacerbated NaIO3-induced retinal degeneration. (A) Fundus camera (white arrows indicate foci of yellow-white exudation). (B) OCT (black arrow indicates the OCT imaging location; red arrows indicate bulging deposits like drusen. Scale bar: 100 µm.). (C) H&E staining (green arrows indicate bulging deposits. Scale bar: 50 µm.). (D) OCT detecting retinal thickness (black circular arrow indicates the OCT imaging location. Scale bar: 100 µm.). (E) The retinal thickness quantitative analysis based on (D). (n = 5; ****P < 0.0001). (F) ERG results. (G) The a-wave quantitative analysis based on (F). (H) The b-wave quantitative analysis based on (F) (n = 5; **P < 0.01, ***P < 0.001, ****P < 0.0001). (I) TUNEL results indicated retina apoptotic cells (white arrows indicate apoptotic cells. Scale bar: 50 µm. Below is the organizational structure of partial enlargement of the green rectangle in above.). (J) Quantitative analysis the number of apoptotic cells based on of (I) (n = 5; **P < 0.01, ****P < 0.0001).
Figure 5.
 
(A) DEGs of overexpression CAMK2D in ARPE-19 cells were shown in a volcano plot (P < 0.05, |log2FC| > log2(1.8)). (B) Venn diagrams show overlapping genes between disease genes of AMD and the DEGs. (C) Western blotting of CFI in ARPE-19 cells of overexpression CAMK2D. (D) Quantification based on (C). (E) Western blotting of CFI in ARPE-19 cells of knockdown CAMK2D. (F) Quantification based on (E) (n = 3 per group. ns: P > 0.05, *P < 0.05, **P < 0.01, ***P < 0.001).
Figure 5.
 
(A) DEGs of overexpression CAMK2D in ARPE-19 cells were shown in a volcano plot (P < 0.05, |log2FC| > log2(1.8)). (B) Venn diagrams show overlapping genes between disease genes of AMD and the DEGs. (C) Western blotting of CFI in ARPE-19 cells of overexpression CAMK2D. (D) Quantification based on (C). (E) Western blotting of CFI in ARPE-19 cells of knockdown CAMK2D. (F) Quantification based on (E) (n = 3 per group. ns: P > 0.05, *P < 0.05, **P < 0.01, ***P < 0.001).
Figure 6.
 
(A) Western blotting of CFI in ARPE-19 cells of CFI knockdown. (B). Quantification based on (A) (n = 3. ns: P > 0.05, **P < 0.01). (C) The total apoptosis rates were statistically analyzed based on (E) the Annexin V-FITC Apoptosis Detection Kit with PI. (D) The total apoptosis rates were statistically analyzed based on (F) the Annexin V-PE Apoptosis Detection Kit with 7AAD. (E) The percentage of cells in each quadrant is presented on the Annexin V-FITC Apoptosis Detection Kit with PI. (F) The percentage of cells in each quadrant is presented on the Annexin V-PE Apoptosis Detection Kit with 7AAD (n = 3. ns: P > 0.05, **P < 0.01, ****P < 0.0001). (G) Western blotting of CFI in RPE-Choroidal tissue of mice. (H) Quantification based on (G) (n = 3. ns: P > 0.05, *P < 0.05, **P < 0.01, ***P < 0.001). (I) The retinal thickness quantitative analysis based on (L). (n = 5; **P < 0.01, ****P < 0.0001). (J) The a-wave quantitative analysis based on (M). (K) The b-wave quantitative analysis based on (M) (n = 5; *P < 0.05, **P < 0.01, ****P < 0.0001). (L) OCT detecting retinal thickness (Scale bar: 100 µm.). (M) ERG results.
Figure 6.
 
(A) Western blotting of CFI in ARPE-19 cells of CFI knockdown. (B). Quantification based on (A) (n = 3. ns: P > 0.05, **P < 0.01). (C) The total apoptosis rates were statistically analyzed based on (E) the Annexin V-FITC Apoptosis Detection Kit with PI. (D) The total apoptosis rates were statistically analyzed based on (F) the Annexin V-PE Apoptosis Detection Kit with 7AAD. (E) The percentage of cells in each quadrant is presented on the Annexin V-FITC Apoptosis Detection Kit with PI. (F) The percentage of cells in each quadrant is presented on the Annexin V-PE Apoptosis Detection Kit with 7AAD (n = 3. ns: P > 0.05, **P < 0.01, ****P < 0.0001). (G) Western blotting of CFI in RPE-Choroidal tissue of mice. (H) Quantification based on (G) (n = 3. ns: P > 0.05, *P < 0.05, **P < 0.01, ***P < 0.001). (I) The retinal thickness quantitative analysis based on (L). (n = 5; **P < 0.01, ****P < 0.0001). (J) The a-wave quantitative analysis based on (M). (K) The b-wave quantitative analysis based on (M) (n = 5; *P < 0.05, **P < 0.01, ****P < 0.0001). (L) OCT detecting retinal thickness (Scale bar: 100 µm.). (M) ERG results.
×
×

This PDF is available to Subscribers Only

Sign in or purchase a subscription to access this content. ×

You must be signed into an individual account to use this feature.

×