Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Adv Exp Med Biol ; 1415: 353-358, 2023.
Article in English | MEDLINE | ID: mdl-37440056

ABSTRACT

The cone photoreceptor cyclic nucleotide-gated (CNG) channel plays a pivotal role in cone phototransduction. Mutations in genes encoding the channel subunits CNGA3 and CNGB3 account for about 80% of all cases of achromatopsia and are associated with progressive cone dystrophies. CNG channel deficiency leads to cellular/endoplasmic reticulum (ER) calcium dysregulation and ER stress-associated cone apoptosis. This work investigated the role of the ER calcium channel ryanodine receptor 1 (Ryr1) in ER stress and cone degeneration in CNG channel deficiency. The AAV-mediated CRISPR/SaCas9 genome editing was used to knock down Ryr1 specifically in cones. CNG channel-deficient mice displayed improved cone survival after subretinal injection of AAV2-SaCas9/gRNA-Ryr1, manifested as increased expression levels of cone proteins M-opsin, S-opsin, and cone arrestin. Knockdown of Ryr1 also led to reduced ER stress and increased expression levels of the ER-associated degradation proteins. This work demonstrates a role of Ryr1 in ER stress and cone degeneration in CNG channel deficiency, and supports strategies targeting ER calcium regulation for cone preservation.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels , Ryanodine Receptor Calcium Release Channel , Animals , Mice , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Calcium/metabolism , Proteolysis , Retinal Cone Photoreceptor Cells/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Opsins/genetics , Nucleotides, Cyclic/metabolism
2.
FASEB J ; 35(6): e21579, 2021 06.
Article in English | MEDLINE | ID: mdl-33960001

ABSTRACT

Endoplasmic reticulum (ER) Ca2+ homeostasis relies on an appropriate balance between efflux- and influx-channel activity responding to dynamic changes of intracellular Ca2+ levels. Dysregulation of this complex signaling network has been shown to contribute to neuronal and photoreceptor death in neuro- and retinal degenerative diseases, respectively. In mice with cone cyclic nucleotide-gated (CNG) channel deficiency, a model of achromatopsia/cone dystrophy, cones display early-onset ER stress-associated apoptosis and protein mislocalization. Cones in these mice also show reduced cytosolic Ca2+ level and subsequent elevation in the ER Ca2+ -efflux-channel activity, specifically the inositol-1,4,5-trisphosphate receptor type 1 (IP3 R1), and deletion of IP3 R1 results in preservation of cones. This work investigated how preservation of ER Ca2+ stores leads to cone protection. We examined the effects of cone specific deletion of IP3 R1 on ER stress responses/cone death, protein localization, and ER proteostasis/ER-associated degradation. We demonstrated that deletion of IP3 R1 improves trafficking of cone-specific proteins M-/S-opsin and phosphodiesterase 6C to cone outer segments and reduces localization to cone inner segments. Consistent with the improved protein localization, deletion of IP3 R1 results in increased ER retrotranslocation protein expression, reduced proteasome subunit expression, reduced ER stress/cone death, and reduced retinal remodeling. We also observed the enhanced ER retrotranslocation in mice that have been treated with a chemical chaperone, supporting the connection between improved ER retrotranslocation/proteostasis and alleviation of ER stress. Findings from this work demonstrate the importance of ER Ca2+ stores in ER proteostasis and protein trafficking/localization in photoreceptors, strengthen the link between dysregulation of ER Ca2+ homeostasis and ER stress/cone degeneration, and support an involvement of improved ER proteostasis in ER Ca2+ preservation-induced cone protection; thereby identifying IP3 R1 as a critical mediator of ER stress and protein mislocalization and as a potential target to preserve cones in CNG channel deficiency.


Subject(s)
Calcium/metabolism , Cyclic Nucleotide-Gated Cation Channels/deficiency , Endoplasmic Reticulum/pathology , Inositol 1,4,5-Trisphosphate Receptors/physiology , Proteostasis , Retina/pathology , Animals , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Transport , Retina/metabolism , Signal Transduction
3.
FASEB J ; 34(5): 6335-6350, 2020 05.
Article in English | MEDLINE | ID: mdl-32173907

ABSTRACT

Photoreceptor cyclic nucleotide-gated (CNG) channels regulate Ca2+ influx in rod and cone photoreceptors. Mutations in cone CNG channel subunits CNGA3 and CNGB3 are associated with achromatopsia and cone dystrophies. Mice lacking functional cone CNG channel show endoplasmic reticulum (ER) stress-associated cone degeneration. The elevated cyclic guanosine monophosphate (cGMP)/cGMP-dependent protein kinase (PKG) signaling and upregulation of the ER Ca2+ channel ryanodine receptor 2 (RyR2) have been implicated in cone degeneration. This work investigates the potential contribution of RyR2 to cGMP/PKG signaling-induced ER stress and cone degeneration. We demonstrated that the expression and activity of RyR2 were highly regulated by cGMP/PKG signaling. Depletion of cGMP by deleting retinal guanylate cyclase 1 or inhibition of PKG using chemical inhibitors suppressed the upregulation of RyR2 in CNG channel deficiency. Depletion of cGMP or deletion of Ryr2 equivalently inhibited unfolded protein response/ER stress, activation of the CCAAT-enhancer-binding protein homologous protein, and activation of the cyclic adenosine monophosphate response element-binding protein, leading to early-onset cone protection. In addition, treatment with cGMP significantly enhanced Ryr2 expression in cultured photoreceptor-derived Weri-Rb1 cells. Findings from this work demonstrate the regulation of cGMP/PKG signaling on RyR2 in the retina and support the role of RyR2 upregulation in cGMP/PKG signaling-induced ER stress and photoreceptor degeneration.


Subject(s)
Cyclic GMP/metabolism , Endoplasmic Reticulum Stress , Proto-Oncogene Proteins c-akt/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/pathology , Ryanodine Receptor Calcium Release Channel/physiology , Animals , Basic-Leucine Zipper Transcription Factors/physiology , Cyclic Nucleotide-Gated Cation Channels/physiology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Eye Proteins/physiology , Guanylate Cyclase/physiology , Mice , Mice, Knockout , Receptors, Cell Surface/physiology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/etiology , Retinal Degeneration/metabolism , Signal Transduction , Unfolded Protein Response
4.
Int J Mol Sci ; 22(1)2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33374621

ABSTRACT

Mutations in the CNGA3 gene, which encodes the A subunit of the cyclic guanosine monophosphate (cGMP)-gated cation channel in cone photoreceptor outer segments, cause total colour blindness, also referred to as achromatopsia. Cones lacking this channel protein are non-functional, accumulate high levels of the second messenger cGMP and degenerate over time after induction of ER stress. The cell death mechanisms that lead to loss of affected cones are only partially understood. Here, we explored the disease mechanisms in the Cnga3 knockout (KO) mouse model of achromatopsia. We found that another important effector of cGMP, the cGMP-dependent protein kinase 2 (Prkg2) is crucially involved in cGMP cytotoxicity of cones in Cnga3 KO mice. Virus-mediated knockdown or genetic ablation of Prkg2 in Cnga3 KO mice counteracted degeneration and preserved the number of cones. Analysis of markers of endoplasmic reticulum stress and unfolded protein response confirmed that induction of these processes in Cnga3 KO cones also depends on Prkg2. In conclusion, we identified Prkg2 as a novel key mediator of cone photoreceptor degeneration in achromatopsia. Our data suggest that this cGMP mediator could be a novel pharmacological target for future neuroprotective therapies.


Subject(s)
Color Vision Defects/etiology , Color Vision Defects/metabolism , Cyclic GMP-Dependent Protein Kinase Type II/metabolism , Cyclic Nucleotide-Gated Cation Channels/deficiency , Retinal Cone Photoreceptor Cells/metabolism , Animals , Biomarkers , Color Vision Defects/pathology , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Disease Models, Animal , Disease Susceptibility , Endoplasmic Reticulum Stress , Fluorescent Antibody Technique , Gene Expression , Mice , Mice, Knockout , Microscopy, Confocal , Models, Biological , Retinal Degeneration/etiology , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Unfolded Protein Response
5.
FASEB J ; : fj201800484RR, 2018 Jun 06.
Article in English | MEDLINE | ID: mdl-29874126

ABSTRACT

Thyroid hormone (TH) signaling has been shown to regulate cone photoreceptor viability. Suppression of TH signaling with antithyroid drug treatment or by targeting iodothyronine deiodinases and TH receptors preserves cones in mouse models of retinal degeneration, including the Leber congenital amaurosis Rpe65-deficient mice. This work investigates the cellular mechanisms underlying how suppressing TH signaling preserves cones in Rpe65-deficient mice, using mice deficient in type 2 iodothyronine deiodinase (Dio2), the enzyme that converts the prohormone thyroxine to the active hormone triiodothyronine (T3). Deficiency of Dio2 improved cone survival and function in Rpe65-/- and Rpe65-deficiency on a cone dominant background ( Rpe65-/-/ Nrl-/-) mice. Analysis of cell death pathways revealed that receptor-interacting serine/threonine-protein kinase (RIPK)/necroptosis activity was increased in Rpe65-/-/ Nrl-/- retinas, and Dio2 deficiency reversed the alterations. Cell-stress analysis showed that the cellular oxidative stress responses were increased in Rpe65-/-/ Nrl-/- retinas, and Dio2 deficiency abolished the elevations. Similarly, antithyroid drug treatment resulted in reduced RIPK/necroptosis activity and oxidative stress responses in Rpe65-/-/ Nrl-/- retinas. Moreover, treatment with T3 significantly induced RIPK/necroptosis activity and oxidative stress responses in the retina. This work shows that suppression of TH signaling reduces cellular RIPK/necroptosis activity and oxidative stress responses in degenerating retinas, suggesting a mechanism underlying the observed cone preservation.-Yang, F., Ma, H., Butler, M. R., Ding, X.-Q. Deficiency of type 2 iodothyronine deiodinase reduces necroptosis activity and oxidative stress responses in retinas of Leber congenital amaurosis model mice.

6.
J Biol Chem ; 292(27): 11189-11205, 2017 07 07.
Article in English | MEDLINE | ID: mdl-28495882

ABSTRACT

Endoplasmic reticulum (ER) stress and mislocalization of improperly folded proteins have been shown to contribute to photoreceptor death in models of inherited retinal degenerative diseases. In particular, mice with cone cyclic nucleotide-gated (CNG) channel deficiency, a model for achromatopsia, display both early-onset ER stress and opsin mistrafficking. By 2 weeks of age, these mice show elevated signaling from all three arms of the ER-stress pathway, and by 1 month, cone opsin is improperly distributed away from its normal outer segment location to other retinal layers. This work investigated the role of Ca2+-release channels in ER stress, protein mislocalization, and cone death in a mouse model of CNG-channel deficiency. We examined whether preservation of luminal Ca2+ stores through pharmacological and genetic suppression of ER Ca2+ efflux protects cones by attenuating ER stress. We demonstrated that the inhibition of ER Ca2+-efflux channels reduced all three arms of ER-stress signaling while improving opsin trafficking to cone outer segments and decreasing cone death by 20-35%. Cone-specific gene deletion of the inositol-1,4,5-trisphosphate receptor type I (IP3R1) also significantly increased cone density in the CNG-channel-deficient mice, suggesting that IP3R1 signaling contributes to Ca2+ homeostasis and cone survival. Consistent with the important contribution of organellar Ca2+ signaling in this achromatopsia mouse model, significant differences in dynamic intraorganellar Ca2+ levels were detected in CNG-channel-deficient cones. These results thus identify a novel molecular link between Ca2+ homeostasis and cone degeneration, thereby revealing novel therapeutic targets to preserve cones in inherited retinal degenerative diseases.


Subject(s)
Calcium Signaling , Color Vision Defects/metabolism , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Ion Channel Gating , Retinal Cone Photoreceptor Cells/metabolism , Animals , Cell Death/genetics , Cell Survival , Color Vision Defects/genetics , Disease Models, Animal , Endoplasmic Reticulum/genetics , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mice , Mice, Knockout , Retinal Cone Photoreceptor Cells/pathology
7.
FASEB J ; 31(8): 3425-3438, 2017 08.
Article in English | MEDLINE | ID: mdl-28428265

ABSTRACT

Thyroid hormone (TH) signaling regulates cell proliferation, differentiation, and metabolism. Recent studies have implicated TH signaling in cone photoreceptor viability. Using mouse models of retinal degeneration, we demonstrated that antithyroid drug treatment and targeting iodothyronine deiodinases (DIOs) to suppress cellular tri-iodothyronine (T3) production or increase T3 degradation preserves cones. In this work, we investigated the effectiveness of inhibition of the TH receptor (TR). Two genes, THRA and THRB, encode TRs; THRB2 has been associated with cone viability. Using TR antagonists and Thrb2 deletion, we examined the effects of TR inhibition. Systemic and ocular treatment with the TR antagonists NH-3 and 1-850 increased cone density by 30-40% in the Rpe65-/- mouse model of Leber congenital amaurosis and reduced the number of TUNEL+ cells. Cone survival was significantly improved in Rpe65-/- and Cpfl1 (a model of achromatopsia with Pde6c defect) mice with Thrb2 deletion. Ventral cone density in Cpfl1/Thrb2-/- and Rpe65-/- /Thrb2-/- mice was increased by 1- to 4-fold, compared with age-matched controls. Moreover, the expression levels of TR were significantly higher in the cone-degeneration retinas, suggesting locally elevated TR signaling. This work shows that the effects of antithyroid treatment or targeting DIOs were likely mediated by TRs and that suppressing TR protects cones. Our findings support the view that inhibition of TR locally in the retina is a therapeutic strategy for retinal degeneration management.-Ma, H., Yang, F., Butler, M. R., Belcher, J., Redmond, T. M., Placzek, A. T., Scanlan, T. S., Ding, X.-Q. Inhibition of thyroid hormone receptor locally in the retina is a therapeutic strategy for retinal degeneration.


Subject(s)
Antithyroid Agents/pharmacology , Methimazole/pharmacology , Receptors, Thyroid Hormone/antagonists & inhibitors , Retina/metabolism , Retinal Degeneration/drug therapy , Animals , Antithyroid Agents/therapeutic use , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Death , Disease Models, Animal , Eye Proteins/genetics , Eye Proteins/metabolism , Gene Deletion , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Methimazole/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenoxyacetates/pharmacology , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinoblastoma , Triiodothyronine , cis-trans-Isomerases/genetics , cis-trans-Isomerases/metabolism
8.
Adv Exp Med Biol ; 1074: 125-131, 2018.
Article in English | MEDLINE | ID: mdl-29721936

ABSTRACT

Leber congenital amaurosis (LCA) is a devastating pediatric retinal degenerative disease, accounting for 20% of blindness in children attending schools for the blind. Mutations in the RPE65 gene, which encodes the retinal pigment epithelium-specific isomerohydrolase RPE65, account for 16% of all LCA cases. Recent findings have linked cone photoreceptor viability to thyroid hormone (TH) signaling. TH signaling regulates cell proliferation, differentiation, and metabolism. At the cellular level, TH action is regulated by the two iodothyronine deiodinases, DIO2 and DIO3. DIO2 converts the prohormone thyroxine (T4) to the bioactive hormone triiodothyronine (T3), and DIO3 inactivates T3 and T4. The present work investigates the effects of overexpression of DIO3 to suppress TH signaling and thereby modulate cone death/survival. Subretinal delivery of AAV5-IRBP/GNAT2-hDIO3 induced robust expression of DIO3 in the mouse retina and significantly reduced the number of TUNEL-positive cells in the cone-dominant LCA model Rpe65 -/- /Nrl -/- mice. Our work shows that suppressing TH signaling by overexpression of DIO3 preserves cones, supporting that suppressing TH signaling locally in the retina may represent a treatment strategy for LCA management.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/therapeutic use , Iodide Peroxidase/therapeutic use , Leber Congenital Amaurosis/therapy , Retinal Cone Photoreceptor Cells/enzymology , cis-trans-Isomerases/deficiency , Animals , Apoptosis , Basic-Leucine Zipper Transcription Factors/deficiency , Eye Proteins/genetics , Gene Expression , Genes, Synthetic , Genetic Vectors/administration & dosage , Heterotrimeric GTP-Binding Proteins/genetics , Injections, Intraocular , Iodide Peroxidase/biosynthesis , Iodide Peroxidase/genetics , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/pathology , Mice , Mice, Knockout , Mutation , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Retinal Cone Photoreceptor Cells/pathology , Retinol-Binding Proteins/genetics , Thyroid Hormones/metabolism , Transduction, Genetic
9.
J Biol Chem ; 291(16): 8721-34, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26893377

ABSTRACT

Cone photoreceptor cyclic nucleotide-gated (CNG) channels play a pivotal role in cone phototransduction, which is a process essential for daylight vision, color vision, and visual acuity. Mutations in the cone channel subunits CNGA3 and CNGB3 are associated with human cone diseases, including achromatopsia, cone dystrophies, and early onset macular degeneration. Mutations in CNGB3 alone account for 50% of reported cases of achromatopsia. This work investigated the role of CNGB3 in cone light response and cone channel structural stability. As cones comprise only 2-3% of the total photoreceptor population in the wild-type mouse retina, we used Cngb3(-/-)/Nrl(-/-) mice with CNGB3 deficiency on a cone-dominant background in our study. We found that, in the absence of CNGB3, CNGA3 was able to travel to the outer segments, co-localize with cone opsin, and form tetrameric complexes. Electroretinogram analyses revealed reduced cone light response amplitude/sensitivity and slower response recovery in Cngb3(-/-)/Nrl(-/-) mice compared with Nrl(-/-) mice. Absence of CNGB3 expression altered the adaptation capacity of cones and severely compromised function in bright light. Biochemical analysis demonstrated that CNGA3 channels lacking CNGB3 were more resilient to proteolysis than CNGA3/CNGB3 channels, suggesting a hindered structural flexibility. Thus, CNGB3 regulates cone light response kinetics and the channel structural flexibility. This work advances our understanding of the biochemical and functional role of CNGB3 in cone photoreceptors.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/metabolism , Light , Retinal Cone Photoreceptor Cells/metabolism , Animals , Cyclic Nucleotide-Gated Cation Channels/genetics , Humans , Mice , Mice, Knockout , Opsins/genetics , Opsins/metabolism , Retinal Cone Photoreceptor Cells/cytology
10.
Hum Mol Genet ; 24(13): 3699-707, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25855802

ABSTRACT

The CNGA3(-/-)/Nrl(-/-) mouse is a cone-dominant model with Cnga3 channel deficiency, which partially mimics the all cone foveal structure of human achromatopsia 2 with CNGA3 mutations. Although subretinal (SR) AAV vector administration can transfect retinal cells efficiently, the injection-induced retinal detachment can cause retinal damage, particularly when SR vector bleb includes the fovea. We therefore explored whether cone function-structure could be rescued in CNGA3(-/-)/Nrl(-/-) mice by intravitreal (IVit) delivery of tyrosine to phenylalanine (Y-F) capsid mutant AAV8. We find that AAV-mediated CNGA3 expression can restore cone function and rescue structure following IVit delivery of AAV8 (Y447, 733F) vector. Rescue was assessed by restoration of the cone-mediated electroretinogram (ERG), optomotor responses, and cone opsin immunohistochemistry. Demonstration of gene therapy in a cone-dominant mouse model by IVit delivery provides a potential alternative vector delivery mode for safely transducing foveal cones in achromatopsia patients and in other human retinal diseases affecting foveal function.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Color Vision Defects/genetics , Color Vision Defects/therapy , Cyclic Nucleotide-Gated Cation Channels/genetics , Eye Proteins/genetics , Genetic Therapy , Retinal Cone Photoreceptor Cells/physiology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Color Vision Defects/metabolism , Color Vision Defects/physiopathology , Cyclic Nucleotide-Gated Cation Channels/metabolism , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Eye Proteins/metabolism , Female , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
FASEB J ; 30(12): 4313-4325, 2016 12.
Article in English | MEDLINE | ID: mdl-27623928

ABSTRACT

Recent studies have implicated thyroid hormone (TH) signaling in cone photoreceptor viability. Using mouse models of retinal degeneration, we found that antithyroid treatment preserves cones. This work investigates the significance of targeting intracellular TH components locally in the retina. The cellular TH level is mainly regulated by deiodinase iodothyronine (DIO)-2 and -3. DIO2 converts thyroxine (T4) to triiodothyronine (T3), which binds to the TH receptor, whereas DIO3 degrades T3 and T4. We examined cone survival after overexpression of DIO3 and inhibition of DIO2 and demonstrated the benefits of these manipulations. Subretinal delivery of AAV5-IRBP/GNAT2-DIO3, which directs expression of human DIO3 specifically in cones, increased cone density by 30-40% in a Rpe65-/- mouse model of Lebers congenital amaurosis (LCA) and in a Cpfl1 mouse with Pde6c defect model of achromatopsia, compared with their respective untreated controls. Intravitreal and topical delivery of the DIO2 inhibitor iopanoic acid also significantly improved cone survival in the LCA model mice. Moreover, the expression levels of DIO2 and Slc16a2 were significantly higher in the diseased retinas, suggesting locally elevated TH signaling. We show that targeting DIOs protects cones, and intracellular inhibition of TH components locally in the retina may represent a novel strategy for retinal degeneration management.-Yang, F., Ma, H., Belcher, J., Butler, M. R., Redmond, T. M., Boye, S. L., Hauswirth, W. W., Ding, X.-Q. Targeting iodothyronine deiodinases locally in the retina is a therapeutic strategy for retinal degeneration.


Subject(s)
Iodide Peroxidase/metabolism , Retina/metabolism , Retinal Degeneration/metabolism , Animals , Cells, Cultured , Mice, Knockout , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/genetics , Signal Transduction/physiology , Thyroid Hormones/metabolism , Triiodothyronine/metabolism
12.
Proc Natl Acad Sci U S A ; 111(9): 3602-7, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24550448

ABSTRACT

Cone phototransduction and survival of cones in the human macula is essential for color vision and for visual acuity. Progressive cone degeneration in age-related macular degeneration, Stargardt disease, and recessive cone dystrophies is a major cause of blindness. Thyroid hormone (TH) signaling, which regulates cell proliferation, differentiation, and apoptosis, plays a central role in cone opsin expression and patterning in the retina. Here, we investigated whether TH signaling affects cone viability in inherited retinal degeneration mouse models. Retinol isomerase RPE65-deficient mice [a model of Leber congenital amaurosis (LCA) with rapid cone loss] and cone photoreceptor function loss type 1 mice (severe recessive achromatopsia) were used to determine whether suppressing TH signaling with antithyroid treatment reduces cone death. Further, cone cyclic nucleotide-gated channel B subunit-deficient mice (moderate achromatopsia) and guanylate cyclase 2e-deficient mice (LCA with slower cone loss) were used to determine whether triiodothyronine (T3) treatment (stimulating TH signaling) causes deterioration of cones. We found that cone density in retinol isomerase RPE65-deficient and cone photoreceptor function loss type 1 mice increased about sixfold following antithyroid treatment. Cone density in cone cyclic nucleotide-gated channel B subunit-deficient and guanylate cyclase 2e-deficient mice decreased about 40% following T3 treatment. The effect of TH signaling on cone viability appears to be independent of its regulation on cone opsin expression. This work demonstrates that suppressing TH signaling in retina dystrophy mouse models is protective of cones, providing insights into cone preservation and therapeutic interventions.


Subject(s)
Color Vision Defects/complications , Leber Congenital Amaurosis/complications , Retinal Cone Photoreceptor Cells/physiology , Retinal Degeneration/prevention & control , Signal Transduction/physiology , Thyroid Hormones/metabolism , Animals , Antithyroid Agents/pharmacology , Color Vision Defects/drug therapy , Cone Opsins/metabolism , Cyclic Nucleotide-Gated Cation Channels/deficiency , Guanylate Cyclase/deficiency , Leber Congenital Amaurosis/drug therapy , Methimazole , Mice , Mice, Knockout , Receptors, Cell Surface/deficiency , Retinal Cone Photoreceptor Cells/drug effects , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/etiology , Retinal Degeneration/physiopathology , Triiodothyronine/pharmacology , cis-trans-Isomerases/deficiency
13.
J Biol Chem ; 290(34): 20880-20892, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26124274

ABSTRACT

Photoreceptor cyclic nucleotide-gated (CNG) channels play a pivotal role in phototransduction. Mutations in the cone CNG channel subunits CNGA3 and CNGB3 are associated with achromatopsia and cone dystrophies. We have shown endoplasmic reticulum (ER) stress-associated apoptotic cone death and increased phosphorylation of the ER Ca(2+) channel inositol 1,4,5-trisphosphate receptor 1 (IP3R1) in CNG channel-deficient mice. We also presented a remarkable elevation of cGMP and an increased activity of the cGMP-dependent protein kinase (protein kinase G, PKG) in CNG channel deficiency. This work investigated whether cGMP/PKG signaling regulates ER stress and IP3R1 phosphorylation in CNG channel-deficient cones. Treatment with PKG inhibitor and deletion of guanylate cyclase-1 (GC1), the enzyme producing cGMP in cones, were used to suppress cGMP/PKG signaling in cone-dominant Cnga3(-/-)/Nrl(-/-) mice. We found that treatment with PKG inhibitor or deletion of GC1 effectively reduced apoptotic cone death, increased expression levels of cone proteins, and decreased activation of Müller glial cells. Furthermore, we observed significantly increased phosphorylation of IP3R1 and reduced ER stress. Our findings demonstrate a role of cGMP/PKG signaling in ER stress and ER Ca(2+) channel regulation and provide insights into the mechanism of cone degeneration in CNG channel deficiency.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide-Gated Cation Channels/genetics , Endoplasmic Reticulum Stress/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Animals , Apoptosis , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , Carbazoles/pharmacology , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic Nucleotide-Gated Cation Channels/deficiency , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Eye Proteins/genetics , Gene Expression Regulation , Guanylate Cyclase/deficiency , Guanylate Cyclase/genetics , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mice , Mice, Knockout , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Retinal Cone Photoreceptor Cells/cytology , Signal Transduction , Thionucleotides/pharmacology
14.
Adv Exp Med Biol ; 854: 613-8, 2016.
Article in English | MEDLINE | ID: mdl-26427466

ABSTRACT

Thyroid hormone (TH) signaling regulates cell proliferation, differentiation, and apoptosis. In the retina, TH signaling plays a central role in cone opsin expression. TH signaling inhibits S opsin expression, stimulates M opsin expression, and promotes dorsal-ventral opsin patterning. TH signaling has also been associated with cone photoreceptor viability. Treatment with thyroid hormone triiodothyronine (T3) or induction of high T3 by deleting the hormone-inactivating enzyme type 3 iodothyronine deiodinase (DIO3) causes cone death in mice. This effect is reversed by deletion of the TH receptor (TR) gene. Consistent with the T3 treatment effect, suppressing TH signaling preserves cones in mouse models of retinal degeneration. The regulation of cone survival by TH signaling appears to be independent of its regulatory role in cone opsin expression. The mechanism by which TH signaling regulates cone viability remains to be identified. The current understanding of TH signaling regulation in photoreceptor viability suggests that suppressing TH signaling locally in the retina may represent a novel strategy for retinal degeneration management.


Subject(s)
Apoptosis/physiology , Retinal Cone Photoreceptor Cells/metabolism , Signal Transduction/physiology , Thyroid Hormones/metabolism , Animals , Apoptosis/genetics , Cell Survival/genetics , Cell Survival/physiology , Humans , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Mice, Knockout , Models, Biological , Signal Transduction/genetics
15.
Hum Mol Genet ; 22(19): 3906-19, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23740940

ABSTRACT

The cone photoreceptor cyclic nucleotide-gated (CNG) channel is essential for central and color vision and visual acuity. Mutations in the channel subunits CNGA3 and CNGB3 are associated with achromatopsia and cone dystrophy. We investigated the gene expression profiles in mouse retina with CNG channel deficiency using whole genome expression microarrays. As cones comprise only 2 to 3% of the total photoreceptor population in the wild-type mouse retina, the mouse lines with CNG channel deficiency on a cone-dominant background, i.e. Cnga3-/-/Nrl-/- and Cngb3-/-/Nrl-/- mice, were used in our study. Comparative data analysis revealed a total of 105 genes altered in Cnga3-/-/Nrl-/- and 92 in Cngb3-/-/Nrl-/- retinas, relative to Nrl-/- retinas, with 27 genes changed in both genotypes. The differentially expressed genes primarily encode proteins associated with cell signaling, cellular function maintenance and gene expression. Ingenuity pathway analysis (IPA) identified 26 and 9 canonical pathways in Cnga3-/-/Nrl-/- and Cngb3-/-/Nrl-/- retinas, respectively, with 6 pathways being shared. The shared pathways include phototransduction, cAMP/PKA-mediated signaling, endothelin signaling, and EIF2/endoplasmic reticulum (ER) stress, whereas the IL-1, CREB, and purine metabolism signaling were found to specifically associate with Cnga3 deficiency. Thus, CNG channel deficiency differentially regulates genes that affect cell processes such as phototransduction, cellular survival and gene expression, and such regulations play a crucial role(s) in the retinal adaptation to impaired cone phototransduction. Though lack of Cnga3 and Cngb3 shares many common pathways, deficiency of Cnga3 causes more significant alterations in gene expression. This work provides insights into how cones respond to impaired phototransduction at the gene expression levels.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/metabolism , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Vision, Ocular/genetics , Animals , Cyclic Nucleotide-Gated Cation Channels/deficiency , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Light Signal Transduction , Mice , Mice, Inbred C57BL , Microarray Analysis , Opsins/genetics , Opsins/metabolism , Receptors, Dopamine D4/genetics , Receptors, Dopamine D4/metabolism , Signal Transduction
16.
J Neurosci ; 33(37): 14939-48, 2013 Sep 11.
Article in English | MEDLINE | ID: mdl-24027293

ABSTRACT

Photoreceptor cyclic nucleotide-gated (CNG) channels regulate Ca(2+) influx in rod and cone photoreceptors. cGMP, the native ligand of the photoreceptor CNG channels, has been associated with cytotoxicity when its levels rise above normal due to defects in photoreceptor phosphodiesterase (PDE6) or regulation of retinal guanylyl cyclase (retGC). We found a massive accumulation of cGMP in CNGA3-deficient retina and investigated whether cGMP accumulation plays a role in cone degeneration in CNG channel deficiency. The time course study showed that the retinal cGMP level in Cnga3(-/-);Nrl(-/-) mice with CNGA3 deficiency on a cone-dominant background was sharply increased at postnatal day 8 (P8), peaked around P10-P15, remained high through P30-P60, and returned to near control level at P90. This elevation pattern correlated with photoreceptor apoptotic death, which peaked around P15-P20. In Cnga3(-/-);Gucy2e(-/-) mice lacking retGC1, cone density and expression levels of cone-specific proteins were significantly increased compared with Cnga3(-/-), consistent with a role of cGMP accumulation as the major contributor to cone death caused by CNG channel deficiency. The activity and expression levels of cGMP-dependent protein kinase G (PKG) were significantly increased in Cnga3(-/-);Nrl(-/-) retina compared with Nrl(-/-), suggesting an involvement of PKG regulation in cell death. Our results indicate that cGMP accumulation in photoreceptors can itself exert cytotoxic effect in cones, independently of CNG channel activity and Ca(2+) influx.


Subject(s)
Cyclic GMP/metabolism , Cyclic Nucleotide-Gated Cation Channels/deficiency , Photoreceptor Cells/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Animals , Animals, Newborn , Carrier Proteins , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide-Gated Cation Channels/genetics , Enzyme-Linked Immunosorbent Assay , Eye Proteins/metabolism , Guanylate Cyclase/deficiency , Guanylate Cyclase/metabolism , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/deficiency , Phosphoric Diester Hydrolases/metabolism , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/metabolism , Receptors, N-Methyl-D-Aspartate , Retina/pathology
17.
J Neurosci ; 33(12): 5182-94, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23516284

ABSTRACT

Mammalian cones respond to light by closing a cGMP-gated channel via a cascade that includes a heterotrimeric G-protein, cone transducin, comprising Gαt2, Gß3 and Gγt2 subunits. The function of Gßγ in this cascade has not been examined. Here, we investigate the role of Gß3 by assessing cone structure and function in Gß3-null mouse (Gnb3(-/-)). We found that Gß3 is required for the normal expression of its partners, because in the Gnb3(-/-) cone outer segments, the levels of Gαt2 and Gγt2 are reduced by fourfold to sixfold, whereas other components of the cascade remain unaltered. Surprisingly, Gnb3(-/-) cones produce stable responses with normal kinetics and saturating response amplitudes similar to that of the wild-type, suggesting that cone phototransduction can function efficiently without a Gß subunit. However, light sensitivity was reduced by approximately fourfold in the knock-out cones. Because the reduction in sensitivity was similar in magnitude to the reduction in Gαt2 level in the cone outer segment, we conclude that activation of Gαt2 in Gnb3(-/-) cones proceeds at a rate approximately proportional to its outer segment concentration, and that activation of phosphodiesterase and downstream cascade components is normal. These results suggest that the main role of Gß3 in cones is to establish optimal levels of transducin heteromer in the outer segment, thereby indirectly contributing to robust response properties.


Subject(s)
Heterotrimeric GTP-Binding Proteins/genetics , Retinal Cone Photoreceptor Cells/physiology , Transducin/genetics , Vision, Ocular/physiology , Animals , Color , Female , GABA Plasma Membrane Transport Proteins/genetics , Green Fluorescent Proteins/genetics , Heterotrimeric GTP-Binding Proteins/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Neurological , Photic Stimulation , Retinal Photoreceptor Cell Outer Segment/physiology , Transducin/physiology , Ultraviolet Rays
18.
Adv Exp Med Biol ; 801: 57-65, 2014.
Article in English | MEDLINE | ID: mdl-24664681

ABSTRACT

Photopic (cone) vision essential for color sensation, central vision, and visual acuity is mediated by the activation of photoreceptor cyclic nucleotide-gated (CNG) channels. Naturally occurring mutations in the cone channel subunits CNGA3 and CNGB3 are associated with achromatopsia and cone dystrophies. This work investigated the functional modulation of cone CNG channel by exploring the channel-interacting proteins. Retinal protein extracts prepared from cone-dominant Nrl (- / -) mice were used in CNGA3 antibody affinity purification, followed by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) separation and matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry analysis. The peptide mass fingerprinting of the tryptic digests and database search identified a number of proteins including spectrin alpha-2, ATPase (Na(+)/K(+) transporting) alpha-3, alpha and beta subunits of ATP synthase (H(+) transporting, mitochondrial F1 complex), and alpha-2 subunit of the guanine nucleotide-binding protein. In addition, the affinity-binding assays demonstrated an interaction between cone CNG channel and calmodulin but not cone Na(+)/Ca(2+)-K(+) exchanger in the mouse retina. Results of this study provide insight into our understanding of cone CNG channel-interacting proteins and the functional modulations.


Subject(s)
Color Vision Defects/physiopathology , Cyclic Nucleotide-Gated Cation Channels/physiology , Retinal Cone Photoreceptor Cells/physiology , Amino Acid Sequence , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Chromatography, Affinity , Cyclic Nucleotide-Gated Cation Channels/analysis , Cyclic Nucleotide-Gated Cation Channels/isolation & purification , Eye Proteins/genetics , Ion Channel Gating/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Protein Interaction Domains and Motifs , Sodium-Calcium Exchanger/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
19.
J Biol Chem ; 287(22): 18018-29, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22493484

ABSTRACT

Cyclic nucleotide-gated (CNG) channels play a pivotal role in phototransduction. Mutations in the cone CNG channel subunits CNGA3 and CNGB3 account for >70% of all known cases of achromatopsia. Cones degenerate in achromatopsia patients and in CNGA3(-/-) and CNGB3(-/-) mice. This work investigates the molecular basis of cone degeneration in CNG channel deficiency. As cones comprise only 2-3% of the total photoreceptor population in the wild-type mouse retina, we generated mouse lines with CNG channel deficiency on a cone-dominant background, i.e. CNGA3(-/-)/Nrl(-/-) and CNGB3(-/-)/Nrl(-/-) mice. The retinal phenotype and potential cell death pathways were examined by functional, biochemical, and immunohistochemical approaches. CNGA3(-/-)/Nrl(-/-) and CNGB3(-/-)/Nrl(-/-) mice showed impaired cone function, opsin mislocalization, and cone degeneration similar to that in the single knock-out mice. The endoplasmic reticulum stress marker proteins, including Grp78/Bip, phospho-eIF2α, phospho-IP(3)R, and CCAAT/enhancer-binding protein homologous protein, were elevated significantly in CNGA3(-/-)/Nrl(-/-) and CNGB3(-/-)/Nrl(-/-) retinas, compared with the age-matched (postnatal 30 days) Nrl(-/-) controls. Along with these, up-regulation of the cysteine protease calpains and cleavage of caspase-12 and caspase-7 were found in the channel-deficient retinas, suggesting an endoplasmic reticulum stress-associated apoptosis. In addition, we observed a nuclear translocation of apoptosis-inducing factor (AIF) and endonuclease G in CNGA3(-/-)/Nrl(-/-) and CNGB3(-/-)/Nrl(-/-) retinas, implying a mitochondrial insult in the endoplasmic reticulum stress-activated cell death process. Taken together, our findings suggest a crucial role of endoplasmic reticulum stress in cone degeneration associated with CNG channel deficiency.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/genetics , Endoplasmic Reticulum/metabolism , Ion Channel Gating , Retinal Cone Photoreceptor Cells/metabolism , Animals , Cell Death , Endoplasmic Reticulum Chaperone BiP , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal
20.
Hum Mol Genet ; 20(16): 3161-75, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21576125

ABSTRACT

Mutations in the CNGB3 gene account for >50% of all known cases of achromatopsia. Although of early onset, its stationary character and the potential for rapid assessment of restoration of retinal function following therapy renders achromatopsia a very attractive candidate for gene therapy. Here we tested the efficacy of an rAAV2/8 vector containing a human cone arrestin promoter and a human CNGB3 cDNA in CNGB3 deficient mice. Following subretinal delivery of the vector, CNGB3 was detected in both M- and S-cones and resulted in increased levels of CNGA3, increased cone density and survival, improved cone outer segment structure and normal subcellular compartmentalization of cone opsins. Therapy also resulted in long-term improvement of retinal function, with restoration of cone ERG amplitudes of up to 90% of wild-type and a significant improvement in visual acuity. Remarkably, successful restoration of cone function was observed even when treatment was initiated at 6 months of age; however, restoration of normal visual acuity was only possible in younger animals (e.g. 2-4 weeks old). This study represents achievement of the most substantial restoration of visual function reported to date in an animal model of achromatopsia using a human gene construct, which has the potential to be utilized in clinical trials.


Subject(s)
Aging/pathology , Color Vision Defects/physiopathology , Color Vision Defects/therapy , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/therapeutic use , Genetic Therapy , Vision, Ocular/physiology , Animals , Arrestins/genetics , Cell Survival , Color Vision Defects/pathology , Cyclic Nucleotide-Gated Cation Channels/deficiency , Cyclic Nucleotide-Gated Cation Channels/metabolism , Disease Models, Animal , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Injections , Mice , Mice, Transgenic , Opsins/metabolism , Organ Specificity , Promoter Regions, Genetic/genetics , Protein Transport , Retina/metabolism , Retina/pathology , Retinal Cone Photoreceptor Cells/pathology , Retinal Cone Photoreceptor Cells/ultrastructure , Time Factors , Visual Acuity/physiology
SELECTION OF CITATIONS
SEARCH DETAIL