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











Publication year range
1.
Cells ; 12(19)2023 09 26.
Article in English | MEDLINE | ID: mdl-37830574

ABSTRACT

Previously, the RXR agonist UAB126 demonstrated therapeutic potential to treat obese mice by controlling blood glucose levels (BGL) and altering the expression of genes associated with lipid metabolism and inflammatory response. The purpose of the study was to assess the effects of UAB126 on the progression of diabetic retinopathy (DR) in rodent models of type 1 diabetes (T1D), streptozotocin-induced, and type 2 diabetes (T2D), in db/db mice. UAB126 treatment was delivered either by oral gavage for 6 weeks or by topical application of eye drops for 2 weeks. At the end of the treatment, the retinal function of diabetic mice was assessed by electroretinography (ERG), and their retinal tissue was harvested for protein and gene expression analyses. Bone-marrow cells were isolated and differentiated into bone marrow-derived macrophages (BMDMs). The glycolysis stress test and the 2-DG glucose uptake analysis were performed. Our results demonstrated that in the UAB126-treated diabetic BMDMs, the ECAR rate and the 2-DG uptake were improved as compared to untreated diabetic BMDMs. In UAB126-treated diabetic mice, hyperglycemia was reduced and associated with the preservation of ERG amplitudes and enhanced AMPK activity. Retinas from diabetic mice treated with topical UAB126 demonstrated an increase in Rxr and Ppar and the expression of genes associated with lipid metabolism. Altogether, our data indicate that RXR activation is beneficial to preclinical models of DR.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Diabetic Retinopathy , Mice , Animals , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/prevention & control , Diabetic Retinopathy/metabolism , Retinoid X Receptors , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal
2.
bioRxiv ; 2023 Aug 04.
Article in English | MEDLINE | ID: mdl-37577690

ABSTRACT

Previously, the RXR agonist UAB126 demonstrated therapeutic potential to treat obese mice by controlling blood glucose levels (BGL) and altering the expression of genes associated with lipid metabolism and inflammatory response. The purpose of the study was to assess UAB126 effect in progression of diabetic retinopathy (DR) in rodent models of Type1 diabetes (T1D), streptozotocin-induced, and Type2 diabetes (T2D), the db/db mice. UAB126 treatment was delivered either by oral gavage for 6 weeks or by topical application of eye drops for 2 weeks. At the end of the treatment, the retinal function of diabetic mice was assessed by electroretinography (ERG), and their retinal tissue was harvested for protein and gene expression analyses. Bone-marrow cells were isolated and differentiated into bone marrow-derived macrophages (BMDMs). The glycolysis stress test and the 2-DG glucose uptake analysis were performed. Our results demonstrated that in the UAB126-treated diabetic BMDMs, the ECAR rate and the 2-DG uptake were improved as compared to untreated diabetic BMDMs. In UAB126-treated diabetic mice, hyperglycemia was reduced and associated with the preservation of ERG amplitudes and enhanced AMPK activity. Retinas from diabetic mice treated with topical UAB126 demonstrated an increase in Rxr and Ppar, and expression of genes associated with lipid metabolism. Altogether, our data indicate that RXR activation is beneficial to preclinical models of DR.

3.
Int J Mol Sci ; 23(22)2022 Nov 09.
Article in English | MEDLINE | ID: mdl-36430227

ABSTRACT

The UPR is sustainably activated in degenerating retinas, leading to translational inhibition via p-eIF2α. Recent findings have demonstrated that ablation of growth arrest and DNA damage-inducible protein 34 (GADD34), a protein phosphatase 1 regulatory subunit permitting translational machinery operation through p-eIF2α elevation, does not impact the rate of translation in fast-degenerating rd16 mice. The current study aimed to validate whether P23H RHO mice degenerating at a slower pace manifest translational attenuation and whether GADD34 ablation impacts the rate of retinal degeneration via further suppression of retinal protein synthesis and apoptotic cell death. For this study, mice were examined with ERG and histological analyses. The molecular assessment was conducted in the naïve and LPS-challenged mice using Western blot and qRT-PCR analyses. Thus, this study demonstrates that the P23H RHO retinas manifest translational attenuation. However, GADD34 ablation resulted in a more prominent p-eIF2a increase without impacting the translation rate. GADD34 deficiency also led to a reduction in scotopic ERG amplitudes and an increased number of TUNEL-positive cells. Molecular analysis revealed that GADD34 deficiency reduces the expression of p-STAT3 and Il-6 while increasing the expression of Tnfa. Overall, the data indicate that GADD34 plays a multifunctional role. Under chronic UPR activation, GADD34 acts as a feedback player, dephosphorylating p-eIF2a, although this role does not seem to be critical. Additionally, GADD34 controls cytokine expression and STAT3 activation. Perhaps these molecular events are particularly important in controlling the pace of retinal degeneration.


Subject(s)
Retinal Degeneration , Animals , Mice , Eukaryotic Initiation Factor-2/metabolism , Mice, Inbred C57BL , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Retina/metabolism , Retinal Degeneration/metabolism
4.
Cell Death Dis ; 12(7): 664, 2021 07 02.
Article in English | MEDLINE | ID: mdl-34215725

ABSTRACT

Various retinal degenerative disorders manifest in alterations of the AKT/mTOR axis. Despite this, consensus on the therapeutic targeting of mTOR in degenerating retinas has not yet been achieved. Therefore, we investigated the role of AKT/mTOR signaling in rd16 retinas, in which we restored the AKT/mTOR axis by genetic ablation of pseudokinase TRB3, known to inhibit phosphorylation of AKT and mTOR. First, we found that TRB3 ablation resulted in preservation of photoreceptor function in degenerating retinas. Then, we learned that the mTOR downstream cellular pathways involved in the homeostasis of photoreceptors were also reprogrammed in rd16 TRB3-/- retinas. Thus, the level of inactivated translational repressor p-4E-BP1 was significantly increased in these mice along with the restoration of translational rate. Moreover, in rd16 mice manifesting decline in p-mTOR at P15, we found elevated expression of Beclin-1 and ATG5 autophagy genes. Thus, these mice showed impaired autophagy flux measured as an increase in LC3 conversion and p62 accumulation. In addition, the RFP-EGFP-LC3 transgene expression in rd16 retinas resulted in statistically fewer numbers of red puncta in photoreceptors, suggesting impaired late autophagic vacuoles. In contrast, TRIB3 ablation in these mice resulted in improved autophagy flux. The restoration of translation rate and the boost in autophagosome formation occurred concomitantly with an increase in total Ub and rhodopsin protein levels and the elevation of E3 ligase Parkin1. We propose that TRB3 may retard retinal degeneration and be a promising therapeutic target to treat various retinal degenerative disorders.


Subject(s)
Cell Cycle Proteins/metabolism , Photoreceptor Cells, Vertebrate/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Retinal Degeneration/enzymology , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagosomes/genetics , Autophagosomes/metabolism , Autophagosomes/pathology , Autophagy , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Beclin-1/genetics , Beclin-1/metabolism , Cell Cycle Proteins/genetics , Disease Models, Animal , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Rhodopsin/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
5.
Diabetes ; 70(8): 1738-1753, 2021 08.
Article in English | MEDLINE | ID: mdl-33975909

ABSTRACT

The current understanding of the molecular pathogenesis of diabetic retinopathy does not provide a mechanistic link between early molecular changes and the subsequent progression of the disease. In this study, we found that human diabetic retinas overexpressed TRIB3 and investigated the role of TRIB3 in diabetic retinal pathobiology in mice. We discovered that TRIB3 controlled major molecular events in early diabetic retinas via HIF1α-mediated regulation of retinal glucose flux, reprogramming cellular metabolism, and governing of inflammatory gene expression. These early molecular events further defined the development of neurovascular deficit observed in mice with diabetic retinopathy. TRIB3 ablation in the streptozotocin-induced mouse model led to significant retinal ganglion cell survival and functional restoration accompanied by a dramatic reduction in pericyte loss and acellular capillary formation. Under hypoxic conditions, TRIB3 contributed to advanced proliferative stages by significant upregulation of GFAP and VEGF expression, thus controlling gliosis and aberrant vascularization in oxygen-induced retinopathy mouse retinas. Overall, our data reveal that TRIB3 is a master regulator of diabetic retinal pathophysiology that may accelerate the onset and progression of diabetic retinopathy to proliferative stages in humans and present TRIB3 as a potentially novel therapeutic target for diabetic retinopathy.


Subject(s)
Cell Cycle Proteins/genetics , Diabetic Retinopathy/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Repressor Proteins/genetics , Retina/metabolism , Animals , Capillaries/metabolism , Capillaries/pathology , Cell Cycle Proteins/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Disease Progression , Humans , Mice , Pericytes/metabolism , Pericytes/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/metabolism , Retina/pathology
6.
Front Endocrinol (Lausanne) ; 12: 799711, 2021.
Article in English | MEDLINE | ID: mdl-35046899

ABSTRACT

Existing animal models with rod-dominant retinas have shown that hyperglycemia injures neurons, but it is not yet clearly understood how blue cone photoreceptors and retinal ganglion cells (RGCs) deteriorate in patients because of compromised insulin tolerance. In contrast, northern tree shrews (Tupaia Belangeri), one of the closest living relatives of primates, have a cone-dominant retina with short wave sensitivity (SWS) and long wave sensitivity (LWS) cones. Therefore, we injected animals with a single streptozotocin dose (175 mg/kg i.p.) to investigate whether sustained hyperglycemia models the features of human diabetic retinopathy (DR). We used the photopic electroretinogram (ERG) to measure the amplitudes of A and B waves and the photopic negative responses (PhNR) to evaluate cone and RGC function. Retinal flat mounts were prepared for immunohistochemical analysis to count the numbers of neurons with antibodies against cone opsins and RGC specific BRN3a proteins. The levels of the proteins TRIB3, ISR-1, and p-AKT/p-mTOR were measured with western blot. The results demonstrated that tree shrews manifested sustained hyperglycemia leading to a slight but significant loss of SWS cones (12%) and RGCs (20%) 16 weeks after streptozotocin injection. The loss of BRN3a-positive RGCs was also reflected by a 30% decline in BRN3a protein expression. These were accompanied by reduced ERG amplitudes and PhNRs. Importantly, the diabetic retinas demonstrated increased expression of TRIB3 and level of p-AKT/p-mTOR axis but reduced level of IRS-1 protein. Therefore, a new non-primate model of DR with SWS cone and RGC dysfunction lays the foundation to better understand retinal pathophysiology at the molecular level and opens an avenue for improving the research on the treatment of human eye diseases.


Subject(s)
Diabetic Retinopathy/physiopathology , Disease Models, Animal , Tupaiidae/physiology , Animals , Diabetic Retinopathy/complications , Diabetic Retinopathy/metabolism , Electroretinography , Hyperglycemia/complications , Hyperglycemia/physiopathology , Male , Signal Transduction
7.
Prog Retin Eye Res ; 79: 100860, 2020 11.
Article in English | MEDLINE | ID: mdl-32272207

ABSTRACT

Physiological equilibrium in the retina depends on coordinated work between rod and cone photoreceptors and can be compromised by the expression of mutant proteins leading to inherited retinal degeneration (IRD). IRD is a diverse group of retinal dystrophies with multifaceted molecular mechanisms that are not fully understood. In this review, we focus on the contribution of chronically activated unfolded protein response (UPR) to inherited retinal pathogenesis, placing special emphasis on studies employing genetically modified animal models. As constitutively active UPR in degenerating retinas may activate pro-apoptotic programs associated with oxidative stress, pro-inflammatory signaling, dysfunctional autophagy, free cytosolic Ca2+ overload, and altered protein synthesis rate in the retina, we focus on the regulatory mechanisms of translational attenuation and approaches to overcoming translational attenuation in degenerating retinas. We also discuss current research on the role of the UPR mediator PERK and its downstream targets in degenerating retinas and highlight the therapeutic benefits of reprogramming PERK signaling in preclinical animal models of IRD. Finally, we describe pharmacological approaches targeting UPR in ocular diseases and consider their potential applications to IRD.


Subject(s)
Disease Management , Endoplasmic Reticulum Stress , Retinal Degeneration/metabolism , Rhodopsin/metabolism , Animals , Autophagy , Humans , Retinal Degeneration/therapy , Signal Transduction , Unfolded Protein Response
8.
Invest Ophthalmol Vis Sci ; 60(14): 4849-4857, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31747684

ABSTRACT

Purpose: We reported previously that retinas of mice with inherited retinal degeneration make less protein than retinas of normal mice. Despite recent studies suggesting that diminished protein synthesis rates may contribute to neurologic disorders, a direct link between protein synthesis rates and the progression of neurodegeneration has not been established. Moreover, it remains unclear whether reduced protein synthesis could be involved in retinal pathogenesis. Dysregulation of AKT/mTOR signaling has been reported in the retina during retinal degeneration, but to what extent this signaling contributes to translational attenuation in these mice remains uncertain. Methods: C57BL/6J and rd16 mice were subcutaneously injected with anisomycin to chronically inhibit protein synthesis rates. An AAV2 construct encoding constitutively active 4ebp1 was subretinally delivered in wildtype animals to lower protein synthesis rates. 4ebp1/2 were knocked out in rd16 mice. Results: Anisomycin treatment lowered retinal translation rates, accelerated retinal degeneration in rd16 mice, and initiated cell death in the retinas of C57BL/6J mice. AAV-mediated transfer of constitutively active 4ebp1-4A into the subretinal space of wildtype animals inhibited protein synthesis, and led to reduced electroretinography amplitudes and fewer ONL nuclei. Finally, we report that restoring protein synthesis rates by knocking out 4ebp1/2 was associated with an approximately 2-fold increase in rhodopsin levels and a delay in retinal degeneration in rd16 mice. Conclusions: Our study indicates that protein synthesis inhibition is likely not a cell defense mechanism in the retina by which deteriorating photoreceptors survive, but may be harmful to degenerating retinas, and that restoring protein synthesis may have therapeutic potential in delaying the progression of retinal degeneration.


Subject(s)
Protein Biosynthesis/physiology , Retina/physiopathology , Retinal Degeneration/physiopathology , Adaptor Proteins, Signal Transducing/genetics , Animals , Anisomycin/pharmacology , Cell Cycle Proteins/genetics , Cell Death , Dependovirus , Electroretinography , Eukaryotic Initiation Factors/genetics , Gene Expression Regulation/physiology , In Situ Nick-End Labeling , Injections, Subcutaneous , Mice , Mice, Inbred C57BL , Mice, Knockout , Parvovirinae/genetics , Protein Synthesis Inhibitors/pharmacology , Retina/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Rhodopsin/metabolism , Transfection
9.
Virus Res ; 274: 197771, 2019 12.
Article in English | MEDLINE | ID: mdl-31577935

ABSTRACT

We compared the phenotypes of three mutant AAV2 viruses containing mutations in arginine amino acids (R585, R588 and R484) previously shown to be involved in AAV2 heparan sulfate binding. The transduction efficiencies of wild type and mutant viruses were determined in the eye, the brain and peripheral organs following subretinal, striatal and intravenous injection, respectively, in mice and rats. We found that each of the three mutants (the single mutant R585A; the double mutant R585, 588A; and the triple mutant R585, 588, 484A) had a unique phenotype compared to wt and each other. R585A was completely defective for transducing peripheral organs via intravenous injection, suggesting that R585A may be useful for targeting peripheral organs by substitution of peptide ligands in the capsid surface. In the brain, all three mutants displayed widespread transduction, with the double mutant R585, 588A displaying the greatest spread and the greatest number of transduced neurons. The double mutant was also extremely efficient for retrograde transport, while the triple mutant was almost completely defective for retrograde transport. This suggested that R484 may be directly involved in interaction with the transport machinery. Finally, the double mutant also displayed improved transduction of the eye compared to wild type and the other mutants.


Subject(s)
Capsid Proteins/genetics , Capsid/metabolism , Heparan Sulfate Proteoglycans/metabolism , Parvovirinae/physiology , Animals , Axonal Transport/genetics , Capsid Proteins/metabolism , Dependovirus , Female , Male , Mice , Mutation , Parvovirinae/genetics , Parvovirinae/metabolism , Phenotype , Protein Binding , Rats , Viral Tropism/genetics
10.
Neuroscience ; 377: 174-183, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29526688

ABSTRACT

Phospholipase D2 (PLD2), an enzyme involved in vesicle trafficking and membrane signaling, interacts with α-synuclein, a protein known to contribute in the development of Parkinson disease (PD). We previously reported that PLD2 overexpression in rat substantia nigra pars compacta (SNc) causes a rapid neurodegeneration of dopamine neurons, and that α-synuclein suppresses PLD2-induced nigral degeneration (Gorbatyuk et al., 2010). Here, we report that PLD2 toxicity is due to its lipase activity. Overexpression of a catalytically inactive mutant (K758R) of PLD2 prevents the loss of dopaminergic neurons in the SNc and does not show signs of toxicity after 10 weeks of overexpression. Further, mutant K758R does not affect dopamine levels in the striatum. In contrast, mutants that prevent PLD2 interaction with dynamin or growth factor receptor bound protein 2 (Grb2) but retained lipase activity, continued to show rapid neurodegeneration. These findings suggest that neither the interaction of PLD2 with dynamin, which has a role in vesicle trafficking, nor the PLD2 interaction with Grb2, which has multiple roles in cell cycle control, chemotaxis and activation of tyrosine kinase complexes, are the primary cause of neurodegeneration. Instead, the synthesis of phosphatidic acid (the product of PLD2), which is a second messenger in multiple cellular pathways, appears to be the key to PLD2 induced neurodegeneration. The fact that α-synuclein is a regulator of PLD2 activity suggests that regulation of PLD2 activity could be important in the progression of PD.


Subject(s)
Nerve Degeneration/enzymology , Parkinsonian Disorders/enzymology , Pars Compacta/enzymology , Phospholipase D/metabolism , Animals , Dynamins/metabolism , GRB2 Adaptor Protein/metabolism , Gene Expression , HEK293 Cells , Humans , Mutation , Nerve Degeneration/pathology , Neurons/enzymology , Neurons/pathology , Parkinsonian Disorders/pathology , Pars Compacta/pathology , Phospholipase D/genetics , Rats , Tyrosine 3-Monooxygenase/metabolism
11.
Front Cell Neurosci ; 11: 410, 2017.
Article in English | MEDLINE | ID: mdl-29326555

ABSTRACT

Activation of the endoplasmic reticulum (ER) stress and ER stress response, also known as the unfolded protein response (UPR), is common to various degenerative disorders. Therefore, signaling components of the UPR are currently emerging as potential targets for intervention and treatment of human diseases. One UPR signaling member, activating transcription factor 4 (ATF4), has been found up-regulated in many pathological conditions, pointing to therapeutic potential in targeting its expression. In cells, ATF4 governs multiple signaling pathways, including autophagy, oxidative stress, inflammation, and translation, suggesting a multifaceted role of ATF4 in the progression of various pathologies. However, ATF4 has been shown to trigger both pro-survival and pro-death pathways, and this, perhaps, can explain the contradictory opinions in current literature regarding targeting ATF4 for clinical application. In this review, we summarized recent published studies from our labs and others that focus on the therapeutic potential of the strategy controlling ATF4 expression in different retinal and neurodegenerative disorders.

12.
Neurosci Lett ; 627: 36-41, 2016 08 03.
Article in English | MEDLINE | ID: mdl-27233218

ABSTRACT

Activating transcription factor 4 (ATF4) is a member of the PERK signaling pathway, which directly binds endoplasmic reticulum stress target genes and plays a crucial role in both adaptations to stress and activation of apoptosis. Previous publications demonstrated conflicting evidence on the role of ATF4 in the pathogenesis of neurodegenerative disorders. In this study, we used recombinant adeno-associate virus (rAAV)-mediated gene transfer to investigate if the sustained up-regulation of ATF4 launches a pro-survival or pro-death trend in the dopamine (DA) cells of the substantia nigra pars compacta in a rat model of Parkinson-like neurodegeneration induced by human alpha-synuclein (αS) overexpression. We showed that ATF4 does not protect nigral DA neurons against an αS-induced pathology. Moreover, the rAAV-mediated overexpression of ATF4 resulted in severe nigra-striatal degeneration via activation of caspases 3/7.


Subject(s)
Activating Transcription Factor 4/metabolism , Apoptosis , Dopaminergic Neurons/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Pars Compacta/pathology , Animals , Caspase 3/metabolism , Caspase 7/metabolism , Disease Models, Animal , Dopaminergic Neurons/metabolism , Female , Humans , Parkinson Disease/genetics , Pars Compacta/metabolism , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism , Up-Regulation , alpha-Synuclein/metabolism
13.
PLoS One ; 11(5): e0154779, 2016.
Article in English | MEDLINE | ID: mdl-27144303

ABSTRACT

T17M rhodopsin expression in rod photoreceptors leads to severe retinal degeneration and is associated with the activation of ER stress related Unfolded Protein Response (UPR) signaling. Here, we show a novel role of a UPR transcription factor, ATF4, in photoreceptor cellular pathology. We demonstrated a pro-death role for ATF4 overexpression during autosomal dominant retinitis pigmentosa (ADRP). Based on our results in ATF4 knockout mice and adeno-associated viral (AAV) delivery of ATF4 to the retina, we validated a novel therapeutic approach targeting ATF4 over the course of retinal degeneration. In T17M rhodopsin retinas, we observed ATF4 overexpression concomitantly with reduction of p62 and elevation of p53 levels. These molecular alterations, together with increased CHOP and caspase-3/7 activity, possibly contributed to the mechanism of photoreceptor cell loss. Conversely, ATF4 knockdown retarded retinal degeneration in 1-month-old T17M Rhodopsin mice and promoted photoreceptor survival, as measured by scotopic and photopic ERGs and photoreceptor nuclei row counts. Similarly, ATF4 knockdown also markedly delayed retinal degeneration in 3-month-old ADRP animals. This delay was accompanied by a dramatic decrease in UPR signaling, the launching of anti-oxidant defense, initiation of autophagy, and improvement of rhodopsin biosynthesis which together perhaps combat the cellular stress associated with T17M rhodopsin. Our data indicate that augmented ATF4 signals during retinal degeneration plays a cytotoxic role by triggering photoreceptor cell death. Future ADRP therapy regulating ATF4 expression can be developed to treat retinal degenerative disorders associated with activated UPR.


Subject(s)
Activating Transcription Factor 4/metabolism , Autophagy/physiology , Endoplasmic Reticulum/metabolism , Retinal Degeneration/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/metabolism , Stress, Physiological/physiology , Animals , Caspase 3/metabolism , Caspase 7/metabolism , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Retina/metabolism , Rhodopsin/metabolism , Transcription Factors/metabolism , Unfolded Protein Response/physiology
14.
Neuroscience ; 328: 1-8, 2016 07 22.
Article in English | MEDLINE | ID: mdl-27109921

ABSTRACT

Neuronatin (NNAT) is a small transmembrane proteolipid that is highly expressed in the embryonic developing brain and several other peripheral tissues. This study is the first to provide evidence that NNAT is detected in the adult retina of various adult rod-dominant mammals, including wild-type (WT) rodents, transgenic rodents expressing mutant S334ter, P23H, or T17M rhodopsin, non-human primates, humans, and cone-dominant tree shrews. Immunohistochemical and quantitative real time polymerase chain reaction (qRT-PCR) analyses were applied to detect NNAT. Confocal microscopy analysis revealed that NNAT immunofluorescence is restricted to the outer segments (OSs) of photoreceptors without evidence of staining in other retinal cell types across all mammalian species. Moreover, in tree shrew retinas, we found NNAT to be co-localized with rhodopsin, indicating its predominant expression in rods. The rod-derived expression of NNAT was further confirmed by qRT-PCR in isolated rod photoreceptor cells. We also used these cells to mimic cellular stress in transgenic retinas by treating them with the endoplasmic reticulum stress inducer, tunicamycin. Thus, our data revealed accumulation of NNAT around the nucleus as compared to dispersed localization of NNAT within control cells. This distribution coincided with the partial intracellular mislocalization of NNAT to the outer nuclear layer observed in transgenic retinas. In addition, stressed retinas demonstrated an increase of NNAT mRNA and protein levels. Therefore, our study demonstrated that NNAT is a novel stress-responsive protein with a potential structural and/or functional role in adult mammalian retinas.


Subject(s)
Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Stress, Physiological/physiology , Animals , Cells, Cultured , Disease Models, Animal , Endoplasmic Reticulum Stress/physiology , Female , Gene Expression Regulation , Humans , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Nerve Tissue Proteins/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Rats, Transgenic , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Rod Photoreceptor Cells/pathology , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Species Specificity , Tunicamycin , Tupaiidae
15.
Neurosci Lett ; 609: 30-5, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26467812

ABSTRACT

An unfolded protein response (UPR) in addition to oxidative stress and the inflammatory response is known to be activated in age-related ocular disorders, such as macular degeneration, diabetic retinopathy, glaucoma, and cataracts. Therefore, we aimed to investigate whether healthy aged retinas display UPR hallmarks, in order to establish a baseline for the activated UPR markers for age-related ocular diseases. Using western blotting, we determined that the hallmarks of the UPR PERK arm, phosphorylated (p) eIF2a, ATF4, and GADD34, were significantly altered in aged vs. young rat retinas. The cleaved pATF6 (50) and CHOP proteins were dramatically upregulated in the aged rodent retinas, indicating the activation of the ATF6 UPR arm. The UPR activation was associated with a drop in rhodopsin expression and in the NRF2 and HO1 levels, suggesting a decline in the anti-oxidant defense in aged retinas. Moreover, we observed down-regulation of anti-inflammatory IL-10 and IL-13 and upregulation of pro-inflammatory RANTES in the healthy aged retinas, as measured using the Bio-plex assay. Our results suggest that cellular homeostasis in normal aged retinas is compromised, resulting in the concomitant activation of the UPR, oxidative stress, and inflammatory signaling. This knowledge brings us closer to understanding the cellular mechanisms of the age-related retinopathies and ocular disorders characterized by an ongoing UPR, and highlight the UPR signaling molecules that should be validated as potential therapeutic targets.


Subject(s)
Aging/metabolism , Retina/metabolism , Unfolded Protein Response , Animals , Biomarkers/metabolism , Inflammation/metabolism , Mice, Inbred C57BL , Oxidative Stress , Rats, Inbred F344 , Rhodopsin/metabolism
16.
Neurobiol Aging ; 36(6): 2213-23, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25863526

ABSTRACT

Age-related structural changes and gradual loss of key enzymes significantly affect the ability of the endoplasmic reticulum (ER) to facilitate proper protein folding and maintain homeostasis. In this work, we present several lines of evidence supporting the hypothesis that the age-related decline in expression of the ER chaperone glucose-regulated protein 78 (GRP78) could be related to the development of Parkinson's disease. We first determined that old (24 months) rats exhibit significantly lower levels of GRP78 protein in the nigrostriatal system as compared with young (2 months) animals. Then using recombinant adeno-associate virus-mediated gene transfer, we found that GRP78 downregulation by specific small interfering RNAs (siRNAs) aggravates alpha-synuclein (α-syn) neurotoxicity in nigral dopamine (DA) neurons. Moreover, the degree of chaperone decline corresponds with the severity of neurodegeneration. Additionally, comparative analysis of nigral tissues obtained from old and young rats revealed that aging affects the capacity of nigral DA cells to upregulate endogenous GRP78 protein in response to human α-syn neurotoxicity. Finally, we demonstrated that a sustained increase of GRP78 protein over the course of 9 months protected aging nigral DA neurons in the α-syn-induced rat model of Parkinson's-like neurodegeneration. Our data indicate that the ER chaperone GRP78 may have therapeutic potential for preventing and/or slowing age-related neurodegeneration.


Subject(s)
Aging/genetics , Dopaminergic Neurons/drug effects , Gene Knockdown Techniques , Heat-Shock Proteins , RNA, Small Interfering , Substantia Nigra/cytology , alpha-Synuclein/toxicity , Aging/metabolism , Aging/pathology , Animals , Disease Models, Animal , Down-Regulation , Endoplasmic Reticulum/physiology , Endoplasmic Reticulum Chaperone BiP , Female , Heat-Shock Proteins/genetics , Heat-Shock Proteins/physiology , Homeostasis , Humans , Male , Molecular Chaperones , Parkinson Disease/genetics , Protein Folding , RNA, Small Interfering/genetics , Rats, Inbred F344
17.
Adv Exp Med Biol ; 801: 455-62, 2014.
Article in English | MEDLINE | ID: mdl-24664731

ABSTRACT

The goal of this study is to validate whether reprogramming of the UPR via modulation of pro-apoptotic caspase-7 and CHOP proteins could be an effective approach to slow down the rate of retinal degeneration in ADRP mice. In order to pursue our goal we created the T17M RHO CASP7 and T17M RHO CHOP mice to study the impact of the CASP7 or CHOP ablations in T17M RHO retina by ERG, SD-OCT, histology and western blot analysis. The scotopic ERG demonstrated that the ablation of the CASP7 in T17M RHO retina leads to significant preservation of the function of photoreceptors compared to control. Surprisingly, the ablation of pro-apoptotic CHOP protein in T17M RHO mice led to a more severe form of retinal degeneration. Results of the SD-OCT and histology were in agreement with the ERG data. The further analysis demonstrated that the preservation of the structure and function or the acceleration of the onset of the T17M RHO photoreceptor degeneration occurred via reprogramming of the UPR. In addition, the CASP7 ablation leads to the inhibition of cJUN mediated apoptosis, while the ablation of CHOP induces an increase in the HDAC. Thus, manipulation with the UPR requires careful examination in order to achieve a therapeutic effect.


Subject(s)
Caspase 7/genetics , Retinal Degeneration/genetics , Retinitis Pigmentosa/genetics , Rhodopsin/genetics , Transcription Factor CHOP/genetics , Unfolded Protein Response/genetics , Animals , Apoptosis/genetics , Caspase 7/metabolism , Disease Models, Animal , Electroretinography , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Rhodopsin/metabolism , Transcription Factor CHOP/metabolism
18.
J Neurosci ; 33(47): 18368-80, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24259562

ABSTRACT

Hormone peptide tyrosine-tyrosine (PYY) is secreted into circulation from the gut L-endocrine cells in response to food intake, thus inducing satiation during interaction with its preferred receptor, Y2R. Clinical applications of systemically administered PYY for the purpose of reducing body weight were compromised as a result of the common side effect of visceral sickness. We describe here a novel approach of elevating PYY in saliva in mice, which, although reliably inducing strong anorexic responses, does not cause aversive reactions. The augmentation of salivary PYY activated forebrain areas known to mediate feeding, hunger, and satiation while minimally affecting brainstem chemoreceptor zones triggering nausea. By comparing neuronal pathways activated by systemic versus salivary PYY, we identified a metabolic circuit associated with Y2R-positive cells in the oral cavity and extending through brainstem nuclei into hypothalamic satiety centers. The discovery of this alternative circuit that regulates ingestive behavior without inducing taste aversion may open the possibility of a therapeutic application of PYY for the treatment of obesity via direct oral application.


Subject(s)
Feeding Behavior/drug effects , Peptide Fragments/pharmacology , Peptide YY/deficiency , Saliva/enzymology , Aminophylline , Animals , Conditioning, Psychological/drug effects , Eating/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glucagon-Like Peptide 1/metabolism , Humans , Iodine Isotopes/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Oxytocin/metabolism , Peptide YY/chemistry , Proto-Oncogene Proteins c-fos/metabolism , Satiation/drug effects , Tyrosine 3-Monooxygenase/metabolism , Vasopressins/metabolism , alpha-MSH/metabolism
19.
Mol Vis ; 19: 1985-98, 2013.
Article in English | MEDLINE | ID: mdl-24068865

ABSTRACT

Recently published literature has provided evidence that the unfolded protein response (UPR) is involved in the development of retinal degeneration. The scope of these studies encompassed diabetic retinopathy, retinopathy of prematurity, glaucoma, retinal detachment, light-induced retinal degeneration, age-related macular degeneration, and inherited retinal degeneration. Subsequent studies investigating the role of individual UPR markers in retinal pathogenesis and examining the therapeutic potential of reprogramming the UPR as a method for modulating the rate of retinal degeneration have been initiated. Manipulation of UPR markers has been made possible by the use of knockout mice, pharmacological agents, and viral vector-mediated augmentation of gene expression. Future research will aim at identifying specific inhibitors and/or inducers of UPR regulatory markers as well as expand the list of UPR-related animal models. Additionally, adeno-associated virus-mediated gene delivery is a safe and effective method for modulating gene expression, and thus is a useful research tool for manipulating individual UPR markers in affected retinas and a promising delivery vector for gene therapy in retinal degenerative disorders.


Subject(s)
Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Unfolded Protein Response , Animals , Biomarkers/metabolism , Disease Models, Animal , Gene Transfer Techniques , Humans , Viruses/metabolism
20.
J Genet Syndr Gene Ther ; 4(2)2013 Mar 11.
Article in English | MEDLINE | ID: mdl-23750325

ABSTRACT

The glucose regulated protein 78 (GRP78), also known as BiP, is the endoplasmatic reticulum (ER) homologue of HSP70, which plays a dual role in the ER by controlling protein folding, in order to prevent aggregation, and by regulating the signaling of the unfolded protein response (UPR). Most neurodegenerative disorders including Parkinson's, Alzheimer's diseases and progressive retinal degeneration are characterized by activation of the UPR and modified expression of GRP78. The expression levels and activity of GRP78 are altered with age raising the question of whether the lack of GRP78 could be a predisposing factor for many neurodegenerative disorders associated with age including PD, Alzheimer and Age-related macular degeneration. Attempts to induce or upregulate GRP78 in animal models of neurodegeneration have recently been made with the help of pharmacological BiP protein Inducer X (BIX) and GRP78 cDNA delivery via adeno-associated virus (AAV) vectors. The results of these studies validate GRP78 as a new therapeutic target for treatments of forebrain ischemia, Parkinson disease and retinal degeneration. These data, together with the results from age-related studies, highlight the importance for developing drugs to induce elevation of endogenous GRP78 in order to increase cellular survival and extend functional longevity.

SELECTION OF CITATIONS
SEARCH DETAIL