Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 812
Filter
1.
Biomolecules ; 14(5)2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38785932

ABSTRACT

Augmenting the natural melanocortin pathway in mouse eyes with uveitis or diabetes protects the retinas from degeneration. The retinal cells are protected from oxidative and apoptotic signals of death. Therefore, we investigated the effects of a therapeutic application of the melanocortin alpha-melanocyte-stimulating hormone (α-MSH) on an ischemia and reperfusion (I/R) model of retinal degenerative disease. Eyes were subjected to an I/R procedure and were treated with α-MSH. Retinal sections were histopathologically scored. Also, the retinal sections were immunostained for viable ganglion cells, activated Muller cells, microglial cells, and apoptosis. The I/R caused retinal deformation and ganglion cell loss that was significantly reduced in I/R eyes treated with α-MSH. While α-MSH treatment marginally reduced the number of GFAP-positive Muller cells, it significantly suppressed the density of Iba1-positive microglial cells in the I/R retinas. Within one hour after I/R, there was apoptosis in the ganglion cell layer, and by 48 h, there was apoptosis in all layers of the neuroretina. The α-MSH treatment significantly reduced and delayed the onset of apoptosis in the retinas of I/R eyes. The results demonstrate that therapeutically augmenting the melanocortin pathways preserves retinal structure and cell survival in eyes with progressive neuroretinal degenerative disease.


Subject(s)
Apoptosis , Homeostasis , Reperfusion Injury , Retina , Retinal Ganglion Cells , alpha-MSH , Animals , alpha-MSH/pharmacology , alpha-MSH/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Mice , Apoptosis/drug effects , Retina/metabolism , Retina/drug effects , Retina/pathology , Homeostasis/drug effects , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Mice, Inbred C57BL , Microglia/metabolism , Microglia/drug effects , Male , Ependymoglial Cells/metabolism , Ependymoglial Cells/drug effects , Ependymoglial Cells/pathology , Disease Models, Animal , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Degeneration/drug therapy
2.
Biomed Pharmacother ; 175: 116711, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38735082

ABSTRACT

Glaucoma, the leading cause of irreversible blindness worldwide, is characterized by neurodegeneration and neuroinflammation with retinal NAD/NADP and GSH decline. Nicotinamide adenine dinucleotide (NAD)/NAD phosphate (NADP) and glutathione (GSH) are two redox reducers in neuronal and glial metabolism. However, therapeutic strategies targeting NAD/NADP or GSH do not exert ideal effects, and the underlying mechanisms are still poorly understood. We assessed morphological changes in retinal ganglion cells (RGCs), the affected neurons in glaucoma, and Müller cells, the major glial cells in the retina, as well as the levels of phosphorylated p38 (p-p38) and Caspase-3 in glaucoma patients. We constructed a modified chronic ocular hypertensive rat model and an oxygen-glucose deprivation (OGD) cell model. After applying NADPH and N-acetylcysteine (NAC), a precursor to cysteine, the rate-limiting substrate in GSH biosynthesis, to cells, apoptosis, axonal damage and peroxidation were reduced in the RGCs of the NAC group and p-p38 levels were decreased in the RGCs of the NADPH group, while in stimulated Müller cells cultured individually or cocultured with RGCs, gliosis and p38/MAPK, rather than JNK/MAPK, activation were inhibited. The results were more synergistic in the rat model, where either NADPH or NAC showed crossover effects on inhibiting peroxidation and p38/MAPK pathway activation. Moreover, the combination of NADPH and NAC ameliorated RGC electrophysiological function and prevented Müller cell gliosis to the greatest extent. These data illustrated conjoined mechanisms in glaucomatous RGC injury and Müller cell gliosis and suggested that NADPH and NAC collaborate as a neuroprotective and anti-inflammatory combination treatment for glaucoma and other underlying human neurodegenerative diseases.


Subject(s)
Acetylcysteine , NADP , Ocular Hypertension , Rats, Sprague-Dawley , Retinal Ganglion Cells , p38 Mitogen-Activated Protein Kinases , Animals , NADP/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Ocular Hypertension/metabolism , Ocular Hypertension/drug therapy , Ocular Hypertension/pathology , Acetylcysteine/pharmacology , Rats , Male , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Glaucoma/metabolism , Glaucoma/pathology , Glaucoma/drug therapy , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Humans , Ependymoglial Cells/drug effects , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Disease Models, Animal , MAP Kinase Signaling System/drug effects , Apoptosis/drug effects , Chronic Disease , Neuroprotective Agents/pharmacology , Cells, Cultured , Lipid Peroxidation/drug effects
3.
Exp Cell Res ; 439(1): 114087, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38735619

ABSTRACT

Diabetic retinopathy (DR) is a common microvascular complication that causes visual impairment or loss. Aquaporin 4 (AQP4) is a regulatory protein involved in water transport and metabolism. In previous studies, we found that AQP4 is related to hypoxia injury in Muller cells. Transient receptor potential cation channel subfamily V member 4 (TRPV4) is a non-selective cation channel protein involved in the regulation of a variety of ophthalmic diseases. However, the effects of AQP4 and TRPV4 on ferroptosis and oxidative stress in high glucose (HG)-treated Muller cells are unclear. In this study, we investigated the functions of AQP4 and TRPV4 in DR. HG was used to treat mouse Muller cells. Reverse transcription quantitative polymerase chain reaction was used to measure AQP4 mRNA expression. Western blotting was used to detect the protein levels of AQP4, PTGS2, GPX4, and TRPV4. Cell count kit-8, flow cytometry, 5,5',6,6'-tetrachloro-1,1,3,3'-tetraethylbenzimidazolyl carbocyanine iodide staining, and glutathione (GSH), superoxide dismutase (SOD), and malondialdehyde (MDA) kits were used to evaluate the function of the Muller cells. Streptozotocin was used to induce DR in rats. Haematoxylin and eosin staining was performed to stain the retina of rats. GSH, SOD, and MDA detection kits, immunofluorescence, and flow cytometry assays were performed to study the function of AQP4 and TRPV4 in DR rats. Results found that AQP4 and TRPV4 were overexpressed in HG-induced Muller cells and streptozotocin-induced DR rats. AQP4 inhibition promoted proliferation and cell cycle progression, repressed cell apoptosis, ferroptosis, and oxidative stress, and alleviated retinal injury in DR rats. Mechanistically, AQP4 positively regulated TRPV4 expression. Overexpression of TRPV4 enhanced ferroptosis and oxidative stress in HG-treated Muller cells, and inhibition of TRPV4 had a protective effect on DR-induced retinal injury in rats. In conclusion, inhibition of AQP4 inhibits the ferroptosis and oxidative stress in Muller cells by downregulating TRPV4, which may be a potential target for DR therapy.


Subject(s)
Aquaporin 4 , Diabetic Retinopathy , Ependymoglial Cells , Ferroptosis , Oxidative Stress , TRPV Cation Channels , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Animals , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Diabetic Retinopathy/genetics , Mice , Aquaporin 4/metabolism , Aquaporin 4/genetics , Rats , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Glucose/metabolism , Glucose/pharmacology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Male , Rats, Sprague-Dawley , Mice, Inbred C57BL
4.
Eur J Med Res ; 29(1): 265, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38698486

ABSTRACT

Diabetic retinopathy (DR), a leading cause of visual impairment, demands a profound comprehension of its cellular mechanisms to formulate effective therapeutic strategies. Our study presentes a comprehensive single-cell analysis elucidating the intricate landscape of Müller cells within DR, emphasizing their nuanced involvement. Utilizing scRNA-seq data from both Sprague-Dawley rat models and human patients, we delineated distinct Müller cell clusters and their corresponding gene expression profiles. These findings were further validated through differential gene expression analysis utilizing human transcriptomic data. Notably, certain Müller cell clusters displayed upregulation of the Rho gene, implying a phagocytic response to damaged photoreceptors within the DR microenvironment. This phenomenon was consistently observed across species. Additionally, the co-expression patterns of RHO and PDE6G within Müller cell clusters provided compelling evidence supporting their potential role in maintaining retinal integrity during DR. Our results offer novel insights into the cellular dynamics of DR and underscore Müller cells as promising therapeutic targets for preserving vision in retinal disorders induced by diabetes.


Subject(s)
Diabetic Retinopathy , Ependymoglial Cells , Rats, Sprague-Dawley , Single-Cell Analysis , Diabetic Retinopathy/pathology , Diabetic Retinopathy/genetics , Ependymoglial Cells/pathology , Ependymoglial Cells/metabolism , Single-Cell Analysis/methods , Animals , Humans , Rats , Transcriptome
5.
FASEB J ; 38(10): e23671, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38752538

ABSTRACT

NLRP3 inflammasome activation has emerged as a critical initiator of inflammatory response in ischemic retinopathy. Here, we identified the effect of a potent, selective NLRP3 inhibitor, MCC950, on autophagy and apoptosis under hypoxia. Neonatal mice were exposed to hyperoxia for 5 days to establish oxygen-induced retinopathy (OIR) model. Intravitreal injection of MCC950 was given, and then autophagy and apoptosis markers were assessed. Retinal autophagy, apoptosis, and related pathways were evaluated by western blot, immunofluorescent labeling, transmission electron microscopy, and TUNEL assay. Autophagic activity in Müller glia after NLRP3 inflammasome inhibition, together with its influence on photoreceptor death, was studied using western blot, immunofluorescence staining, mRFP-GFP-LC3 adenovirus transfection, cell viability, proliferation, and apoptosis assays. Results showed that activation of NLRP3 inflammasome in Müller glia was detected in OIR model. MCC950 could improve impaired retinal autophagic flux and attenuate retinal apoptosis while it regulated the retinal AMPK/mTOR/ULK-1 pathway. Suppressed autophagy and depressed proliferation capacity resulting from hypoxia was promoted after MCC950 treatment in Müller glia. Inhibition of AMPK and ULK-1 pathway significantly interfered with the MCC950-induced autophagy activity, indicating MCC950 positively modulated autophagy through AMPK/mTOR/ULK-1 pathway in Müller cells. Furthermore, blockage of autophagy in Müller glia significantly induced apoptosis in the cocultured 661W photoreceptor cells, whereas MCC950 markedly preserved the density of photoreceptor cells. These findings substantiated the therapeutic potential of MCC950 against impaired autophagy and subsequent apoptosis under hypoxia. Such protective effect might involve the modulation of AMPK/mTOR/ULK-1 pathway. Targeting NLRP3 inflammasome in Müller glia could be beneficial for photoreceptor survival under hypoxic conditions.


Subject(s)
Apoptosis , Autophagy , Ependymoglial Cells , Furans , Indenes , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Sulfonamides , Animals , Autophagy/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice , Apoptosis/drug effects , Sulfonamides/pharmacology , Inflammasomes/metabolism , Furans/pharmacology , Ependymoglial Cells/metabolism , Ependymoglial Cells/drug effects , Indenes/pharmacology , Mice, Inbred C57BL , Hypoxia/metabolism , Cyclic S-Oxides/pharmacology , Sulfones/pharmacology , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/pathology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Photoreceptor Cells/metabolism , Photoreceptor Cells/drug effects , Signal Transduction/drug effects
6.
J Exp Biol ; 227(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38690647

ABSTRACT

Hibernation is an extreme state of seasonal energy conservation, reducing metabolic rate to as little as 1% of the active state. During the hibernation season, many species of hibernating mammals cycle repeatedly between the active (aroused) and hibernating (torpid) states (T-A cycling), using brown adipose tissue (BAT) to drive cyclical rewarming. The regulatory mechanisms controlling this process remain undefined but are presumed to involve thermoregulatory centres in the hypothalamus. Here, we used the golden hamster (Mesocricetus auratus), and high-resolution monitoring of BAT, core body temperature and ventilation rate, to sample at precisely defined phases of the T-A cycle. Using c-fos as a marker of cellular activity, we show that although the dorsomedial hypothalamus is active during torpor entry, neither it nor the pre-optic area shows any significant changes during the earliest stages of spontaneous arousal. Contrastingly, in three non-neuronal sites previously linked to control of metabolic physiology over seasonal and daily time scales - the choroid plexus, pars tuberalis and third ventricle tanycytes - peak c-fos expression is seen at arousal initiation. We suggest that through their sensitivity to factors in the blood or cerebrospinal fluid, these sites may mediate metabolic feedback-based initiation of the spontaneous arousal process.


Subject(s)
Arousal , Choroid Plexus , Ependymoglial Cells , Hibernation , Proto-Oncogene Proteins c-fos , Torpor , Animals , Proto-Oncogene Proteins c-fos/metabolism , Arousal/physiology , Torpor/physiology , Hibernation/physiology , Ependymoglial Cells/metabolism , Ependymoglial Cells/physiology , Choroid Plexus/metabolism , Choroid Plexus/physiology , Mesocricetus , Male , Adipose Tissue, Brown/physiology , Adipose Tissue, Brown/metabolism , Cricetinae
7.
Biomolecules ; 14(5)2024 May 10.
Article in English | MEDLINE | ID: mdl-38785974

ABSTRACT

Diabetic retinopathy (DR) affects over 140 million people globally. The mechanisms that lead to blindness are still enigmatic but there is evidence that sustained inflammation and hypoxia contribute to vascular damage. Despite efforts to understand the role of inflammation and microglia in DR's pathology, the contribution of astrocytes to hypoxic responses is less clear. To investigate the role of astrocytes in hypoxia-induced retinopathy, we utilized a 7-day systemic hypoxia model using the GFAP-CreERT2:Rosa26iDTR transgenic mouse line. This allows for the induction of inflammatory reactive astrogliosis following tamoxifen and diphtheria toxin administration. We hypothesize that DTx-induced astrogliosis is neuroprotective during hypoxia-induced retinopathy. Glial, neuronal, and vascular responses were quantified using immunostaining, with antibodies against GFAP, vimentin, IBA-1, NeuN, fibrinogen, and CD31. Cytokine responses were measured in both the brain and serum. We report that while both DTx and hypoxia induced a phenotype of reduced microglia morphological activation, DTx, but not hypoxia, induced an increase in the Müller glia marker vimentin. We did not observe that the combination of DTx and hypoxic treatments exacerbated the signs of reactive glial cells, nor did we observe a significant change in the expression immunomodulatory mediators IL-1ß, IL2, IL-4, IL-5, IL-6, IL-10, IL-18, CCL17, TGF-ß1, GM-CSF, TNF-α, and IFN-γ. Overall, our results suggest that, in this hypoxia model, reactive astrogliosis does not alter the inflammatory responses or cause vascular damage in the retina.


Subject(s)
Disease Models, Animal , Ependymoglial Cells , Gliosis , Mice, Transgenic , Microglia , Animals , Gliosis/pathology , Gliosis/metabolism , Gliosis/chemically induced , Mice , Microglia/metabolism , Microglia/pathology , Microglia/drug effects , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Ependymoglial Cells/drug effects , Retina/metabolism , Retina/pathology , Retina/drug effects , Hypoxia/metabolism , Hypoxia/pathology , Astrocytes/metabolism , Astrocytes/pathology , Astrocytes/drug effects , Glial Fibrillary Acidic Protein/metabolism , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Cytokines/metabolism , Vimentin/metabolism , Vimentin/genetics , Diphtheria Toxin
8.
Mol Vis ; 30: 167-174, 2024.
Article in English | MEDLINE | ID: mdl-38601015

ABSTRACT

Purpose: To examine whether increased ephrin type-B receptor 1 (EphB1) leads to inflammatory mediators in retinal Müller cells. Methods: Diabetic human and mouse retinal samples were examined for EphB1 protein levels. Rat Müller cells (rMC-1) were grown in culture and treated with EphB1 siRNA or ephrin B1-Fc to explore inflammatory mediators in cells grown in high glucose. An EphB1 overexpression adeno-associated virus (AAV) was used to increase EphB1 in Müller cells in vivo. Ischemia/reperfusion (I/R) was performed on mice treated with the EphB1 overexpression AAV to explore the actions of EphB1 on retinal neuronal changes in vivo. Results: EphB1 protein levels were increased in diabetic human and mouse retinal samples. Knockdown of EphB1 reduced inflammatory mediator levels in Müller cells grown in high glucose. Ephrin B1-Fc increased inflammatory proteins in rMC-1 cells grown in normal and high glucose. Treatment of mice with I/R caused retinal thinning and loss of cell numbers in the ganglion cell layer. This was increased in mice exposed to I/R and treated with the EphB1 overexpressing AAVs. Conclusions: EphB1 is increased in the retinas of diabetic humans and mice and in high glucose-treated Müller cells. This increase leads to inflammatory proteins. EphB1 also enhanced retinal damage in response to I/R. Taken together, inhibition of EphB1 may offer a new therapeutic option for diabetic retinopathy.


Subject(s)
Diabetic Retinopathy , Ephrin-B1 , Retinal Diseases , Animals , Humans , Mice , Rats , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Ependymoglial Cells/metabolism , Ephrin-B1/genetics , Ephrin-B1/metabolism , Glucose/metabolism , Inflammation Mediators/metabolism , Retina/metabolism , Retinal Diseases/metabolism
9.
Cell Biochem Funct ; 42(4): e4024, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38666564

ABSTRACT

Diabetic retinopathy (DR) is a significant complication of diabetes that often leads to blindness, impacting Müller cells, the primary retinal macroglia involved in DR pathogenesis. Reactive oxygen species (ROS) play a crucial role in the development of DR. The objective of this study was to investigate the involvement of sestrin2 in DR using a high-glucose (HG)-induced Müller cell model and assessing cell proliferation with 5-ethynyl-2-deoxyuridine (EdU) labeling. Following this, sestrin2 was upregulated in Müller cells to investigate its effects on ROS, tube formation, and inflammation both in vitro and in vivo, as well as its interaction with the nuclear factor erythroid2-related factor 2 (Nrf2) signaling pathway. The findings demonstrated a gradual increase in the number of EdU-positive cells over time, with a subsequent decrease after 72 h of exposure to high glucose levels. Additionally, the expression of sestrin2 exhibited a progressive increase over time, followed by a decrease at 72 h. The rh-sestrin2 treatment suppressed the injury of Müller cells, decreased ROS level, and inhibited the tube formation. Rh-sestrin2 treatment enhanced the expression of sestrin2, Nrf2, heme oxygenase-1 (HO-1), and glutamine synthetase (GS); however, the ML385 treatment reversed the protective effect of rh-sestrin2. Finally, we evaluated the effect of sestrin2 in a DR rat model. Sestrin2 overexpression treatment improved the pathological injury of retina and attenuated the oxidative damage and inflammatory reaction. Our results highlighted the inhibitory effect of sestrin2 in the damage of retina, thus presenting a novel therapeutic sight for DR.


Subject(s)
Diabetic Retinopathy , Reactive Oxygen Species , Sestrins , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Animals , Reactive Oxygen Species/metabolism , Rats , Male , Rats, Sprague-Dawley , NF-E2-Related Factor 2/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Glucose/metabolism , Cell Proliferation/drug effects , Ependymoglial Cells/metabolism , Ependymoglial Cells/drug effects , Ependymoglial Cells/pathology , Signal Transduction/drug effects , Peroxidases/metabolism , Cells, Cultured
10.
Biomed Pharmacother ; 174: 116538, 2024 May.
Article in English | MEDLINE | ID: mdl-38579401

ABSTRACT

Glaucoma is considered a neurodegenerative disease characterized by progressive visual field defects that may lead to blindness. Although controlling intraocular pressure (IOP) is the mainstay of glaucoma treatment, some glaucoma patients have unmet needs due to unclear pathogenic mechanisms. Recently, there has been growing evidence that neuroinflammation is a potential target for the development of novel antiglaucoma agents. In this study, we investigated the protective effects and cellular mechanisms of H7E, a novel small molecule inhibits HDAC8, using in vitro and in vivo glaucoma-like models. Importantly, H7E mitigated extracellular MMP-9 activity and MCP-1 levels in glutamate- or S100B-stimulated reactive Müller glia. In addition, H7E inhibited the upregulation of inflammation- and proliferation-related signaling pathways, particularly the ERK and JNK MAPK pathways. Under conditions of oxidative damage, H7E prevents retinal cell death and reduces extracellular glutamate released from stressed Müller glia. In a mouse model of NMDA-induced retinal degeneration, H7E alleviated functional and structural defects within the inner retina as assessed by electroretinography and optical coherence tomography. Our results demonstrated that the newly identified compound H7E protects against glaucoma damage by specifically targeting HDAC8 activity in the retina. This protective effect is attributed to the inhibition of Müller glial activation and the prevention of retinal cell death caused by oxidative stress.


Subject(s)
Ependymoglial Cells , Glaucoma , Histone Deacetylase Inhibitors , Histone Deacetylases , Mice, Inbred C57BL , Oxidative Stress , Animals , Oxidative Stress/drug effects , Glaucoma/drug therapy , Glaucoma/metabolism , Glaucoma/pathology , Histone Deacetylase Inhibitors/pharmacology , Ependymoglial Cells/drug effects , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Mice , Histone Deacetylases/metabolism , Retina/drug effects , Retina/metabolism , Retina/pathology , Disease Models, Animal , Neuroprotective Agents/pharmacology , Male , Retinal Degeneration/drug therapy , Retinal Degeneration/pathology , Retinal Degeneration/metabolism , Retinal Degeneration/prevention & control
11.
Mol Metab ; 84: 101940, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38641253

ABSTRACT

OBJECTIVE: The liver releases glucose into the blood using the glucose-6-phosphatase (G6Pase) system, a multiprotein complex located in the endoplasmic reticulum (ER). Here, we show for the first time that the G6Pase system is also expressed in hypothalamic tanycytes, and it is required to regulate energy balance. METHODS: Using automatized qRT-PCR and immunohistochemical analyses, we evaluated the expression of the G6Pase system. Fluorescent glucose analogue (2-NBDG) uptake was evaluated by 4D live-cell microscopy. Glucose release was tested using a glucose detection kit and high-content live-cell analysis instrument, Incucyte s3. In vivo G6pt knockdown in tanycytes was performed by AAV1-shG6PT-mCherry intracerebroventricular injection. Body weight gain, adipose tissue weight, food intake, glucose metabolism, c-Fos, and neuropeptide expression were evaluated at 4 weeks post-transduction. RESULTS: Tanycytes sequester glucose-6-phosphate (G6P) into the ER through the G6Pase system and release glucose in hypoglycaemia via facilitative glucose transporters (GLUTs). Strikingly, in vivo tanycytic G6pt knockdown has a powerful peripheral anabolic effect observed through decreased body weight, white adipose tissue (WAT) tissue mass, and strong downregulation of lipogenesis genes. Selective deletion of G6pt in tanycytes also decreases food intake, c-Fos expression in the arcuate nucleus (ARC), and Npy mRNA expression in fasted mice. CONCLUSIONS: The tanycyte-associated G6Pase system is a central mechanism involved in controlling metabolism and energy balance.


Subject(s)
Energy Metabolism , Ependymoglial Cells , Glucose-6-Phosphatase , Glucose , Hypoglycemia , Hypothalamus , Animals , Glucose-6-Phosphatase/metabolism , Glucose-6-Phosphatase/genetics , Mice , Hypothalamus/metabolism , Glucose/metabolism , Male , Hypoglycemia/metabolism , Ependymoglial Cells/metabolism , Mice, Inbred C57BL , Endoplasmic Reticulum/metabolism
12.
Exp Eye Res ; 243: 109890, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615833

ABSTRACT

Phosphodiesterase (PDE) inhibitors - such as vardenafil - are used primarily for treating erectile dysfunction via increasing cyclic guanosine monophosphate (cGMP) levels. Recent studies have also demonstrated their significant cardioprotective effects in several diseases, including diabetes, upon long-term, continuous application. However, PDE inhibitors are not specific for PDE5 and also inhibit the retinal isoform. A sustained rise in cGMP in photoreceptors is known to be toxic; therefore, we hypothesized that long-term vardenafil treatment might result in retinotoxicity. The hypothesis was tested in a clinically relevant animal model of type 2 diabetes mellitus. Histological experiments were performed on lean and diabetic Zucker Diabetic Fatty rats. Half of the animals were treated with vardenafil for six months, and the retinal effects were evaluated. Vardenafil treatment alleviated rod outer segment degeneration but decreased rod numbers in some positions and induced changes in the interphotoreceptor matrix, even in control animals. Vardenafil treatment decreased total retinal thickness in the control and diabetic groups and reduced the number of nuclei in the outer nuclear layer. Müller cell activation was detectable even in the vardenafil-treated control animals, and vardenafil did not improve gliosis in the diabetic group. Vardenafil-treated animals showed complex retinal alterations with improvements in some parameters while deterioration in others. Our results point towards the retinotoxicity of vardenafil, even without diabetes, which raises doubts about the retinal safety of long-term continuous vardenafil administration. This effect needs to be considered when approving PDE inhibitors for alternative indications.


Subject(s)
Diabetes Mellitus, Experimental , Phosphodiesterase 5 Inhibitors , Rats, Zucker , Vardenafil Dihydrochloride , Vardenafil Dihydrochloride/pharmacology , Vardenafil Dihydrochloride/toxicity , Animals , Rats , Phosphodiesterase 5 Inhibitors/pharmacology , Male , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/pathology , Retina/drug effects , Retina/pathology , Ependymoglial Cells/drug effects , Ependymoglial Cells/pathology , Ependymoglial Cells/metabolism
13.
Cell Rep ; 43(5): 114143, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38676924

ABSTRACT

Cellular retinaldehyde-binding protein (CRALBP) supports production of 11-cis-retinaldehyde and its delivery to photoreceptors. It is found in the retinal pigment epithelium (RPE) and Müller glia (MG), but the relative functional importance of these two cellular pools is debated. Here, we report RPE- and MG-specific CRALBP knockout (KO) mice and examine their photoreceptor and visual cycle function. Bulk visual chromophore regeneration in RPE-KO mice is 15-fold slower than in controls, accounting for their delayed rod dark adaptation and protection against retinal phototoxicity, whereas MG-KO mice have normal bulk visual chromophore regeneration and retinal light damage susceptibility. Cone pigment regeneration is significantly impaired in RPE-KO mice but mildly affected in MG-KO mice, disclosing an unexpectedly strong reliance of cone photoreceptors on the RPE-based visual cycle. These data reveal a dominant role for RPE-CRALBP in supporting rod and cone function and highlight the importance of RPE cell targeting for CRALBP gene therapies.


Subject(s)
Carrier Proteins , Mice, Knockout , Retinal Cone Photoreceptor Cells , Retinal Pigment Epithelium , Animals , Retinal Pigment Epithelium/metabolism , Mice , Carrier Proteins/metabolism , Carrier Proteins/genetics , Retinal Cone Photoreceptor Cells/metabolism , Ependymoglial Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Mice, Inbred C57BL , Retinal Pigments/metabolism
14.
Exp Mol Med ; 56(4): 975-986, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38609519

ABSTRACT

We explored the genomic events underlying central neurocytoma (CN), a rare neoplasm of the central nervous system, via multiomics approaches, including whole-exome sequencing, bulk and single-nuclei RNA sequencing, and methylation sequencing. We identified FGFR3 hypomethylation leading to FGFR3 overexpression as a major event in the ontogeny of CN that affects crucial downstream events, such as aberrant PI3K-AKT activity and neuronal development pathways. Furthermore, we found similarities between CN and radial glial cells based on analyses of gene markers and CN tumor cells and postulate that CN tumorigenesis is due to dysregulation of radial glial cell differentiation into neurons. Our data demonstrate the potential role of FGFR3 as one of the leading drivers of tumorigenesis in CN.


Subject(s)
DNA Methylation , Ependymoglial Cells , Neurocytoma , Receptor, Fibroblast Growth Factor, Type 3 , Humans , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Neurocytoma/genetics , Neurocytoma/pathology , Neurocytoma/metabolism , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Gene Expression Regulation, Neoplastic
15.
Proc Natl Acad Sci U S A ; 121(13): e2314802121, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38498715

ABSTRACT

The molecular basis for cortical expansion during evolution remains largely unknown. Here, we report that fibroblast growth factor (FGF)-extracellular signal-regulated kinase (ERK) signaling promotes the self-renewal and expansion of cortical radial glial (RG) cells. Furthermore, FGF-ERK signaling induces bone morphogenic protein 7 (Bmp7) expression in cortical RG cells, which increases the length of the neurogenic period. We demonstrate that ERK signaling and Sonic Hedgehog (SHH) signaling mutually inhibit each other in cortical RG cells. We provide evidence that ERK signaling is elevated in cortical RG cells during development and evolution. We propose that the expansion of the mammalian cortex, notably in human, is driven by the ERK-BMP7-GLI3R signaling pathway in cortical RG cells, which participates in a positive feedback loop through antagonizing SHH signaling. We also propose that the relatively short cortical neurogenic period in mice is partly due to mouse cortical RG cells receiving higher SHH signaling that antagonizes ERK signaling.


Subject(s)
Ependymoglial Cells , Extracellular Signal-Regulated MAP Kinases , Animals , Mice , Humans , Extracellular Signal-Regulated MAP Kinases/metabolism , Ependymoglial Cells/metabolism , Cell Proliferation , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Signal Transduction , Fibroblast Growth Factors , Mammals/metabolism
16.
PLoS One ; 19(3): e0300370, 2024.
Article in English | MEDLINE | ID: mdl-38536827

ABSTRACT

Anti-VEGF (vascular endothelial growth factor) drugs such as aflibercept (AFL) and bevacizumab (BVZ) inhibit pathological neo-angiogenesis and vascular permeability in retinal vascular diseases. As cytokines and growth factors are produced by Müller glial cells under stressful and pathological conditions, we evaluated the in vitro effect of AFL (Eylea®, 0.5 mg/mL) and BVZ (Avastin®, 0.5 mg/mL) on cell viability/metabolism, and cytokine/growth factor production by Müller cells (MIO-M1) under cobalt chloride (CoCl2)-induced hypoxia after 24h, 48h and 72h. Cell viability/metabolism were analyzed by Trypan Blue and MTT assays and cytokine/growth factors in supernatants by Luminex xMAP-based multiplex bead-based immunoassay. Cell viability increased with AFL at 48h and 72h and decreased with BVZ or hypoxia at 24h. BVZ-treated cells showed lower cell viability than AFL at all exposure times. Cell metabolism increased with AFL but decreased with BVZ (72h) and hypoxia (48h and72h). As expected, AFL and BVZ decreased VEGF levels. AFL increased PDGF-BB, IL-6 and TNF-α (24h) and BVZ increased PDGF-BB (72h). Hypoxia reduced IL-1ß, -6, -8, TNF-α and PDGF-BB at 24h, and its suppressive effect was more prominent than AFL (EGF, PDGF-BB, IL-1ß, IL-6, IL-8, and TNF-α) and BVZ (PDGF-BB and IL-6) effects. Hypoxia increased bFGF levels at 48h and 72h, even when combined with anti-VEGFs. However, the stimulatory effect of BVZ predominated over hypoxia for IL-8 and TNF-α (24h), as well as for IL-1ß (72h). Thus, AFL and BVZ exhibit distinct exposure times effects on MIO-M1 cells viability, metabolism, and cytokines/growth factors. Hypoxia and BVZ decreased MIO-M1 cell viability/metabolism, whereas AFL likely induced gliosis. Hypoxia resulted in immunosuppression, and BVZ stimulated inflammation in hypoxic MIO-M1 cells. These findings highlight the complexity of the cellular response as well as the interplay between anti-VEGF treatments and the hypoxic microenvironment.


Subject(s)
Ependymoglial Cells , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins , Vascular Endothelial Growth Factor A , Humans , Bevacizumab/pharmacology , Bevacizumab/metabolism , Vascular Endothelial Growth Factor A/metabolism , Ependymoglial Cells/metabolism , Cell Survival , Becaplermin/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukin-8/metabolism , Interleukin-6/metabolism , Vascular Endothelial Growth Factors/metabolism , Cytokines/metabolism , Hypoxia/metabolism , Neovascularization, Pathologic/pathology , Inflammation/pathology
17.
Glia ; 72(6): 1067-1081, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38497356

ABSTRACT

Alzheimer's Disease (AD) pathogenesis is thought to begin up to 20 years before cognitive symptoms appear, suggesting the need for more sensitive diagnostic biomarkers of AD. In this report, we demonstrated pathological changes in retinal Müller glia significantly earlier than amyloid pathology in AD mouse models. By utilizing the knock-in NLGF mouse model, we surprisingly discovered an increase in reticulon 3 (RTN3) protein levels in the NLGF retina as early as postnatal day 30 (P30). Despite RTN3 being a canonically neuronal protein, this increase was noted in the retinal Müller glia, confirmed by immunohistochemical characterization. Further unbiased transcriptomic assays of the P30 NLGF retina revealed that retinal Müller glia were the most sensitive responding cells in this mouse retina, compared with other cell types including photoreceptor cells and ganglion neurons. Pathway analyses of differentially expressed genes in glia cells showed activation of ER stress response via the upregulation of unfolded protein response (UPR) proteins such as ATF4 and CHOP. Early elevation of RTN3 in response to challenges by toxic Aß likely facilitated UPR. Altogether, these findings suggest that Müller glia act as a sentinel for AD pathology in the retina and should aid for both intervention and diagnosis.


Subject(s)
Alzheimer Disease , Animals , Mice , Alzheimer Disease/pathology , Retina , Neurons/metabolism , Disease Models, Animal , Amyloidogenic Proteins/metabolism , Neuroglia/metabolism , Brain/metabolism , Ependymoglial Cells/metabolism
18.
Biol Open ; 13(4)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38526172

ABSTRACT

In contrast to mammals, zebrafish undergo successful neural regeneration following spinal cord injury. Spinal cord ependymo-radial glia (ERG) undergo injury-induced proliferation and neuronal differentiation to replace damaged cells and restore motor function. However, the molecular cues driving these processes remain elusive. Here, we demonstrate that the evolutionarily conserved P2X7 receptors are widely distributed on neurons and ERG within the zebrafish spinal cord. At the protein level, the P2X7 receptor expressed in zebrafish is a truncated splice variant of the full-length variant found in mammals. The protein expression of this 50 kDa isoform was significantly downregulated at 7 days post-injury (dpi) but returned to basal levels at 14 dpi when compared to naïve controls. Pharmacological activation of P2X7 following SCI resulted in a greater number of proliferating cells around the central canal by 7 dpi but did not affect neuronal differentiation at 14 dpi. Our findings suggest that unlike in mammals, P2X7 signaling may not play a maladaptive role following SCI in adult zebrafish and may also work to curb the proliferative response of ERG following injury.


Subject(s)
Spinal Cord Injuries , Zebrafish , Animals , Ependymoglial Cells/metabolism , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism , Cell Proliferation , Mammals
19.
Cells ; 13(5)2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38474393

ABSTRACT

CD40 induces pro-inflammatory responses in endothelial and Müller cells and is required for the development of diabetic retinopathy (DR). CD40 is upregulated in these cells in patients with DR. CD40 upregulation is a central feature of CD40-driven inflammatory disorders. What drives CD40 upregulation in the diabetic retina remains unknown. We examined the role of advanced glycation end products (AGEs) in CD40 upregulation in endothelial cells and Müller cells. Human endothelial cells and Müller cells were incubated with unmodified or methylglyoxal (MGO)-modified fibronectin. CD40 expression was assessed by flow cytometry. The expression of ICAM-1 and CCL2 was examined by flow cytometry or ELISA after stimulation with CD154 (CD40 ligand). The expression of carboxymethyl lysine (CML), fibronectin, and laminin as well as CD40 in endothelial and Müller cells from patients with DR was examined by confocal microscopy. Fibronectin modified by MGO upregulated CD40 in endothelial and Müller cells. CD40 upregulation was functionally relevant. MGO-modified fibronectin enhanced CD154-driven upregulation of ICAM-1 and CCL2 in endothelial and Müller cells. Increased CD40 expression in endothelial and Müller cells from patients with DR was associated with increased CML expression in fibronectin and laminin. These findings identify AGEs as inducers of CD40 upregulation in endothelial and Müller cells and enhancers of CD40-dependent pro-inflammatory responses. CD40 upregulation in these cells is associated with higher CML expression in fibronectin and laminin in patients with DR. This study revealed that CD40 and AGEs, two important drivers of DR, are interconnected.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Humans , Diabetic Retinopathy/metabolism , Intercellular Adhesion Molecule-1/metabolism , Fibronectins/metabolism , Ependymoglial Cells/metabolism , Endothelial Cells/metabolism , Magnesium Oxide/metabolism , Retina/metabolism , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Laminin/metabolism , Glycation End Products, Advanced/metabolism , Diabetes Mellitus/metabolism
20.
Nat Cell Biol ; 26(5): 698-709, 2024 May.
Article in English | MEDLINE | ID: mdl-38548890

ABSTRACT

The human neocortex has undergone strong evolutionary expansion, largely due to an increased progenitor population, the basal radial glial cells. These cells are responsible for the production of a diversity of cell types, but the successive cell fate decisions taken by individual progenitors remain unknown. Here we developed a semi-automated live/fixed correlative imaging method to map basal radial glial cell division modes in early fetal tissue and cerebral organoids. Through the live analysis of hundreds of dividing progenitors, we show that basal radial glial cells undergo abundant symmetric amplifying divisions, and frequent self-consuming direct neurogenic divisions, bypassing intermediate progenitors. These direct neurogenic divisions are more abundant in the upper part of the subventricular zone. We furthermore demonstrate asymmetric Notch activation in the self-renewing daughter cells, independently of basal fibre inheritance. Our results reveal a remarkable conservation of fate decisions in cerebral organoids, supporting their value as models of early human neurogenesis.


Subject(s)
Cell Lineage , Neocortex , Neural Stem Cells , Neurogenesis , Organoids , Humans , Neocortex/cytology , Neocortex/embryology , Neocortex/metabolism , Organoids/cytology , Organoids/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Cell Differentiation , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Receptors, Notch/metabolism , Receptors, Notch/genetics , Cell Division , Cell Proliferation
SELECTION OF CITATIONS
SEARCH DETAIL
...