Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.357
Filtrar
1.
FASEB J ; 38(10): e23671, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38752538

RESUMEN

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.


Asunto(s)
Apoptosis , Autofagia , Células Ependimogliales , Furanos , Indenos , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Sulfonamidas , Animales , Autofagia/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ratones , Apoptosis/efectos de los fármacos , Sulfonamidas/farmacología , Inflamasomas/metabolismo , Furanos/farmacología , Células Ependimogliales/metabolismo , Células Ependimogliales/efectos de los fármacos , Indenos/farmacología , Ratones Endogámicos C57BL , Hipoxia/metabolismo , Óxidos S-Cíclicos/farmacología , Sulfonas/farmacología , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Células Fotorreceptoras de Vertebrados/patología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
2.
Eur J Med Res ; 29(1): 265, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38698486

RESUMEN

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.


Asunto(s)
Retinopatía Diabética , Células Ependimogliales , Ratas Sprague-Dawley , Análisis de la Célula Individual , Retinopatía Diabética/patología , Retinopatía Diabética/genética , Células Ependimogliales/patología , Células Ependimogliales/metabolismo , Análisis de la Célula Individual/métodos , Animales , Humanos , Ratas , Transcriptoma
3.
Biomolecules ; 14(5)2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38785932

RESUMEN

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.


Asunto(s)
Apoptosis , Homeostasis , Daño por Reperfusión , Retina , Células Ganglionares de la Retina , alfa-MSH , Animales , alfa-MSH/farmacología , alfa-MSH/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Ratones , Apoptosis/efectos de los fármacos , Retina/metabolismo , Retina/efectos de los fármacos , Retina/patología , Homeostasis/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/efectos de los fármacos , Masculino , Células Ependimogliales/metabolismo , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/patología , Modelos Animales de Enfermedad , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Degeneración Retiniana/tratamiento farmacológico
4.
Biomolecules ; 14(5)2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38785974

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Células Ependimogliales , Gliosis , Ratones Transgénicos , Microglía , Animales , Gliosis/patología , Gliosis/metabolismo , Gliosis/inducido químicamente , Ratones , Microglía/metabolismo , Microglía/patología , Microglía/efectos de los fármacos , Células Ependimogliales/metabolismo , Células Ependimogliales/patología , Células Ependimogliales/efectos de los fármacos , Retina/metabolismo , Retina/patología , Retina/efectos de los fármacos , Hipoxia/metabolismo , Hipoxia/patología , Astrocitos/metabolismo , Astrocitos/patología , Astrocitos/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Citocinas/metabolismo , Vimentina/metabolismo , Vimentina/genética , Toxina Diftérica
5.
J Exp Biol ; 227(10)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38690647

RESUMEN

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.


Asunto(s)
Nivel de Alerta , Plexo Coroideo , Células Ependimogliales , Hibernación , Proteínas Proto-Oncogénicas c-fos , Letargo , Animales , Proteínas Proto-Oncogénicas c-fos/metabolismo , Nivel de Alerta/fisiología , Letargo/fisiología , Hibernación/fisiología , Células Ependimogliales/metabolismo , Células Ependimogliales/fisiología , Plexo Coroideo/metabolismo , Plexo Coroideo/fisiología , Mesocricetus , Masculino , Tejido Adiposo Pardo/fisiología , Tejido Adiposo Pardo/metabolismo , Cricetinae
6.
Mol Metab ; 84: 101940, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38641253

RESUMEN

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.


Asunto(s)
Metabolismo Energético , Células Ependimogliales , Glucosa-6-Fosfatasa , Glucosa , Hipoglucemia , Hipotálamo , Animales , Glucosa-6-Fosfatasa/metabolismo , Glucosa-6-Fosfatasa/genética , Ratones , Hipotálamo/metabolismo , Glucosa/metabolismo , Masculino , Hipoglucemia/metabolismo , Células Ependimogliales/metabolismo , Ratones Endogámicos C57BL , Retículo Endoplásmico/metabolismo
7.
Biomed Pharmacother ; 174: 116538, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38579401

RESUMEN

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.


Asunto(s)
Células Ependimogliales , Glaucoma , Inhibidores de Histona Desacetilasas , Histona Desacetilasas , Ratones Endogámicos C57BL , Estrés Oxidativo , Animales , Estrés Oxidativo/efectos de los fármacos , Glaucoma/tratamiento farmacológico , Glaucoma/metabolismo , Glaucoma/patología , Inhibidores de Histona Desacetilasas/farmacología , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/metabolismo , Células Ependimogliales/patología , Ratones , Histona Desacetilasas/metabolismo , Retina/efectos de los fármacos , Retina/metabolismo , Retina/patología , Modelos Animales de Enfermedad , Fármacos Neuroprotectores/farmacología , Masculino , Degeneración Retiniana/tratamiento farmacológico , Degeneración Retiniana/patología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/prevención & control
8.
Exp Eye Res ; 243: 109890, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38615833

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Experimental , Inhibidores de Fosfodiesterasa 5 , Ratas Zucker , Diclorhidrato de Vardenafil , Diclorhidrato de Vardenafil/farmacología , Diclorhidrato de Vardenafil/toxicidad , Animales , Ratas , Inhibidores de Fosfodiesterasa 5/farmacología , Masculino , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/patología , Retina/efectos de los fármacos , Retina/patología , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/patología , Células Ependimogliales/metabolismo
9.
Mol Vis ; 30: 167-174, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38601015

RESUMEN

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.


Asunto(s)
Retinopatía Diabética , Efrina-B1 , Enfermedades de la Retina , Animales , Humanos , Ratones , Ratas , Retinopatía Diabética/genética , Retinopatía Diabética/metabolismo , Células Ependimogliales/metabolismo , Efrina-B1/genética , Efrina-B1/metabolismo , Glucosa/metabolismo , Mediadores de Inflamación/metabolismo , Retina/metabolismo , Enfermedades de la Retina/metabolismo
10.
Exp Mol Med ; 56(4): 975-986, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38609519

RESUMEN

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.


Asunto(s)
Metilación de ADN , Células Ependimogliales , Neurocitoma , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos , Humanos , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Neurocitoma/genética , Neurocitoma/patología , Neurocitoma/metabolismo , Células Ependimogliales/metabolismo , Células Ependimogliales/patología , Regulación Neoplásica de la Expresión Génica
11.
Cell Biochem Funct ; 42(4): e4024, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38666564

RESUMEN

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.


Asunto(s)
Retinopatía Diabética , Especies Reactivas de Oxígeno , Sestrinas , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Animales , Especies Reactivas de Oxígeno/metabolismo , Ratas , Masculino , Ratas Sprague-Dawley , Factor 2 Relacionado con NF-E2/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Glucosa/metabolismo , Proliferación Celular/efectos de los fármacos , Células Ependimogliales/metabolismo , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/patología , Transducción de Señal/efectos de los fármacos , Peroxidasas/metabolismo , Células Cultivadas
12.
Glia ; 72(7): 1236-1258, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38515287

RESUMEN

The purpose of this study was to investigate how ID factors regulate the ability of Müller glia (MG) to reprogram into proliferating MG-derived progenitor cells (MGPCs) in the chick retina. We found that ID1 is transiently expressed by maturing MG (mMG), whereas ID4 is maintained in mMG in embryonic retinas. In mature retinas, ID4 was prominently expressed by resting MG, but following retinal damage ID4 was rapidly upregulated and then downregulated in MGPCs. By contrast, ID1, ID2, and ID3 were low in resting MG and then upregulated in MGPCs. Inhibition of ID factors following retinal damage decreased numbers of proliferating MGPCs. Inhibition of IDs, after MGPC proliferation, significantly increased numbers of progeny that differentiated as neurons. In damaged or undamaged retinas inhibition of IDs increased levels of p21Cip1 in MG. In response to damage or insulin+FGF2 levels of CDKN1A message and p21Cip1 protein were decreased, absent in proliferating MGPCs, and elevated in MG returning to a resting phenotype. Inhibition of notch- or gp130/Jak/Stat-signaling in damaged retinas increased levels of ID4 but not p21Cip1 in MG. Although ID4 is the predominant isoform expressed by MG in the chick retina, id1 and id2a are predominantly expressed by resting MG and downregulated in activated MG and MGPCs in zebrafish retinas. We conclude that ID factors have a significant impact on regulating the responses of MG to retinal damage, controlling the ability of MG to proliferate by regulating levels of p21Cip1, and suppressing the neurogenic potential of MGPCs.


Asunto(s)
Proliferación Celular , Células Ependimogliales , Proteínas Inhibidoras de la Diferenciación , Retina , Animales , Proliferación Celular/fisiología , Proliferación Celular/efectos de los fármacos , Proteínas Inhibidoras de la Diferenciación/metabolismo , Proteínas Inhibidoras de la Diferenciación/genética , Retina/metabolismo , Retina/citología , Células Ependimogliales/metabolismo , Células Ependimogliales/fisiología , Neurogénesis/fisiología , Neurogénesis/efectos de los fármacos , Embrión de Pollo , Células-Madre Neurales/metabolismo , Pollos , Neuroglía/metabolismo , Células Madre/metabolismo , Células Madre/fisiología
13.
Curr Biol ; 34(5): R209-R211, 2024 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-38471452

RESUMEN

In many species, metabolic and reproductive functions are coupled to the seasons. Tanycytes, specialized glial cells in the hypothalamus, play an important function in these physiological changes. A new study now shows that light exposure drastically alters the formation of sensory cilia on tanycytes.


Asunto(s)
Células Ependimogliales , Hipotálamo , Células Ependimogliales/metabolismo , Estaciones del Año , Hipotálamo/metabolismo , Neuroglía/metabolismo , Biología
14.
Nat Cell Biol ; 26(5): 698-709, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38548890

RESUMEN

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.


Asunto(s)
Linaje de la Célula , Neocórtex , Células-Madre Neurales , Neurogénesis , Organoides , Humanos , Neocórtex/citología , Neocórtex/embriología , Neocórtex/metabolismo , Organoides/citología , Organoides/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Diferenciación Celular , Células Ependimogliales/citología , Células Ependimogliales/metabolismo , Receptores Notch/metabolismo , Receptores Notch/genética , División Celular , Proliferación Celular
15.
PLoS One ; 19(3): e0300370, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38536827

RESUMEN

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.


Asunto(s)
Células Ependimogliales , Receptores de Factores de Crecimiento Endotelial Vascular , Proteínas Recombinantes de Fusión , Factor A de Crecimiento Endotelial Vascular , Humanos , Bevacizumab/farmacología , Bevacizumab/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Células Ependimogliales/metabolismo , Supervivencia Celular , Becaplermina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-8/metabolismo , Interleucina-6/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo , Citocinas/metabolismo , Hipoxia/metabolismo , Neovascularización Patológica/patología , Inflamación/patología
16.
Nature ; 628(8009): 826-834, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38538787

RESUMEN

Empirical evidence suggests that heat exposure reduces food intake. However, the neurocircuit architecture and the signalling mechanisms that form an associative interface between sensory and metabolic modalities remain unknown, despite primary thermoceptive neurons in the pontine parabrachial nucleus becoming well characterized1. Tanycytes are a specialized cell type along the wall of the third ventricle2 that bidirectionally transport hormones and signalling molecules between the brain's parenchyma and ventricular system3-8. Here we show that tanycytes are activated upon acute thermal challenge and are necessary to reduce food intake afterwards. Virus-mediated gene manipulation and circuit mapping showed that thermosensing glutamatergic neurons of the parabrachial nucleus innervate tanycytes either directly or through second-order hypothalamic neurons. Heat-dependent Fos expression in tanycytes suggested their ability to produce signalling molecules, including vascular endothelial growth factor A (VEGFA). Instead of discharging VEGFA into the cerebrospinal fluid for a systemic effect, VEGFA was released along the parenchymal processes of tanycytes in the arcuate nucleus. VEGFA then increased the spike threshold of Flt1-expressing dopamine and agouti-related peptide (Agrp)-containing neurons, thus priming net anorexigenic output. Indeed, both acute heat and the chemogenetic activation of glutamatergic parabrachial neurons at thermoneutrality reduced food intake for hours, in a manner that is sensitive to both Vegfa loss-of-function and blockage of vesicle-associated membrane protein 2 (VAMP2)-dependent exocytosis from tanycytes. Overall, we define a multimodal neurocircuit in which tanycytes link parabrachial sensory relay to the long-term enforcement of a metabolic code.


Asunto(s)
Tronco Encefálico , Células Ependimogliales , Conducta Alimentaria , Calor , Hipotálamo , Vías Nerviosas , Neuronas , Animales , Femenino , Masculino , Ratones , Proteína Relacionada con Agouti/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/citología , Tronco Encefálico/citología , Tronco Encefálico/fisiología , Dopamina/metabolismo , Ingestión de Alimentos/fisiología , Células Ependimogliales/citología , Células Ependimogliales/fisiología , Conducta Alimentaria/fisiología , Ácido Glutámico/metabolismo , Hipotálamo/citología , Hipotálamo/fisiología , Vías Nerviosas/metabolismo , Neuronas/metabolismo , Núcleos Parabraquiales/citología , Núcleos Parabraquiales/metabolismo , Núcleos Parabraquiales/fisiología , Sensación Térmica/fisiología , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/líquido cefalorraquídeo , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Cells ; 13(5)2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38474393

RESUMEN

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.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Humanos , Retinopatía Diabética/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Fibronectinas/metabolismo , Células Ependimogliales/metabolismo , Células Endoteliales/metabolismo , Óxido de Magnesio/metabolismo , Retina/metabolismo , Antígenos CD40/metabolismo , Ligando de CD40/metabolismo , Laminina/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Diabetes Mellitus/metabolismo
18.
Biol Open ; 13(4)2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38526172

RESUMEN

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.


Asunto(s)
Traumatismos de la Médula Espinal , Pez Cebra , Animales , Células Ependimogliales/metabolismo , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/metabolismo , Proliferación Celular , Mamíferos
19.
Exp Eye Res ; 242: 109872, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38514024

RESUMEN

X-linked retinoschisis (XLRS) is an early onset degenerative retinal disease characterized by cystic lesions in the middle layers of the retina. These structural changes are accompanied by a loss of visual acuity and decreased contrast sensitivity. XLRS is caused by mutations in the gene Rs1 which encodes the secreted protein Retinoschisin 1. Young Rs1-mutant mouse models develop key hallmarks of XLRS including intraretinal schisis and abnormal electroretinograms. The electroretinogram (ERG) comprises activity of multiple cellular generators, and it is not known how and when each of these is impacted in Rs1 mutant mice. Here we use an ex vivo ERG system and pharmacological blockade to determine how ERG components generated by photoreceptors, ON-bipolar, and Müller glial cells are impacted in Rs1 mutants and to determine the time course of these changes. We report that ERG abnormalities begin near eye-opening and that all ERG components are involved.


Asunto(s)
Moléculas de Adhesión Celular , Modelos Animales de Enfermedad , Electrorretinografía , Proteínas del Ojo , Retinosquisis , Animales , Retinosquisis/genética , Retinosquisis/fisiopatología , Ratones , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Ratones Endogámicos C57BL , Mutación , Células Ependimogliales/patología , Células Ependimogliales/metabolismo , Masculino , Células Bipolares de la Retina/patología , Células Bipolares de la Retina/metabolismo
20.
Proc Natl Acad Sci U S A ; 121(13): e2314802121, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38498715

RESUMEN

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.


Asunto(s)
Células Ependimogliales , Quinasas MAP Reguladas por Señal Extracelular , Animales , Ratones , Humanos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células Ependimogliales/metabolismo , Proliferación Celular , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Transducción de Señal , Factores de Crecimiento de Fibroblastos , Mamíferos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...