Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 11.619
Filtrar
1.
Mol Biol Rep ; 51(1): 606, 2024 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-38704498

RESUMEN

BACKGROUND: Recent in vitro studies using RB1+/- fibroblasts and MSCs have shown molecular and functional disruptions without the need for biallelic loss of RB1. However, this was not reflected in the recent in vitro studies employing RB1+/- retinal organoids. To gain further insights into the molecular disruptions in the RB1+/- retinal organoids, we performed a high throughput RNA sequencing analysis. METHODS AND RESULTS: iPSCs were generated from RB1+/+ and RB1+/- OAMSCs derived from retinoblastoma patients. RB1+/+ and RB1+/- iPSCs were subjected to a step-wise retinal differentiation protocol. Retinal differentiation was evaluated by Real-time PCR and flow cytometry analysis of the retinal markers. To gain further insights into the molecular differences in RB1+/- retinal organoids, a high throughput RNA sequencing followed by differential gene expression analysis and gene set enrichment analysis (GSEA) was performed. The analysis revealed a shift from the regular metabolic process of glycolysis to oxidative phosphorylation in the RB1+/- retinal organoids. To investigate further, we performed assays to determine the levels of pyruvate, lactate and ATP in the retinal organoids. The results revealed significant increase in ATP and pyruvate levels in RB1+/- retinal organoids of day 120 compared to that of the RB1+/+. The results thus revealed enhanced ATP production in the RB1+/- retinal organoids. CONCLUSION: The study provides novel insights into the metabolic phenotype of heterozygous RB1 mutant suggesting dysregulation of energy metabolism and glycolytic pathways to be first step even before the changes in cellular proliferation or other phenotypic consequences ensue.


Asunto(s)
Adenosina Trifosfato , Diferenciación Celular , Células Madre Pluripotentes Inducidas , Mutación , Organoides , Retina , Retinoblastoma , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Organoides/metabolismo , Retina/metabolismo , Retina/citología , Retinoblastoma/genética , Retinoblastoma/metabolismo , Adenosina Trifosfato/metabolismo , Diferenciación Celular/genética , Mutación/genética , Heterocigoto , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Glucólisis/genética , Proteínas de Unión a Retinoblastoma/genética , Proteínas de Unión a Retinoblastoma/metabolismo
2.
Methods Mol Biol ; 2800: 67-74, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38709478

RESUMEN

The study of cell signaling within tissues can be enhanced using highly multiplexed immunohistochemistry to localize the presence and spatial distribution of numerous pathways of interest simultaneously. Additional data can also be gained by placing the identified proteins into the context of adjacent structures, stroma, and interacting partners. Here, we outline a protocol for using the recently described IBEX method on tissues. This is an open and simple cyclic immunohistochemistry approach suited to this application. We describe a simplified protocol and provide guidance on the method, using a 12-marker panel on human retina to demonstrate the approach.


Asunto(s)
Inmunohistoquímica , Retina , Transducción de Señal , Humanos , Inmunohistoquímica/métodos , Retina/metabolismo , Retina/citología , Biomarcadores , Imagen Molecular/métodos
3.
Int J Mol Sci ; 25(9)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38732096

RESUMEN

Alterations in intraocular and external pressure critically involve the pathogenesis of glaucoma, traumatic retinal injury (TRI), and other retinal disorders, and retinal neurons have been reported to express multiple mechanical-sensitive channels (MSCs) in recent decades. However, the role of MSCs in visual functions and pressure-related retinal conditions has been unclear. This review will focus on the variety and functional significance of the MSCs permeable to K+, Na+, and Ca2+, primarily including the big potassium channel (BK); the two-pore domain potassium channels TRAAK and TREK; Piezo; the epithelial sodium channel (ENaC); and the transient receptor potential channels vanilloid TRPV1, TRPV2, and TRPV4 in retinal photoreceptors, bipolar cells, horizontal cells, amacrine cells, and ganglion cells. Most MSCs do not directly mediate visual signals in vertebrate retinas. On the other hand, some studies have shown that MSCs can open in physiological conditions and regulate the activities of retinal neurons. While these data reasonably predict the crossing of visual and mechanical signals, how retinal light pathways deal with endogenous and exogenous mechanical stimulation is uncertain.


Asunto(s)
Canales Iónicos , Neuronas Retinianas , Humanos , Animales , Canales Iónicos/metabolismo , Neuronas Retinianas/metabolismo , Mecanotransducción Celular , Retina/metabolismo , Retina/citología
4.
Glia ; 72(7): 1356-1370, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38591270

RESUMEN

Astrocytes throughout the central nervous system are heterogeneous in both structure and function. This diversity leads to tissue-specific specialization where morphology is adapted to the surrounding neuronal circuitry, as seen in Bergman glia of the cerebellum and Müller glia of the retina. Because morphology can be a differentiating factor for cellular classification, we recently developed a mouse where glial-fibrillary acidic protein (GFAP)-expressing cells stochastically label for full membranous morphology. Here we utilize this tool to investigate whether morphological and electrophysiological features separate types of mouse retinal astrocytes. In this work, we report on a novel glial population found in the inner plexiform layer and ganglion cell layer which expresses the canonical astrocyte markers GFAP, S100ß, connexin-43, Sox2 and Sox9. Apart from their retinal layer localization, these cells are unique in their radial distribution. They are notably absent from the mid-retina but are heavily concentrated near the optic nerve head, and to a lesser degree the peripheral retina. Additionally, their morphology is distinct from both nerve fiber layer astrocytes and Müller glia, appearing more similar to amacrine cells. Despite this structural similarity, these cells lack protein expression of common neuronal markers. Additionally, they do not exhibit action potentials, but rather resemble astrocytes and Müller glia in their small amplitude, graded depolarization to both light onset and offset. Their structure, protein expression, physiology, and intercellular connections suggest that these cells are astrocytic, displaced from their counterparts in the nerve fiber layer. As such, we refer to these cells as displaced retinal astrocytes.


Asunto(s)
Astrocitos , Ratones Transgénicos , Retina , Animales , Astrocitos/metabolismo , Astrocitos/fisiología , Retina/citología , Retina/metabolismo , Retina/fisiología , Ratones , Proteína Ácida Fibrilar de la Glía/metabolismo , Ratones Endogámicos C57BL , Potenciales de Acción/fisiología
5.
Nat Commun ; 15(1): 3567, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38670973

RESUMEN

The emergence of retinal progenitor cells and differentiation to various retinal cell types represent fundamental processes during retinal development. Herein, we provide a comprehensive single cell characterisation of transcriptional and chromatin accessibility changes that underline retinal progenitor cell specification and differentiation over the course of human retinal development up to midgestation. Our lineage trajectory data demonstrate the presence of early retinal progenitors, which transit to late, and further to transient neurogenic progenitors, that give rise to all the retinal neurons. Combining single cell RNA-Seq with spatial transcriptomics of early eye samples, we demonstrate the transient presence of early retinal progenitors in the ciliary margin zone with decreasing occurrence from 8 post-conception week of human development. In retinal progenitor cells, we identified a significant enrichment for transcriptional enhanced associate domain transcription factor binding motifs, which when inhibited led to loss of cycling progenitors and retinal identity in pluripotent stem cell derived organoids.


Asunto(s)
Diferenciación Celular , Retina , Análisis de la Célula Individual , Células Madre , Humanos , Análisis de la Célula Individual/métodos , Retina/citología , Retina/metabolismo , Células Madre/citología , Células Madre/metabolismo , Organoides/metabolismo , Organoides/citología , Regulación del Desarrollo de la Expresión Génica , Cromatina/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , RNA-Seq , Linaje de la Célula , Transcriptoma
6.
Glia ; 72(7): 1217-1235, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38511347

RESUMEN

Brain function is critically dependent on correct circuit assembly. Microglia are well-known for their important roles in immunological defense and neural plasticity, but whether they can also mediate experience-induced correction of miswired circuitry is unclear. Ten-m3 knockout (KO) mice display a pronounced and stereotyped visuotopic mismapping of ipsilateral retinal inputs in their visual thalamus, providing a useful model to probe circuit correction mechanisms. Environmental enrichment (EE) commenced around birth, but not later in life, can drive a partial correction of the most mismapped retinal inputs in Ten-m3 KO mice. Here, we assess whether enrichment unlocks the capacity for microglia to selectively engulf and remove miswired circuitry, and the timing of this effect. Expression of the microglial-associated lysosomal protein CD68 showed a clear enrichment-driven, spatially restricted change which had not commenced at postnatal day (P)18, was evident at P21, more robust at P25, and had ceased by P30. This was observed specifically at the corrective pruning site and was absent at a control site. An engulfment assay at the corrective pruning site in P25 mice showed EE-driven microglial-uptake of the mismapped axon terminals. This was temporally and spatially specific, as no enrichment-driven microglial engulfment was seen in P18 KO mice, nor the control locus. The timecourse of the EE-driven corrective pruning as determined anatomically, aligned with this pattern of microglia reactivity and engulfment. Collectively, these findings show experience can drive targeted microglial engulfment of miswired neural circuitry during a restricted postnatal window. This may have important therapeutic implications for neurodevelopmental conditions involving aberrant neural connectivity.


Asunto(s)
Animales Recién Nacidos , Ratones Noqueados , Microglía , Animales , Microglía/metabolismo , Microglía/fisiología , Ratones Endogámicos C57BL , Ratones , Plasticidad Neuronal/fisiología , Antígenos CD/metabolismo , Vías Visuales/fisiología , Antígenos de Diferenciación Mielomonocítica/metabolismo , Retina/fisiología , Retina/citología , Retina/metabolismo , Ambiente , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/deficiencia , Molécula CD68
7.
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
8.
Stem Cells Dev ; 33(9-10): 201-213, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38390839

RESUMEN

Because derivation of retinal organoids (ROs) and transplantation are frequently split between geographically distant locations, we developed a special shipping device and protocol capable of the organoids' delivery to any location. Human embryonic stem cell (hESC)-derived ROs were differentiated from the hESC line H1 (WA01), shipped overnight to another location, and then transplanted into the subretinal space of blind immunodeficient retinal degeneration (RD) rats. Development of transplants was monitored by spectral-domain optical coherence tomography. Visual function was accessed by optokinetic tests and superior colliculus (SC) electrophysiology. Cryostat sections through transplants were stained with hematoxylin and eosin; or processed for immunohistochemistry to label human donor cells, retinal cell types, and synaptic markers. After transplantation, ROs integrated into the host RD retina, formed functional photoreceptors, and improved vision in rats with advanced RD. The survival and vision improvement are comparable with our previous results of hESC-ROs without a long-distance delivery. Furthermore, for the first time in the stem cell transplantation field, we demonstrated that the response heatmap on the SC showed a similar shape to the location of the transplant in the host retina, which suggested the point-to-point projection of the transplant from the retina to SC. In conclusion, our results showed that using our special device and protocol, the hESC-derived ROs can be shipped over long distance and are capable of survival and visual improvement after transplantation into the RD rats. Our data provide a proof-of-concept for stem cell replacement as a therapy for RD patients.


Asunto(s)
Células Madre Embrionarias Humanas , Organoides , Retina , Degeneración Retiniana , Animales , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/trasplante , Degeneración Retiniana/terapia , Degeneración Retiniana/patología , Humanos , Organoides/citología , Organoides/trasplante , Ratas , Retina/citología , Retina/patología , Diferenciación Celular , Trasplante de Células Madre/métodos , Supervivencia Celular , Tomografía de Coherencia Óptica
9.
Nature ; 626(8001): 1084-1093, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38355799

RESUMEN

The house mouse (Mus musculus) is an exceptional model system, combining genetic tractability with close evolutionary affinity to humans1,2. Mouse gestation lasts only 3 weeks, during which the genome orchestrates the astonishing transformation of a single-cell zygote into a free-living pup composed of more than 500 million cells. Here, to establish a global framework for exploring mammalian development, we applied optimized single-cell combinatorial indexing3 to profile the transcriptional states of 12.4 million nuclei from 83 embryos, precisely staged at 2- to 6-hour intervals spanning late gastrulation (embryonic day 8) to birth (postnatal day 0). From these data, we annotate hundreds of cell types and explore the ontogenesis of the posterior embryo during somitogenesis and of kidney, mesenchyme, retina and early neurons. We leverage the temporal resolution and sampling depth of these whole-embryo snapshots, together with published data4-8 from earlier timepoints, to construct a rooted tree of cell-type relationships that spans the entirety of prenatal development, from zygote to birth. Throughout this tree, we systematically nominate genes encoding transcription factors and other proteins as candidate drivers of the in vivo differentiation of hundreds of cell types. Remarkably, the most marked temporal shifts in cell states are observed within one hour of birth and presumably underlie the massive physiological adaptations that must accompany the successful transition of a mammalian fetus to life outside the womb.


Asunto(s)
Animales Recién Nacidos , Embrión de Mamíferos , Desarrollo Embrionario , Gástrula , Análisis de la Célula Individual , Imagen de Lapso de Tiempo , Animales , Femenino , Ratones , Embarazo , Animales Recién Nacidos/embriología , Animales Recién Nacidos/genética , Diferenciación Celular/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Desarrollo Embrionario/genética , Gástrula/citología , Gástrula/embriología , Gastrulación/genética , Riñón/citología , Riñón/embriología , Mesodermo/citología , Mesodermo/enzimología , Neuronas/citología , Neuronas/metabolismo , Retina/citología , Retina/embriología , Somitos/citología , Somitos/embriología , Factores de Tiempo , Factores de Transcripción/genética , Transcripción Genética , Especificidad de Órganos/genética
10.
Sci Rep ; 14(1): 684, 2024 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-38182646

RESUMEN

Stargardt disease type 1 (STGD1), the most common form of hereditary macular dystrophy, can be caused by biallelic combinations of over 2200 variants in the ABCA4 gene. This leads to reduced or absent ABCA4 protein activity, resulting in toxic metabolite accumulation in the retina and damage of the retinal pigment epithelium and photoreceptors. Approximately 21% of all ABCA4 variants that contribute to disease influence ABCA4 pre-mRNA splicing. This emphasizes the need for therapies to restore disrupted ABCA4 splicing and halt STGD1 progression. Previously, QR-1011, an antisense oligonucleotide (AON), successfully corrected splicing abnormalities and restored normal ABCA4 protein translation in human retinal organoids carrying the prevalent disease-causing variant c.5461-10T>C in ABCA4. Here, we investigated whether QR-1011 could also correct splicing in four less common non-canonical splice site (NCSS) variants flanking ABCA4 exon 39: c.5461-8T>G, c.5461-6T>C, c.5584+5G>A and c.5584+6T>C. We administered QR-1011 and three other AONs to midigene-transfected cells and demonstrate that QR-1011 had the most pronounced effect on splicing compared to the others. Moreover, QR-1011 significantly increased full-length ABCA4 transcript levels for c.5461-8T>G and c.5584+6T>C. Splicing restoration could not be achieved in the other two variants, suggesting their more severe effect on splicing. Overall, QR-1011, initially developed for a single ABCA4 variant, exhibited potent splice correction capabilities for two additional severe NCSS variants nearby. This suggests the possibility of a broader therapeutic impact of QR-1011 extending beyond its original target and highlights the potential for treating a larger population of STGD1 patients affected by multiple severe ABCA4 variants with a single AON.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Oligodesoxirribonucleótidos Antisentido , Organoides , Enfermedad de Stargardt , Humanos , Transportadoras de Casetes de Unión a ATP/genética , Exones , Retina/citología , Empalme del ARN/efectos de los fármacos , Enfermedad de Stargardt/tratamiento farmacológico , Enfermedad de Stargardt/genética , Oligodesoxirribonucleótidos Antisentido/farmacología , Organoides/efectos de los fármacos
11.
J Cell Biol ; 223(2)2024 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-38126997

RESUMEN

Lattice cells (LCs) in the developing Drosophila retina change shape before attaining final form. Previously, we showed that repeated contraction and expansion of apical cell contacts affect these dynamics. Here, we describe another factor, the assembly of a Rho1-dependent medioapical actomyosin ring formed by nodes linked by filaments that contract the apical cell area. Cell area contraction alternates with relaxation, generating pulsatile changes in cell area that exert force on neighboring LCs. Moreover, Rho1 signaling is sensitive to mechanical changes, becoming active when tension decreases and cells expand, while the negative regulator RhoGAP71E accumulates when tension increases and cells contract. This results in cycles of cell area contraction and relaxation that are reciprocally synchronized between adjacent LCs. Thus, mechanically sensitive Rho1 signaling controls pulsatile medioapical actomyosin contraction and coordinates cell behavior across the epithelium. Disrupting the kinetics of pulsing can lead to developmental errors, suggesting this process controls cell shape and tissue integrity during epithelial morphogenesis of the retina.


Asunto(s)
Actomiosina , Drosophila , Ojo , Animales , Citoesqueleto de Actina/fisiología , Actomiosina/fisiología , Citocinesis , Drosophila/embriología , Morfogénesis , Ojo/embriología , Proteínas de Unión al GTP rho/fisiología , Proteínas de Drosophila/fisiología , Retina/citología
12.
Nature ; 624(7991): 415-424, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38092908

RESUMEN

The basic plan of the retina is conserved across vertebrates, yet species differ profoundly in their visual needs1. Retinal cell types may have evolved to accommodate these varied needs, but this has not been systematically studied. Here we generated and integrated single-cell transcriptomic atlases of the retina from 17 species: humans, two non-human primates, four rodents, three ungulates, opossum, ferret, tree shrew, a bird, a reptile, a teleost fish and a lamprey. We found high molecular conservation of the six retinal cell classes (photoreceptors, horizontal cells, bipolar cells, amacrine cells, retinal ganglion cells (RGCs) and Müller glia), with transcriptomic variation across species related to evolutionary distance. Major subclasses were also conserved, whereas variation among cell types within classes or subclasses was more pronounced. However, an integrative analysis revealed that numerous cell types are shared across species, based on conserved gene expression programmes that are likely to trace back to an early ancestral vertebrate. The degree of variation among cell types increased from the outer retina (photoreceptors) to the inner retina (RGCs), suggesting that evolution acts preferentially to shape the retinal output. Finally, we identified rodent orthologues of midget RGCs, which comprise more than 80% of RGCs in the human retina, subserve high-acuity vision, and were previously believed to be restricted to primates2. By contrast, the mouse orthologues have large receptive fields and comprise around 2% of mouse RGCs. Projections of both primate and mouse orthologous types are overrepresented in the thalamus, which supplies the primary visual cortex. We suggest that midget RGCs are not primate innovations, but are descendants of evolutionarily ancient types that decreased in size and increased in number as primates evolved, thereby facilitating high visual acuity and increased cortical processing of visual information.


Asunto(s)
Evolución Biológica , Neuronas , Retina , Vertebrados , Visión Ocular , Animales , Humanos , Neuronas/clasificación , Neuronas/citología , Neuronas/fisiología , Retina/citología , Retina/fisiología , Células Ganglionares de la Retina/clasificación , Análisis de Expresión Génica de una Sola Célula , Vertebrados/fisiología , Visión Ocular/fisiología , Especificidad de la Especie , Células Amacrinas/clasificación , Células Fotorreceptoras/clasificación , Células Ependimogliales/clasificación , Células Bipolares de la Retina/clasificación , Percepción Visual
13.
J Biol Chem ; 299(12): 105461, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37977220

RESUMEN

Müller glial cells, which are the most predominant glial subtype in the retina, play multiple important roles, including the maintenance of structural integrity, homeostasis, and physiological functions of the retina. We have previously found that the Rax homeoprotein is expressed in postnatal and mature Müller glial cells in the mouse retina. However, the function of Rax in postnatal and mature Müller glial cells remains to be elucidated. In the current study, we first investigated Rax function in retinal development using retroviral lineage analysis and found that Rax controls the specification of late-born retinal cell types, including Müller glial cells in the postnatal retina. We next generated Rax tamoxifen-induced conditional KO (Rax iCKO) mice, where Rax can be depleted in mTFP-labeled Müller glial cells upon tamoxifen treatment, by crossing Raxflox/flox mice with Rlbp1-CreERT2 mice, which we have produced. Immunohistochemical analysis showed a characteristic of reactive gliosis and enhanced gliosis of Müller glial cells in Rax iCKO retinas under normal and stress conditions, respectively. We performed RNA-seq analysis on mTFP-positive cells purified from the Rax iCKO retina and found significantly reduced expression of suppressor of cytokinesignaling-3 (Socs3). Reporter gene assays showed that Rax directly transactivates the Socs3 promoter. We observed decreased expression of Socs3 in Müller glial cells of Rax iCKO retinas by immunostaining. Taken together, the present results suggest that Rax suppresses inflammation in Müller glial cells by transactivating Socs3. This study sheds light on the transcriptional regulatory mechanisms underlying retinal Müller glial cell homeostasis.


Asunto(s)
Células Ependimogliales , Proteínas del Ojo , Proteínas de Homeodominio , Homeostasis , Retina , Factores de Transcripción , Animales , Ratones , Células Ependimogliales/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Gliosis/genética , Gliosis/metabolismo , Gliosis/patología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Homeostasis/genética , Retina/citología , Retina/crecimiento & desarrollo , Retina/metabolismo , Retina/patología , RNA-Seq , Tamoxifeno/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional
14.
Nature ; 623(7986): 381-386, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37880369

RESUMEN

To maintain a stable and clear image of the world, our eyes reflexively follow the direction in which a visual scene is moving. Such gaze-stabilization mechanisms reduce image blur as we move in the environment. In non-primate mammals, this behaviour is initiated by retinal output neurons called ON-type direction-selective ganglion cells (ON-DSGCs), which detect the direction of image motion and transmit signals to brainstem nuclei that drive compensatory eye movements1. However, ON-DSGCs have not yet been identified in the retina of primates, raising the possibility that this reflex is mediated by cortical visual areas. Here we mined single-cell RNA transcriptomic data from primate retina to identify a candidate ON-DSGC. We then combined two-photon calcium imaging, molecular identification and morphological analysis to reveal a population of ON-DSGCs in the macaque retina. The morphology, molecular signature and GABA (γ-aminobutyric acid)-dependent mechanisms that underlie direction selectivity in primate ON-DSGCs are highly conserved with those in other mammals. We further identify a candidate ON-DSGC in human retina. The presence of ON-DSGCs in primates highlights the need to examine the contribution of subcortical retinal mechanisms to normal and aberrant gaze stabilization in the developing and mature visual system.


Asunto(s)
Movimientos Oculares , Macaca , Retina , Células Ganglionares de la Retina , Animales , Humanos , Movimientos Oculares/fisiología , Estimulación Luminosa , Retina/citología , Retina/fisiología , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/fisiología , Movimiento (Física) , Análisis de Expresión Génica de una Sola Célula , Ácido gamma-Aminobutírico/metabolismo , Señalización del Calcio , Fijación Ocular/fisiología
15.
Sci Rep ; 13(1): 13239, 2023 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-37580467

RESUMEN

The endosomal-lysosomal system (ELS), which carries out cellular processes such as cellular waste degradation via autophagy, is essential for cell homeostasis. ELS inefficiency leads to augmented levels of damaged organelles and intracellular deposits. Consequently, the modulation of autophagic flux has been recognized as target to remove damaging cell waste. Recently, we showed that cysteinyl leukotriene receptor 1 (CysLTR1) antagonist application increases the autophagic flux in the retinal pigment epithelial cell line ARPE-19. Consequently, we investigated the effect of CysLTR1 inhibition-driven autophagy induction on aggregated proteins in ARPE-19 cells using flow cytometry analysis. A subset of ARPE-19 cells expressed CysLTR1 on the surface (SE+); these cells showed increased levels of autophagosomes, late endosomes/lysosomes, aggregated proteins, and autophagy as well as decreased reactive oxygen species (ROS) formation. Furthermore, CysLTR1 inhibition for 24 h using the antagonist zafirlukast decreased the quantities of autophagosomes, late endosomes/lysosomes, aggregated proteins and ROS in CysLTR1 SE- and SE+ cells. We concluded that high levels of plasma membrane-localized CysLTR1 indicate an increased amount of aggregated protein, which raises the rate of autophagic flux. Furthermore, CysLTR1 antagonist application potentially mimics the physiological conditions observed in CysLTR1 SE+ cells and can be considered as strategy to dampen cellular aging.


Asunto(s)
Autofagosomas , Autofagia , Células Epiteliales , Autofagosomas/metabolismo , Células Epiteliales/metabolismo , Lisosomas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Retina/citología
16.
Nature ; 620(7974): 615-624, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37558872

RESUMEN

The concomitant occurrence of tissue growth and organization is a hallmark of organismal development1-3. This often means that proliferating and differentiating cells are found at the same time in a continuously changing tissue environment. How cells adapt to architectural changes to prevent spatial interference remains unclear. Here, to understand how cell movements that are key for growth and organization are orchestrated, we study the emergence of photoreceptor neurons that occur during the peak of retinal growth, using zebrafish, human tissue and human organoids. Quantitative imaging reveals that successful retinal morphogenesis depends on the active bidirectional translocation of photoreceptors, leading to a transient transfer of the entire cell population away from the apical proliferative zone. This pattern of migration is driven by cytoskeletal machineries that differ depending on the direction: microtubules are exclusively required for basal translocation, whereas actomyosin is involved in apical movement. Blocking the basal translocation of photoreceptors induces apical congestion, which hampers the apical divisions of progenitor cells and leads to secondary defects in lamination. Thus, photoreceptor migration is crucial to prevent competition for space, and to allow concurrent tissue growth and lamination. This shows that neuronal migration, in addition to its canonical role in cell positioning4, can be involved in coordinating morphogenesis.


Asunto(s)
Movimiento Celular , Morfogénesis , Células Fotorreceptoras , Retina , Animales , Humanos , Actomiosina/metabolismo , Competencia Celular , Diferenciación Celular , Movimiento Celular/fisiología , Proliferación Celular , Microtúbulos/metabolismo , Morfogénesis/fisiología , Organoides/citología , Organoides/embriología , Células Fotorreceptoras/citología , Células Fotorreceptoras/fisiología , Retina/citología , Retina/embriología , Pez Cebra/embriología
17.
Tohoku J Exp Med ; 261(3): 211-219, 2023 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-37635064

RESUMEN

Cell division control protein 42 (CDC42) modulates insulin secretion and angiogenesis to participate in the pathology of diabetic complications and retinal vascular-associated diseases. This study intended to explore the role of CDC42 in the progression of diabetic retinopathy, and the underlying mechanism. Human retinal microvascular endothelial cells (hRMECs) were cultured in 5.5 mM glucose (normal glucose) or 25 mM glucose (high glucose; HG) medium, respectively. CDC42 overexpression plasmid and small interference RNA (oe-CDC42 and si-CDC42) or corresponding negative controls (oe-NC and si-NC) were transfected into hRMECs under HG. Then, platelet-activating factor C-16 (C16-PAF) (MEK/ERK pathway activator) was added to si-CDC42 or si-NC transfected hRMECs under HG. Our study showed that HG increased CDC42 mRNA and protein, cell viability, invasive cell count, branch points, and tube length but reduced cell apoptosis in hRMECs. CDC42 upregulation enhanced cell viability, invasive cell count, branch points, tube length, p-MEK, and p-ERK, but attenuated cell apoptosis. Downregulation of CDC42 exhibited opposite trends. In addition, C16-PAF also increased cell viability, invasive cell count, branch points, and tube length, p-MEK, and p-ERK, but retarded cell apoptosis. Notably, C16-PAF diminished the effect of CDC42 downregulation on the above-mentioned functions in hRMECs under HG. Conclusively, CDC42 promotes HG-induced hRMEC viability and invasion, as well as angiogenesis, but inhibits apoptosis by activating the MEK/ERK pathway, which may be responsible for the progression of diabetic retinopathy.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Humanos , División Celular , Diabetes Mellitus/patología , Retinopatía Diabética/genética , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Células Endoteliales/metabolismo , Glucosa/metabolismo , Sistema de Señalización de MAP Quinasas , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/farmacología , Retina/citología , Retina/metabolismo
18.
Biosystems ; 225: 104867, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36792004

RESUMEN

Perception of color by humans and other primates is a complex problem, studied by neurophysiology, psychophysiology, psycholinguistics, and even philosophy. Being mostly trichromats, simian primates have three types of opsin proteins, expressed in cone neurons in the eye, which allow for the sensing of color as the physical wavelength of light. Further, in neural networks of the retina, the coding principle changes from three types of sensor proteins to two opponent channels: activity of one type of neuron encode the evolutionarily ancient blue-yellow axis of color stimuli, and another more recent evolutionary channel, encoding the axis of red-green color stimuli. Both color channels are distinctive in neural organization at all levels from the eye to the neocortex, where it is thought that the perception of color (as philosophical qualia) emerges from the activity of some neuron ensembles. Here, using data from neurophysiology as a starting point, we propose a hypothesis on how the perception of color can be encoded in the activity of certain neurons in the neocortex. These conceptual neurons, herein referred to as 'color neurons', code only the hue of the color of visual stimulus, similar to place cells and number neurons, already described in primate brains. A case study with preliminary, but direct, evidence for existing conceptual color neurons in the human brain was published in 2008. We predict that the upcoming studies in non-human primates will be more extensive and provide a more detailed description of conceptual color neurons.


Asunto(s)
Neocórtex , Primates , Percepción Visual , Animales , Neocórtex/citología , Neocórtex/fisiología , Primates/fisiología , Color , Retina/citología , Retina/fisiología , Evolución Biológica
19.
Nucleic Acids Res ; 51(5): 2151-2176, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36715342

RESUMEN

How the diverse neural cell types emerge from multipotent neural progenitor cells during central nervous system development remains poorly understood. Recent scRNA-seq studies have delineated the developmental trajectories of individual neural cell types in many neural systems including the neural retina. Further understanding of the formation of neural cell diversity requires knowledge about how the epigenetic landscape shifts along individual cell lineages and how key transcription factors regulate these changes. In this study, we dissect the changes in the epigenetic landscape during early retinal cell differentiation by scATAC-seq and identify globally the enhancers, enriched motifs, and potential interacting transcription factors underlying the cell state/type specific gene expression in individual lineages. Using CUT&Tag, we further identify the enhancers bound directly by four key transcription factors, Otx2, Atoh7, Pou4f2 and Isl1, including those dependent on Atoh7, and uncover the sequential and combinatorial interactions of these factors with the epigenetic landscape to control gene expression along individual retinal cell lineages such as retinal ganglion cells (RGCs). Our results reveal a general paradigm in which transcription factors collaborate and compete to regulate the emergence of distinct retinal cell types such as RGCs from multipotent retinal progenitor cells (RPCs).


Asunto(s)
Retina , Factores de Transcripción , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Retina/citología , Retina/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
FEBS Lett ; 597(3): 427-436, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36349512

RESUMEN

Trimethylation of histone H3 at lysine 36 (H3K36me3) is associated with active transcription. We used mouse retinal explant cultures and shRNA to investigate the roles of Setd2 and Setd5, which encode H3K36me3 methyltransferases, in retinal development. We found that shSetd5 caused abnormal retinal structures and reduced rods and Müller cells, whereas shSetd2 did not cause any abnormalities. The mutant SETD5 lacking the SET domain failed to reverse the phenotypes observed in the shSetd5-expressing retinas, while SETD5S1257*, which does not interact with HDAC3 and PAF1 complexes, rescued proliferation, but not apoptosis, induced by shSetd5. Taken together, we found that Setd5, but not Setd2, is essential for sustaining retinal cell survival and proliferation, and the SET domain of SETD5 is pivotal for both functions.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Histonas , Retina , Animales , Ratones , Proliferación Celular , Supervivencia Celular , Histonas/genética , Neuronas , Retina/citología , Retina/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA