Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 17.152
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(6): 1387-1401.e13, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38412859

RESUMEN

The Crumbs homolog 1 (CRB1) gene is associated with retinal degeneration, most commonly Leber congenital amaurosis (LCA) and retinitis pigmentosa (RP). Here, we demonstrate that murine retinas bearing the Rd8 mutation of Crb1 are characterized by the presence of intralesional bacteria. While normal CRB1 expression was enriched in the apical junctional complexes of retinal pigment epithelium and colonic enterocytes, Crb1 mutations dampened its expression at both sites. Consequent impairment of the outer blood retinal barrier and colonic intestinal epithelial barrier in Rd8 mice led to the translocation of intestinal bacteria from the lower gastrointestinal (GI) tract to the retina, resulting in secondary retinal degeneration. Either the depletion of bacteria systemically or the reintroduction of normal Crb1 expression colonically rescued Rd8-mutation-associated retinal degeneration without reversing the retinal barrier breach. Our data elucidate the pathogenesis of Crb1-mutation-associated retinal degenerations and suggest that antimicrobial agents have the potential to treat this devastating blinding disease.


Asunto(s)
Proteínas del Tejido Nervioso , Degeneración Retiniana , Animales , Ratones , Traslocación Bacteriana , Proteínas del Ojo/genética , Amaurosis Congénita de Leber/genética , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Retina/metabolismo , Degeneración Retiniana/genética , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología
2.
Cell ; 187(11): 2767-2784.e23, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38733989

RESUMEN

The vasculature of the central nervous system is a 3D lattice composed of laminar vascular beds interconnected by penetrating vessels. The mechanisms controlling 3D lattice network formation remain largely unknown. Combining viral labeling, genetic marking, and single-cell profiling in the mouse retina, we discovered a perivascular neuronal subset, annotated as Fam19a4/Nts-positive retinal ganglion cells (Fam19a4/Nts-RGCs), directly contacting the vasculature with perisomatic endfeet. Developmental ablation of Fam19a4/Nts-RGCs led to disoriented growth of penetrating vessels near the ganglion cell layer (GCL), leading to a disorganized 3D vascular lattice. We identified enriched PIEZO2 expression in Fam19a4/Nts-RGCs. Piezo2 loss from all retinal neurons or Fam19a4/Nts-RGCs abolished the direct neurovascular contacts and phenocopied the Fam19a4/Nts-RGC ablation deficits. The defective vascular structure led to reduced capillary perfusion and sensitized the retina to ischemic insults. Furthermore, we uncovered a Piezo2-dependent perivascular granule cell subset for cerebellar vascular patterning, indicating neuronal Piezo2-dependent 3D vascular patterning in the brain.


Asunto(s)
Cerebelo , Neuronas , Retina , Animales , Femenino , Masculino , Ratones , Cerebelo/metabolismo , Cerebelo/irrigación sanguínea , Cerebelo/citología , Canales Iónicos/metabolismo , Ratones Endogámicos C57BL , Neuronas/metabolismo , Retina/citología , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Vasos Retinianos/metabolismo
3.
Nat Immunol ; 24(4): 700-713, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36807640

RESUMEN

Non-neuronal cells are key to the complex cellular interplay that follows central nervous system insult. To understand this interplay, we generated a single-cell atlas of immune, glial and retinal pigment epithelial cells from adult mouse retina before and at multiple time points after axonal transection. We identified rare subsets in naive retina, including interferon (IFN)-response glia and border-associated macrophages, and delineated injury-induced changes in cell composition, expression programs and interactions. Computational analysis charted a three-phase multicellular inflammatory cascade after injury. In the early phase, retinal macroglia and microglia were reactivated, providing chemotactic signals concurrent with infiltration of CCR2+ monocytes from the circulation. These cells differentiated into macrophages in the intermediate phase, while an IFN-response program, likely driven by microglia-derived type I IFN, was activated across resident glia. The late phase indicated inflammatory resolution. Our findings provide a framework to decipher cellular circuitry, spatial relationships and molecular interactions following tissue injury.


Asunto(s)
Macrófagos , Retina , Animales , Ratones , Retina/lesiones , Retina/metabolismo , Microglía , Sistema Nervioso Central , Monocitos
4.
Cell ; 182(6): 1623-1640.e34, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32946783

RESUMEN

Human organoids recapitulating the cell-type diversity and function of their target organ are valuable for basic and translational research. We developed light-sensitive human retinal organoids with multiple nuclear and synaptic layers and functional synapses. We sequenced the RNA of 285,441 single cells from these organoids at seven developmental time points and from the periphery, fovea, pigment epithelium and choroid of light-responsive adult human retinas, and performed histochemistry. Cell types in organoids matured in vitro to a stable "developed" state at a rate similar to human retina development in vivo. Transcriptomes of organoid cell types converged toward the transcriptomes of adult peripheral retinal cell types. Expression of disease-associated genes was cell-type-specific in adult retina, and cell-type specificity was retained in organoids. We implicate unexpected cell types in diseases such as macular degeneration. This resource identifies cellular targets for studying disease mechanisms in organoids and for targeted repair in human retinas.


Asunto(s)
Diferenciación Celular/genética , Organoides/citología , Organoides/metabolismo , Retina/citología , Retina/metabolismo , Análisis de la Célula Individual/métodos , Sinapsis/fisiología , Transcriptoma/genética , Técnicas de Cultivo de Célula/métodos , Línea Celular , Electrofisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Predisposición Genética a la Enfermedad/genética , Humanos , Hibridación in Situ , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Microscopía Electrónica , Familia de Multigenes , Naftoquinonas , Organoides/efectos de la radiación , Organoides/ultraestructura , Retina/patología , Retina/efectos de la radiación
5.
Cell ; 177(2): 243-255.e15, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30827682

RESUMEN

Mammals cannot see light over 700 nm in wavelength. This limitation is due to the physical thermodynamic properties of the photon-detecting opsins. However, the detection of naturally invisible near-infrared (NIR) light is a desirable ability. To break this limitation, we developed ocular injectable photoreceptor-binding upconversion nanoparticles (pbUCNPs). These nanoparticles anchored on retinal photoreceptors as miniature NIR light transducers to create NIR light image vision with negligible side effects. Based on single-photoreceptor recordings, electroretinograms, cortical recordings, and visual behavioral tests, we demonstrated that mice with these nanoantennae could not only perceive NIR light, but also see NIR light patterns. Excitingly, the injected mice were also able to differentiate sophisticated NIR shape patterns. Moreover, the NIR light pattern vision was ambient-daylight compatible and existed in parallel with native daylight vision. This new method will provide unmatched opportunities for a wide variety of emerging bio-integrated nanodevice designs and applications. VIDEO ABSTRACT.


Asunto(s)
Nanopartículas/uso terapéutico , Células Fotorreceptoras de Vertebrados/fisiología , Visión Ocular/fisiología , Animales , Femenino , Rayos Infrarrojos , Inyecciones/métodos , Luz , Masculino , Mamíferos/fisiología , Ratones , Ratones Endogámicos C57BL , Opsinas/metabolismo , Retina/metabolismo , Retina/fisiología , Células Fotorreceptoras Retinianas Conos/fisiología , Visión Ocular/genética
6.
Cell ; 175(1): 71-84.e18, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30173913

RESUMEN

Light exerts a range of powerful biological effects beyond image vision, including mood and learning regulation. While the source of photic information affecting mood and cognitive functions is well established, viz. intrinsically photosensitive retinal ganglion cells (ipRGCs), the central mediators are unknown. Here, we reveal that the direct effects of light on learning and mood utilize distinct ipRGC output streams. ipRGCs that project to the suprachiasmatic nucleus (SCN) mediate the effects of light on learning, independently of the SCN's pacemaker function. Mood regulation by light, on the other hand, requires an SCN-independent pathway linking ipRGCs to a previously unrecognized thalamic region, termed perihabenular nucleus (PHb). The PHb is integrated in a distinctive circuitry with mood-regulating centers and is both necessary and sufficient for driving the effects of light on affective behavior. Together, these results provide new insights into the neural basis required for light to influence mood and learning.


Asunto(s)
Afecto/efectos de la radiación , Aprendizaje/efectos de la radiación , Luz , Afecto/fisiología , Animales , Encéfalo/fisiología , Ritmo Circadiano , Aprendizaje/fisiología , Ratones , Ratones Endogámicos C57BL , Fototerapia/métodos , Retina/metabolismo , Retina/fisiología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/fisiología , Células Ganglionares de la Retina/efectos de la radiación , Transducción de Señal/fisiología , Núcleo Supraquiasmático/metabolismo , Visión Ocular/fisiología , Vías Visuales/metabolismo , Percepción Visual/fisiología
7.
Cell ; 173(4): 851-863.e16, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29576452

RESUMEN

Hibernating mammals survive hypothermia (<10°C) without injury, a remarkable feat of cellular preservation that bears significance for potential medical applications. However, mechanisms imparting cold resistance, such as cytoskeleton stability, remain elusive. Using the first iPSC line from a hibernating mammal (13-lined ground squirrel), we uncovered cellular pathways critical for cold tolerance. Comparison between human and ground squirrel iPSC-derived neurons revealed differential mitochondrial and protein quality control responses to cold. In human iPSC-neurons, cold triggered mitochondrial stress, resulting in reactive oxygen species overproduction and lysosomal membrane permeabilization, contributing to microtubule destruction. Manipulations of these pathways endowed microtubule cold stability upon human iPSC-neurons and rat (a non-hibernator) retina, preserving its light responsiveness after prolonged cold exposure. Furthermore, these treatments significantly improved microtubule integrity in cold-stored kidneys, demonstrating the potential for prolonging shelf-life of organ transplants. Thus, ground squirrel iPSCs offer a unique platform for bringing cold-adaptive strategies from hibernators to humans in clinical applications. VIDEO ABSTRACT.


Asunto(s)
Adaptación Fisiológica , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/metabolismo , Animales , Diferenciación Celular , Frío , Humanos , Células Madre Pluripotentes Inducidas/citología , Riñón/efectos de los fármacos , Riñón/metabolismo , Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Neuronas/citología , Estrés Oxidativo , Inhibidores de Proteasas/farmacología , Ratas , Especies Reactivas de Oxígeno/metabolismo , Retina/metabolismo , Sciuridae , Transcriptoma , Tubulina (Proteína)/química , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
8.
Annu Rev Cell Dev Biol ; 35: 567-589, 2019 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-31553635

RESUMEN

Deafness or hearing deficits are debilitating conditions. They are often caused by loss of sensory hair cells or defects in their function. In contrast to mammals, nonmammalian vertebrates robustly regenerate hair cells after injury. Studying the molecular and cellular basis of nonmammalian vertebrate hair cell regeneration provides valuable insights into developing cures for human deafness. In this review, we discuss the current literature on hair cell regeneration in the context of other models for sensory cell regeneration, such as the retina and the olfactory epithelium. This comparison reveals commonalities with, as well as differences between, the different regenerating systems, which begin to define a cellular and molecular blueprint of regeneration. In addition, we propose how new technical advances can address outstanding questions in the field.


Asunto(s)
Células Madre Adultas/metabolismo , Oído Interno/metabolismo , Células Ciliadas Auditivas/fisiología , Mucosa Olfatoria/metabolismo , Regeneración/fisiología , Retina/metabolismo , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Citocinas/metabolismo , Oído Interno/citología , Células Ciliadas Auditivas/citología , Células Ciliadas Auditivas/metabolismo , Humanos , Inflamación/genética , Inflamación/metabolismo , Macrófagos/metabolismo , Regeneración/genética , Retina/citología , Transducción de Señal/genética , Transducción de Señal/fisiología , Heridas y Lesiones/genética , Heridas y Lesiones/metabolismo
9.
Cell ; 161(4): 817-32, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25957687

RESUMEN

Rod-derived cone viability factor (RdCVF) is an inactive thioredoxin secreted by rod photoreceptors that protects cones from degeneration. Because the secondary loss of cones in retinitis pigmentosa (RP) leads to blindness, the administration of RdCVF is a promising therapy for this untreatable neurodegenerative disease. Here, we investigated the mechanism underlying the protective role of RdCVF in RP. We show that RdCVF acts through binding to Basigin-1 (BSG1), a transmembrane protein expressed specifically by photoreceptors. BSG1 binds to the glucose transporter GLUT1, resulting in increased glucose entry into cones. Increased glucose promotes cone survival by stimulation of aerobic glycolysis. Moreover, a missense mutation of RdCVF results in its inability to bind to BSG1, stimulate glucose uptake, and prevent secondary cone death in a model of RP. Our data uncover an entirely novel mechanism of neuroprotection through the stimulation of glucose metabolism.


Asunto(s)
Proteínas del Ojo/metabolismo , Glucólisis , Tiorredoxinas/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Basigina/genética , Basigina/metabolismo , Proteínas del Ojo/genética , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Ratones , Mutación Missense , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/citología , Células Fotorreceptoras Retinianas Conos/metabolismo , Retinitis Pigmentosa/metabolismo , Tiorredoxinas/genética
10.
Cell ; 159(3): 584-96, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25417109

RESUMEN

Vascular and nervous systems, two major networks in mammalian bodies, show a high degree of anatomical parallelism and functional crosstalk. During development, neurons guide and attract blood vessels, and consequently this parallelism is established. Here, we identified a noncanonical neurovascular interaction in eye development and disease. VEGFR2, a critical endothelial receptor for VEGF, was more abundantly expressed in retinal neurons than in endothelial cells, including endothelial tip cells. Genetic deletion of VEGFR2 in neurons caused misdirected angiogenesis toward neurons, resulting in abnormally increased vascular density around neurons. Further genetic experiments revealed that this misdirected angiogenesis was attributable to an excessive amount of VEGF protein around neurons caused by insufficient engulfment of VEGF by VEGFR2-deficient neurons. Moreover, absence of neuronal VEGFR2 caused misdirected regenerative angiogenesis in ischemic retinopathy. Thus, this study revealed neurovascular crosstalk and unprecedented cellular regulation of VEGF: retinal neurons titrate VEGF to limit neuronal vascularization. PAPERFLICK:


Asunto(s)
Neovascularización Fisiológica , Neuronas/metabolismo , Retina/crecimiento & desarrollo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Endocitosis , Técnicas de Sustitución del Gen , Ratones , Ratones Noqueados , Neurogénesis , Retina/metabolismo , Retina/patología , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
11.
Cell ; 158(3): 534-48, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25018104

RESUMEN

Depending on endoplasmic reticulum (ER) stress levels, the ER transmembrane multidomain protein IRE1α promotes either adaptation or apoptosis. Unfolded ER proteins cause IRE1α lumenal domain homo-oligomerization, inducing trans autophosphorylation that further drives homo-oligomerization of its cytosolic kinase/endoribonuclease (RNase) domains to activate mRNA splicing of adaptive XBP1 transcription factor. However, under high/chronic ER stress, IRE1α surpasses an oligomerization threshold that expands RNase substrate repertoire to many ER-localized mRNAs, leading to apoptosis. To modulate these effects, we developed ATP-competitive IRE1α Kinase-Inhibiting RNase Attenuators-KIRAs-that allosterically inhibit IRE1α's RNase by breaking oligomers. One optimized KIRA, KIRA6, inhibits IRE1α in vivo and promotes cell survival under ER stress. Intravitreally, KIRA6 preserves photoreceptor functional viability in rat models of ER stress-induced retinal degeneration. Systemically, KIRA6 preserves pancreatic ß cells, increases insulin, and reduces hyperglycemia in Akita diabetic mice. Thus, IRE1α powerfully controls cell fate but can itself be controlled with small molecules to reduce cell degeneration.


Asunto(s)
Estrés del Retículo Endoplásmico , Endorribonucleasas/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Regulación Alostérica , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Endorribonucleasas/química , Endorribonucleasas/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Retina/metabolismo , Ribonucleasas/antagonistas & inhibidores
12.
Cell ; 154(2): 351-64, 2013 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-23870124

RESUMEN

Neuronal growth cones select synaptic partners through interactions with multiple cell surfaces in their environment. Many of these interactions are adhesive, yet it is unclear how growth cones integrate adhesive cues to direct their movements. Here, we examine the mechanisms that enable photoreceptors in the Drosophila visual system to choose synaptic partners. We demonstrate that the classical cadherin, N-cadherin, and an atypical cadherin, Flamingo, act redundantly to instruct the targeting choices made by every photoreceptor axon. These molecules gradually bias the spatial distribution of growth cone filopodia, polarizing each growth cone toward its future synaptic target before direct contact with the target occurs. We demonstrate that these molecules are localized to distinct subcellular domains and create a network of adhesive interactions distributed across many growth cones. Because this network comprises multiple redundant interactions, a complex wiring diagram can be constructed with extraordinary fidelity, suggesting a general principle.


Asunto(s)
Cadherinas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Conos de Crecimiento , Células Fotorreceptoras de Invertebrados/metabolismo , Sinapsis , Animales , Axones/metabolismo , Conos de Crecimiento/metabolismo , Retina/metabolismo
13.
Nature ; 606(7913): 351-357, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35545677

RESUMEN

Death is defined as the irreversible cessation of circulatory, respiratory or brain activity. Many peripheral human organs can be transplanted from deceased donors using protocols to optimize viability. However, tissues from the central nervous system rapidly lose viability after circulation ceases1,2, impeding their potential for transplantation. The time course and mechanisms causing neuronal death and the potential for revival remain poorly defined. Here, using the retina as a model of the central nervous system, we systemically examine the kinetics of death and neuronal revival. We demonstrate the swift decline of neuronal signalling and identify conditions for reviving synchronous in vivo-like trans-synaptic transmission in postmortem mouse and human retina. We measure light-evoked responses in human macular photoreceptors in eyes removed up to 5 h after death and identify modifiable factors that drive reversible and irreversible loss of light signalling after death. Finally, we quantify the rate-limiting deactivation reaction of phototransduction, a model G protein signalling cascade, in peripheral and macular human and macaque retina. Our approach will have broad applications and impact by enabling transformative studies in the human central nervous system, raising questions about the irreversibility of neuronal cell death, and providing new avenues for visual rehabilitation.


Asunto(s)
Fototransducción , Rehabilitación Neurológica , Cambios Post Mortem , Retina , Animales , Autopsia , Muerte Celular/efectos de la radiación , Sistema Nervioso Central/efectos de la radiación , Humanos , Fototransducción/efectos de la radiación , Macaca , Ratones , Retina/metabolismo , Retina/efectos de la radiación , Factores de Tiempo
14.
Trends Biochem Sci ; 48(2): 172-186, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36163145

RESUMEN

Visual phototransduction is the most extensively studied G protein-coupled receptor (GPCR) signaling pathway because of its quantifiable stimulus, non-redundancy of genes, and immense importance in vision. We summarize recent discoveries that have advanced our understanding of rod outer segment (ROS) morphology and the pathological basis of retinal diseases. We have combined recently published cryo-electron tomography (cryo-ET) data on the ROS with structural knowledge on individual proteins to define the precise spatial limitations under which phototransduction occurs. Although hypothetical, the reconstruction of the rod phototransduction system highlights the potential roles of phosphodiesterase 6 (PDE6) and guanylate cyclases (GCs) in maintaining the spacing between ROS discs, suggesting a plausible mechanism by which intrinsic optical signals are generated in the retina.


Asunto(s)
Retina , Segmento Externo de la Célula en Bastón , Segmento Externo de la Célula en Bastón/metabolismo , Segmento Externo de la Célula en Bastón/patología , Especies Reactivas de Oxígeno/metabolismo , Retina/metabolismo , Transducción de Señal , Receptores Acoplados a Proteínas G/metabolismo
15.
Trends Genet ; 40(8): 718-729, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38782642

RESUMEN

Intimate links between epigenome modifications and metabolites allude to a crucial role of cellular metabolism in transcriptional regulation. Retina, being a highly metabolic tissue, adapts by integrating inputs from genetic, epigenetic, and extracellular signals. Precise global epigenomic signatures guide development and homeostasis of the intricate retinal structure and function. Epigenomic and metabolic realignment are hallmarks of aging and highlight a link of the epigenome-metabolism nexus with aging-associated multifactorial traits affecting the retina, including age-related macular degeneration and glaucoma. Here, we focus on emerging principles of epigenomic and metabolic control of retinal gene regulation, with emphasis on their contribution to human disease. In addition, we discuss potential mitigation strategies involving lifestyle changes that target the epigenome-metabolome relationship for maintaining retinal function.


Asunto(s)
Envejecimiento , Epigénesis Genética , Epigenoma , Retina , Humanos , Envejecimiento/genética , Envejecimiento/metabolismo , Epigenoma/genética , Retina/metabolismo , Degeneración Macular/genética , Degeneración Macular/metabolismo , Animales , Regulación de la Expresión Génica/genética , Epigenómica , Glaucoma/genética , Glaucoma/metabolismo , Metilación de ADN/genética
16.
EMBO J ; 42(14): e112657, 2023 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-37184124

RESUMEN

Correct nervous system development depends on the timely differentiation of progenitor cells into neurons. While the output of progenitor differentiation is well investigated at the population and clonal level, how stereotypic or variable fate decisions are during development is still more elusive. To fill this gap, we here follow the fate outcome of single neurogenic progenitors in the zebrafish retina over time using live imaging. We find that neurogenic progenitor divisions produce two daughter cells, one of deterministic and one of probabilistic fate. Interference with the deterministic branch of the lineage affects lineage progression. In contrast, interference with fate probabilities of the probabilistic branch results in a broader range of fate possibilities than in wild-type and involves the production of any neuronal cell type even at non-canonical developmental stages. Combining the interference data with stochastic modelling of fate probabilities revealed that a simple gene regulatory network is able to predict the observed fate decision probabilities during wild-type development. These findings unveil unexpected lineage flexibility that could ensure robust development of the retina and other tissues.


Asunto(s)
Retina , Pez Cebra , Animales , Pez Cebra/genética , Retina/metabolismo , Diferenciación Celular/fisiología , Neurogénesis/fisiología , Células Madre/metabolismo , Linaje de la Célula
17.
Genome Res ; 34(2): 243-255, 2024 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-38355306

RESUMEN

Dozens of variants in the gene for the homeodomain transcription factor (TF) cone-rod homeobox (CRX) are linked with human blinding diseases that vary in their severity and age of onset. How different variants in this single TF alter its function in ways that lead to a range of phenotypes is unclear. We characterized the effects of human disease-causing variants on CRX cis-regulatory function by deploying massively parallel reporter assays (MPRAs) in mouse retina explants carrying knock-ins of two variants, one in the DNA-binding domain (p.R90W) and the other in the transcriptional effector domain (p.E168d2). The degree of reporter gene dysregulation in these mutant Crx retinas corresponds with their phenotypic severity. The two variants affect similar sets of enhancers, and p.E168d2 has distinct effects on silencers. Cis-regulatory elements (CREs) near cone photoreceptor genes are enriched for silencers that are derepressed in the presence of p.E168d2. Chromatin environments of CRX-bound loci are partially predictive of episomal MPRA activity, and distal elements whose accessibility increases later in retinal development are enriched for CREs with silencer activity. We identified a set of potentially pleiotropic regulatory elements that convert from silencers to enhancers in retinas that lack a functional CRX effector domain. Our findings show that phenotypically distinct variants in different domains of CRX have partially overlapping effects on its cis-regulatory function, leading to misregulation of similar sets of enhancers while having a qualitatively different impact on silencers.


Asunto(s)
Proteínas de Homeodominio , Transactivadores , Animales , Humanos , Ratones , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética
18.
Development ; 151(2)2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38095299

RESUMEN

Binocular vision requires the segregation of retinal ganglion cell (RGC) axons extending from the retina into the ipsilateral and contralateral optic tracts. RGC axon segregation occurs at the optic chiasm, which forms at the ventral diencephalon midline. Using expression analyses, retinal explants and genetically modified mice, we demonstrate that CXCL12 (SDF1) is required for axon segregation at the optic chiasm. CXCL12 is expressed by the meninges bordering the optic pathway, and CXCR4 by both ipsilaterally and contralaterally projecting RGCs. CXCL12 or ventral diencephalon meninges potently promoted axon outgrowth from both ipsilaterally and contralaterally projecting RGCs. Further, a higher proportion of axons projected ipsilaterally in mice lacking CXCL12 or its receptor CXCR4 compared with wild-type mice as a result of misrouting of presumptive contralaterally specified RGC axons. Although RGCs also expressed the alternative CXCL12 receptor ACKR3, the optic chiasm developed normally in mice lacking ACKR3. Our data support a model whereby meningeal-derived CXCL12 helps drive axon growth from CXCR4-expressing RGCs towards the diencephalon midline, enabling contralateral axon growth. These findings further our understanding of the molecular and cellular mechanisms controlling optic pathway development.


Asunto(s)
Quiasma Óptico , Células Ganglionares de la Retina , Animales , Ratones , Axones/metabolismo , Diencéfalo , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Vías Visuales
19.
Development ; 151(2)2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38108453

RESUMEN

A growing wealth of data suggest that reactive oxygen species (ROS) signalling might be crucial in conferring embryonic or adult stem cells their specific properties. However, how stem cells control ROS production and scavenging, and how ROS in turn contribute to stemness, remain poorly understood. Using the Xenopus retina as a model system, we first investigated the redox status of retinal stem cells (RSCs). We discovered that they exhibit higher ROS levels compared with progenitors and retinal neurons, and express a set of specific redox genes. We next addressed the question of ROS functional involvement in these cells. Using pharmacological or genetic tools, we demonstrate that inhibition of NADPH oxidase-dependent ROS production increases the proportion of quiescent RSCs. Surprisingly, this is accompanied by an apparent acceleration of the mean division speed within the remaining proliferating pool. Our data further unveil that such impact on RSC cell cycling is achieved by modulation of the Wnt/Hedgehog signalling balance. Altogether, we highlight that RSCs exhibit distinctive redox characteristics and exploit NADPH oxidase signalling to limit quiescence and fine-tune their proliferation rate.


Asunto(s)
Células Madre Adultas , Células-Madre Neurales , Animales , Xenopus laevis/metabolismo , Especies Reactivas de Oxígeno , Proliferación Celular , Proteínas Hedgehog , Retina/metabolismo , Células Madre Adultas/metabolismo , NADPH Oxidasas/genética , Vía de Señalización Wnt
20.
Development ; 151(13)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38994775

RESUMEN

Super-enhancers (SEs) are expansive regions of genomic DNA that regulate the expression of genes involved in cell identity and cell fate. We recently identified developmental stage- and cell type-specific modules within the murine Vsx2 SE. Here, we show that the human VSX2 SE modules have similar developmental stage- and cell type-specific activity in reporter gene assays. By inserting the human sequence of one VSX2 SE module into a mouse with microphthalmia, eye size was rescued. To understand the function of these SE modules during human retinal development, we deleted individual modules in human embryonic stem cells and generated retinal organoids. Deleting one module results in small organoids, recapitulating the small-eyed phenotype of mice with microphthalmia, while deletion of the other module led to disruptions in bipolar neuron development. This prototypical SE serves as a model for understanding developmental stage- and cell type-specific effects of neurogenic transcription factors with complex expression patterns. Moreover, by elucidating the gene regulatory mechanisms, we can begin to examine how dysregulation of these mechanisms contributes to phenotypic diversity and disease.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio , Retina , Factores de Transcripción , Animales , Humanos , Ratones , Elementos de Facilitación Genéticos/genética , Evolución Molecular , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/citología , Microftalmía/genética , Microftalmía/patología , Neurogénesis/genética , Organoides/metabolismo , Retina/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA