Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 4119, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37433773

RESUMEN

The accumulation of atypical, cytotoxic 1-deoxysphingolipids (1-dSLs) has been linked to retinal diseases such as diabetic retinopathy and Macular Telangiectasia Type 2. However, the molecular mechanisms by which 1-dSLs induce toxicity in retinal cells remain poorly understood. Here, we integrate bulk and single-nucleus RNA-sequencing to define biological pathways that modulate 1-dSL toxicity in human retinal organoids. Our results demonstrate that 1-dSLs differentially activate signaling arms of the unfolded protein response (UPR) in photoreceptor cells and Müller glia. Using a combination of pharmacologic activators and inhibitors, we show that sustained PERK signaling through the integrated stress response (ISR) and deficiencies in signaling through the protective ATF6 arm of the UPR are implicated in 1-dSL-induced photoreceptor toxicity. Further, we demonstrate that pharmacologic activation of ATF6 mitigates 1-dSL toxicity without impacting PERK/ISR signaling. Collectively, our results identify new opportunities to intervene in 1-dSL linked diseases through targeting different arms of the UPR.


Asunto(s)
Retinopatía Diabética , Telangiectasia Retiniana , Humanos , Esfingolípidos , Respuesta de Proteína Desplegada
2.
Glia ; 71(10): 2372-2382, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37335016

RESUMEN

In the retina, microglia are resident immune cells that are essential for development and function. Retinal microglia play a central role in mediating pathological degeneration in diseases such as glaucoma, retinitis pigmentosa, age-related neurodegeneration, ischemic retinopathy, and diabetic retinopathy. Current models of mature human retinal organoids (ROs) derived from iPS cell (hiPSC) do not contain resident microglia integrated into retinal layers. Increasing cellular diversity in ROs by including resident microglia would more accurately represent the native retina and better model diseases in which microglia play a key role. In this study, we develop a new 3D in vitro tissue model of microglia-containing retinal organoids by co-culturing ROs and hiPSC-derived macrophage precursor cells (MPCs). We optimized the parameters for successful integration of MPCs into retinal organoids. We show that while in the ROs, MPCs migrate to the equivalent of the outer plexiform layer where retinal microglia cells reside in healthy retinal tissue. While there, they develop a mature morphology characterized by small cell bodies and long branching processes which is only observed in vivo. During this maturation process these MPCs cycle through an activated phase followed by a stable mature microglial phase as seen by the down regulation of pro-inflammatory cytokines and upregulation of anti-inflammatory cytokines. Finally, we characterized mature ROs with integrated MPCs using RNAseq showing an enrichment of cell-type specific microglia markers. We propose that this co-culture system may be useful for understanding the pathogenesis of retinal diseases involving retinal microglia and for drug discovery directly in human tissue.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedades de la Retina , Humanos , Células Madre Pluripotentes Inducidas/patología , Microglía/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Retina , Enfermedades de la Retina/patología , Organoides/patología , Macrófagos/patología , Citocinas/metabolismo
3.
J Clin Invest ; 133(9)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37115691

RESUMEN

Patient-derived induced pluripotent stem cells (iPSCs) provide a powerful tool for identifying cellular and molecular mechanisms of disease. Macular telangiectasia type 2 (MacTel) is a rare, late-onset degenerative retinal disease with an extremely heterogeneous genetic architecture, lending itself to the use of iPSCs. Whole-exome sequencing screens and pedigree analyses have identified rare causative mutations that account for less than 5% of cases. Metabolomic surveys of patient populations and GWAS have linked MacTel to decreased circulating levels of serine and elevated levels of neurotoxic 1-deoxysphingolipids (1-dSLs). However, retina-specific, disease-contributing factors have yet to be identified. Here, we used iPSC-differentiated retinal pigmented epithelial (iRPE) cells derived from donors with or without MacTel to screen for novel cell-intrinsic pathological mechanisms. We show that MacTel iRPE cells mimicked the low serine levels observed in serum from patients with MacTel. Through RNA-Seq and gene set enrichment pathway analysis, we determined that MacTel iRPE cells are enriched in cellular stress pathways and dysregulation of central carbon metabolism. Using respirometry and mitochondrial stress testing, we functionally validated that MacTel iRPE cells had a reduction in mitochondrial function that was independent of defects in serine biosynthesis and 1-dSL accumulation. Thus, we identified phenotypes that may constitute alternative disease mechanisms beyond the known serine/sphingolipid pathway.


Asunto(s)
Retinopatía Diabética , Células Madre Pluripotentes Inducidas , Telangiectasia Retiniana , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Telangiectasia Retiniana/metabolismo , Telangiectasia Retiniana/patología , Retinopatía Diabética/metabolismo , Mitocondrias/metabolismo , Células Epiteliales/metabolismo , Serina/metabolismo
4.
Nature ; 557(7705): 375-380, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29743677

RESUMEN

The transcriptional programs that establish neuronal identity evolved to produce the rich diversity of neuronal cell types that arise sequentially during development. Remarkably, transient expression of certain transcription factors can also endow non-neural cells with neuronal properties. The relationship between reprogramming factors and the transcriptional networks that produce neuronal identity and diversity remains largely unknown. Here, from a screen of 598 pairs of transcription factors, we identify 76 pairs of transcription factors that induce mouse fibroblasts to differentiate into cells with neuronal features. By comparing the transcriptomes of these induced neuronal cells (iN cells) with those of endogenous neurons, we define a 'core' cell-autonomous neuronal signature. The iN cells also exhibit diversity; each transcription factor pair produces iN cells with unique transcriptional patterns that can predict their pharmacological responses. By linking distinct transcription factor input 'codes' to defined transcriptional outputs, this study delineates cell-autonomous features of neuronal identity and diversity and expands the reprogramming toolbox to facilitate engineering of induced neurons with desired patterns of gene expression and related functional properties.


Asunto(s)
Reprogramación Celular/genética , Neuronas/citología , Neuronas/metabolismo , Animales , Fibroblastos/citología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Neuronas/efectos de los fármacos , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Transcriptoma/genética
5.
EMBO J ; 34(11): 1445-55, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-25908841

RESUMEN

The nervous system is comprised of a vast diversity of distinct neural cell types. Differences between neuronal subtypes drive the assembly of neuronal circuits and underlie the subtype specificity of many neurological diseases. Yet, because neurons are irreversibly post-mitotic and not readily available from patients, it has not been feasible to study specific subtypes of human neurons in larger numbers. A powerful means to study neuronal diversity and neurological disease is to establish methods to produce desired neuronal subtypes in vitro. Traditionally this has been accomplished by treating pluripotent or neural stem cells with growth factors and morphogens that recapitulate exogenous developmental signals. These approaches often require extended periods of culture, which can limit their utility. However, more recently, it has become possible to produce neurons directly from fibroblasts using transcription factors and/or microRNAs. This technique referred to as direct reprogramming or transdifferentiation has proven to be a rapid, robust, and reproducible method to generate mature neurons of many different subtypes from multiple cell sources. Here, we highlight recent advances in generating neurons of specific subtypes using direct reprogramming and outline various scenarios in which induced neurons may be applied to studies of neuronal function and neurological disease.


Asunto(s)
Ingeniería Celular/métodos , Reprogramación Celular , Fibroblastos , MicroARNs , Células-Madre Neurales , Neuronas , Factores de Transcripción , Animales , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , MicroARNs/biosíntesis , MicroARNs/genética , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/terapia , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
6.
Nat Neurosci ; 18(1): 25-35, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25420069

RESUMEN

Humans and mice detect pain, itch, temperature, pressure, stretch and limb position via signaling from peripheral sensory neurons. These neurons are divided into three functional classes (nociceptors/pruritoceptors, mechanoreceptors and proprioceptors) that are distinguished by their selective expression of TrkA, TrkB or TrkC receptors, respectively. We found that transiently coexpressing Brn3a with either Ngn1 or Ngn2 selectively reprogrammed human and mouse fibroblasts to acquire key properties of these three classes of sensory neurons. These induced sensory neurons (iSNs) were electrically active, exhibited distinct sensory neuron morphologies and matched the characteristic gene expression patterns of endogenous sensory neurons, including selective expression of Trk receptors. In addition, we found that calcium-imaging assays could identify subsets of iSNs that selectively responded to diverse ligands known to activate itch- and pain-sensing neurons. These results offer a simple and rapid means for producing genetically diverse human sensory neurons suitable for drug screening and mechanistic studies.


Asunto(s)
Fibroblastos/fisiología , Sistema Nervioso Periférico/citología , Células Receptoras Sensoriales/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Femenino , Fibroblastos/ultraestructura , Humanos , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Nociceptores/ultraestructura , Técnicas de Placa-Clamp , Sistema Nervioso Periférico/ultraestructura , Embarazo , Receptor trkC/genética , Células Receptoras Sensoriales/ultraestructura , Factor de Transcripción Brn-3A/genética , Factor de Transcripción Brn-3A/fisiología
7.
PLoS Genet ; 8(2): e1002501, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22383890

RESUMEN

During neurogenesis, transcription factors combinatorially specify neuronal fates and then differentiate subtype identities by inducing subtype-specific gene expression profiles. But how is neuronal subtype identity maintained in mature neurons? Modeling this question in two Drosophila neuronal subtypes (Tv1 and Tv4), we test whether the subtype transcription factor networks that direct differentiation during development are required persistently for long-term maintenance of subtype identity. By conditional transcription factor knockdown in adult Tv neurons after normal development, we find that most transcription factors within the Tv1/Tv4 subtype transcription networks are indeed required to maintain Tv1/Tv4 subtype-specific gene expression in adults. Thus, gene expression profiles are not simply "locked-in," but must be actively maintained by persistent developmental transcription factor networks. We also examined the cross-regulatory relationships between all transcription factors that persisted in adult Tv1/Tv4 neurons. We show that certain critical cross-regulatory relationships that had existed between these transcription factors during development were no longer present in the mature adult neuron. This points to key differences between developmental and maintenance transcriptional regulatory networks in individual neurons. Together, our results provide novel insight showing that the maintenance of subtype identity is an active process underpinned by persistently active, combinatorially-acting, developmental transcription factors. These findings have implications for understanding the maintenance of all long-lived cell types and the functional degeneration of neurons in the aging brain.


Asunto(s)
Diferenciación Celular/fisiología , Redes Reguladoras de Genes , Neuronas/citología , Neuronas/fisiología , Factores de Transcripción/fisiología , Envejecimiento/fisiología , Animales , Drosophila , Regulación del Desarrollo de la Expresión Génica , Sistema Nervioso/citología , Sistema Nervioso/embriología , Fenómenos Fisiológicos del Sistema Nervioso , Transducción de Señal
8.
J Neurosci ; 29(12): 3852-64, 2009 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-19321782

RESUMEN

The terminal differentiation of many developing neurons occurs after they innervate their target cells and is triggered by secreted target-derived signals that are transduced by presynaptic cognate receptors. Such retrograde signaling induces the expression of genes that are often distinctive markers of neuronal phenotype and function. However, whether long-term maintenance of neuronal phenotype requires persistent retrograde signaling remains poorly understood. Previously, we demonstrated that retrograde bone morphogenetic protein (BMP) signaling induces expression of a phenotypic marker of Drosophila Tv neurons, the neuropeptide FMRFamide (FMRFa). Here, we used a genetic technique that spatiotemporally targets transgene expression in Drosophila to test the role of persistent BMP signaling in the maintenance of Tv phenotype. We show that expression of dominant blockers of BMP signaling selectively in adult Tv neurons dramatically downregulated FMRFa expression. Moreover, adult-onset expression of mutant Glued, which blocks dynein/dynactin-mediated retrograde axonal transport, eliminated retrograde BMP signaling and dramatically downregulated FMRFa expression. Finally, we found that BMP deprivation did not affect Tv neuron survival and that FMRFa expression fully recovered to control levels after the termination of BMP blockade or Glued expression. Our results show that persistent retrograde BMP signaling is required to induce and to subsequently maintain the expression of a stably expressed phenotypic marker in a subset of mature Drosophila neurons. We postulate that retrograde maintenance of neuronal phenotype is conserved in vertebrates, and as a consequence, neuronal phenotype is likely vulnerable to neurodegenerative disease pathologies that disrupt neuronal connectivity or axonal transport.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Drosophila/metabolismo , Neuronas/citología , Animales , Transporte Axonal , Biomarcadores/metabolismo , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Proteínas Morfogenéticas Óseas/genética , Supervivencia Celular , Regulación hacia Abajo , Drosophila/citología , Drosophila/genética , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/genética , FMRFamida/biosíntesis , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Asociadas a Microtúbulos/genética , Sistema Nervioso/citología , Neuronas/metabolismo , Fenotipo , Transducción de Señal , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA