Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Exp Neurol ; 379: 114877, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38944331

RESUMEN

In an attempt to repair injured central nervous system (CNS) nerves/tracts, immune cells are recruited into the injury site, but endogenous response in adult mammals is insufficient for promoting regeneration of severed axons. Here, we found that a portion of retinal ganglion cell (RGC) CNS projection neurons that survive after optic nerve crush (ONC) injury are enriched for and upregulate fibronectin (Fn)-interacting integrins Itga5 and ItgaV, and that Fn promotes long-term survival and long-distance axon regeneration of a portion of axotomized adult RGCs in culture. We then show that, Fn is developmentally downregulated in the axonal tracts of optic nerve and spinal cord, but injury-activated macrophages/microglia upregulate Fn while axon regeneration-promoting zymosan augments their recruitment (and thereby increases Fn levels) in the injured optic nerve. Finally, we found that Fn's RGD motif, established to interact with Itga5 and ItgaV, promotes long-term survival and long-distance axon regeneration of adult RGCs after ONC in vivo, with some axons reaching the optic chiasm when co-treated with Rpl7a gene therapy. Thus, experimentally augmenting Fn levels in the injured CNS is a promising approach for therapeutic neuroprotection and axon regeneration of at least a portion of neurons.

2.
Exp Neurol ; 375: 114741, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38395216

RESUMEN

Nuclear factor erythroid 2 like (Nfe2l) gene family members 1-3 mediate cellular response to oxidative stress, including in the central nervous system (CNS). However, neuronal functions of Nfe2l3 are unknown. Here, we comparatively evaluated expression of Nfe2l1, Nfe2l2, and Nfe2l3 in singe cell RNA-seq (scRNA-seq)-profiled cortical and retinal ganglion cell (RGC) CNS projection neurons, investigated whether Nfe2l3 regulates neuroprotection and axon regeneration after CNS injury in vivo, and characterized a gene network associated with Nfe2l3 in neurons. We showed that, Nfe2l3 expression transiently peaks in developing immature cortical and RGC projection neurons, but is nearly abolished in adult neurons and is not upregulated after injury. Furthermore, within the retina, Nfe2l3 is enriched in RGCs, primarily neonatally, and not upregulated in injured RGCs, whereas Nfe2l1 and Nfe2l2 are expressed robustly in other retinal cell types as well and are upregulated in injured RGCs. We also found that, expressing Nfe2l3 in injured RGCs through localized intralocular viral vector delivery promotes neuroprotection and long-distance axon regeneration after optic nerve injury in vivo. Moreover, Nfe2l3 provided a similar extent of neuroprotection and axon regeneration as viral vector-targeting of Pten and Klf9, which are prominent regulators of neuroprotection and long-distance axon regeneration. Finally, we bioinformatically characterized a gene network associated with Nfe2l3 in neurons, which predicted the association of Nfe2l3 with established mechanisms of neuroprotection and axon regeneration. Thus, Nfe2l3 is a novel neuroprotection and axon regeneration-promoting factor with a therapeutic potential for treating CNS injury and disease.


Asunto(s)
Axones , Traumatismos del Nervio Óptico , Humanos , Axones/fisiología , Neuroprotección , Regeneración Nerviosa/fisiología , Células Ganglionares de la Retina/metabolismo , Retina/metabolismo , Traumatismos del Nervio Óptico/metabolismo
3.
Neurosci Lett ; 823: 137662, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38286398

RESUMEN

Numerous micro-RNAs (miRNAs) affect neurodevelopment and neuroprotection, but potential roles of many miRNAs in regulating these processes are still unknown. Here, we used the retinal ganglion cell (RGC) central nervous system (CNS) projection neuron and optic nerve crush (ONC) injury model, to optimize a mature miRNA arm-specific quantification method for characterizing the developmental regulation of miR-1247-5p in RGCs, investigated whether injury affects its expression, and tested whether upregulating miR-1247-5p-mimic in RGCs promotes neuroprotection and axon regeneration. We found that, miR-1247-5p is developmentally-downregulated in RGCs, and is further downregulated after ONC. Importantly, RGC-specific upregulation of miR-1247-5p promoted neuroprotection and axon regeneration after injury in vivo. To gain insight into the underlying mechanisms, we analyzed by bulk-mRNA-seq embryonic and adult RGCs, along with adult RGCs transduced by miR-1247-5p-expressing viral vector, and identified developmentally-regulated cilial and mitochondrial biological processes, which were reinstated to their embryonic levels in adult RGCs by upregulation of miR-1247-5p. Since axon growth is also a developmentally-regulated process, in which mitochondrial dynamics play important roles, it is possible that miR-1247-5p promoted neuroprotection and axon regeneration through regulating mitochondrial functions.


Asunto(s)
MicroARNs , Traumatismos del Nervio Óptico , Humanos , Neuroprotección/fisiología , Axones/metabolismo , Regulación hacia Arriba , Regeneración Nerviosa/genética , Traumatismos del Nervio Óptico/genética , Traumatismos del Nervio Óptico/metabolismo , MicroARNs/genética , MicroARNs/metabolismo
4.
Exp Neurol ; 368: 114510, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37633482

RESUMEN

Ribosomal proteins are involved in neurodevelopment and central nervous system (CNS) disease and injury. However, the roles of specific ribosomal protein subunits in developmental axon growth, and their potential as therapeutic targets for treating CNS injuries, are still poorly understood. Here, we show that ribosomal protein large (Rpl) and small (Rps) subunit genes are substantially (56-fold) enriched amongst the genes, which are downregulated during maturation of retinal ganglion cell (RGC) CNS projection neurons. We also show that Rpl and Rps subunits are highly co-regulated in RGCs, and partially re-upregulated after optic nerve crush (ONC). Because developmental downregulation of ribosomal proteins coincides with developmental decline in neuronal intrinsic axon growth capacity, we hypothesized that Rpl/Rps incomplete re-upregulation after injury may be a part of the cellular response which attempts to reactivate intrinsic axon growth mechanisms. We found that experimentally upregulating Rpl7 and Rpl7A promoted axon regeneration after ONC in vivo. Finally, we characterized gene networks associated with Rpl/Rps, and showed that Rpl7 and Rpl7A belong to the cluster of genes, which are shared between translational and neurodevelopmental biological processes (based on gene-ontology) that are co-downregulated in maturing RGCs during the decline in intrinsic axon growth capacity.


Asunto(s)
Axones , Regeneración Nerviosa , Regulación hacia Arriba , Regeneración Nerviosa/genética , Activación Transcripcional , Proteínas Ribosómicas/genética
5.
Brain Res ; 1811: 148377, 2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37121423

RESUMEN

Analysis of retinal ganglion cells (RGCs) by scRNA-seq is emerging as a state-of-the-art method for studying RGC biology and subtypes, as well as for studying the mechanisms of neuroprotection and axon regeneration in the central nervous system (CNS). Rbpms has been established as a pan-RGC marker, and Spp1 has been established as an αRGC type and macrophage marker. Here, we analyzed by scRNA-seq retinal microglia and macrophages, and found Rbpms+ subpopulations of retinal microglia/macrophages, which pose a potential pitfall in scRNA-seq studies involving RGCs. We performed comparative analysis of cellular identity of the presumed RGC cells isolated in recent scRNA-seq studies, and found that Rbpms+ microglia/macrophages confounded identification of RGCs. We also showed using immunohistological analysis that, Rbpms protein localizes to stress granules in a subpopulation of retinal microglia after optic nerve injury, which was further supported by bioinformatics analysis identifying stress granule-associated genes enriched in the Rbpms+ microglia/macrophages. Our findings suggest that the identification of Rbpms+ RGCs by immunostaining after optic nerve injury should exclude cells in which Rbpms signal is restricted to a subcellular granule, and include only those cells in which the Rbpms signal is labeling cell soma diffusely. Finally, we provide solutions for circumventing this potential pitfall of Rbpms-expressing microglia/macrophages in scRNA-seq studies, by including in RGC and αRGC selection criteria other pan-RGC and αRGC markers.


Asunto(s)
Traumatismos del Nervio Óptico , Células Ganglionares de la Retina , Humanos , Células Ganglionares de la Retina/metabolismo , Traumatismos del Nervio Óptico/metabolismo , Microglía/metabolismo , Axones/metabolismo , Transcriptoma , Regeneración Nerviosa , Macrófagos/metabolismo , Proteínas de Unión al ARN/metabolismo
6.
Brain Res ; 1809: 148368, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37059258

RESUMEN

Collapsin response mediator proteins (Crmps) play roles in neuronal development and axon growth. However, neuronal-specific roles of Crmp1, Crmp4, and Crmp5 in regeneration of injured central nervous system (CNS) axons in vivo are unclear. Here, we analyzed developmental and subtype-specific expression of Crmp genes in retinal ganglion cells (RGCs), tested whether overexpressing Crmp1, Crmp4, or Crmp5 in RGCs through localized intralocular AAV2 delivery promotes axon regeneration after optic nerve injury in vivo, and characterized developmental co-regulation of gene-concept networks associated with Crmps. We found that all Crmp genes are developmentally downregulated in RGCs during maturation. However, while Crmp1, Crmp2, and Crmp4 were expressed to a varying degree in most RGC subtypes, Crmp3 and Crmp5 were expressed only in a small subset of RGC subtypes. We then found that after optic nerve injury, Crmp1, Crmp4, and Crmp5 promote RGC axon regeneration to varying extents, with Crmp4 promoting the most axon regeneration and also localizing to axons. We also found that Crmp1 and Crmp4, but not Crmp5, promote RGC survival. Finally, we found that Crmp1, Crmp2, Crmp4, and Crmp5's ability to promote axon regeneration is associated with neurodevelopmental mechanisms, which control RGC's intrinsic axon growth capacity.


Asunto(s)
Traumatismos del Nervio Óptico , Células Ganglionares de la Retina , Humanos , Células Ganglionares de la Retina/metabolismo , Axones/metabolismo , Traumatismos del Nervio Óptico/metabolismo , Regeneración Nerviosa/fisiología , Expresión Génica , Supervivencia Celular
7.
Development ; 150(8)2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-37039265

RESUMEN

Central nervous system projection neurons fail to spontaneously regenerate injured axons. Targeting developmentally regulated genes in order to reactivate embryonic intrinsic axon growth capacity or targeting pro-growth tumor suppressor genes such as Pten promotes long-distance axon regeneration in only a small subset of injured retinal ganglion cells (RGCs), despite many RGCs regenerating short-distance axons. A recent study identified αRGCs as the primary type that regenerates short-distance axons in response to Pten inhibition, but the rare types which regenerate long-distance axons, and cellular features that enable such response, remained unknown. Here, we used a new method for capturing specifically the rare long-distance axon-regenerating RGCs, and also compared their transcriptomes with embryonic RGCs, in order to answer these questions. We found the existence of adult non-α intrinsically photosensitive M1 RGC subtypes that retained features of embryonic cell state, and showed that these subtypes partially dedifferentiated towards an embryonic state and regenerated long-distance axons in response to Pten inhibition. We also identified Pten inhibition-upregulated mitochondria-associated genes, Dynlt1a and Lars2, which promote axon regeneration on their own, and thus present novel therapeutic targets.


Asunto(s)
Aminoacil-ARNt Sintetasas , Traumatismos del Nervio Óptico , Aminoacil-ARNt Sintetasas/metabolismo , Axones/fisiología , Mitocondrias , Regeneración Nerviosa/fisiología , Traumatismos del Nervio Óptico/genética , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Células Ganglionares de la Retina/metabolismo
8.
Development ; 150(8)2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36971369

RESUMEN

Failure of central nervous system projection neurons to spontaneously regenerate long-distance axons underlies irreversibility of white matter pathologies. A barrier to axonal regenerative research is that the axons regenerating in response to experimental treatments stall growth before reaching post-synaptic targets. Here, we test the hypothesis that the interaction of regenerating axons with live oligodendrocytes, which were absent during developmental axon growth, contributes to stalling axonal growth. To test this hypothesis, first, we used single cell RNA-seq (scRNA-seq) and immunohistology to investigate whether post-injury born oligodendrocytes incorporate into the glial scar after optic nerve injury. Then, we administered demyelination-inducing cuprizone and stimulated axon regeneration by Pten knockdown (KD) after optic nerve crush. We found that post-injury born oligodendrocyte lineage cells incorporate into the glial scar, where they are susceptible to the demyelination diet, which reduced their presence in the glial scar. We further found that the demyelination diet enhanced Pten KD-stimulated axon regeneration and that localized cuprizone injection promoted axon regeneration. We also present a resource for comparing the gene expression of scRNA-seq-profiled normal and injured optic nerve oligodendrocyte lineage cells.


Asunto(s)
Axones , Enfermedades Desmielinizantes , Humanos , Axones/fisiología , Gliosis/metabolismo , Gliosis/patología , Cuprizona , Regeneración Nerviosa/fisiología , Células Ganglionares de la Retina/metabolismo , Oligodendroglía , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/metabolismo
9.
bioRxiv ; 2023 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-36747805

RESUMEN

Analysis of retinal ganglion cells (RGCs) by scRNA-seq is emerging as a state-of-the-art method for studying RGC biology and subtypes, as well as for studying the mechanisms of neuroprotection and axon regeneration in the central nervous system (CNS). Rbpms has been established as a pan-RGC marker, and Spp1 has been established as an αRGC type marker. Here, we analyzed by scRNA-seq retinal microglia and macrophages, and found Rbpms+ and Spp1+ subpopulations of retinal microglia/macrophages, which pose a potential pitfall in scRNA-seq studies involving RGCs. We performed comparative analysis of cellular identity of the presumed RGC cells isolated in recent scRNA-seq studies, and found that Rbpms+ and Spp1+ microglia/macrophages confounded identification of RGCs. We also provide solutions for circumventing this potential pitfall in scRNA-seq studies, by including in RGC and αRGC selection criteria other pan-RGC and αRGC markers.

10.
Neurosci Lett ; 765: 136260, 2021 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-34560191

RESUMEN

Projection neurons of the mammalian central nervous system (CNS) do not spontaneously regenerate axons which have been damaged by an injury or disease, often leaving patients with permanent disabilities that affect motor, cognitive, or sensory functions. Although several molecular targets which promote some extent of axon regeneration in animal models have been identified, the resulting recovery is very limited, and the molecular mechanisms underlying the axonal regenerative failure in the CNS are still poorly understood. One of the most studied targets for axon regeneration in the CNS is the mTOR pathway. A number of developmentally regulated genes also have been found to play a role in CNS axon regeneration. Here, we found that Transcriptional Elongation Factor A Like 3 (Tceal3), belonging to the Bex/Tceal transcriptional regulator family, which also modulates the mTOR pathway, is developmentally upregulated in retinal ganglion cell (RGCs) projection CNS neurons, and suppresses their capacity to regenerate axons after injury.


Asunto(s)
Axones , Regeneración Nerviosa , Traumatismos del Nervio Óptico , Factores de Elongación Transcripcional , Animales , Humanos , Ratones , Axones/fisiología , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Regeneración Nerviosa/genética , Traumatismos del Nervio Óptico/fisiopatología , Células Ganglionares de la Retina/fisiología , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/metabolismo , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo , Regulación hacia Arriba
11.
J Neurosci Res ; 99(3): 731-749, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33197966

RESUMEN

The cornea is the most innervated tissue in the human body. Myelinated axons upon inserting into the peripheral corneal stroma lose their myelin sheaths and continue into the central cornea wrapped by only nonmyelinating corneal Schwann cells (nm-cSCs). This anatomical organization is believed to be important for central vision. Here we employed single-cell RNA sequencing (scRNA-seq), microscopy, and transgenics to characterize these nm-cSCs of the central cornea. Using principal component analysis, uniform manifold approximation and projection, and unsupervised hierarchal cell clustering of scRNA-seq data derived from central corneal cells of male rabbits, we successfully identified several clusters representing different corneal cell types, including a unique cell cluster representing nm-cSCs. To confirm protein expression of cSC genes, we performed cross-species validation, employing corneal whole-mount immunostaining with confocal microscopy in mouse corneas. The expression of several representative proteins of nm-cSCs were validated. As the proteolipid protein 1 (PLP1) gene was also expressed in nm-cSCs, we explored the Plp1-eGFP transgenic reporter mouse line to visualize cSCs. Specific and efficient eGFP expression was observed in cSCs in adult mice of different ages. Of several putative cornea-specific SC genes identified, Dickkopf-related protein 1 was shown to be present in nm-cSCs. Taken together, our findings, for the first time, identify important insights and tools toward the study nm-cSCs in isolated tissue and adult animals. We expect that our results will advance the future study of nm-cSCs in applications of nerve repair, and provide a resource for the study of corneal sensory function.


Asunto(s)
Córnea/metabolismo , Expresión Génica/genética , Células de Schwann/metabolismo , Animales , Biomarcadores , Femenino , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Proteolipídica de la Mielina/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Conejos , Factores de Transcripción SOXE/metabolismo , Análisis de la Célula Individual , Sindecano-3/metabolismo , Transcriptoma , Canales de Sodio Activados por Voltaje/metabolismo
12.
Nat Commun ; 9(1): 3203, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30087343

RESUMEN

The original version of the Supplementary Information file associated with this Article contained an error in Supplementary Fig. 2. In panel c, the graph was inadvertently replaced with a duplicate of the graph in panel a. The error has now been fixed and the corrected version Supplementary Information PDF is available to download from the HTML version of the Article.

13.
Sci Rep ; 8(1): 9839, 2018 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-29959434

RESUMEN

The failure of mature central nervous system (CNS) projection neurons to regenerate axons over long distances drastically limits the recovery of functions lost after various CNS injuries and diseases. Although a number of manipulations that stimulate some degree of axon regeneration that overcomes the inhibitory environment after CNS injury have been discovered, the extent of regeneration remains very limited, emphasizing the need for improved therapies. Regenerating axons need nerve tissue environment capable of supporting their growth, and severe extra-axonal tissue damage and remodeling after injury may disrupt such environment. Here, we used traumatic injury to the mouse optic nerve as a model system to investigate how the extent of extra-axonal tissue damage affects experimental axon regeneration. Axon regeneration was stimulated by the shRNA-mediated knockdown (KD) of Pten gene expression in the retinal ganglion cells, and the extent of extra-axonal tissue damage was varied by changing the duration of optic nerve crush. Although no axons were spared using either 1 or 5 seconds crush, we found that Pten KD-stimulated axon regeneration was significantly reduced in 5 seconds compared with 1 second crush. The more severe extra-axonal tissue damage did not cause tissue atrophy, but led to significantly higher upregulation of axon growth-inhibiting chondroitin sulfate proteoglycan (CSPG) in the glial scar and also enlarged glial scar size, compared with less severely damaged tissue. Thus, the success of axon-regenerating approaches that target neuronal intrinsic mechanisms of axon growth is dependent on the preservation of appropriate extra-axonal tissue environment, which may need to be co-concurrently repaired by tissue remodeling methods.


Asunto(s)
Axones/fisiología , Sistema Nervioso Central/fisiología , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Regeneración Nerviosa , Neuroglía/patología , Fosfohidrolasa PTEN/metabolismo , Células Ganglionares de la Retina/citología , Animales , Sistema Nervioso Central/citología , Proteoglicanos Tipo Condroitín Sulfato/administración & dosificación , Proteoglicanos Tipo Condroitín Sulfato/genética , Masculino , Ratones , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfohidrolasa PTEN/genética , Células Ganglionares de la Retina/fisiología , Regulación hacia Arriba
14.
Nat Commun ; 9(1): 2759, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30018341

RESUMEN

Retinal ganglion cells (RGCs) convey the major output of information collected from the eye to the brain. Thirty subtypes of RGCs have been identified to date. Here, we analyze 6225 RGCs (average of 5000 genes per cell) from right and left eyes by single-cell RNA-seq and classify them into 40 subtypes using clustering algorithms. We identify additional subtypes and markers, as well as transcription factors predicted to cooperate in specifying RGC subtypes. Zic1, a marker of the right eye-enriched subtype, is validated by immunostaining in situ. Runx1 and Fst, the markers of other subtypes, are validated in purified RGCs by fluorescent in situ hybridization (FISH) and immunostaining. We show the extent of gene expression variability needed for subtype segregation, and we show a hierarchy in diversification from a cell-type population to subtypes. Finally, we present a website for comparing the gene expression of RGC subtypes.


Asunto(s)
Linaje de la Célula/genética , Proteínas del Ojo/genética , Células Ganglionares de la Retina/clasificación , Células Ganglionares de la Retina/metabolismo , Transcriptoma , Animales , Animales Recién Nacidos , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Proteínas del Ojo/metabolismo , Folistatina/genética , Folistatina/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Hibridación Fluorescente in Situ , Ratones , Ratones Endogámicos C57BL , Células Ganglionares de la Retina/citología , Análisis de la Célula Individual , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
Exp Neurol ; 300: 22-29, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29106981

RESUMEN

The inability of axons to regenerate over long-distances in the central nervous system (CNS) limits the recovery of sensory, motor, and cognitive functions after various CNS injuries and diseases. Although pre-clinical studies have identified a number of manipulations that stimulate some degree of axon growth after CNS damage, the extent of recovery remains quite limited, emphasizing the need for improved therapies. Here, we used traumatic injury to the mouse optic nerve as a model system to test the effects of combining several treatments that have recently been found to promote axon regeneration without the risks associated with manipulating known tumor suppressors or oncogenes. The treatments tested here include TPEN, a chelator of mobile (free) zinc (Zn2+); shRNA against the axon growth-suppressing transcription factor Klf9; and the atypical growth factor oncomodulin combined with a cAMP analog. Whereas some combinatorial treatments produced only marginally stronger effects than the individual treatments alone, co-treatment with TPEN and Klf9 knockdown had a substantially stronger effect on axon regeneration than either one alone. This combination also promoted a high level of cell survival at longer time points. Thus, Zn2+ chelation in combination with Klf9 suppression holds therapeutic potential for promoting axon regeneration after optic nerve injury, and may also be effective for treating other CNS injuries and diseases.


Asunto(s)
Axones/fisiología , Técnicas de Silenciamiento del Gen/métodos , Factores de Transcripción de Tipo Kruppel/antagonistas & inhibidores , Regeneración Nerviosa/fisiología , Traumatismos del Nervio Óptico/metabolismo , Zinc/metabolismo , Animales , Quelantes/metabolismo , Quelantes/farmacología , Etilenodiaminas/metabolismo , Etilenodiaminas/farmacología , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Traumatismos del Nervio Óptico/genética , Traumatismos del Nervio Óptico/terapia
16.
Dev Neurobiol ; 77(4): 419-437, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-26999672

RESUMEN

Serotonin (5HT) is present in a subpopulation of amacrine cells, which form synapses with retinal ganglion cells (RGCs), but little is known about the physiological role of retinal serotonergic circuitry. We found that the 5HT receptor 2C (5HTR2C) is upregulated in RGCs after birth. Amacrine cells generate 5HT and about half of RGCs respond to 5HTR2C agonism with calcium elevation. We found that there are on average 83 5HT+ amacrine cells randomly distributed across the adult mouse retina, all negative for choline acetyltransferase and 90% positive for tyrosine hydroxylase. We also investigated whether 5HTR2C and 5HTR5A affect RGC neurite growth. We found that both suppress neurite growth, and that RGCs from the 5HTR2C knockout (KO) mice grow longer neurites. Furthermore, 5HTR2C is subject to post-transcriptional editing, and we found that only the edited isoform's suppressive effect on neurite growth could be reversed by a 5HTR2C inverse agonist. Next, we investigated the physiological role of 5HTR2C in the retina, and found that 5HTR2C KO mice showed increased amplitude on pattern electroretinogram. Finally, RGC transcriptional profiling and pathways analysis suggested partial developmental compensation for 5HTR2C absence. Taken together, our findings demonstrate that 5HTR2C regulates neurite growth and RGC activity and is necessary for normal amplitude of RGC response to physiologic stimuli, and raise the hypothesis that these functions are modulated by a subset of 5HT+/ChAT-/TH+ amacrine cells as part of retinal serotonergic circuitry. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 419-437, 2017.


Asunto(s)
Células Amacrinas/fisiología , Neuritas/fisiología , Neurogénesis/fisiología , Receptor de Serotonina 5-HT2C/fisiología , Células Ganglionares de la Retina/fisiología , Agonistas del Receptor de Serotonina 5-HT2/farmacología , Visión Ocular/fisiología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neurogénesis/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Serotonina
17.
Sci Rep ; 6: 32249, 2016 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-27577089

RESUMEN

Balance in the transcriptome is regulated by coordinated synthesis and degradation of RNA molecules. Here we investigated whether mammalian cell types intrinsically differ in global coordination of gene splicing and expression levels. We analyzed RNA-seq transcriptome profiles of 8 different purified mouse cell types. We found that different cell types vary in proportion of highly expressed genes and the number of alternatively spliced transcripts expressed per gene, and that the cell types that express more variants of alternatively spliced transcripts per gene are those that have higher proportion of highly expressed genes. Cell types segregated into two clusters based on high or low proportion of highly expressed genes. Biological functions involved in negative regulation of gene expression were enriched in the group of cell types with low proportion of highly expressed genes, and biological functions involved in regulation of transcription and RNA splicing were enriched in the group of cell types with high proportion of highly expressed genes. Our findings show that cell types differ in proportion of highly expressed genes and the number of alternatively spliced transcripts expressed per gene, which represent distinct properties of the transcriptome and may reflect intrinsic differences in global coordination of synthesis, splicing, and degradation of RNA molecules.


Asunto(s)
Empalme Alternativo , Transcriptoma , Animales , Células Cultivadas , Células Endoteliales/fisiología , Regulación de la Expresión Génica , Ratones Transgénicos , Neuroglía/fisiología , Neuronas/fisiología , Análisis de Secuencia de ARN
18.
J Biol Chem ; 290(21): 13417-26, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-25833944

RESUMEN

Set-ß protein plays different roles in neurons, but the diversity of Set-ß neuronal isoforms and their functions have not been characterized. The expression and subcellular localization of Set-ß are altered in Alzheimer disease, cleavage of Set-ß leads to neuronal death after stroke, and the full-length Set-ß regulates retinal ganglion cell (RGC) and hippocampal neuron axon growth and regeneration in a subcellular localization-dependent manner. Here we used various biochemical approaches to investigate Set-ß isoforms and their role in the CNS, using the same type of neurons, RGCs, across studies. We found multiple alternatively spliced isoforms expressed from the Set locus in purified RGCs. Set transcripts containing the Set-ß-specific exon were the most highly expressed isoforms. We also identified a novel, alternatively spliced Set-ß transcript lacking the nuclear localization signal and demonstrated that the full-length (∼39-kDa) Set-ß is localized predominantly in the nucleus, whereas a shorter (∼25-kDa) Set-ß isoform is localized predominantly in the cytoplasm. Finally, we show that an N-terminal Set-ß cleavage product can induce neuronal death.


Asunto(s)
Empalme Alternativo/genética , Apoptosis , Proteínas Portadoras/metabolismo , Neuronas/patología , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Células Ganglionares de la Retina/patología , Animales , Animales Recién Nacidos , Western Blotting , Proteínas Portadoras/genética , Proliferación Celular , Células Cultivadas , Proteínas de Unión al ADN , Técnica del Anticuerpo Fluorescente , Chaperonas de Histonas , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Isoformas de Proteínas , ARN Mensajero/genética , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Ganglionares de la Retina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
J Neurosci ; 34(21): 7361-74, 2014 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-24849368

RESUMEN

The failure of the CNS neurons to regenerate axons after injury or stroke is a major clinical problem. Transcriptional regulators like Set-ß are well positioned to regulate intrinsic axon regeneration capacity, which declines developmentally in maturing CNS neurons. Set-ß also functions at cellular membranes and its subcellular localization is disrupted in Alzheimer's disease, but many of its biological mechanisms have not been explored in neurons. We found that Set-ß was upregulated postnatally in CNS neurons, and was primarily localized to the nucleus but was also detected in the cytoplasm and adjacent to the plasma membrane. Remarkably, nuclear Set-ß suppressed, whereas Set-ß localized to cytoplasmic membranes promoted neurite growth in rodent retinal ganglion cells and hippocampal neurons. Mimicking serine 9 phosphorylation, as found in Alzheimer's disease brains, delayed nuclear import and furthermore blocked the ability of nuclear Set-ß to suppress neurite growth. We also present data on gene regulation and protein binding partner recruitment by Set-ß in primary neurons, raising the hypothesis that nuclear Set-ß may preferentially regulate gene expression whereas Set-ß at cytoplasmic membranes may regulate unique cofactors, including PP2A, which we show also regulates axon growth in vitro. Finally, increasing recruitment of Set-ß to cellular membranes promoted adult rat optic nerve axon regeneration after injury in vivo. Thus, Set-ß differentially regulates axon growth and regeneration depending on subcellular localization and phosphorylation.


Asunto(s)
Axones/metabolismo , Membrana Celular/metabolismo , Regeneración Nerviosa/genética , Neuronas/ultraestructura , Traumatismos del Nervio Óptico/fisiopatología , Proteínas del Complejo de Iniciación de Transcripción Pol1/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Células Cultivadas , Modelos Animales de Enfermedad , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/citología , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Traumatismos del Nervio Óptico/patología , Proteínas del Complejo de Iniciación de Transcripción Pol1/genética , Ratas , Ratas Sprague-Dawley , Retina/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...