Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Dev Dyn ; 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37909656

RESUMEN

BACKGROUND: Post-translational histone modifications are among the most common epigenetic modifications that orchestrate gene expression, playing a pivotal role during embryonic development and in various pathological conditions. Among histone lysine demethylases, KDM7A, also known as KIAA1718 or JHDM1D, catalyzes the demethylation of H3K9me1/2 and H3K27me1/2, leading to transcriptional regulation. Previous data suggest that KDM7A plays a central role in several biological processes, including cell proliferation, commitment, differentiation, apoptosis, and maintenance. However, information on the expression pattern of KDM7A in whole organisms is limited, and its functional role is still unclear. RESULTS: In Xenopus development, kdm7a is expressed early, undergoing spatiotemporal regulation in various organs and tissues, including the central nervous system and the eye. Focusing on retinal development, we found that kdm7a overexpression does not affect the expression of genes critically involved in early neural development and eye-field specification, whereas unbalances the distribution of neural cell subtypes in the mature retina by disfavoring the development of ganglion cells while promoting that of horizontal cells. CONCLUSIONS: Kdm7a is dynamically expressed during embryonic development, and its overexpression influences late retinal development, suggesting a potential involvement in the molecular machinery regulating the spatiotemporally ordered generation of retinal neuronal subtypes.

2.
Cell Mol Life Sci ; 79(1): 28, 2021 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-34936031

RESUMEN

Microgravity and space radiation (SR) are two highly influential factors affecting humans in space flight (SF). Many health problems reported by astronauts derive from endothelial dysfunction and impaired homeostasis. Here, we describe the adaptive response of human, capillary endothelial cells to SF. Reference samples on the ground and at 1g onboard permitted discrimination between the contribution of microgravity and SR within the combined responses to SF. Cell softening and reduced motility occurred in SF cells, with a loss of actin stress fibers and a broader distribution of microtubules and intermediate filaments within the cytoplasm than in control cells. Furthermore, in space the number of primary cilia per cell increased and DNA repair mechanisms were found to be activated. Transcriptomics revealed the opposing effects of microgravity from SR for specific molecular pathways: SR, unlike microgravity, stimulated pathways for endothelial activation, such as hypoxia and inflammation, DNA repair and apoptosis, inhibiting autophagic flux and promoting an aged-like phenotype. Conversely, microgravity, unlike SR, activated pathways for metabolism and a pro-proliferative phenotype. Therefore, we suggest microgravity and SR should be considered separately to tailor effective countermeasures to protect astronauts' health.


Asunto(s)
Autofagia , Capilares/citología , Radiación Cósmica , Células Endoteliales/efectos de la radiación , Transducción de Señal , Ingravidez , Apoptosis , Biomarcadores/metabolismo , Línea Celular , Supervivencia Celular , Cromosomas Humanos/metabolismo , Citoesqueleto/metabolismo , Daño del ADN , Fluorescencia , Regulación de la Expresión Génica , Genoma Humano , Humanos , Masculino , Mecanotransducción Celular , Modelos Biológicos , Transducción de Señal/efectos de la radiación , Vuelo Espacial , Estrés Fisiológico , Homeostasis del Telómero , Transcriptoma/genética
3.
Int J Mol Sci ; 24(1)2022 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-36613611

RESUMEN

Haploinsufficiency of the SETD5 gene, encoding a SET domain-containing histone methyltransferase, has been identified as a cause of intellectual disability and Autism Spectrum Disorder (ASD). Recently, the zebrafish has emerged as a valuable model to study neurodevelopmental disorders because of its genetic tractability, robust behavioral traits and amenability to high-throughput drug screening. To model human SETD5 haploinsufficiency, we generated zebrafish setd5 mutants using the CRISPR/Cas9 technology and characterized their morphological, behavioral and molecular phenotypes. According to our observation that setd5 is expressed in adult zebrafish brain, including those areas controlling social behavior, we found that setd5 heterozygous mutants exhibit defective aggregation and coordination abilities required for shoaling interactions, as well as indifference to social stimuli. Interestingly, impairment in social interest is rescued by risperidone, an antipsychotic drug used to treat behavioral traits in ASD individuals. The molecular analysis underscored the downregulation of genes encoding proteins involved in the synaptic structure and function in the adult brain, thus suggesting that brain hypo-connectivity could be responsible for the social impairments of setd5 mutant fishes. The zebrafish setd5 mutants display ASD-like features and are a promising setd5 haploinsufficiency model for drug screening aimed at reversing the behavioral phenotypes.


Asunto(s)
Trastorno del Espectro Autista , Metiltransferasas , Animales , Humanos , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Encéfalo/metabolismo , Sistemas CRISPR-Cas , Metiltransferasas/genética , Metiltransferasas/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Conducta Social
4.
Hum Mol Genet ; 27(5): 761-779, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29281027

RESUMEN

P23H is the most common mutation in the RHODOPSIN (RHO) gene leading to a dominant form of retinitis pigmentosa (RP), a rod photoreceptor degeneration that invariably causes vision loss. Specific disruption of the disease P23H RHO mutant while preserving the wild-type (WT) functional allele would be an invaluable therapy for this disease. However, various technologies tested in the past failed to achieve effective changes and consequently therapeutic benefits. We validated a CRISPR/Cas9 strategy to specifically inactivate the P23H RHO mutant, while preserving the WT allele in vitro. We, then, translated this approach in vivo by delivering the CRISPR/Cas9 components in murine Rho+/P23H mutant retinae. Targeted retinae presented a high rate of cleavage in the P23H but not WT Rho allele. This gene manipulation was sufficient to slow photoreceptor degeneration and improve retinal functions. To improve the translational potential of our approach, we tested intravitreal delivery of this system by means of adeno-associated viruses (AAVs). To this purpose, the employment of the AAV9-PHP.B resulted the most effective in disrupting the P23H Rho mutant. Finally, this approach was translated successfully in human cells engineered with the homozygous P23H RHO gene mutation. Overall, this is a significant proof-of-concept that gene allele specific targeting by CRISPR/Cas9 technology is specific and efficient and represents an unprecedented tool for treating RP and more broadly dominant genetic human disorders affecting the eye, as well as other tissues.


Asunto(s)
Marcación de Gen/métodos , Vectores Genéticos , Retina/fisiología , Degeneración Retiniana/terapia , Rodopsina/genética , Alelos , Animales , Sistemas CRISPR-Cas , Electroporación/métodos , Fibroblastos , Terapia Genética/métodos , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Mutación , ARN Guía de Kinetoplastida , Retina/patología , Degeneración Retiniana/genética
5.
Cell Mol Life Sci ; 75(7): 1255-1267, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29098325

RESUMEN

Glaucoma and other optic neuropathies are characterized by a loss of retinal ganglion cells (RGCs), a cell layer located in the posterior eye segment. Several preclinical studies demonstrate that neurotrophins (NTs) prevent RGC loss. However, NTs are rarely investigated in the clinic due to various issues, such as difficulties in reaching the retina, the very short half-life of NTs, and the need for multiple injections. We demonstrate that NTs can be conjugated to magnetic nanoparticles (MNPs), which act as smart drug carriers. This combines the advantages of the self-localization of the drug in the retina and drug protection from fast degradation. We tested the nerve growth factor and brain-derived neurotrophic factor by comparing the neuroprotection of free versus conjugated proteins in a model of RGC loss induced by oxidative stress. Histological data demonstrated that the conjugated proteins totally prevented RGC loss, in sharp contrast to the equivalent dose of free proteins, which had no effect. The overall data suggest that the nanoscale MNP-protein hybrid is an excellent tool in implementing ocular drug delivery strategies for neuroprotection and therapy.


Asunto(s)
Nanopartículas/química , Factores de Crecimiento Nervioso/farmacología , Neuroprotección/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Retina/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/administración & dosificación , Factor Neurotrófico Derivado del Encéfalo/química , Factor Neurotrófico Derivado del Encéfalo/farmacología , Sistemas de Liberación de Medicamentos , Glaucoma/metabolismo , Glaucoma/patología , Humanos , Factor de Crecimiento Nervioso/administración & dosificación , Factor de Crecimiento Nervioso/química , Factor de Crecimiento Nervioso/farmacología , Factores de Crecimiento Nervioso/administración & dosificación , Factores de Crecimiento Nervioso/química , Células PC12 , Ratas , Retina/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Células Tumorales Cultivadas
6.
Stem Cells ; 33(8): 2496-508, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25913744

RESUMEN

It has long been known that the depletion of bone morphogenetic protein (BMP) is one of the key factors necessary for the development of anterior neuroectodermal structures. However, the precise molecular mechanisms that underlie forebrain regionalization are still not completely understood. Here, we show that Noggin1 is involved in the regionalization of anterior neural structures in a dose-dependent manner. Low doses of Noggin1 expand prosencephalic territories, while higher doses specify diencephalic and retinal regions at the expense of telencephalic areas. A similar dose-dependent mechanism determines the ability of Noggin1 to convert pluripotent cells in prosencephalic or diencephalic/retinal precursors, as shown by transplant experiments and molecular analyses. At a molecular level, the strong inhibition of BMP signaling exerted by high doses of Noggin1 reinforces the Nodal/transforming growth factor (TGF)ß signaling pathway, leading to activation of Gli1 and Gli2 and subsequent activation of Sonic Hedgehog (SHH) signaling. We propose a new role for Noggin1 in determining specific anterior neural structures by the modulation of TGFß and SHH signaling.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Portadoras/metabolismo , Células Madre Pluripotentes/metabolismo , Retina/embriología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Portadoras/genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Retina/citología , Telencéfalo/citología , Telencéfalo/embriología , Factor de Crecimiento Transformador beta/genética , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis
7.
Dev Dyn ; 243(10): 1352-61, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24801179

RESUMEN

BACKGROUND: The transcription factor Rx1, also known as Rax, controls key properties of retinal precursors including migration behavior, proliferation, and maintenance of multipotency. However, Rx1 effector genes are largely unknown. RESULTS: To identify genes controlled by Rx1 in early retinal precursors, we compared the transcriptome of Xenopus embryos overexpressing Rx1 to that of embryos in which Rx1 was knocked-down. In particular, we selected 52 genes coherently regulated, i.e., actived in Rx1 gain of function and repressed in Rx1 loss of function experiments, or vice versa. RT-qPCR and in situ hybridization confirmed the trend of regulation predicted by microarray data for the selected genes. Most of the genes upregulated by Rx1 are coexpressed with this transcription factor, while downregulated genes are either not expressed or expressed at very low levels in the early developing retina. Putative direct Rx1 target genes, activated by GR-Rx1 in the absence of protein synthesis, include Ephrin B1 and Sh2d3c, an interactor of ephrinB1 receptor, which represent candidate novel effectors for the migration promoting activity of Rx1. CONCLUSIONS: This study identifies previously undescribed Rx1 regulated genes mainly involved in transcription regulation, cell migration/adhesion, and cell proliferation that contribute to delineate the molecular mechanisms underlying Rx1 activities.


Asunto(s)
Proteínas del Ojo/fisiología , Regulación del Desarrollo de la Expresión Génica , Retina/embriología , Transcriptoma , Proteínas de Xenopus/fisiología , Animales , Animales Modificados Genéticamente , Diferenciación Celular/genética , Embrión no Mamífero , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Perfilación de la Expresión Génica , Análisis por Micromatrices , Retina/metabolismo , Xenopus/embriología , Xenopus/genética , Xenopus laevis/embriología , Xenopus laevis/genética
8.
Stem Cells ; 31(12): 2842-7, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24038725

RESUMEN

The molecular mechanisms underlying the acquisition of retinal precursor identity are scarcely defined. Although the homeobox gene Rx1 (also known as Rax) plays a major role in specifying retinal precursors and maintaining their multipotent state, the involved mechanisms remain to be largely deciphered. Here, following a highthroughput screen for genes regulated by Rx1, we found that this transcription factor specifies the fate of retinal progenitors by repressing genes normally activated in adjacent ectodermal territories. Unexpectedly, we also observed that Rx1, mainly through the activation of the transcriptional repressors TLE2 and Hes4, is necessary and sufficient to inhibit endomesodermal gene expression in retinal precursors of the eye field. In particular, Rx1 knockdown leads retinogenic blastomeres to adopt an endomesodermal fate, indicating a previously undescribed function for Rx1 in preventing the expression of endomesoderm determinants known to inhibit retinal fate. Altogether these data suggest that an essential requirement to establish a retinal precursor identity is the active inhibition of pathways leading to alternative fates.


Asunto(s)
Proteínas del Ojo/metabolismo , Proteínas Represoras/metabolismo , Retina/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Xenopus/metabolismo , Animales , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Retina/citología , Xenopus laevis
9.
Cells ; 13(10)2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38786093

RESUMEN

Vision starts in retinal photoreceptors when specialized proteins (opsins) sense photons via their covalently bonded vitamin A derivative 11cis retinaldehyde (11cis-RAL). The reaction of non-enzymatic aldehydes with amino groups lacks specificity, and the reaction products may trigger cell damage. However, the reduced synthesis of 11cis-RAL results in photoreceptor demise and suggests the need for careful control over 11cis-RAL handling by retinal cells. This perspective focuses on retinoid(s) synthesis, their control in the adult retina, and their role during retina development. It also explores the potential importance of 9cis vitamin A derivatives in regulating retinoid synthesis and their impact on photoreceptor development and survival. Additionally, recent advancements suggesting the pivotal nature of retinoid synthesis regulation for cone cell viability are discussed.


Asunto(s)
Retinoides , Animales , Humanos , Retina/metabolismo , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Retinaldehído/metabolismo , Retinoides/metabolismo , Vitamina A/metabolismo
10.
Genes (Basel) ; 15(4)2024 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-38674426

RESUMEN

Haploinsufficiency of the PRR12 gene is implicated in a human neuro-ocular syndrome. Although identified as a nuclear protein highly expressed in the embryonic mouse brain, PRR12 molecular function remains elusive. This study explores the spatio-temporal expression of zebrafish PRR12 co-orthologs, prr12a and prr12b, as a first step to elucidate their function. In silico analysis reveals high evolutionary conservation in the DNA-interacting domains for both orthologs, with significant syntenic conservation observed for the prr12b locus. In situ hybridization and RT-qPCR analyses on zebrafish embryos and larvae reveal distinct expression patterns: prr12a is expressed early in zygotic development, mainly in the central nervous system, while prr12b expression initiates during gastrulation, localizing later to dopaminergic telencephalic and diencephalic cell clusters. Both transcripts are enriched in the ganglion cell and inner neural layers of the 72 hpf retina, with prr12b widely distributed in the ciliary marginal zone. In the adult brain, prr12a and prr12b are found in the cerebellum, amygdala and ventral telencephalon, which represent the main areas affected in autistic patients. Overall, this study suggests PRR12's potential involvement in eye and brain development, laying the groundwork for further investigations into PRR12-related neurobehavioral disorders.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas de la Membrana , Proteínas del Tejido Nervioso , Proteínas de Pez Cebra , Animales , Encéfalo/metabolismo , Encéfalo/crecimiento & desarrollo , Retina/metabolismo , Retina/crecimiento & desarrollo , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas de la Membrana/genética
11.
Int J Dev Biol ; 68(2): 85-91, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39016375

RESUMEN

The tRNA-histidine guanylyltransferase 1-like (THG1L), also known as induced in high glucose-1 (IHG-1), encodes for an essential mitochondria-associated protein highly conserved throughout evolution, that catalyses the 3'-5' addition of a guanine to the 5'-end of tRNA-histidine (tRNAHis). Previous data indicated that THG1L plays a crucial role in the regulation of mitochondrial biogenesis and dynamics, in ATP production, and is critically involved in the modulation of apoptosis, cell-cycle progression and survival, as well as in cellular stress responses and redox homeostasis. Dysregulations of THG1L expression play a central role in various pathologies, including nephropathies, and neurodevelopmental disorders often characterized by developmental delay and cerebellar ataxia. Despite the essential role of THG1L, little is known about its expression during vertebrate development. Herein, we examined the detailed spatio-temporal expression of this gene in the developing Xenopus laevis. Our results show that thg1l is maternally inherited and its temporal expression suggests a role during the earliest stages of embryogenesis. Spatially, thg1l mRNA localizes in the ectoderm and marginal zone mesoderm during early stages of development. Then, at tadpole stages, thg1l transcripts mostly localise in neural crests and their derivatives, somites, developing kidney and central nervous system, therefore largely coinciding with territories displaying intense energy metabolism during organogenesis in Xenopus.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas de Xenopus , Xenopus laevis , Animales , Xenopus laevis/metabolismo , Xenopus laevis/embriología , Xenopus laevis/genética , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Embrión no Mamífero/metabolismo , Embrión no Mamífero/embriología , Desarrollo Embrionario/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
12.
Zebrafish ; 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38963004

RESUMEN

The 4th Italian Zebrafish Meeting took place in Palermo from February 7 to 9, 2024. The primary aim of this meeting was to bring together a diverse group of principal investigators, young researchers, facility managers, commercial vendors, and others to provide an important forum for presentation and discussion of the most innovative and exciting scientific research currently ongoing in Italy using the zebrafish model. Nonetheless, the meeting program has been conceived to allow the dissemination of cutting-edge scientific research across a wide range of topics and to shed light on its future directions, without geographical boundaries. Indeed, people from various parts of the world joined the meeting, and 210 participants presented their latest work in talks and posters. Importantly, the meeting had designated time to foster open scientific exchange and informal networking opportunities among participants of all career stages, thus allowing initiation of new collaborations and strengthening of existing partnerships. The meeting was a tremendous success as testified by the highest participation ever since the first meeting of the series in 2017, coupled with the highly positive satisfaction rating expressed by the attendants. The full program and detailed information about the meeting can be found on the dedicated website at https://itazebrafishmeeting.wixsite.com/izm2024.

13.
Proc Natl Acad Sci U S A ; 107(14): 6352-7, 2010 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-20308548

RESUMEN

In the developing central nervous system, the cell cycle clock plays a crucial role in determining cell fate specification. A second clock, the circadian oscillator, generates daily rhythms of cell cycle progression. Although these two clocks interact, the mechanisms linking circadian cell cycle progression and cell fate determination are still poorly understood. A convenient system to address this issue is the pineal organ of lower vertebrates, which contains only two neuronal types, photoreceptors and projection neurons. In particular, photoreceptors constitute the core of the pineal circadian system, being able to transduce daily light inputs into the rhythmical production of melatonin. However, the genetic program leading to photoreceptor fate largely remains to be deciphered. Here, we report a previously undescribed function for the homeobox gene Bsx in controlling pineal proliferation and photoreceptor fate in Xenopus. We show that Xenopus Bsx (Xbsx) is expressed rhythmically in postmitotic photoreceptor precursors, reaching a peak during the night, with a cycle that is complementary to the daily rhythms of S-phase entry displayed by pineal cells. Xbsx knockdown results in increased night levels of pineal proliferation, whereas activation of a GR-Xbsx protein flattens the daily rhythms of S-phase entry to the lowest level. Furthermore, evidence is presented that Xbsx is necessary and sufficient to promote a photoreceptor fate. Altogether, these data indicate that Xbsx plays a dual role in contributing to shape the profile of the circadian cell cycle progression and in the specification of pineal photoreceptors, thus acting as a unique link between these two events.


Asunto(s)
Ciclo Celular , Linaje de la Célula , Ritmo Circadiano , Células Fotorreceptoras/metabolismo , Glándula Pineal/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Animales , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Células Fotorreceptoras/citología , Glándula Pineal/citología , Glándula Pineal/embriología , Factores de Transcripción/genética , Proteínas de Xenopus/genética , Xenopus laevis/embriología , Xenopus laevis/genética
14.
Sci Rep ; 13(1): 6025, 2023 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-37055439

RESUMEN

In proliferating multipotent retinal progenitors, transcription factors dynamics set the fate of postmitotic daughter cells, but postmitotic cell fate plasticity driven by extrinsic factors remains controversial. Transcriptome analysis reveals the concurrent expression by postmitotic rod precursors of genes critical for the Müller glia cell fate, which are rarely generated from terminally-dividing progenitors as a pair with rod precursors. By combining gene expression and functional characterisation in single cultured rod precursors, we identified a time-restricted window where increasing cell culture density switches off the expression of genes critical for Müller glial cells. Intriguingly, rod precursors in low cell culture density maintain the expression of genes of rod and glial cell fate and develop a mixed rod/Muller glial cells electrophysiological fingerprint, revealing rods derailment toward a hybrid rod-glial phenotype. The notion of cell culture density as an extrinsic factor critical for preventing rod-fated cells diversion toward a hybrid cell state may explain the occurrence of hybrid rod/MG cells in the adult retina and provide a strategy to improve engraftment yield in regenerative approaches to retinal degenerative disease by stabilising the fate of grafted rod precursors.


Asunto(s)
Neuroglía , Retina , Retina/metabolismo , Neuroglía/metabolismo , Diferenciación Celular/genética , Factores de Transcripción/metabolismo , Técnicas de Cultivo de Célula
15.
Cells ; 10(9)2021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-34572137

RESUMEN

Inherited retinal degenerations (IRD) affecting either photoreceptors or pigment epithelial cells cause progressive visual loss and severe disability, up to complete blindness. Retinal organoids (ROs) technologies opened up the development of human inducible pluripotent stem cells (hiPSC) for disease modeling and replacement therapies. However, hiPSC-derived ROs applications to IRD presently display limited maturation and functionality, with most photoreceptors lacking well-developed outer segments (OS) and light responsiveness comparable to their adult retinal counterparts. In this review, we address for the first time the microenvironment where OS mature, i.e., the subretinal space (SRS), and discuss SRS role in photoreceptors metabolic reprogramming required for OS generation. We also address bioengineering issues to improve culture systems proficiency to promote OS maturation in hiPSC-derived ROs. This issue is crucial, as satisfying the demanding metabolic needs of photoreceptors may unleash hiPSC-derived ROs full potential for disease modeling, drug development, and replacement therapies.


Asunto(s)
Bioingeniería/métodos , Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Organoides/citología , Degeneración Retiniana/terapia , Epitelio Pigmentado de la Retina/citología , Animales , Humanos , Degeneración Retiniana/patología
16.
Stem Cells ; 27(9): 2146-52, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19591225

RESUMEN

Driving specific differentiation pathways in multipotent stem cells is a main goal of cell therapy. Here we exploited the differentiating potential of Xenopus animal cap embryonic stem (ACES) cells to investigate the factors necessary to drive multipotent stem cells toward retinal fates. ACES cells are multipotent, and can be diverged from their default ectodermal fate to give rise to cell types from all three germ layers. We found that a single secreted molecule, Noggin, is sufficient to elicit retinal fates in ACES cells. Reverse-transcription polymerase chain reaction, immunohistochemistry, and in situ hybridization experiments showed that high doses of Noggin are able to support the expression of terminal differentiation markers of the neural retina in ACES cells in vitro. Following in vivo transplantation, ACES cells expressing high Noggin doses form eyes, both in the presumptive eye field region and in ectopic posterior locations. The eyes originating from the transplants in the eye field region are functionally equivalent to normal eyes, as seen by electrophysiology and c-fos expression in response to light. Our data show that in Xenopus embryos, proper doses of a single molecule, Noggin, can drive ACES cells toward retinal cell differentiation without additional cues. This makes Xenopus ACES cells a suitable model system to direct differentiation of stem cells toward retinal fates and encourages further studies on the role of Noggin in the retinal differentiation of mammalian stem cells.


Asunto(s)
Proteínas Portadoras/metabolismo , Diferenciación Celular , Células Madre Embrionarias/citología , Retina/citología , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Animales , Proteínas Portadoras/genética , Regulación del Desarrollo de la Expresión Génica/genética , Inmunohistoquímica , Hibridación in Situ , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Xenopus laevis/metabolismo
17.
Sci Rep ; 10(1): 17358, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33060638

RESUMEN

The plant-derived natural alkaloid berberine displays therapeutic potential to treat several pathological conditions, including dyslipidemias, diabetes and cardiovascular disorders. However, data on berberine effects during embryonic development are scarce and in part controversial. In this study, using zebrafish embryos as vertebrate experimental model, we address the effects of berberine treatment on cardiovascular system development and functionality. Starting from the observation that berberine induces developmental toxicity and pericardial edema in a time- and concentration-dependent manner, we found that treated embryos display cardiac looping defects and, at later stages, present an abnormal heart characterized by a stretched morphology and atrial endocardial/myocardial detachment. Furthermore, berberine affected cardiac functionality of the embryos, promoting bradycardia and reducing the cardiac output, the atrial shortening fraction percentage and the atrial stroke volume. We also found that, during development, berberine interferes with the angiogenic process, without altering vascular permeability. These alterations are associated with increased levels of vascular endothelial growth factor aa (vegfaa) mRNA, suggesting an important role for Vegfaa as mediator of berberine-induced cardiovascular defects. Altogether, these data indicate that berberine treatment during vertebrate development leads to an impairment of cardiovascular system morphogenesis and functionality, suggesting a note of caution in its use during pregnancy and lactation.


Asunto(s)
Berberina/toxicidad , Sistema Cardiovascular/embriología , Morfogénesis/efectos de los fármacos , Pez Cebra/embriología , Animales , Teratógenos/toxicidad
18.
J Exp Clin Cancer Res ; 39(1): 103, 2020 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-32498717

RESUMEN

BACKGROUND: Understanding the signalling pathways involved in angiogenesis, and developing anti-angiogenic drugs are one of the major focuses on cancer research. Herein, we assessed the effect of CPTH6, a lysine acetyltransferase inhibitor and anti-tumoral compound, on angiogenesis-related properties of both endothelial and cancer cells. METHODS: The in vitro effect of CPTH6 on protein acetylation and anti-angiogenic properties on endothelial and lung cancer cells was evaluated via wound healing, trans-well invasion and migration, tube formation, immunoblotting and immunofluorescence. Matrigel plug assay, zebrafish embryo and mouse xenograft models were used to evaluate in vivo anti-angiogenic effect of CPTH6. RESULTS: CPTH6 impaired in vitro endothelial angiogenesis-related functions, and decreased the in vivo vascularization both in mice xenografts and zebrafish embryos. Mechanistically, CPTH6 reduced α-tubulin acetylation and induced accumulation of acetylated microtubules in the perinuclear region of endothelial cells. Interestingly, CPTH6 also affected the angiogenesis-related properties of lung cancer cells, and conditioned media derived from CPTH6-treated lung cancer cells impaired endothelial cells morphogenesis. CPTH6 also modulated the VEGF/VEGFR2 pathway, and reshaped cytoskeletal organization of lung cancer cells. Finally, anti-migratory effect of CPTH6, dependent on α-tubulin acetylation, was also demonstrated by genetic approaches in lung cancer cells. CONCLUSION: Overall, this study indicates that α-tubulin acetylation could play a role in the anti-angiogenic effect of CPTH6 and, more in general, it adds information to the role of histone acetyltransferases in tumor angiogenesis, and proposes the inhibition of these enzymes as an antiangiogenic therapy of cancer.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Neoplasias Pulmonares/irrigación sanguínea , Lisina Acetiltransferasas/antagonistas & inhibidores , Neovascularización Patológica/tratamiento farmacológico , Tiazoles/farmacología , Animales , Apoptosis , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Transducción de Señal , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Birth Defects Res C Embryo Today ; 87(3): 284-95, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19750521

RESUMEN

The specification of retinal cell fate is a multistep process that begins during early development and results from the spatio-temporal coordination of cell cycle, cell differentiation, and morphogenesis. This review focuses on recent advances in understanding the molecular mechanisms underlying the distinct steps of retinal specification. Emphasis is placed on key regulatory events that control the multipotency of retinal progenitors, the generation of cell diversity, and the establishment of the clock that determines the ordered generation of retinal cell types. These basic studies have paved the way to the latest progress on the isolation and in vitro generation of retinal stem cells, which is presented in the light of possible therapeutic applications.


Asunto(s)
Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Retina/citología , Células Madre , Animales , Embrión de Mamíferos/citología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , MicroARNs , Retina/metabolismo , Células Madre/citología , Células Madre/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vertebrados/embriología , Xenopus/embriología
20.
PLoS Biol ; 4(9): e272, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16903786

RESUMEN

The reason why different types of vertebrate nerve cells are generated in a particular sequence is still poorly understood. In the vertebrate retina, homeobox genes play a crucial role in establishing different cell identities. Here we provide evidence of a cellular clock that sequentially activates distinct homeobox genes in embryonic retinal cells, linking the identity of a retinal cell to its time of generation. By in situ expression analysis, we found that the three Xenopus homeobox genes Xotx5b, Xvsx1, and Xotx2 are initially transcribed but not translated in early retinal progenitors. Their translation requires cell cycle progression and is sequentially activated in photoreceptors (Xotx5b) and bipolar cells (Xvsx1 and Xotx2). Furthermore, by in vivo lipofection of "sensors" in which green fluorescent protein translation is under control of the 3' untranslated region (UTR), we found that the 3' UTRs of Xotx5b, Xvsx1, and Xotx2 are sufficient to drive a spatiotemporal pattern of translation matching that of the corresponding proteins and consistent with the time of generation of photoreceptors (Xotx5b) and bipolar cells (Xvsx1 and Xotx2). The block of cell cycle progression of single early retinal progenitors impairs their differentiation as photoreceptors and bipolar cells, but is rescued by the lipofection of Xotx5b and Xvsx1 coding sequences, respectively. This is the first evidence to our knowledge that vertebrate homeobox proteins can work as effectors of a cellular clock to establish distinct cell identities.


Asunto(s)
Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/fisiología , Retina/citología , Animales , Relojes Biológicos/genética , Ciclo Celular/fisiología , Diferenciación Celular/genética , Linaje de la Célula , Células Cultivadas , Factores de Transcripción E2F/metabolismo , Factores de Transcripción E2F/fisiología , Proteínas del Ojo/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Datos de Secuencia Molecular , Factores de Transcripción Otx/metabolismo , Biosíntesis de Proteínas , Células Bipolares de la Retina/metabolismo , Transfección , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Xenopus laevis/fisiología , Proteinas GADD45
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA