Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Development ; 146(16)2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31405994

RESUMEN

Retinal ganglion cell (RGC) degeneration is a hallmark of glaucoma, the most prevalent cause of irreversible blindness. Thus, therapeutic strategies are needed to protect and replace these projection neurons. One innovative approach is to promote de novo genesis of RGCs via manipulation of endogenous cell sources. Here, we demonstrate that the pluripotency regulator gene Krüppel-like factor 4 (Klf4) is sufficient to change the potency of lineage-restricted retinal progenitor cells to generate RGCs in vivo Transcriptome analysis disclosed that the overexpression of Klf4 induces crucial regulators of RGC competence and specification, including Atoh7 and Eya2 In contrast, loss-of-function studies in mice and zebrafish demonstrated that Klf4 is not essential for generation or differentiation of RGCs during retinogenesis. Nevertheless, induced RGCs (iRGCs) generated upon Klf4 overexpression migrate to the proper layer and project axons aligned with endogenous fascicles that reach the optic nerve head. Notably, iRGCs survive for up to 30 days after in vivo generation. We identified Klf4 as a promising candidate for reprogramming retinal cells and regenerating RGCs in the retina.This article has an associated 'The people behind the papers' interview.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/fisiología , Neurogénesis , Células Ganglionares de la Retina/fisiología , Animales , Ciclo Celular , Femenino , Proteínas de Homeodominio/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regeneración Nerviosa , Células-Madre Neurales/fisiología , Ratas , Factor de Transcripción Brn-3A/metabolismo , Factor de Transcripción Brn-3B/metabolismo , Pez Cebra , Proteínas de Pez Cebra/fisiología
2.
An Acad Bras Cienc ; 92(4): e20191517, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32844990

RESUMEN

Pathogenic microbial detection and control in laboratory animal facilities is essential to guarantee animal welfare, data validity and reproducibility. Helicobacter spp. are known to affect mice health, what may interfere with experimental outcomes. This study aimed to screen for Helicobacter spp. in mice from animal facilities in Rio de Janeiro, Brazil using a PCR-based method. Primers designed to specifically identify Helicobacter spp. were used to amplify feces or intestine DNA extracted of mice from four different animal facilities. The expected 375 base pairs (bp) amplicon was purified, sequenced and a similarity of 95% was observed when compared to deposited sequences of H. hepaticus and H. bilis. In our screening, Helicobacter spp. was detected in ~59% of fecal and ~70% of intestine samples. Our study is the first to screen for Helicobacter spp. in mouse facilities of a Rio de Janeiro University using a low cost, rapid molecular diagnostic test. Although Helicobacter spp. screening is not mandatory according to Brazilian animal welfare regulation it is recommended by institutional animal health monitoring programs guidelines worldwide, including ARRIVE, AAALAC and FELASA.


Asunto(s)
Infecciones por Helicobacter , Helicobacter , Animales , Animales de Laboratorio , Brasil , ADN Bacteriano , Helicobacter/aislamiento & purificación , Infecciones por Helicobacter/diagnóstico , Infecciones por Helicobacter/veterinaria , Ratones , Reacción en Cadena de la Polimerasa , Reproducibilidad de los Resultados , Universidades
3.
Neurochem Res ; 44(7): 1780, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31104195

RESUMEN

The authors regret that they neglected to cite their conference report on the technical part of a 'preliminary study' presented at, and published in, the Biomedical Sciences and Engineering Conference (BSEC), 2010, May 25-26 (Fully automated segmentation and characterization of the dendritic trees of retinal horizontal neurons -DOI: 10.1109/BSEC.2010.5510843 ), as it related to the larger dataset presented as validation of the method in the Neurochemical Research article (Automated Tracing of Horizontal Neuron Processes During Retinal Development- Neurochem Res. 2011 Apr;36(4):583-93). This resulted in the lack of transparency on the re-use and duplication of introductory text, which should have been cited. No figures or tables were reproduced, but rather larger confirmatory data and different set of results were reported. Appropriate authors were cited in both papers.

4.
Dev Biol ; 429(1): 105-117, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28716713

RESUMEN

Myc proto-oncogenes regulate diverse cellular processes during development, but their roles during morphogenesis of specific tissues are not fully understood. We found that c-myc regulates cell proliferation in mouse lens development and previous genome-wide studies suggested functional roles for N-myc in developing lens. Here, we examined the role of N-myc in mouse lens development. Genetic inactivation of N-myc in the surface ectoderm or lens vesicle impaired eye and lens growth, while "late" inactivation in lens fibers had no effect. Unexpectedly, defective growth of N-myc-deficient lenses was not associated with alterations in lens progenitor cell proliferation or survival. Notably, N-myc-deficient lens exhibited a delay in degradation of DNA in terminally differentiating lens fiber cells. RNA-sequencing analysis of N-myc-deficient lenses identified a cohort of down-regulated genes associated with fiber cell differentiation that included DNaseIIß. Further, an integrated analysis of differentially expressed genes in N-myc-deficient lens using normal lens expression patterns of iSyTE, N-myc-binding motif analysis and molecular interaction data from the String database led to the derivation of an N-myc-based gene regulatory network in the lens. Finally, analysis of N-myc and c-myc double-deficient lens demonstrated that these Myc genes cooperate to drive lens growth prior to lens vesicle stage. Together, these findings provide evidence for exclusive and cooperative functions of Myc transcription factors in mouse lens development and identify novel mechanisms by which N-myc regulates cell differentiation during eye morphogenesis.


Asunto(s)
Diferenciación Celular , Cristalino/citología , Cristalino/crecimiento & desarrollo , Proteína Proto-Oncogénica N-Myc/metabolismo , Animales , Diferenciación Celular/genética , Núcleo Celular/metabolismo , Proliferación Celular/genética , Supervivencia Celular/genética , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Cristalino/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transcripción Genética , Transcriptoma/genética
5.
Proc Biol Sci ; 284(1852)2017 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-28381624

RESUMEN

The unique eyes of the four-eyed fish Anableps anableps have long intrigued biologists. Key features associated with the bulging eye of Anableps include the expanded frontal bone and the duplicated pupils and cornea. Furthermore, the Anableps retina expresses different photoreceptor genes in dorsal and ventral regions, potentially associated with distinct aerial and aquatic stimuli. To gain insight into the developmental basis of the Anableps unique eye, we examined neurocranium and eye ontogeny, as well as photoreceptor gene expression during larval stages. First, we described six larval stages during which duplication of eye structures occurs. Our osteological analysis of neurocranium ontogeny revealed another distinctive Anablepid feature: an ossified interorbital septum partially separating the orbital cavities. Furthermore, we identified the onset of differences in cell proliferation and cell layer density between dorsal and ventral regions of the retina. Finally, we show that differential photoreceptor gene expression in the retina initiates during development, suggesting that it is inherited and not environmentally determined. In sum, our results shed light on the ontogenetic steps leading to the highly derived Anableps eye.


Asunto(s)
Ciprinodontiformes/embriología , Ojo/embriología , Retina/fisiología , Animales , Cráneo , Visión Ocular
6.
An Acad Bras Cienc ; 87(2 Suppl): 1323-48, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26397828

RESUMEN

Genome modification technologies are powerful tools for molecular biology and related areas. Advances in animal transgenesis and genome editing technologies during the past three decades allowed systematic interrogation of gene function that can help model how the genome influences cellular physiology. Genetic engineering via homologous recombination (HR) has been the standard method to modify genomic sequences. Nevertheless, nuclease-guided genome editing methods that were developed recently, such as ZFN, TALEN and CRISPR/Cas, opened new perspectives for biomedical research. Here, we present a brief historical perspective of genome modification methods, focusing on transgenic mice models. Moreover, we describe how new techniques were discovered and improved, present the paradigm shifts and discuss their limitations and applications for biomedical research as well as possible future directions.


Asunto(s)
Animales Modificados Genéticamente/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Ingeniería Genética/métodos , Dedos de Zinc/genética , Animales , Marcación de Gen/métodos , Ratones , Ratas
7.
Proc Natl Acad Sci U S A ; 108(52): 21111-6, 2011 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-22160703

RESUMEN

Neuronal differentiation with respect to the acquisition of synaptic competence needs to be regulated precisely during neurogenesis to ensure proper formation of circuits at the right place and time in development. This regulation is particularly important for synaptic triads among photoreceptors, horizontal cells (HCs), and bipolar cells in the retina, because HCs are among the first cell types produced during development, and bipolar cells are among the last. HCs undergo a dramatic transition from vertically oriented neurites that form columnar arbors to overlapping laminar dendritic arbors with differentiation. However, how this process is regulated and coordinated with differentiation of photoreceptors and bipolar cells remains unknown. Previous studies have suggested that the retinoblastoma (Rb) tumor suppressor gene may play a role in horizontal cell differentiation and synaptogenesis. By combining genetic mosaic analysis of individual synaptic triads with neuroanatomic analyses and multiphoton live imaging of developing HCs, we found that Rb plays a cell-autonomous role in the reorganization of horizontal cell neurites as they differentiate. Aberrant vertical processes in Rb-deficient HCs form ectopic synapses with rods in the outer nuclear layer but lack bipolar dendrites. Although previous reports indicate that photoreceptor abnormalities can trigger formation of ectopic synapses, our studies now demonstrate that defects in a postsynaptic partner contribute to the formation of ectopic photoreceptor synapses in the mammalian retina.


Asunto(s)
Diferenciación Celular/fisiología , Dendritas/fisiología , Neurogénesis/fisiología , Células Horizontales de la Retina/citología , Proteína de Retinoblastoma/metabolismo , Sinapsis/fisiología , Animales , Ratones , Microscopía Confocal , Microscopía Electrónica de Transmisión , Proteína de Retinoblastoma/genética
8.
J Biol Chem ; 287(42): 35506-35515, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-22910907

RESUMEN

The prion protein (PrP(C)) is a cell surface protein expressed mainly in the nervous system. In addition to the role of its abnormal conformer in transmissible spongiform encephalopathies, normal PrP(C) may be implicated in other degenerative conditions often associated with inflammation. PrP(C) is also present in cells of hematopoietic origin, including T cells, dendritic cells, and macrophages, and it has been shown to modulate their functions. Here, we investigated the impact of inflammation and stress on the expression and function of PrP(C) in neutrophils, a cell type critically involved in both acute and chronic inflammation. We found that systemic injection of LPS induced transcription and translation of PrP(C) in mouse neutrophils. Up-regulation of PrP(C) was dependent on the serum content of TGF-ß and glucocorticoids (GC), which, in turn, are contingent on the activation of the hypothalamic-pituitary-adrenal axis in response to systemic inflammation. GC and TGF-ß, either alone or in combination, directly up-regulated PrP(C) in neutrophils, and accordingly, the blockade of GC receptors in vivo curtailed the LPS-induced increase in the content of PrP(C). Moreover, GC also mediated up-regulation of PrP(C) in neutrophils following noninflammatory restraint stress. Finally, neutrophils with up-regulated PrP(C) presented enhanced peroxide-dependent cytotoxicity to endothelial cells. The data demonstrate a novel interplay of the nervous, endocrine, and immune systems upon both the expression and function of PrP(C) in neutrophils, which may have a broad impact upon the physiology and pathology of various organs and systems.


Asunto(s)
Regulación de la Expresión Génica , Sistema Hipotálamo-Hipofisario/metabolismo , Neutrófilos/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Proteínas PrPC/biosíntesis , Estrés Fisiológico , Animales , Glucocorticoides/genética , Glucocorticoides/inmunología , Glucocorticoides/metabolismo , Sistema Hipotálamo-Hipofisario/inmunología , Sistema Hipotálamo-Hipofisario/patología , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos/farmacología , Ratones , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/patología , Sistema Hipófiso-Suprarrenal/inmunología , Sistema Hipófiso-Suprarrenal/patología , Proteínas PrPC/genética , Proteínas PrPC/inmunología , Enfermedades por Prión/genética , Enfermedades por Prión/inmunología , Enfermedades por Prión/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/genética , Biosíntesis de Proteínas/inmunología , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Transcripción Genética/inmunología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo
9.
Vaccines (Basel) ; 11(11)2023 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-38006064

RESUMEN

Mucosal vaccination appears to be suitable to protect against SARS-CoV-2 infection. In this study, we tested an intranasal mucosal vaccine candidate for COVID-19 that consisted of a cationic liposome containing a trimeric SARS-CoV-2 spike protein and CpG-ODNs, a Toll-like receptor 9 agonist, as an adjuvant. In vitro and in vivo experiments indicated the absence of toxicity following the intranasal administration of this vaccine formulation. First, we found that subcutaneous or intranasal vaccination protected hACE-2 transgenic mice from infection with the wild-type (Wuhan) SARS-CoV-2 strain, as shown by weight loss and mortality indicators. However, when compared with subcutaneous administration, the intranasal route was more effective in the pulmonary clearance of the virus and induced higher neutralizing antibodies and anti-S IgA titers. In addition, the intranasal vaccination afforded protection against gamma, delta, and omicron virus variants of concern. Furthermore, the intranasal vaccine formulation was superior to intramuscular vaccination with a recombinant, replication-deficient chimpanzee adenovirus vector encoding the SARS-CoV-2 spike glycoprotein (Oxford/AstraZeneca) in terms of virus lung clearance and production of neutralizing antibodies in serum and bronchial alveolar lavage (BAL). Finally, the intranasal liposomal formulation boosted heterologous immunity induced by previous intramuscular vaccination with the Oxford/AstraZeneca vaccine, which was more robust than homologous immunity.

10.
Neurochem Res ; 36(4): 583-93, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21221777

RESUMEN

In the developing mammalian retina, horizontal neurons undergo a dramatic reorganization of their processes shortly after they migrate to their appropriate laminar position. This is an important process because it is now understood that the apical processes are important for establishing the regular mosaic of horizontal cells in the retina and proper reorganization during lamination is required for synaptogenesis with photoreceptors and bipolar neurons. However, this process is difficult to study because the analysis of horizontal neuron anatomy is labor intensive and time-consuming. In this paper, we present a computational method for automatically tracing the three-dimensional (3-D) dendritic structure of horizontal retinal neurons in two-photon laser scanning microscope (TPLSM) imagery. Our method is based on 3-D skeletonization and is thus able to preserve the complex structure of the dendritic arbor of these cells. We demonstrate the effectiveness of our approach by comparing our tracing results against two sets of semi-automated traces over a set of 10 horizontal neurons ranging in age from P1 to P5. We observe an average agreement level of 81% between our automated trace and the manual traces. This automated method will serve as an important starting point for further refinement and optimization.


Asunto(s)
Neuronas/fisiología , Retina/embriología , Animales , Ratones , Ratones Transgénicos , Retina/crecimiento & desarrollo
11.
Front Cell Dev Biol ; 9: 731308, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34805142

RESUMEN

Several inherited human syndromes that severely affect organogenesis and other developmental processes are caused by mutations in replication stress response (RSR) genes. Although the molecular machinery of RSR is conserved, disease-causing mutations in RSR-genes may have distinct tissue-specific outcomes, indicating that progenitor cells may differ in their responses to RSR inactivation. Therefore, understanding how different cell types respond to replication stress is crucial to uncover the mechanisms of RSR-related human syndromes. Here, we review the ocular manifestations in RSR-related human syndromes and summarize recent findings investigating the mechanisms of RSR during eye development in vivo. We highlight a remarkable heterogeneity of progenitor cells responses to RSR inactivation and discuss its implications for RSR-related human syndromes.

12.
Int J Dev Biol ; 65(4-5-6): 245-250, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33372686

RESUMEN

Vertebrate eyes share the same general organization, though species have evolved morphological and functional adaptations to diverse environments. Cave-adapted animals are characterized by a variety of features including eye reduction, loss of body pigmentation, and enhanced non-visual sensory systems. Species that live in perpetual darkness have also evolved sensory mechanisms that are independent of light stimuli. The subterranean catfish Phreatobius cisternarum lives in the Amazonian phreatic zone and displays a diversity of morphological features that are similar to those observed in cavefish and appear to be adaptations to life in the dark. Here we combine histological and transcriptome analyses to characterize sensory adaptations of P. cisternarum to the subterranean environment. Histological analysis showed that the vestigial eyes of P. cisternarum contain a rudimentary lens. Transcriptome analysis revealed a repertoire of eleven visual and non-visual opsins and the expression of 36 genes involved in lens development and maintenance. In contrast to other cavefish species, such as Astyanax mexicanus, Phreatichthys andruzzii, Sinocyclocheilus anophthalmus and Sinocyclocheilus microphthalmus, DASPEI neuromast staining patterns did not show an increase in the number of sensory hair cells. Our work reveals unique adaptations in the visual system of P. cisternarum to underground habitats and helps to shed light into troglomorphic attributes of subterranean animals.


Asunto(s)
Adaptación Fisiológica , Bagres , Ojo/crecimiento & desarrollo , Animales , Evolución Biológica , Cuevas
13.
Eur J Neurosci ; 32(3): 311-21, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20646049

RESUMEN

During retinal development, cell proliferation and exit from the cell cycle must be precisely regulated to ensure the generation of the appropriate numbers and proportions of the various retinal cell types. Previously, we showed that pituitary adenylyl cyclase-activating polypeptide (PACAP) exerts a neuroprotective effect in the developing retina of rats, through the cAMP-cAMP-dependent protein kinase (protein kinase A) (PKA) pathway. Here, we show that PACAP also regulates the proliferation of retinal progenitor cells. PACAP, PACAP-specific receptor (PAC1), and the receptors activated by both PACAP and vasoactive intestinal peptide (VIP), VPAC1 and VPAC2, are expressed during embryonic and postnatal development of the rat retina. Treatment of retinal explants with PACAP38 reduced the incorporation of [(3)H]thymidine as well as the number of 5-bromo-2'-deoxyuridine-positive and cyclin D1-positive cells. Pharmacological experiments indicated that PACAP triggers this antiproliferative effect through the activation of both PAC1 and VPACs, and the cAMP-PKA pathway. In addition, PACAP receptor activation decreased both cyclin D1 mRNA and protein content. Altogether, the data support the hypothesis that PACAP is a cell-extrinsic regulator with multiple roles during retinal development, including the regulation of proliferation in a subpopulation of retinal progenitor cells.


Asunto(s)
Proliferación Celular , Ciclina D1/metabolismo , Regulación hacia Abajo , Neurogénesis/fisiología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Retina/metabolismo , Neuronas Retinianas/metabolismo , Análisis de Varianza , Animales , Western Blotting , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ciclina D1/genética , Inmunohistoquímica , Microscopía Confocal , Fosforilación , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Receptores de Péptido Intestinal Vasoactivo/genética , Receptores de Péptido Intestinal Vasoactivo/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre , Técnicas de Cultivo de Tejidos , Péptido Intestinal Vasoactivo/metabolismo
14.
Genes (Basel) ; 11(7)2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32630049

RESUMEN

The precise replication of DNA and the successful segregation of chromosomes are essential for the faithful transmission of genetic information during the cell cycle. Alterations in the dynamics of genome replication, also referred to as DNA replication stress, may lead to DNA damage and, consequently, mutations and chromosomal rearrangements. Extensive research has revealed that DNA replication stress drives genome instability during tumorigenesis. Over decades, genetic studies of inherited syndromes have established a connection between the mutations in genes required for proper DNA repair/DNA damage responses and neurological diseases. It is becoming clear that both the prevention and the responses to replication stress are particularly important for nervous system development and function. The accurate regulation of cell proliferation is key for the expansion of progenitor pools during central nervous system (CNS) development, adult neurogenesis, and regeneration. Moreover, DNA replication stress in glial cells regulates CNS tumorigenesis and plays a role in neurodegenerative diseases such as ataxia telangiectasia (A-T). Here, we review how replication stress generation and replication stress response (RSR) contribute to the CNS development, homeostasis, and disease. Both cell-autonomous mechanisms, as well as the evidence of RSR-mediated alterations of the cellular microenvironment in the nervous system, were discussed.


Asunto(s)
Replicación del ADN , Homeostasis , Enfermedades del Sistema Nervioso/genética , Sistema Nervioso/metabolismo , Animales , Daño del ADN , Inestabilidad Genómica , Humanos , Enfermedades del Sistema Nervioso/metabolismo
15.
Front Cell Dev Biol ; 8: 711, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32850831

RESUMEN

Genomic instability in the central nervous system (CNS) is associated with defective neurodevelopment and neurodegeneration. Congenital human syndromes that affect the CNS development originate from mutations in genes of the DNA damage response (DDR) pathways. RINT1 (Rad50-interacting protein 1) is a partner of RAD50, that participates in the cellular responses to DNA double-strand breaks (DSB). Recently, we showed that Rint1 regulates cell survival in the developing brain and its loss led to premature lethality associated with genomic stability. To bypass the lethality of Rint1 inactivation in the embryonic brain and better understand the roles of RINT1 in CNS development, we conditionally inactivated Rint1 in retinal progenitor cells (RPCs) during embryogenesis. Rint1 loss led to accumulation of endogenous DNA damage, but RINT1 was not necessary for the cell cycle checkpoint activation in these neural progenitor cells. As a consequence, proliferating progenitors and postmitotic neurons underwent apoptosis causing defective neurogenesis of retinal ganglion cells, malformation of the optic nerve and blindness. Notably, inactivation of Trp53 prevented apoptosis of the RPCs and rescued the generation of retinal neurons and vision loss. Together, these results revealed an essential role for TRP53-mediated apoptosis in the malformations of the visual system caused by RINT1 loss and suggests that defective responses to DNA damage drive retinal malformations.

16.
Cell Death Dis ; 11(10): 923, 2020 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-33110058

RESUMEN

The maintenance of genomic stability during the cell cycle of progenitor cells is essential for the faithful transmission of genetic information. Mutations in genes that ensure genome stability lead to human developmental syndromes. Mutations in Ataxia Telangiectasia and Rad3-related (ATR) or in ATR-interacting protein (ATRIP) lead to Seckel syndrome, which is characterized by developmental malformations and short life expectancy. While the roles of ATR in replicative stress response and chromosomal segregation are well established, it is unknown how ATRIP contributes to maintaining genomic stability in progenitor cells in vivo. Here, we generated the first mouse model to investigate ATRIP function. Conditional inactivation of Atrip in progenitor cells of the CNS and eye led to microcephaly, microphthalmia and postnatal lethality. To understand the mechanisms underlying these malformations, we used lens progenitor cells as a model and found that ATRIP loss promotes replicative stress and TP53-dependent cell death. Trp53 inactivation in Atrip-deficient progenitor cells rescued apoptosis, but increased mitotic DNA damage and mitotic defects. Our findings demonstrate an essential role of ATRIP in preventing DNA damage accumulation during unchallenged replication.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Daño del ADN/genética , Replicación del ADN/genética , Proteínas de Unión al ADN/genética , Células Madre/metabolismo , Animales , Proliferación Celular , Humanos , Ratones
17.
Dis Model Mech ; 13(10)2020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-32994318

RESUMEN

Seckel syndrome is a type of microcephalic primordial dwarfism (MPD) that is characterized by growth retardation and neurodevelopmental defects, including reports of retinopathy. Mutations in key mediators of the replication stress response, the mutually dependent partners ATR and ATRIP, are among the known causes of Seckel syndrome. However, it remains unclear how their deficiency disrupts the development and function of the central nervous system (CNS). Here, we investigated the cellular and molecular consequences of ATRIP deficiency in different cell populations of the developing murine neural retina. We discovered that conditional inactivation of Atrip in photoreceptor neurons did not affect their survival or function. In contrast, Atrip deficiency in retinal progenitor cells (RPCs) led to severe lamination defects followed by secondary photoreceptor degeneration and loss of vision. Furthermore, we showed that RPCs lacking functional ATRIP exhibited higher levels of replicative stress and accumulated endogenous DNA damage that was accompanied by stabilization of TRP53. Notably, inactivation of Trp53 prevented apoptosis of Atrip-deficient progenitor cells and was sufficient to rescue retinal dysplasia, neurodegeneration and loss of vision. Together, these results reveal an essential role of ATRIP-mediated replication stress response in CNS development and suggest that the TRP53-mediated apoptosis of progenitor cells might contribute to retinal malformations in Seckel syndrome and other MPD disorders.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Anomalías Múltiples/patología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al ADN/metabolismo , Degeneración Nerviosa/patología , Displasia Retiniana/patología , Células Madre/patología , Animales , Apoptosis , Ceguera/patología , Muerte Celular , Proliferación Celular , Daño del ADN , Modelos Animales de Enfermedad , Embrión de Mamíferos/patología , Desarrollo Embrionario , Ratones , Degeneración Nerviosa/complicaciones , Neurogénesis , Células Fotorreceptoras de Vertebrados/patología , Retina/patología , Displasia Retiniana/complicaciones , Síndrome , Proteína p53 Supresora de Tumor/metabolismo , Visión Ocular
18.
Brain Res ; 1192: 37-60, 2008 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-17597590

RESUMEN

In the developing vertebrate retina, precise coordination of retinal progenitor cell proliferation and cell-cycle exit is essential for the formation of a functionally mature retina. Unregulated or disrupted cell proliferation may lead to dysplasia, retinal degeneration or retinoblastoma. Both cell-intrinsic and -extrinsic factors regulate the proliferation of progenitor cells during CNS development. There is now growing evidence that in the developing vertebrate retina, both slow and fast neurotransmitter systems modulate the proliferation of retinal progenitor cells. Classic neurotransmitters, such as GABA (gamma-amino butyric acid), glycine, glutamate, ACh (acetylcholine) and ATP (adenosine triphosphate) are released, via vesicular or non-vesicular mechanisms, into the immature retinal environment. Furthermore, these neurotransmitters signal through functional receptors even before synapses are formed. Recent evidence indicates that the activation of purinergic and muscarinic receptors may regulate the cell-cycle machinery and consequently the expansion of the retinal progenitor pool. Interestingly, GABA and glutamate appear to have opposing roles, inducing retinal progenitor cell-cycle exit. In this review, we present recent findings that begin to elucidate the roles of neurotransmitters as regulators of progenitor cell proliferation at early stages of retinal development. These studies also raise several new questions, including how these neurotransmitters regulate specific cell-cycle pathways and the mechanisms by which retinal progenitor cells integrate the signals from neurotransmitters and other exogenous factors during vertebrate retina development.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular , Neuronas/metabolismo , Neurotransmisores/metabolismo , Retina/embriología , Células Madre/metabolismo , Animales , Ciclo Celular/fisiología , Humanos , Neuronas/citología , Retina/citología , Retina/crecimiento & desarrollo , Transducción de Señal/fisiología , Células Madre/citología
19.
Transl Oncol ; 10(5): 726-733, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28734226

RESUMEN

Esophageal cancer is the sixth most common cause of cancer-related death worldwide. Current chemotherapy regimens include a combination of 5-fluorouracil (5-FU) and cisplatin, but more efficient therapy strategies are needed to increase 5-year survival. Alterations in the signaling pathway of the tumor suppressor gene Rb-1, which encodes a phosphoprotein (pRB) that negatively regulates the G1/S transition of the cell cycle, are present in 70% of all tumors, but its role in esophageal cancer is still unclear. Most of these are alterations leading to up-regulation of the activity of cyclin-dependent kinases (CDKs) to phosphorylate pRB, which suggests that keeping the wild type pRB phosphorylated might be advantageous. Besides proliferation, pRB also regulates apoptosis induced by tumor necrosis factor-alpha (TNF-α) and DNA-damage. We investigated the status of phosphorylation of pRB along esophageal tumorigenesis stages, as well as whether hyperphosphorylation of pRB could suppress apoptosis induced by cisplatin, 5-FU, or TNF-α in esophageal cancer cells. pRB phosphorylation increased progressively from normal esophageal tissue to metaplasia and adenocarcinoma, suggesting that pRB phosphorylation increases along esophageal tumor stages. When RB-1 was knocked down or CDK inhibitors reduced the levels of phosphorylated pRB, opposite apoptotic effects were observed, depending on the combination of drugs tested: whereas TNF-α- and cisplatin-induced apoptosis increased, 5-FU-induced apoptosis decreased. Taken together, these data suggest that pRB plays a role in esophageal adenocarcinoma and that, depending on the type of anti-cancer treatment, combining CDK inhibitors and chemotherapy has the potential to increase the sensitivity of esophageal cancer cells to cell death.

20.
Prog Retin Eye Res ; 24(4): 457-91, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15845345

RESUMEN

It has long been known that a barrage of signals from neighboring and connecting cells, as well as components of the extracellular matrix, control cell survival. Given the extensive repertoire of retinal neurotransmitters, neuromodulators and neurotrophic factors, and the exhuberant interconnectivity of retinal interneurons, it is likely that various classes of released neuroactive substances may be involved in the control of sensitivity to retinal cell death. The aim of this article is to review evidence that neurotransmitters and neuropeptides control the sensitivity to programmed cell death in the developing retina. Whereas the best understood mechanism of execution of cell death is that of caspase-mediated apoptosis, current evidence shows that not only there are many parallel pathways to apoptotic cell death, but non-apoptotic programs of execution of cell death are also available, and may be triggered either in isolation or combined with apoptosis. The experimental data show that many upstream signaling pathways can modulate cell death, including those dependent on the second messengers cAMP-PKA, calcium and nitric oxide. Evidence for anterograde neurotrophic control is provided by a variety of models of the central nervous system, and the data reviewed here indicate that an early function of certain neurotransmitters, such as glutamate and dopamine, as well as neuropeptides such as pituitary adenylyl cyclase-activating polypeptide and vasoactive intestinal peptide is the trophic support of cell populations in the developing retina. This may have implications both regarding the mechanisms of retinal organogenesis, as well as pathological conditions leading to retinal dystrophies and to dysfunctional cellular behavior.


Asunto(s)
Apoptosis/fisiología , Neuropéptidos/fisiología , Neurotransmisores/fisiología , Retina/crecimiento & desarrollo , Animales , Supervivencia Celular , Humanos , Retina/citología , Retina/metabolismo , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA