Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Nucleic Acids Res ; 52(8): 4167-4184, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38324473

RESUMEN

Sam68 and SLM2 are paralog RNA binding proteins (RBPs) expressed in the cerebral cortex and display similar splicing activities. However, their relative functions during cortical development are unknown. We found that these RBPs exhibit an opposite expression pattern during development. Sam68 expression declines postnatally while SLM2 increases after birth, and this developmental pattern is reinforced by hierarchical control of Sam68 expression by SLM2. Analysis of Sam68:Slm2 double knockout (Sam68:Slm2dko) mice revealed hundreds of exons that respond to joint depletion of these proteins. Moreover, parallel analysis of single and double knockout cortices indicated that exons regulated mainly by SLM2 are characterized by a dynamic splicing pattern during development, whereas Sam68-dependent exons are spliced at relatively constant rates. Dynamic splicing of SLM2-sensitive exons is completely suppressed in the Sam68:Slm2dko developing cortex. Sam68:Slm2dko mice die perinatally with defects in neurogenesis and in neuronal differentiation, and develop a hydrocephalus, consistent with splicing alterations in genes related to these biological processes. Thus, our study reveals that developmental control of separate Sam68 and Slm2 paralog genes encoding homologous RBPs enables the orchestration of a dynamic splicing program needed for brain development and viability, while ensuring a robust redundant mechanism that supports proper cortical development.


Asunto(s)
Corteza Cerebral , Empalme del ARN , Proteínas de Unión al ARN , Animales , Ratones , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Corteza Cerebral/metabolismo , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Exones/genética , Regulación del Desarrollo de la Expresión Génica , Ratones Noqueados , Neurogénesis/genética , Neuronas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
2.
Hum Mutat ; 43(1): 67-73, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34747546

RESUMEN

Biallelic mutations in the BRAT1 gene, encoding BRCA1-associated ATM activator 1, result in variable phenotypes, from rigidity and multifocal seizure syndrome, lethal neonatal to neurodevelopmental disorder, and cerebellar atrophy with or without seizures, without obvious genotype-phenotype associations. We describe two families at the mildest end of the spectrum, differing in clinical presentation despite a common genotype at the BRAT1 locus. Two siblings displayed nonprogressive congenital ataxia and shrunken cerebellum on magnetic resonance imaging. A third unrelated patient showed normal neurodevelopment, adolescence-onset seizures, and ataxia, shrunken cerebellum, and ultrastructural abnormalities on skin biopsy, representing the mildest form of NEDCAS hitherto described. Exome sequencing identified the c.638dup and the novel c.1395G>A BRAT1 variants, the latter causing exon 10 skippings. The p53-MCL test revealed normal ATM kinase activity. Our findings broaden the allelic and clinical spectrum of BRAT1-related disease, which should be suspected in presence of nonprogressive cerebellar signs, even without a neurodevelopmental disorder.


Asunto(s)
Proteínas Nucleares , Convulsiones , Estudios de Asociación Genética , Genotipo , Humanos , Mutación , Proteínas Nucleares/genética , Fenotipo , Convulsiones/genética
3.
Cell Mol Life Sci ; 78(19-20): 6431-6451, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34406416

RESUMEN

In the past two decades, mounting evidence has modified the classical view of the cerebellum as a brain region specifically involved in the modulation of motor functions. Indeed, clinical studies and engineered mouse models have highlighted cerebellar circuits implicated in cognitive functions and behavior. Furthermore, it is now clear that insults occurring in specific time windows of cerebellar development can affect cognitive performance later in life and are associated with neurological syndromes, such as Autism Spectrum Disorder. Despite its almost homogenous cytoarchitecture, how cerebellar circuits form and function is not completely elucidated yet. Notably, the apparently simple neuronal organization of the cerebellum, in which Purkinje cells represent the only output, hides an elevated functional diversity even within the same neuronal population. Such complexity is the result of the integration of intrinsic morphogenetic programs and extracellular cues from the surrounding environment, which impact on the regulation of the transcriptome of cerebellar neurons. In this review, we briefly summarize key features of the development and structure of the cerebellum before focusing on the pathways involved in the acquisition of the cerebellar neuron identity. We focus on gene expression and mRNA processing programs, including mRNA methylation, trafficking and splicing, that are set in motion during cerebellar development and participate to its physiology. These programs are likely to add new layers of complexity and versatility that are fundamental for the adaptability of cerebellar neurons.


Asunto(s)
Cerebelo/fisiología , Transcriptoma/genética , Animales , Trastorno del Espectro Autista/genética , Humanos , Neurogénesis/genética , Neuronas/fisiología , Células de Purkinje/fisiología
4.
Int J Mol Sci ; 23(20)2022 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-36293427

RESUMEN

Meiosis is the unique division of germ cells resulting in the recombination of the maternal and paternal genomes and the production of haploid gametes. In mammals, it begins during the fetal life in females and during puberty in males. In both cases, entering meiosis requires a timely switch from the mitotic to the meiotic cell cycle and the transition from a potential pluripotent status to meiotic differentiation. Revealing the molecular mechanisms underlying these interrelated processes represents the essence in understanding the beginning of meiosis. Meiosis facilitates diversity across individuals and acts as a fundamental driver of evolution. Major differences between sexes and among species complicate the understanding of how meiosis begins. Basic meiotic research is further hindered by a current lack of meiotic cell lines. This has been recently partly overcome with the use of primordial-germ-cell-like cells (PGCLCs) generated from pluripotent stem cells. Much of what we know about this process depends on data from model organisms, namely, the mouse; in mice, the process, however, appears to differ in many aspects from that in humans. Identifying the mechanisms and molecules controlling germ cells to enter meiosis has represented and still represents a major challenge for reproductive medicine. In fact, the proper execution of meiosis is essential for fertility, for maintaining the integrity of the genome, and for ensuring the normal development of the offspring. The main clinical consequences of meiotic defects are infertility and, probably, increased susceptibility to some types of germ-cell tumors. In the present work, we report and discuss data mainly concerning the beginning of meiosis in mammalian female germ cells, referring to such process in males only when pertinent. After a brief account of this process in mice and humans and an historical chronicle of the major hypotheses and progress in this topic, the most recent results are reviewed and discussed.


Asunto(s)
Meiosis , Células Madre Pluripotentes , Humanos , Masculino , Femenino , Ratones , Animales , Meiosis/genética , Células Germinativas/metabolismo , Diferenciación Celular , Mamíferos/genética
5.
J Cell Mol Med ; 25(1): 383-396, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33236849

RESUMEN

STRA8 (Stimulated by Retinoic Acid Gene 8) controls the crucial decision of germ cells to engage meiotic division up and down-regulating genes involved in the meiotic programme. It has been proven as an amplifier of genes involved in cell cycle control and chromosome events, however, how STRA8 functions as negative regulator are not well understood. In this study, we demonstrate that STRA8 can interact with itself and with other basic Helix-Loop-Helix (bHLH) transcription factors through its HLH domain and that this domain is important for its ability to negatively interfere with the Ebox-mediated transcriptional activity of bHLH transcription factors. Significantly, we show that STRA8 interacts with TCF3/E47, a class I bHLH transcription factors, and with SOHLH1, a gonadal-specific bHLH, in male germ cells obtained from prepuberal mouse testis. We demonstrated that STRA8, indirectly, is able to exert a negative control on the SOHLH1-dependent stimulation of c-KIT expression in late differentiating spermatogonia and preleptotene spermatocytes. Although part of this results were obtained only 'in vitro', they support the notion that STRA8 interacting with different transcription factors, besides its established role as 'amplifier' of meiotic programme, is able to finely modulate the balance between spermatogonia proliferation, differentiation and acquisition of meiotic competence.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Western Blotting , Femenino , Técnica del Anticuerpo Fluorescente , Células HEK293 , Humanos , Masculino , Unión Proteica , Proteínas Proto-Oncogénicas c-kit/genética
6.
Cells ; 13(14)2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-39056758

RESUMEN

Autism spectrum disorders (ASDs) are complex neurodevelopmental conditions characterized by deficits in social interaction and communication, as well as repetitive behaviors. Although the etiology of ASD is multifactorial, with both genetic and environmental factors contributing to its development, a strong genetic basis is widely recognized. Recent research has identified numerous genetic mutations and genomic rearrangements associated with ASD-characterizing genes involved in brain development. Alterations in developmental programs are particularly harmful during critical periods of brain development. Notably, studies have indicated that genetic disruptions occurring during the second trimester of pregnancy affect cortical development, while disturbances in the perinatal and early postnatal period affect cerebellar development. The developmental defects must be viewed in the context of the role of the cerebellum in cognitive processes, which is now well established. The present review emphasizes the genetic complexity and neuropathological mechanisms underlying ASD and aims to provide insights into the cerebellar involvement in the disorder, focusing on recent advances in the molecular landscape governing its development in humans. Furthermore, we highlight when and in which cerebellar neurons the ASD-associated genes may play a role in the development of cortico-cerebellar circuits. Finally, we discuss improvements in protocols for generating cerebellar organoids to recapitulate the long period of development and maturation of this organ. These models, if generated from patient-induced pluripotent stem cells (iPSC), could provide a valuable approach to elucidate the contribution of defective genes to ASD pathology and inform diagnostic and therapeutic strategies.


Asunto(s)
Trastorno del Espectro Autista , Cerebelo , Humanos , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/patología , Cerebelo/patología , Cerebelo/crecimiento & desarrollo , Animales
7.
Biol Reprod ; 88(6): 145, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23636811

RESUMEN

In the present study, we demonstrate that minimal concentrations (≤ 1 nM) of retinoic acid (RA), equivalent to the quantity contaminating serum-containing culture medium, are sufficient to promote meiotic entry and progression through meiotic prophase I (MPI) stages in isolated 12.5-days postcoitum (dpc) XX and XY mouse primordial germ cells (PGCs) in culture. Similarly, we found that the same low RA concentration up-regulated or induced stimulation by retinoic acid 8 (Stra8) in such cells, both at mRNA and protein level. In preleptotene/leptotene germ cells, STRA8 was localized in nuclear dots that disappeared at later MPI stages. In addition to Stra8, other meiotic genes such as Dmc1 and Rec8 appeared stimulated by RA directly in PGCs with similar concentration-dependent trends. Finally, we found that RA induced Stra8, Sycp3, Dmc1, and Rec8 transcripts, promoting meiotic entry in culture also in pregonadal 10.5-dpc PGCs of both sexes. When cultured isolated from somatic cells, such PGCs, however, were unable to progress through MPI stages, while after entering meiosis, they progressed through MPI when cultured within aorta/gonad/mesonephros tissues. We conclude that besides RA, germ cell intrinsic factors and other exogenous signals from the surrounding somatic cells are probably necessary for meiotic entry and progression in mouse PGCs.


Asunto(s)
Células Germinativas/efectos de los fármacos , Meiosis/efectos de los fármacos , Proteínas/metabolismo , Tretinoina/farmacología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Células Cultivadas , Células Madre Embrionarias , Células Germinativas/metabolismo , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión a Fosfato , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas/genética
8.
Transl Psychiatry ; 13(1): 114, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-37019889

RESUMEN

Autism spectrum disorder (ASD) includes a set of highly heritable neurodevelopmental syndromes characterized by social and communication impairment, repetitive behaviour, and intellectual disability. Although mutations in multiple genes have been associated to ASD, most patients lack detectable genetic alterations. For this reason, environmental factors are commonly thought to also contribute to ASD aetiology. Transcriptome analyses have revealed that autistic brains possess distinct gene expression signatures, whose elucidation can provide insights about the mechanisms underlying the effects of ASD-causing genetic and environmental factors. Herein, we have identified a coordinated and temporally regulated programme of gene expression in the post-natal development of cerebellum, a brain area whose defects are strongly associated with ASD. Notably, this cerebellar developmental programme is significantly enriched in ASD-linked genes. Clustering analyses highlighted six different patterns of gene expression modulated during cerebellar development, with most of them being enriched in functional processes that are frequently dysregulated in ASD. By using the valproic acid mouse model of ASD, we found that ASD-linked genes are dysregulated in the developing cerebellum of ASD-like mice, a defect that correlates with impaired social behaviour and altered cerebellar cortical morphology. Moreover, changes in transcript levels were reflected in aberrant protein expression, indicating the functional relevance of these alterations. Thus, our work uncovers a complex ASD-related transcriptional programme regulated during cerebellar development and highlight genes whose expression is dysregulated in this brain area of an ASD mouse model.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Ratones , Animales , Trastorno Autístico/metabolismo , Ácido Valproico , Trastorno del Espectro Autista/genética , Cerebelo/metabolismo , Encéfalo
9.
Reprod Biol Endocrinol ; 9: 136, 2011 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-21978455

RESUMEN

BACKGROUND: Currently, there are no suitable assays available to evaluate the embryotoxicity of leached components from restorative dental materials. METHODS: The effect of the medium conditioned by composites and amalgam on mouse blastocysts in vitro was tested. The materials were also subcutaneously implanted, and the effect of the medium supplemented with serum from the host blood was evaluated in the embryotoxicity assay. The embryo implantation rate in the material-transplanted mothers was also evaluated. RESULTS: The results show that while the culture in media conditioned by amalgams did not affect blastocyst development, the medium conditioned by composites caused blastocyst degeneration and apoptosis. The development of blastocysts in a medium containing serum obtained from animals after transplantation was, however, without effect. Finally, inconsistent reduction in the implantation rate in transplanted mothers was observed. CONCLUSIONS: In this study, we provide examples of in vitro and in vivo tests that may be used to evaluate embryotoxicity for dental materials. Our results show that leached components from our composite-material induced embryotoxicity in vitro, however, no toxicity was observed when subcutaneously implanted in vivo. This highlights the necessity of integrated in vitro and in vivo tests for valuable predictive estimation of embryotoxicity for complex materials.


Asunto(s)
Blastocisto/efectos de los fármacos , Materiales Dentales/toxicidad , Restauración Dental Permanente/efectos adversos , Desarrollo Embrionario/efectos de los fármacos , Teratógenos/toxicidad , Pruebas de Toxicidad , Animales , Apoptosis/efectos de los fármacos , Blastocisto/patología , Resinas Compuestas/química , Resinas Compuestas/toxicidad , Medios de Cultivo Condicionados/química , Amalgama Dental/química , Amalgama Dental/toxicidad , Materiales Dentales/química , Ectogénesis/efectos de los fármacos , Técnicas de Cultivo de Embriones , Implantación del Embrión/efectos de los fármacos , Transferencia de Embrión , Femenino , Ensayo de Materiales , Ratones , Reproducibilidad de los Resultados , Solubilidad , Teratógenos/química
10.
Exp Cell Res ; 316(10): 1716-27, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20380832

RESUMEN

We report here that in the mouse embryonic gonads in addition to gonadal somatic cells, primordial germ cells (PGCs) the precursors of adult gametes, express estrogen receptor alpha (ERalpha) and that through this receptor, 17-beta-estradiol (E2) is able to modulate in such cells molecular signalling known to be crucial for their development. We demonstrated that PGCs from 11.5 to 12.5 days post coitum (dpc) mouse embryos express ERalpha transcripts and protein and that at concentrations of 10(-8)M E2 stimulates rapid (within 20 min) about 4-fold AKT (Ser473) and 3-fold ERK2 (Thr202/Tyr204) and SRC (Tyr418) phosphorylation. In addition, the E2 stimulatory effects were associated with increased phosphorylation of the KIT receptor (Tyr568/570). While the ER antagonist ICI182780 was able to abolish these effects, AKT phosphorylation induced by E2 was also inhibited by the PI3K inhibitor LY294002 and the SRC family inhibitor PP2. This latter was also able to abolish the increased phosphorylation of KIT and ERKs caused by E2. Taken together these results suggest that E2 may modulate via ERalpha non-genomic signalling/phosphorylation cascade in mouse PGCs. This was also supported by the finding that PGCs express MNAR, a scaffold protein that regulate ER activation in other cell types. Finally, we found that when PGCs were cultured in the presence of 10(-8)M E2 a significant ICI inhibitable increase of their number occurred. The present study provides evidence for novel direct non-genomic actions of estrogens on PGCs and suggests that these cells can represent a potential target for estrogens and estrogenic compounds during the early stages of embryo development in mammals.


Asunto(s)
Células Madre Embrionarias/metabolismo , Estrógenos/metabolismo , Células Germinativas/metabolismo , Animales , Secuencia de Bases , Cartilla de ADN/genética , Células Madre Embrionarias/efectos de los fármacos , Estradiol/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Células Germinativas/efectos de los fármacos , Edad Gestacional , Técnicas In Vitro , Masculino , Ratones , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/metabolismo
11.
Front Cell Dev Biol ; 9: 723388, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34557491

RESUMEN

Previous studies have shown that nicotine could impair the germ cell cyst breakdown and the primordial follicle assembly by autophagy. In this paper, we discovered that luteinizing hormone (LH) and follicle-stimulating hormone (FSH) could counteract the damage caused by nicotine of mouse germ cell cyst breakdown. The neonatal mice were separately intraperitoneally injected with nicotine, nicotine plus LH, nicotine plus FSH, and saline (control) for 4 days. Compared with the nicotine group, the quality of oocytes and the number of follicles were remarkably increased in the nicotine plus LH group or nicotine plus FSH group. LH and FSH could alleviate nicotine-induced oocyte autophagy by different pathways. LH reduced the nicotine-induced autophagy by restoring the phosphorylation level of adenosine 5'-monophosphate-activated protein kinase α-1, while FSH by downregulating the phosphorylation level of Forkhead box class O 1. In addition, in a subsequent study of 6-week mice in different treated groups, we found that LH and FSH supplementation significantly improved normal maturation rates, fertilization rates, and embryo's developmental potential of oocytes in oocytes exposed to nicotine. Taken together, these results suggested that LH and FSH could counteract the damage caused by nicotine and finally ensure normal germ cell cyst breakdown and early embryo development.

12.
J Biol Chem ; 284(51): 35781-93, 2009 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-19805549

RESUMEN

Stra8 (stimulated by retinoic acid 8) encodes a protein crucial for mammalian germ cells entering into premeiotic stages. Here, to elucidate the still unknown STRA8 molecular functions, we studied the cellular localization of the protein in several cell types, including premeiotic mouse germ cells and stem cell lines. We reported distinct STRA8 localization in germ and stem cell types and a heterogeneous protein distribution in the cytoplasm and nucleus of such cells suggesting that the protein can shuttle between these two compartments. Moreover, we identified specific protein motifs determining its nuclear import/export. Furthermore, we demonstrated that in transfected cell lines the nuclear import of STRA8 is an active process depending on an N-terminal basic nuclear localization signal. Moreover, its nuclear export is mainly mediated by the Exportin1 (XPO1) recognition of a nuclear export signal. Significantly, we also demonstrated that STRA8 associates with DNA and possesses transcriptional activity. These observations strongly suggest that STRA8 can exert important functions in the nucleus rather than in the cytoplasm as believed previously, likely depending on the cell type and regulated by its nuclear-cytoplasmic shuttling.


Asunto(s)
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células Germinativas/metabolismo , Proteínas/metabolismo , Células Madre/metabolismo , Transcripción Genética/fisiología , Transporte Activo de Núcleo Celular/fisiología , Proteínas Adaptadoras Transductoras de Señales , Secuencias de Aminoácidos/fisiología , Animales , Núcleo Celular/genética , Citoplasma/genética , ADN/genética , ADN/metabolismo , Células Germinativas/citología , Carioferinas/genética , Carioferinas/metabolismo , Meiosis/fisiología , Ratones , Señales de Localización Nuclear/genética , Señales de Localización Nuclear/metabolismo , Proteínas/genética , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Células Madre/citología , Proteína Exportina 1
13.
Cell Death Dis ; 11(4): 240, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32303676

RESUMEN

Homologous recombination and chromosome segregation in meiosis rely on the timely expression of two splice variants of the endonuclease SPO11, named α and ß, which respectively skip or include exon 2. However, in spite of its physiological importance, the mechanism underlying Spo11 alternative splicing in meiosis is still unknown. By screening the activity of factors that are predicted to bind the alternatively spliced region of Spo11, we identified hnRNPH as a key regulator of SPO11α splicing in mouse spermatocytes. Although hnRNPH was not upregulated in meiosis concomitantly with the switch in splicing, its recruitment to Spo11 pre-mRNA was favored by selective modulation of RNA polymerase II (RNAPII) phosphorylation and processivity in proximity of exon 2. The hnRNPH binding sites were localized near those of splicing factors that promote SPO11ß splicing, suggesting that hnRNPH favors exon 2 skipping by competing out positive regulators. Indeed, hnRNPH binds proximal to a consensus motif for Sam68, a positive regulator of SPO11ß splicing in vitro and in vivo, and it interferes with Sam68 binding to the Spo11 pre-mRNA. Thus, our work reveals that modulation of RNAPII dynamics in concert with hnRNPH recruitment exerts a combinatorial control of the timely regulated Spo11 splicing during meiosis.


Asunto(s)
Empalme Alternativo/genética , Endodesoxirribonucleasas/metabolismo , Meiosis/genética , ARN Polimerasa II/genética , Espermatocitos/metabolismo , Espermatogénesis/genética , Animales , Humanos , Masculino , Ratones , ARN Polimerasa II/metabolismo , Factores de Empalme de ARN
14.
Cell Rep ; 31(9): 107703, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32492419

RESUMEN

Tight coordination of gene expression in the developing cerebellum is crucial for establishment of neuronal circuits governing motor and cognitive function. However, transcriptional changes alone do not explain all of the switches underlying neuronal differentiation. Here we unveiled a widespread and highly dynamic splicing program that affects synaptic genes in cerebellar neurons. The motifs enriched in modulated exons implicated the splicing factor Sam68 as a regulator of this program. Sam68 controls splicing of exons with weak branchpoints by directly binding near the 3' splice site and competing with U2AF recruitment. Ablation of Sam68 disrupts splicing regulation of synaptic genes associated with neurodevelopmental diseases and impairs synaptic connections and firing of Purkinje cells, resulting in motor coordination defects, ataxia, and abnormal social behavior. These findings uncover an unexpectedly dynamic splicing regulatory network that shapes the synapse in early life and establishes motor and cognitive circuitry in the developing cerebellum.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cerebelo/metabolismo , Empalme del ARN , Proteínas de Unión al ARN/metabolismo , Sinapsis/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Conducta Animal , Cerebelo/citología , Cerebelo/crecimiento & desarrollo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Células de Purkinje/metabolismo , Sitios de Empalme de ARN , Proteínas de Unión al ARN/genética , Factor de Empalme U2AF/metabolismo
15.
Cell Rep ; 26(11): 2929-2941.e5, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30865884

RESUMEN

Male germ cells express the widest repertoire of transcript variants in mammalian tissues. Nevertheless, factors and mechanisms underlying such pronounced diversity are largely unknown. The splicing regulator Sam68 is highly expressed in meiotic cells, and its ablation results in defective spermatogenesis. Herein, we uncover an extensive splicing program operated by Sam68 across meiosis, primarily characterized by alternative last exon (ALE) regulation in genes of functional relevance for spermatogenesis. Lack of Sam68 preferentially causes premature transcript termination at internal polyadenylation sites, a feature observed also upon depletion of the spliceosomal U1snRNP in somatic cells. Notably, Sam68-regulated ALEs are characterized by proximity between U1snRNP and Sam68 binding motifs. We demonstrate a physical association between Sam68 and U1snRNP and show that U1snRNP recruitment to Sam68-regulated ALEs is impaired in Sam68-/- germ cells. Thus, our study reveals an unexpected cooperation between Sam68 and U1snRNP that insures proper processing of transcripts essential for male fertility.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Empalme del ARN , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Espermatogénesis , Espermatogonias/metabolismo , Terminación de la Transcripción Genética , Regiones no Traducidas 3' , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Proteínas de Unión al ARN/genética , Ribonucleoproteínas Nucleares Pequeñas/genética , Espermatogonias/citología
16.
Chem Biol Interact ; 169(1): 1-14, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17537412

RESUMEN

Lindane (gamma-HCH) is a persistent environmental pollutant that may act as endocrine disrupter, affecting the nervous, immune and reproductive system, possibly through endocrine-mediated mechanisms. Since both estrogen receptors (ER-alpha and -beta) have shown to be target for endocrine disruption, we investigated the role of gamma-HCH on the development of female reproductive system. For an in vivo evaluation of gamma-HCH effects during prenatal period, pregnant CD1 mice were treated p.o. on gestational days 9-16 with 15 mg/kg bw/day of gamma-HCH and vehicle. The in vivo findings in treated F1 pups - in the absence of signs of systemic toxicity - included increase in the absolute and relative and absolute uterus weight revealed on post-natal day 22, earlier vaginal patency and reduced diameters of primary oocytes at fully sexual maturity. No effects on steroid hormone metabolism (aromatase, testosterone catabolism) were observed. Thus, gamma-HCH elicited subtle effects on female reproductive development likely mediated by ER-beta-mediated pathway(s), without a concurrent impairment of steroid hormone metabolism. Furthermore, to verify whether the endocrine interference of gamma-HCH is attributable to stimulation of ER-beta-mediated pathway(s), its effect has been evaluated in vitro on a cell line, LNCaP, expressing only functional ER-beta. In vitro treatments revealed a concentration-related effect on LNCaP cell viability and proliferation. Significantly, the contemporary addition of a pure anti-estrogen, the ER antagonist ICI 182,780, completely reversed gamma-HCH effects indicating an ER-beta-mediated action. Our findings indicate that gamma-HCH may act as endocrine disruptor during the female reproductive system development and ER-beta as a potential target for this compound and other endocrine disrupting chemicals as well.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Hexaclorociclohexano/farmacología , Desarrollo Sexual/efectos de los fármacos , Animales , Aromatasa/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Receptor beta de Estrógeno/genética , Femenino , Regulación de la Expresión Génica , Hormonas Esteroides Gonadales/metabolismo , Humanos , Hígado/metabolismo , Ratones , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Maduración Sexual/efectos de los fármacos , Testosterona/metabolismo
17.
Cell Death Dis ; 8(11): e3168, 2017 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-29120411

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is characterized by extremely poor prognosis. The standard chemotherapeutic drug, gemcitabine, does not offer significant improvements for PDAC management due to the rapid acquisition of drug resistance by patients. Recent evidence indicates that epithelial-to-mesenchymal transition (EMT) of PDAC cells is strictly associated to early metastasization and resistance to chemotherapy. However, it is not exactly clear how EMT is related to drug resistance or how chemotherapy influences EMT. Herein, we found that ZEB1 is the only EMT-related transcription factor that clearly segregates mesenchymal and epithelial PDAC cell lines. Gemcitabine treatment caused upregulation of ZEB1 protein through post-transcriptional mechanisms in mesenchymal PDAC cells within a context of global inhibition of protein synthesis. The increase in ZEB1 protein correlates with alternative polyadenylation of the transcript, leading to shortening of the 3' untranslated region (UTR) and deletion of binding sites for repressive microRNAs. Polysome profiling indicated that shorter ZEB1 transcripts are specifically retained on the polysomes of PDAC cells during genotoxic stress, while most mRNAs, including longer ZEB1 transcripts, are depleted. Thus, our findings uncover a novel layer of ZEB1 regulation through 3'-end shortening of its transcript and selective association with polysomes under genotoxic stress, strongly suggesting that PDAC cells rely on upregulation of ZEB1 protein expression to withstand hostile environments.


Asunto(s)
Carcinoma Ductal Pancreático/fisiopatología , Neoplasias Pancreáticas/fisiopatología , Poliadenilación , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Regiones no Traducidas 3' , Secuencia de Bases , Sitios de Unión , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Humanos , MicroARNs/química , MicroARNs/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Polirribosomas/metabolismo , Ribonucleoproteínas/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Gemcitabina
18.
Front Biosci ; 10: 542-51, 2005 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-15574391

RESUMEN

In the present article we will focus on the adhesion molecules expressed by mouse primordial germ cells (PGCs) and will discuss the role that they play, or are believed to play, in two crucial processes of PGC development, namely cell lineage specification and migration into the gonadal ridges. Recent findings indicate that the adhesion-dependent allocation of the PGC precursors to a niche within the epiblast and the forming extraembryonic mesoderm during the pre-gastrulation period is crucial for their commitment. Subsequently, PGC migration and homing within the gonadal ridges require integrated signals involving contact of PGCs with extracellular matrix molecules and cellular substrates or repulsion from them, adhesion among PGCs themselves and attraction by the developing gonads. A number of adhesion, or putative adhesion molecules, have been identified in mammalian PGCs, mainly in the mouse. These molecules belong to three adhesion molecule families such as cadherins (E-P- and N-cadherins), integrins and the IgG superfamily (PECAM-1). Moreover oligosaccarides (LewisX) and growth factor receptors (c-Kit) can also play adhesive roles in some stages of PGC development. An understanding of how genes encoding adhesive molecules are regulated in PGCs and the molecular pathways associated with the functions of adhesion receptors is crucial in furthering our knowledge of PGC biology. Adhesion molecules might once again turn out to be crucial in controlling not only the germ cell lineage and PGC migration but also the PGC differentiation fate itself.


Asunto(s)
Células Germinativas/metabolismo , Animales , Cadherinas/metabolismo , Adhesión Celular , Moléculas de Adhesión Celular , Linaje de la Célula , Movimiento Celular , Matriz Extracelular/metabolismo , Humanos , Integrinas/metabolismo , Ratones , Oligosacáridos/química
19.
Mech Dev ; 136: 53-63, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25684074

RESUMEN

In the present paper, starting from the observation of heterogeneous expression of the GOF-18ΔPE-GFP Pou5f1 (Oct3/4) transgene in putative mouse PGC populations settled in the aorta-gonad-mesonephros (AGM) region, we identified various OCT3/4 positive populations showing distinct expression of PGC markers (BLIMP-1, AP, TG-1, STELLA) and co-expressing several proteins (CD-34, CD-41, FLK-1) and genes (Brachyury, Hox-B4, Scl/Tal-1 and Gata-2) of hematopoietic precursors. Moreover, we found that Oct3/4-GFP(weak) CD-34(weak/high) cells possess robust hematopoietic colony forming activity (CFU) in vitro. These data indicate that the cell population usually considered PGCs moving toward the gonadal ridges encompasses a subset of cells co-expressing several germ cell and hematopoietic markers and possessing hematopoietic activity. These results are discussed within of the current model of germline segregation.


Asunto(s)
Células Germinativas/metabolismo , Gónadas/metabolismo , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Mesonefro/metabolismo , Animales , Células Germinativas/citología , Gónadas/citología , Células Madre Hematopoyéticas/citología , Mesonefro/citología , Ratones , Factor 3 de Transcripción de Unión a Octámeros/metabolismo
20.
Endocrinology ; 144(4): 1631-43, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12639948

RESUMEN

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide that elicits the increase of intracellular cAMP levels and protein kinase A activity in various cell systems. Here we show that the pattern of cAMP elevation triggered by PACAP is critical for the fate of LNCaP prostate cancer cells. We demonstrate that these cells express PACAP and its type 1 receptor. A short-term stimulation with PACAP, which generates a transient cAMP rise, induces proliferation of LNCaP cells through a protein kinase A-dependent activation of the MAPK cascade. On the contrary, we observed that chronic PACAP stimulation, giving rise to a sustained cAMP accumulation, leads to proliferation arrest and neuroendocrine differentiation. Moreover, PACAP stimulates phosphory-lation and activation of the cAMP response element binding transcription factor (CREB), and MAPK activation is necessary for its full transcriptional activity, indicating a direct involvement of cAMP response element in PACAP action. These findings demonstrate that a crucial event determining the outcome of prostatic cancer cells progression is the sustained vs. transient intracellular cAMP increase.


Asunto(s)
Mitógenos/farmacología , Neuropéptidos/farmacología , Sistemas Neurosecretores/citología , Neoplasias de la Próstata , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , ADN/biosíntesis , Expresión Génica/fisiología , Humanos , Masculino , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuropéptidos/genética , Fosforilación , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria , Receptores de la Hormona Hipofisaria/genética , Transcripción Genética/fisiología , Células Tumorales Cultivadas/citología , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA