Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Hum Mol Genet ; 30(17): 1649-1665, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34100078

RESUMEN

Megalencephalic Leukoencephalopathy with subcortical Cysts (MLC) is a type of vacuolating leukodystrophy, which is mainly caused by mutations in MLC1 or GLIALCAM. The two MLC-causing genes encode for membrane proteins of yet unknown function that have been linked to the regulation of different chloride channels such as the ClC-2 and VRAC. To gain insight into the role of MLC proteins, we have determined the brain GlialCAM interacting proteome. The proteome includes different transporters and ion channels known to be involved in the regulation of brain homeostasis, proteins related to adhesion or signaling as several G protein-coupled receptors (GPCRs), including the orphan GPRC5B and the proposed prosaposin receptor GPR37L1. Focusing on these two GPCRs, we could validate that they interact directly with MLC proteins. The inactivation of Gpr37l1 in mice upregulated MLC proteins without altering their localization. Conversely, a reduction of GPRC5B levels in primary astrocytes downregulated MLC proteins, leading to an impaired activation of ClC-2 and VRAC. The interaction between the GPCRs and MLC1 was dynamically regulated upon changes in the osmolarity or potassium concentration. We propose that GlialCAM and MLC1 associate with different integral membrane proteins modulating their functions and acting as a recruitment site for various signaling components as the GPCRs identified here. We hypothesized that the GlialCAM/MLC1 complex is working as an adhesion molecule coupled to a tetraspanin-like molecule performing regulatory effects through direct binding or influencing signal transduction events.


Asunto(s)
Quistes/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Receptores Acoplados a Proteínas G/genética , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Moléculas de Adhesión Celular Neurona-Glia/genética , Moléculas de Adhesión Celular Neurona-Glia/metabolismo , Proteínas de Ciclo Celular/genética , Canales de Cloruro/genética , Quistes/metabolismo , Células HEK293 , Células HeLa , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/metabolismo , Humanos , Leucoencefalopatías/genética , Leucoencefalopatías/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Malformaciones del Sistema Nervioso/metabolismo , Transporte de Proteínas , Receptores Acoplados a Proteínas G/metabolismo
2.
Int J Mol Sci ; 23(8)2022 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-35457105

RESUMEN

The vertebrate G protein-coupled receptor 37 and G protein-coupled receptor 37-like 1 (GPR37 and GPR37L1) proteins have amino acid sequence homology to endothelin and bombesin-specific receptors. The prosaposin glycoprotein, its derived peptides, and analogues have been reported to interact with and activate both putative receptors. The GPR37 and GPR37L1 genes are highly expressed in human and rodent brains. GPR37 transcripts are most abundant in oligodendrocytes and in the neurons of the substantia nigra and hippocampus, while the GPR37L1 gene is markedly expressed in cerebellar Bergmann glia astrocytes. The human GPR37 protein is a substrate of parkin, and its insoluble form accumulates in brain samples from patients of inherited juvenile Parkinson's disease. Several Gpr37 and Gpr37l1 mouse mutant strains have been produced and applied to extensive in vivo and ex vivo analyses of respective receptor functions and involvement in brain and other organ pathologies. The genotypic and phenotypic characteristics of the different mouse strains so far published are reported and discussed, and their current and proposed applications to human disease modeling are highlighted.


Asunto(s)
Trastornos Parkinsonianos , Receptores Acoplados a Proteínas G , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Humanos , Ratones , Neuronas/metabolismo , Trastornos Parkinsonianos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
3.
J Cell Sci ; 132(5)2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30745336

RESUMEN

Ataxia telangiectasia is a rare, multi system disease caused by ATM kinase deficiency. Atm-knockout mice recapitulate premature aging, immunodeficiency, cancer predisposition, growth retardation and motor defects, but not cerebellar neurodegeneration and ataxia. We explored whether Atm loss is responsible for skeletal muscle defects by investigating myofiber morphology, oxidative/glycolytic activity, myocyte ultrastructural architecture and neuromuscular junctions. Atm-knockout mice showed reduced muscle and fiber size. Atrophy, protein synthesis impairment and a switch from glycolytic to oxidative fibers were detected, along with an increase of in expression of slow and fast myosin types (Myh7, and Myh2 and Myh4, respectively) in tibialis anterior and solei muscles isolated from Atm-knockout mice. Transmission electron microscopy of tibialis anterior revealed misalignments of Z-lines and sarcomeres and mitochondria abnormalities that were associated with an increase in reactive oxygen species. Moreover, neuromuscular junctions appeared larger and more complex than those in Atm wild-type mice, but with preserved presynaptic terminals. In conclusion, we report for the first time that Atm-knockout mice have clear morphological skeletal muscle defects that will be relevant for the investigation of the oxidative stress response, motor alteration and the interplay with peripheral nervous system in ataxia telangiectasia.


Asunto(s)
Envejecimiento Prematuro/genética , Proteínas de la Ataxia Telangiectasia Mutada/genética , Ataxia Telangiectasia/metabolismo , Síndromes de Inmunodeficiencia/genética , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Neoplasias/genética , Animales , Ataxia Telangiectasia/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Mitocondrias/ultraestructura , Músculo Esquelético/anomalías , Músculo Esquelético/ultraestructura , Especies Reactivas de Oxígeno/metabolismo , Sarcómeros/ultraestructura
4.
J Neurosci Res ; 2020 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-33350496

RESUMEN

Mammalian cerebellar astrocytes critically regulate the differentiation and maturation of neuronal Purkinje cells and granule precursors. The G protein-coupled receptor 37-like 1 (Gpr37l1) is expressed by Bergmann astrocytes and interacts with patched 1 (Ptch1) at peri-ciliary membranes. Cerebellar primary astrocyte cultures from wild-type and Gpr37l1 null mutant mouse pups were established and studied. Primary cilia were produced by cultures of both genotypes, as well as Ptch1 and smoothened (Smo) components of the sonic hedgehog (Shh) mitogenic pathway. Compared to wild-type cells, Gpr37l1-/- astrocytes displayed striking increases in proliferative activity, Ptch1 protein expression and internalization, intracellular cholesterol content, ciliary localization of Smo, as well as a marked production of active Shh. Similar effects were reproduced by treating wild-type astrocytes with a putative prosaptide ligand of Gpr37l1. These findings indicate that Gpr37l1-Ptch1 interactions specifically regulate Ptch1 internalization and trafficking, with consequent stimulation of Shh production and activation of proliferative signaling.

5.
Cell Mol Neurobiol ; 37(1): 145-154, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26935062

RESUMEN

Cellular primary cilia crucially sense and transduce extracellular physicochemical stimuli. Cilium-mediated developmental signaling is tissue and cell type specific. Primary cilia are required for cerebellar differentiation and sonic hedgehog (Shh)-dependent proliferation of neuronal granule precursors. The mammalian G-protein-coupled receptor 37-like 1 is specifically expressed in cerebellar Bergmann glia astrocytes and participates in regulating postnatal cerebellar granule neuron proliferation/differentiation and Bergmann glia and Purkinje neuron maturation. The mouse receptor protein interacts with the patched 1 component of the cilium-associated Shh receptor complex. Mice heterozygous for patched homolog 1 mutations, like heterozygous patched 1 humans, have a higher incidence of Shh subgroup medulloblastoma (MB) and other tumors. Cerebellar cells bearing primary cilia were identified during postnatal development and in adulthood in two mouse strains with altered Shh signaling: a G-protein-coupled receptor 37-like 1 null mutant and an MB-susceptible, heterozygous patched homolog 1 mutant. In addition to granule and Purkinje neurons, primary cilia were also expressed by Bergmann glia astrocytes in both wild-type and mutant animals, from birth to adulthood. Variations in ciliary number and length were related to the different levels of neuronal and glial cell proliferation and maturation, during postnatal cerebellar development. Primary cilia were also detected in pre-neoplastic MB lesions in heterozygous patched homolog 1 mutant mice and they could represent specific markers for the development and analysis of novel cerebellar oncogenic models.


Asunto(s)
Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Cilios/genética , Cilios/patología , Meduloblastoma/genética , Meduloblastoma/patología , Animales , Animales Recién Nacidos , Cerebelo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética
6.
FASEB J ; 29(5): 2059-69, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25609427

RESUMEN

The mammalian G-protein-coupled receptor 37 (GPR37) is expressed in brain, in adult testis, and during the early phase of gonad differentiation. Somatic Sertoli cells (SCs) are located within the seminiferous tubules where they support the germinal epithelium. An adequate number of SCs is required for the complete prepubertal differentiation of germ cells and adult fertility. This study shows that Gpr37 and its ligand prosaposin are both postnatally expressed by SCs, whose proliferation and maturation are affected in Gpr37-null mutant mice during postnatal testicular development. Mutant pups show a delayed timing in sperm cell development, with a partial arrest of spermatocytes at the meiotic pachytene (e.g., 1.5-fold increase in Gpr37(-/-) P21 pups) and their increased apoptosis (e.g., 1.8-fold and 3.5-fold increase in Gpr37(-/-) P21 and adult mice, respectively). Mutant adults have reduced testis weight (wild type, 299 ± 5 mg; knockout, 258 ± 16 mg; P < 0.05) and epididymal sperm count and motility (e.g., 1.5-fold and 1.45-fold decrease in Gpr37(-/-) mice, respectively). Lack of Gpr37 results in the reduction in androgen receptor levels during prepubertal testis development, alongside the altered expression of SC maturation markers. It also affects the prepubertal testis expression of desert hedgehog (Dhh) mitogenic cascade components (Dhh, 1.3-fold increase in Gpr37(-/-) P10 and P21 pups; Gli2, 1.4-fold and 1.6-fold increase in Gpr37(-/-) P10 and P21 pups, respectively) including patched homolog 1 (1.3-fold increase in Gpr37(-/-) P10 and P21 pups), which is found localized in prepubertal SCs and is associated with Gpr37 in cultured primary SC samples. These results indicate that Gpr37 is a specific modulator of murine testis Dhh mitogenic signaling and SC proliferation and maturation.


Asunto(s)
Regulación de la Expresión Génica , Proteínas Hedgehog/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Saposinas/metabolismo , Células de Sertoli/metabolismo , Espermatogénesis/fisiología , Testículo/metabolismo , Animales , Apoptosis , Western Blotting , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Proteínas Hedgehog/genética , Técnicas para Inmunoenzimas , Inmunoprecipitación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Patched , Receptor Patched-1 , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células de Sertoli/citología , Transducción de Señal , Testículo/citología
7.
Proc Natl Acad Sci U S A ; 110(41): 16486-91, 2013 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-24062445

RESUMEN

In the developing cerebellum, the proliferation and differentiation of glial and neuronal cell types depend on the modulation of the sonic hedgehog (Shh) signaling pathway. The vertebrate G-protein-coupled receptor 37-like 1 (GPR37L1) gene encodes a putative G-protein-coupled receptor that is expressed in newborn and adult cerebellar Bergmann glia astrocytes. This study shows that the ablation of the murine Gpr37l1 gene results in premature down-regulation of proliferation of granule neuron precursors and precocious maturation of Bergmann glia and Purkinje neurons. These alterations are accompanied by improved adult motor learning and coordination. Gpr37l1(-/-) mice also exhibit specific modifications of the Shh signaling cascade. Specific assays show that in Bergmann glia cells Gpr37l1 is associated with primary cilium membranes and it specifically interacts and colocalizes with the Shh primary receptor, patched 1. These findings indicate that the patched 1-associated Gpr37l1 receptor participates in the regulation of postnatal cerebellum development by modulating the Shh pathway.


Asunto(s)
Cerebelo/crecimiento & desarrollo , Neuroglía/fisiología , Desempeño Psicomotor/fisiología , Células de Purkinje/fisiología , Receptores Acoplados a Proteínas G/genética , Animales , Western Blotting , Proliferación Celular , Cerebelo/citología , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Eliminación de Gen , Vectores Genéticos/genética , Proteínas Hedgehog/metabolismo , Inmunoprecipitación , Hibridación in Situ , Ratones , Ratones Noqueados , Mitógenos/metabolismo , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/metabolismo
8.
Cells ; 13(12)2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38920681

RESUMEN

Odad3 gene loss-of-function mutation leads to Primary Ciliary Dyskinesia (PCD), a disease caused by motile cilia dysfunction. Previously, we demonstrated that knockout of the Odad3 gene in mice replicates several features of PCD, such as hydrocephalus, defects in left-right body symmetry, and male infertility, with a complete absence of sperm in the reproductive tract. The majority of Odad3 knockout animals die before sexual maturation due to severe hydrocephalus and failure to thrive, which precludes fertility studies. Here, we performed the expression analysis of the Odad3 gene during gonad development and in adult testes. We showed that Odad3 starts its expression during the first wave of spermatogenesis, specifically at the meiotic stage, and that its expression is restricted to the germ cells in the adult testes, suggesting that Odad3 plays a role in spermatozoa formation. Subsequently, we conditionally deleted the Odad3 gene in adult males and demonstrated that even partial ablation of the Odad3 gene leads to asthenoteratozoospermia with multiple morphological abnormalities of sperm flagella (MMAF) in mice. The analysis of the seminiferous tubules in Odad3-deficient mice revealed defects in spermatogenesis with accumulation of seminiferous tubules at the spermiogenesis and spermiation phases. Furthermore, analysis of fertility in heterozygous Odad3+/- knockout mice revealed a reduction in sperm count and motility as well as abnormal sperm morphology. Additionally, Odad3+/- males exhibited a shorter fertile lifespan. Overall, these results suggest the important role of Odad3 and Odad3 gene dosage in male fertility. These findings may have an impact on the genetic and fertility counseling practice of PCD patients carrying Odad3 loss-of-function mutations.


Asunto(s)
Fertilidad , Ratones Noqueados , Espermatogénesis , Espermatozoides , Animales , Masculino , Espermatogénesis/genética , Fertilidad/genética , Ratones , Espermatozoides/metabolismo , Testículo/metabolismo , Testículo/patología , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Ratones Endogámicos C57BL
9.
Growth Factors ; 29(1): 21-35, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21222515

RESUMEN

Despite numerous studies on the role of growth hormone (GH), its function in skeletal muscle apoptosis secondary to various stimuli is poorly understood. In this study, we used rodent muscle cell lines to analyse cell growth and survival as well as the morphological and molecular markers of cell death in C2C12 and L6C5 myoblasts. These cells were treated either in the presence or absence of GH under serum starvation conditions or in the pro-apoptotic concentrations of hydrogen peroxide (H2O2). Although the cells were responsive to the presence of GH, we did not observe GH modulation of cell growth and survival. The presence of GH did not affect the cell death programme or the expression of apoptotic markers in basal conditions or under oxidative stress. In conclusion, this study indicated that GH "by itself" is not effective in modulating the intracellular pathways leading to cell survival or cell death induced by apoptotic stimuli.


Asunto(s)
Apoptosis/efectos de los fármacos , Hormona de Crecimiento Humana/farmacología , Músculo Esquelético/efectos de los fármacos , Mioblastos/efectos de los fármacos , Mioblastos/fisiología , Animales , División Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Mioblastos/citología , Estrés Oxidativo
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 Neurosci ; 15: 703431, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867197

RESUMEN

Glioblastomas (GBM) are the most aggressive tumors originating in the brain. Histopathologic features include circuitous, disorganized, and highly permeable blood vessels with intermittent blood flow. These features contribute to the inability to direct therapeutic agents to tumor cells. Known targets for anti-angiogenic therapies provide minimal or no effect in overall survival of 12-15 months following diagnosis. Identification of novel targets therefore remains an important goal for effective treatment of highly vascularized tumors such as GBM. We previously demonstrated in zebrafish that a balanced level of expression of the transmembrane protein TMEM230/C20ORF30 was required to maintain normal blood vessel structural integrity and promote proper vessel network formation. To investigate whether TMEM230 has a role in the pathogenesis of GBM, we analyzed its prognostic value in patient tumor gene expression datasets and performed cell functional analysis. TMEM230 was found necessary for growth of U87-MG cells, a model of human GBM. Downregulation of TMEM230 resulted in loss of U87 migration, substratum adhesion, and re-passaging capacity. Conditioned media from U87 expressing endogenous TMEM230 induced sprouting and tubule-like structure formation of HUVECs. Moreover, TMEM230 promoted vascular mimicry-like behavior of U87 cells. Gene expression analysis of 702 patients identified that TMEM230 expression levels distinguished high from low grade gliomas. Transcriptomic analysis of patients with gliomas revealed molecular pathways consistent with properties observed in U87 cell assays. Within low grade gliomas, elevated TMEM230 expression levels correlated with reduced overall survival independent from tumor subtype. Highest level of TMEM230 correlated with glioblastoma and ATP-dependent microtubule kinesin motor activity, providing a direction for future therapeutic intervention. Our studies support that TMEM230 has both glial tumor and endothelial cell intracellular and extracellular functions. Elevated levels of TMEM230 promote glial tumor cell migration, extracellular scaffold remodeling, and hypervascularization and abnormal formation of blood vessels. Downregulation of TMEM230 expression may inhibit both low grade glioma and glioblastoma tumor progression and promote normalization of abnormally formed blood vessels. TMEM230 therefore is both a promising anticancer and antiangiogenic therapeutic target for inhibiting GBM tumor cells and tumor-driven angiogenesis.

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 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
14.
Exp Neurol ; 312: 33-42, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30452905

RESUMEN

The G-protein coupled receptor 37-like 1 (Gpr37l1) is specifically expressed in most astrocytic glial cells, including cerebellar Bergmann astrocytes and interacts with patched 1 (Ptch1), a co-receptor of the sonic hedgehog (Shh)-smoothened (Smo) signaling complex. Gpr37l1 null mutant mice exhibit precocious post-natal cerebellar development, with altered Shh-Smo mitogenic cascade and premature down-regulation of granule cell precursor (GCP) proliferation. Gpr37l1 expression is downregulated in medulloblastoma (MB) and upregulated in glioma and glioblastoma tumors. Shh-associated MBs originate postnatally, from dysregulated hyperproliferation of GCPs in developing cerebellum's external granular layer (EGL), as shown in heterozygous Ptch1+/- knock-out mouse strains that model human MB occurrence and progression. This study investigates cerebellar MB phenotypes in newly produced Gpr37l1, Ptch1 double mutant mice. Natural history analysis shows that Gpr37l1 genetic ablation, in Ptch1+/- model animals, results in marked deferment of post-natal tumor occurrence and decreased incidence of more aggressive tumor types. It is also associated with the delayed and diminished presence of more severe types of hyperplastic lesions in Ptch1+/- mice. Consistently, during early post-natal development Gpr37l1-/-;Ptch1+/- pups exhibit reduction in cerebellar GCP proliferation and EGL thickness and a precocious, sustained expression of wingless-type MMTV integration site member 3 (Wnt3), a specific inhibitor of Shh-induced neuronal mitogenesis, in comparison with Ptch1+/- heterozygous single mutants. These findings highlight the specific involvement of Gpr37l1 in modulating postnatal cerebellar Shh-Ptch1-Smo mitogenic signaling in both normal and pathological conditions. The novel Gpr37l1-/-;Ptch1+/- mouse models may thus be instrumental in the detailed characterization of the initial phases of Shh-associated MB insurgence and development.


Asunto(s)
Carcinogénesis/metabolismo , Neoplasias Cerebelosas/metabolismo , Meduloblastoma/metabolismo , Receptor Patched-1/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Animales , Carcinogénesis/genética , Proliferación Celular/fisiología , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Femenino , Masculino , Meduloblastoma/genética , Meduloblastoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor Patched-1/genética , Receptores Acoplados a Proteínas G/genética
15.
Front Cell Neurosci ; 13: 226, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31178699

RESUMEN

The Niemann-Pick type C1 (NPC1) is a rare genetic disease characterized by the accumulation of endocytosed cholesterol and other lipids in the endosome/lysosome compartments. In the brain, the accumulation/mislocalization of unesterified cholesterol, gangliosides and sphingolipids is responsible for the appearance of neuropathological hallmarks, and progressive neurological decline in patients. The imbalance of unesterified cholesterol and other lipids, including GM2 and GM3 gangliosides, alters a number of signaling mechanisms impacting on the overall homeostasis of neurons. In particular, lipid depletion experiments have shown that lipid rafts regulate the cell surface expression of dopamine transporter (DAT) and modulate its activity. Dysregulated dopamine transporter's function results in imbalanced dopamine levels at synapses and severely affects dopamine-induced locomotor responses and dopamine receptor-mediated synaptic signaling. Recent studies begin to correlate dopaminergic stimulation with the length and function of the primary cilium, a non-motile organelle that coordinates numerous signaling pathways. In particular, the absence of dopaminergic D2 receptor stimulation induces the elongation of dorso-striatal neuron's primary cilia. This study has used a mouse model of the NPC1 disease to correlate cholesterol dyshomeostasis with dorso-striatal anomalies in terms of DAT expression and primary cilium (PC) length and morphology. We found that juvenile Npc1nmf164 mice display a reduction of dorso-striatal DAT expression, with associated alterations of PC number, length-frequency distribution, and tortuosity.

16.
Int J Dev Biol ; 54(4): 717-22, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20209442

RESUMEN

We and others have reported that mouse embryonic testes contain a subpopulation of somatic cells expressing estrogen receptor alpha (ERalpha). In order to provide evidence for a possible direct estrogen effect on mammalian testes from the early stage of their differentiation, here we devised a method for the in vitro culture of the ERalpha-expressing cells from 12.5 days post coitum mouse testes and their transfection with plasmids containing the classical estrogen responsive element (ERE) or the alternative estrogen AP-1 responsive element upstream of the luciferase reporter gene (ERE-Luc and AP-1-Luc). StAR immunopositivity of the most part of the ERalpha+ cells grown in culture and subjected to the estrogenic assay, allowed their identification as embryonic Leydig cells. Maximum induction of the ERE-Luc activity was achieved with 10 nM 17-beta estradiol (E2), from 1.7 to 3-fold in such cells and from 2.3 to 5.7-fold in MCF-7 cells used for comparison; the anti-estrogen ICI 182.780 abolished such effects. AP-1-Luc was less sensitive to E2 in both cell types (10 nM E2, 1.2 to 2.7-fold increase in embryonic Leydig cells; about 3-fold in MCF-7 cells) and the effect was not ICI-dependent. Eventually, we stimulated the transfected cells with various xenoestrogens such as lindane, bisphenol A or mono-(2-ethylhexyl) pthalate and with the phytoestrogen zeralenone obtaining evidence for ERE-Luc, but not AP-1-Luc stimulation in embryonic Leydig cells. These results represent evidence of functional ERalpha-dependent genomic pathways in embryonic Leydig cells and describe an in vitro assay suitable for evaluating the activity of putative estrogenic compounds on such cells.


Asunto(s)
Bioensayo , Antagonistas de Estrógenos/farmacología , Estrógenos/farmacología , Células Intersticiales del Testículo/metabolismo , Testículo/embriología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Estradiol/análogos & derivados , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor alfa de Estrógeno/fisiología , Estrógenos/genética , Estrona/genética , Femenino , Fulvestrant , Genes Reporteros/efectos de los fármacos , Humanos , Luciferasas/genética , Masculino , Ratones , Fitoestrógenos/farmacología , Elementos de Respuesta/genética , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Transfección
17.
Toxicol Sci ; 108(2): 445-51, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19221147

RESUMEN

Lindane (gamma-HCH) was examined for its effect on primordial germ cell (PGC) development in the mouse embryo. We found that exposure by gavage of pregnant mice to 15 or 30 mg/kg/bw lindane during the period of PGC migration and gonad colonization (from 8.5 to 11.5 days post coitum, dpc) resulted in a significant reduction of the number of germ cells within 12.5 dpc testis and ovaries (a maximum of about 25 and 40%, respectively). Similarly, lindane caused a dose-dependent decrease of the PGC number in an in vitro culture model. Further experiments showed that in such model, lindane induced features of apoptotic cell death in PGCs such as increase in caspase-3 activity, poly-ADP-ribose polymerase cleavage, and terminal dUTP nick-end labeling (TUNEL) positivity. A marked increase of the number of PGCs positive for TUNEL staining was also observed in 12.5 dpc gonads of embryos from pregnant mice subjected one day before to acute lindane treatment (60 mg/kg/bw). Finally, we show that a brief incubation of isolated PGCs with 10(-5)M lindane resulted in a marked decrease of the basal and kit-ligand-induced phosphorylation level of the AKT kinase, known to be crucial for PGC survival. Taken together these results demonstrate that embryo exposure to lindane during early stages of gametogenesis can severely impair the number of germ cells in the fetal gonads; the compound appears to affect PGC survival through a direct proapoptotic action likely resulting from its adverse effect on AKT activity in such cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Germinativas/efectos de los fármacos , Hexaclorociclohexano/toxicidad , Insecticidas/toxicidad , Animales , Antimetabolitos , Western Blotting , Bromodesoxiuridina , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Gametogénesis/efectos de los fármacos , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Proteína Oncogénica v-akt/biosíntesis , Proteína Oncogénica v-akt/genética , Embarazo
18.
Toxicol Sci ; 103(1): 57-67, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18281260

RESUMEN

The widespread use of diets containing estrogenic compounds raises questions on how relevant the presence of phytoestrogens may be, in order to allow a correct development of the reproductive ability and sexual maturity in humans and animals. The isoflavone genistein is the most estrogenically active molecule present in soy. Here we show that genistein, through an estrogen receptor (ER)-mediated action, modulates gene expression in the whole body of male mice in a dose- and time-dependent manner, at all ages. By luciferase bioassays, we show that genistein-induced ER activation is present in reproductive and nonreproductive organs of the transgenic mice Estrogen Responsive Element (ERE)-tK-LUC, although to an extent that is lower than what observed with the administration of estradiol. Peak activity was registered at genistein doses of 500-5000 microg/kg, at 12 h from the administration by gavage. In the liver, ER-alpha and ER-beta messenger RNAs and two target genes, CYP17 and the progesterone receptor, were modulated by genistein. CYP17 and PR time-dependent induction was similar to that of luciferase. ER-alpha protein level followed an opposite regulation by genistein and estradiol. Genistein passed from the lactating mother to the suckling offspring at levels sufficient to activate gene expression in reproductive and nonreproductive tissues of the pups, with maximal upregulation at 16-24 h. We also followed responsiveness to genistein in the testis, from early development to adult age. Testis are well responsive to genistein as well as to estradiol already at day 14.5 of fetal development, as determined by exposing organotypic cultures from mouse fetus testis. Ovaries were not responsive under the same conditions. Activation of luciferase correlates with an activation of cell proliferation in testis, but not in the ovaries. Prolonged exposure (15 days) to genistein also decreases prostate weight like estradiol. In conclusion, our results show that genistein affects reproductive and nonreproductive organs of male mice in a dose- and time-dependent manner, at all developmental ages.


Asunto(s)
Genisteína/farmacología , Animales , Femenino , Gónadas/efectos de los fármacos , Gónadas/embriología , Gónadas/fisiología , Lactancia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Mensajero/genética , Receptores de Estrógenos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Dev Biol ; 306(2): 572-83, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17467686

RESUMEN

Using a Transwell chamber as migration assay for mouse primordial germ cells (PGCs), we show here that these cells posses directional migration in the absence of somatic cell and defined matrix support and in response to a Kit ligand (KL) gradient or medium conditioned by Aorta/Gonad/Mesonephros and gonadal ridges. Other putative PGC chemoattractants such as SDF1 and TGFbeta did not exert any attractive action on PGCs. The chemoattractant activity of KL and conditioned medium was also evidenced by their ability to stimulate actin reorganization in PGCs. In the aim to identify downstream signaling pathways governing KL chemoattraction on PGCs, we demonstrated that in such cells KL rapidly (5 min) increased autophosphorylation of its receptor c-Kit and caused phosphorylation of the serine-threonine kinase AKT through the action of PI3K. 740Y-P peptide, a direct activator of PI3 kinase, stimulated PGC migration at levels similar to those elicited by KL. LY294002 (a specific inhibitor of PI3K) abolished KL-dependent PGC migration or the chemoattractant activity of the conditioned medium and inhibited AKT phosphorylation; Src kinase inhibitors PP2 and SU6656, caused significant reduction of the KL-dependent PGC migration and AKT phosphorylation, while U0126, a selective inhibitor of the MEK/ERK protein kinase cascade, reduced PGC migration and AKT phosphorylation at lesser extent. SU6656 completely abolished the chemoattractant activity of the conditioned medium. Finally, SB202190 (a p38 inhibitor) and rapamycin (mTOR inhibitor) did not affect PGC migration. In addition, to demonstrate that somatic cells are not essential for PGC motility and directional migration, we evidenced a novel role for KL as PGC chemoattractant and for PI3K/AKT and Src kinase, as players involved in the activation of the PGC migratory machinery and likely important for their directional movement towards the gonadal ridges.


Asunto(s)
Factores Quimiotácticos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factor de Células Madre/metabolismo , Animales , Adhesión Celular , Movimiento Celular , Quimiotaxis , Células Germinativas/citología , Células Germinativas/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Péptidos/química , Fosforilación , Proteínas Proto-Oncogénicas c-kit/química , Transducción de Señal , Familia-src Quinasas/metabolismo
20.
Dev Biol ; 285(1): 49-56, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16139834

RESUMEN

It is known that mammalian primordial germ cells (PGCs), the precursors of oocytes and prospermatogonia, depend for survival and proliferation on specific growth factors and other undetermined compounds. Adhesion to neighboring somatic cells is also believed to be crucial for preventing PGC apoptosis occurring when they lose appropriate cell to cell contacts. This explains the current impossibility to maintain isolated mouse PGCs in culture for periods longer than a few hours in the absence of suitable cell feeder layers producing soluble factors and expressing surface molecules necessary for preventing PGTC apoptosis and stimulating their proliferation. In the present paper, we identified a cocktail of soluble growth factors, namely KL, LIF, BMP-4, SDF-1, bFGF and compounds (N-acetyl-L-cysteine, forskolin, retinoic acid) able to sustain the survival and self-renewal of mouse PGCs in the absence of somatic cell support. We show that under culture conditions allowing PGC adhesion to an acellular substrate, such growth factors and compounds were able to prevent the occurrence of significant levels of apoptosis in PGCs for two days, stimulate their proliferation and, when LIF was omitted from the cocktail, allow most of them to enter into and progress through meiotic prophase I. These results consent for the first time to establish culture conditions for purified mammalian PGCs in the absence of somatic cell support and should make easier the molecular dissection of the processes governing the development of such cells crucial for early gametogenesis.


Asunto(s)
Células Germinativas/citología , Células Germinativas/efectos de los fármacos , Sustancias de Crecimiento/farmacología , Células Madre/citología , Células Madre/efectos de los fármacos , Acetilcisteína/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Colforsina/farmacología , Femenino , Técnicas In Vitro , Interleucina-6/farmacología , Factor Inhibidor de Leucemia , Masculino , Meiosis/efectos de los fármacos , Ratones , Oocitos/citología , Oocitos/efectos de los fármacos , Oogénesis/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Espermatogonias/citología , Espermatogonias/efectos de los fármacos , Tretinoina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA