Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Genome Res ; 28(1): 25-36, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29162641

RESUMEN

Translation initiation generally occurs at AUG codons in eukaryotes, although it has been shown that non-AUG or noncanonical translation initiation can also occur. However, the evidence for noncanonical translation initiation sites (TISs) is largely indirect and based on ribosome profiling (Ribo-seq) studies. Here, using a strategy specifically designed to enrich N termini of proteins, we demonstrate that many human proteins are translated at noncanonical TISs. The large majority of TISs that mapped to 5' untranslated regions were noncanonical and led to N-terminal extension of annotated proteins or translation of upstream small open reading frames (uORF). It has been controversial whether the amino acid corresponding to the start codon is incorporated at the TIS or methionine is still incorporated. We found that methionine was incorporated at almost all noncanonical TISs identified in this study. Comparison of the TISs determined through mass spectrometry with ribosome profiling data revealed that about two-thirds of the novel annotations were indeed supported by the available ribosome profiling data. Sequence conservation across species and a higher abundance of noncanonical TISs than canonical ones in some cases suggests that the noncanonical TISs can have biological functions. Overall, this study provides evidence of protein translation initiation at noncanonical TISs and argues that further studies are required for elucidation of functional implications of such noncanonical translation initiation.


Asunto(s)
Regiones no Traducidas 5' , Espectrometría de Masas , Sistemas de Lectura Abierta , Iniciación de la Cadena Peptídica Traduccional , Ribosomas/metabolismo , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Dominios Proteicos , Ribosomas/genética
2.
Proc Natl Acad Sci U S A ; 109(46): 18962-7, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23112146

RESUMEN

Stroke is a major cause of mortality and morbidity worldwide. Extracellular glutamate accumulation leading to overstimulation of the ionotropic glutamate receptors mediates neuronal injury in stroke and in neurodegenerative disorders. Here we show that miR-223 controls the response to neuronal injury by regulating the functional expression of the glutamate receptor subunits GluR2 and NR2B in brain. Overexpression of miR-223 lowers the levels of GluR2 and NR2B by targeting 3'-UTR target sites (TSs) in GluR2 and NR2B, inhibits NMDA-induced calcium influx in hippocampal neurons, and protects the brain from neuronal cell death following transient global ischemia and excitotoxic injury. MiR-223 deficiency results in higher levels of NR2B and GluR2, enhanced NMDA-induced calcium influx, and increased miniature excitatory postsynaptic currents in hippocampal neurons. In addition, the absence of MiR-223 leads to contextual, but not cued memory deficits and increased neuronal cell death following transient global ischemia and excitotoxicity. These data identify miR-223 as a major regulator of the expression of GluR2 and NR2B, and suggest a therapeutic role for miR-223 in stroke and other excitotoxic neuronal disorders.


Asunto(s)
Señalización del Calcio , Potenciales Postsinápticos Excitadores , Hipocampo/metabolismo , MicroARNs/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Receptores AMPA/biosíntesis , Receptores de N-Metil-D-Aspartato/biosíntesis , Regiones no Traducidas 3'/genética , Animales , Calcio/metabolismo , Células HEK293 , Hipocampo/patología , Humanos , Ratones , MicroARNs/genética , N-Metilaspartato/genética , N-Metilaspartato/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/terapia , Neuronas/patología , Receptores AMPA/genética , Receptores de N-Metil-D-Aspartato/genética , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/terapia
3.
Nat Rev Neurosci ; 10(12): 837-41, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19904280

RESUMEN

Interest in the functions of microRNAs (miRNAs) in the nervous system has recently expanded to include their roles in neurodegeneration. Investigations have begun to reveal the influence of miRNAs on both neuronal survival and the accumulation of toxic proteins that are associated with neurodegeneration, and are providing clues as to how these toxic proteins can influence miRNA expression.


Asunto(s)
MicroARNs/genética , Degeneración Nerviosa/genética , Animales , Química Encefálica/genética , Química Encefálica/fisiología , Supervivencia Celular/fisiología , Humanos , MicroARNs/fisiología , Degeneración Nerviosa/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología
4.
medRxiv ; 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38853970

RESUMEN

Background: Cytogenetic analysis encompasses a suite of standard-of-care diagnostic testing methods that is routinely applied in cases of acute myeloid leukemia (AML) to assess chromosomal changes that are clinically relevant for risk classification and treatment decisions. Objective: In this study, we assess the use of Genomic Proximity Mapping (GPM) for cytogenomic analysis of AML diagnostic specimens for detection of cytogenetic risk variants included in the European Leukemia Network (ELN) risk stratification guidelines. Methods: Archival patient samples (N=48) from the Fred Hutchinson Cancer Center leukemia bank with historical clinical cytogenetic data were processed for GPM and analyzed with the CytoTerra® cloud-based analysis platform. Results: GPM showed 100% concordance for all specific variants that have associated impacts on risk stratification as defined by ELN 2022 criteria, and a 72% concordance rate when considering all variants reported by the FH cytogenetic lab. GPM identified 39 additional variants, including variants of known clinical impact, not observed by cytogenetics. Conclusions: GPM is an effective solution for the evaluation of known AML-associated risk variants and a source for biomarker discovery.

5.
eNeuro ; 8(6)2021.
Artículo en Inglés | MEDLINE | ID: mdl-34759048

RESUMEN

The G2019S mutation in leucine-rich repeat kinase 2 (LRRK2) causes familial Parkinson's disease (PD) and is also found in a subset of idiopathic cases. Prior studies in Drosophila and human induced pluripotent stem cell (iPSC)-derived dopamine neurons uncovered a pronounced effect of G2019S LRRK2 on mRNA translation. It was previously reported that G2019S LRRK2 promotes translation of mRNAs with complex 5' untranslated region (UTR) secondary structure, resulting in increased expression of calcium channels and dysregulated calcium homeostasis in human dopamine neurons. Here, we show that dysregulated translation occurs in the brains of mammalian LRRK2 models in vivo Through ribosome profiling studies of global translation, we observe that mRNAs with complex 5'UTR structure are also preferentially translated in the G2019S LRRK2-expressing mouse brain. Reporter assays suggest that this 5'UTR preference is independent of translation initiation factors. Conversely, translation of mRNAs with complex 5'UTR secondary structure is downregulated in LRRK2 knock-out (KO) mouse brain, indicating a robust link between LRRK2 kinase activity and translation of mRNA with complex 5'UTR structure. Further, substantia nigra pars compacta (SNpc) dopamine neurons in the G2019S LRRK2-expressing brain exhibit increased calcium influx, which is consistent with the previous report from human dopamine neurons. These results collectively suggest that LRRK2 plays a mechanistic role in translational regulation, and the G2019S mutation in LRRK2 causes translational defects leading to calcium dysregulation in the mammalian brain.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad de Parkinson , Animales , Encéfalo/metabolismo , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Enfermedad de Parkinson/genética , Biosíntesis de Proteínas
6.
G3 (Bethesda) ; 11(2)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33598705

RESUMEN

Mummy berry disease, caused by the fungal pathogen Monilinia vaccinii-corymbosi (Mvc), is one of the most economically important diseases of blueberries in North America. Mvc is capable of inducing two separate blighting stages during its life cycle. Infected fruits are rendered mummified and unmarketable. Genomic data for this pathogen is lacking, but could be useful in understanding the reproductive biology of Mvc and the mechanisms it deploys to facilitate host infection. In this study, PacBio sequencing and Hi-C interaction data were utilized to create a chromosome-scale reference genome for Mvc. The genome comprises nine chromosomes with a total length of 30 Mb, an N50 length of 4.06 Mb, and an average 413X sequence coverage. A total of 9399 gene models were predicted and annotated, and BUSCO analysis revealed that 98% of 1,438 searched conserved eukaryotic genes were present in the predicted gene set. Potential effectors were identified, and the mating-type (MAT) locus was characterized. Biotrophic effectors allow the pathogen to avoid recognition by the host plant and evade or mitigate host defense responses during the early stages of fruit infection. Following locule colonization, necrotizing effectors promote the mummification of host tissues. Potential biotrophic effectors utilized by Mvc include chorismate mutase for reducing host salicylate and necrotrophic effectors include necrosis-inducing proteins and hydrolytic enzymes for macerating host tissue. The MAT locus sequences indicate the potential for homothallism in the reference genome, but a deletion allele of the MAT locus, characterized in a second isolate, indicates heterothallism. Further research is needed to verify the roles of individual effectors in virulence and to determine the role of the MAT locus in outcrossing and population genotypic diversity.


Asunto(s)
Ascomicetos/genética , Arándanos Azules (Planta) , Enfermedades de las Plantas , Frutas , América del Norte , Enfermedades de las Plantas/microbiología
7.
Cell Stem Cell ; 27(4): 633-645.e7, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32846140

RESUMEN

The G2019S mutation in leucine-rich repeat kinase 2 (LRRK2) is a common cause of familial Parkinson's disease (PD). This mutation results in dopaminergic neurodegeneration via dysregulated protein translation, although how alterations in protein synthesis contribute to neurodegeneration in human neurons is not known. Here we define the translational landscape in LRRK2-mutant dopaminergic neurons derived from human induced pluripotent stem cells (hiPSCs) via ribosome profiling. We found that mRNAs that have complex secondary structure in the 5' untranslated region (UTR) are translated more efficiently in G2019S LRRK2 neurons. This leads to the enhanced translation of multiple genes involved in Ca2+ regulation and to increased Ca2+ influx and elevated intracellular Ca2+ levels, a major contributor to PD pathogenesis. This study reveals a link between dysregulated translation control and Ca2+ homeostasis in G2019S LRRK2 human dopamine neurons, which potentially contributes to the progressive and selective dopaminergic neurotoxicity in PD.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad de Parkinson , Calcio , Neuronas Dopaminérgicas/metabolismo , Homeostasis , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Mutación/genética , Enfermedad de Parkinson/genética , Biosíntesis de Proteínas
8.
G3 (Bethesda) ; 10(10): 3479-3488, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32859687

RESUMEN

Social wasps of the genus Vespula have spread to nearly all landmasses worldwide and have become significant pests in their introduced ranges, affecting economies and biodiversity. Comprehensive genome assemblies and annotations for these species are required to develop the next generation of control strategies and monitor existing chemical control. We sequenced and annotated the genomes of the common wasp (Vespula vulgaris), German wasp (Vespula germanica), and the western yellowjacket (Vespula pensylvanica). Our chromosome-level Vespula assemblies each contain 176-179 Mb of total sequence assembled into 25 scaffolds, with 10-200 unanchored scaffolds, and 16,566-18,948 genes. We annotated gene sets relevant to the applied management of invasive wasp populations, including genes associated with spermatogenesis and development, pesticide resistance, olfactory receptors, immunity and venom. These genomes provide evidence for active DNA methylation in Vespidae and tandem duplications of venom genes. Our genomic resources will contribute to the development of next-generation control strategies, and monitoring potential resistance to chemical control.


Asunto(s)
Avispas , Animales , Genómica , Avispas/genética
9.
Mol Endocrinol ; 22(3): 623-35, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18032697

RESUMEN

The male sex steroid, testosterone (T), is synthesized from cholesterol in the testicular Leydig cell under control of the pituitary gonadotropin LH. Unlike most cells that use cholesterol primarily for membrane synthesis, steroidogenic cells have additional requirements for cholesterol, because it is the essential precursor for all steroid hormones. Little is known about how Leydig cells satisfy their specialized cholesterol requirements for steroid synthesis. We show that in mice with a unique hypomorphic androgen mutation, which disrupts the feedback loop governing T synthesis, that genes involved in cholesterol biosynthesis/uptake and steroid biosynthesis are up-regulated. We identify LH as the central regulatory molecule that controls both steroidogenesis and Leydig cell cholesterol homeostasis in vivo. In addition to the primary defect caused by high levels of LH, absence of T signaling exacerbates the lipid homeostasis defect in Leydig cells by eliminating a short feedback loop. We show that T signaling can affect the synthesis of steroids and modulates the expression of genes involved in de novo cholesterol synthesis. Surprisingly, accumulation of active sterol response element-binding protein 2 is not required for up-regulation of genes involved in cholesterol biosynthesis and uptake in Leydig cells.


Asunto(s)
Colesterol/metabolismo , Hormona Luteinizante/biosíntesis , Receptores Androgénicos/metabolismo , Testículo/metabolismo , Testosterona/biosíntesis , Animales , Northern Blotting , Células Cultivadas , Colesterol/biosíntesis , Colesterol/genética , AMP Cíclico/farmacología , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/metabolismo , Hormona Luteinizante/sangre , Masculino , Ratones , Oxidorreductasas/biosíntesis , Oxidorreductasas/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/biosíntesis , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores Androgénicos/biosíntesis , Receptores Androgénicos/genética , Receptores de LDL/biosíntesis , Receptores de LDL/genética , Receptores Depuradores de Clase B/biosíntesis , Receptores Depuradores de Clase B/genética , Organismos Libres de Patógenos Específicos , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Testículo/citología , Testosterona/sangre , Regulación hacia Arriba
10.
Mol Endocrinol ; 21(4): 895-907, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17244764

RESUMEN

The androgen receptor (AR) is a transcription factor that plays a critical role in male sexual development, spermatogenesis, and maintenance of hormonal homeostasis. Despite the extensive knowledge of the phenotypic consequences of mutations in Ar, very little is known about the transcriptional targets of AR within the testis. To identify potential targets of androgen signaling in the testis, we have analyzed the transcriptional profile of adult testes from Ar hypomorphs alone or in combination with Sertoli cell-specific Ar ablation. Using Affymetrix MOE430A mouse genome arrays we interrogated more than 22,000 transcripts. We found the expression level of 62 transcripts in the Ar mutants differed by greater than 2-fold compared with wild type. We also found that more transcripts were up-regulated than down-regulated, highlighting AR's role as a transcriptional repressor in the testis. Twelve transcripts were uniquely affected, and 16 transcripts were more severely affected in Sertoli cell-specific Ar ablation compared with hypomorphic Ar mutants. Using a comparative genomic approach, we analyzed the 6 kb around the transcriptional start sites of affected transcripts for conserved AREs (androgen response elements). We identified at least one conserved ARE in 65% of the genes misregulated in our microarray analysis where clear mouse-human orthologs were available. We used a reporter assay in cell culture to functionally verify the AREs for the kallikrein 27 gene. This suggests that the majority of the misregulated transcripts have a high probability of being direct AR targets. The transcripts affected by these Ar mutations encode a diverse array of proteins whose molecular functions support the contention that AR supports spermatogenesis in both a permissive and instructive fashion.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Receptores Androgénicos/fisiología , Proteínas Represoras/fisiología , Espermatogénesis/genética , Testículo/metabolismo , Andrógenos/metabolismo , Animales , Perfilación de la Expresión Génica , Calicreínas/genética , Masculino , Ratones , Ratones Mutantes , Mutación , Péptido Hidrolasas/genética , Receptores Androgénicos/genética , Proteínas Represoras/genética , Elementos de Respuesta/genética , Transducción de Señal/genética , Espermatozoides/metabolismo , Transcripción Genética
11.
Sci Transl Med ; 8(333): 333ra48, 2016 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-27053772

RESUMEN

Translating neuroprotective treatments from discovery in cell and animal models to the clinic has proven challenging. To reduce the gap between basic studies of neurotoxicity and neuroprotection and clinically relevant therapies, we developed a human cortical neuron culture system from human embryonic stem cells or human inducible pluripotent stem cells that generated both excitatory and inhibitory neuronal networks resembling the composition of the human cortex. This methodology used timed administration of retinoic acid to FOXG1(+) neural precursor cells leading to differentiation of neuronal populations representative of the six cortical layers with both excitatory and inhibitory neuronal networks that were functional and homeostatically stable. In human cortical neuronal cultures, excitotoxicity or ischemia due to oxygen and glucose deprivation led to cell death that was dependent on N-methyl-D-aspartate (NMDA) receptors, nitric oxide (NO), and poly(ADP-ribose) polymerase (PARP) (a cell death pathway called parthanatos that is distinct from apoptosis, necroptosis, and other forms of cell death). Neuronal cell death was attenuated by PARP inhibitors that are currently in clinical trials for cancer treatment. This culture system provides a new platform for the study of human cortical neurotoxicity and suggests that PARP inhibitors may be useful for ameliorating excitotoxic and ischemic cell death in human neurons.


Asunto(s)
Corteza Cerebral/citología , Interneuronas/citología , Inhibición Neural/efectos de los fármacos , Neurotoxinas/toxicidad , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Separación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Factores de Transcripción Forkhead/metabolismo , Glucosa/deficiencia , Proteínas Hedgehog/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Modelos Biológicos , N-Metilaspartato/farmacología , Red Nerviosa/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Óxido Nítrico/metabolismo , Oxígeno , Transducción de Señal/efectos de los fármacos , Tretinoina/farmacología
12.
eNeuro ; 3(5)2016.
Artículo en Inglés | MEDLINE | ID: mdl-27822494

RESUMEN

Neuronal loss caused by ischemic injury, trauma, or disease can lead to devastating consequences for the individual. With the goal of limiting neuronal loss, a number of cell death pathways have been studied, but there may be additional contributors to neuronal death that are yet unknown. To identify previously unknown cell death mediators, we performed a high-content genome-wide screening of short, interfering RNA (siRNA) with an siRNA library in murine neural stem cells after exposure to N-methyl-N-nitroso-N'-nitroguanidine (MNNG), which leads to DNA damage and cell death. Eighty genes were identified as key mediators for cell death. Among them, 14 are known cell death mediators and 66 have not previously been linked to cell death pathways. Using an integrated approach with functional and bioinformatics analysis, we provide possible molecular networks, interconnected pathways, and/or protein complexes that may participate in cell death. Of the 66 genes, we selected CCR3 for further evaluation and found that CCR3 is a mediator of neuronal injury. CCR3 inhibition or deletion protects murine cortical cultures from oxygen-glucose deprivation-induced cell death, and CCR3 deletion in mice provides protection from ischemia in vivo. Taken together, our findings suggest that CCR3 is a previously unknown mediator of cell death. Future identification of the neural cell death network in which CCR3 participates will enhance our understanding of the molecular mechanisms of neural cell death.


Asunto(s)
Muerte Celular/fisiología , Neuronas/metabolismo , Receptores CCR3/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Muerte Celular/efectos de los fármacos , Hipoxia de la Célula/fisiología , Células Cultivadas , Biología Computacional , Modelos Animales de Enfermedad , Glucosa/deficiencia , Infarto de la Arteria Cerebral Media , Masculino , Metilnitronitrosoguanidina/toxicidad , Ratones Noqueados , Actividad Motora/fisiología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Interferencia de ARN , Receptores CCR3/antagonistas & inhibidores , Receptores CCR3/genética , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología
13.
Science ; 354(6308)2016 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-27846469

RESUMEN

Inhibition or genetic deletion of poly(ADP-ribose) (PAR) polymerase-1 (PARP-1) is protective against toxic insults in many organ systems. The molecular mechanisms underlying PARP-1-dependent cell death involve release of mitochondrial apoptosis-inducing factor (AIF) and its translocation to the nucleus, which results in chromatinolysis. We identified macrophage migration inhibitory factor (MIF) as a PARP-1-dependent AIF-associated nuclease (PAAN). AIF was required for recruitment of MIF to the nucleus, where MIF cleaves genomic DNA into large fragments. Depletion of MIF, disruption of the AIF-MIF interaction, or mutation of glutamic acid at position 22 in the catalytic nuclease domain blocked MIF nuclease activity and inhibited chromatinolysis, cell death induced by glutamate excitotoxicity, and focal stroke. Inhibition of MIF's nuclease activity is a potential therapeutic target for diseases caused by excessive PARP-1 activation.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Apoptosis , División del ADN , Daño del ADN , ADN de Cadena Simple/metabolismo , Desoxirribonucleasas/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Apoptosis/genética , Factor Inductor de la Apoptosis/genética , Secuencia de Bases , Dominio Catalítico , Núcleo Celular/enzimología , Cromatina/metabolismo , Daño del ADN/genética , Fragmentación del ADN , Desoxirribonucleasas/química , Desoxirribonucleasas/genética , Ácido Glutámico/química , Ácido Glutámico/genética , Ácido Glutámico/toxicidad , Células HeLa , Humanos , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/química , Factores Inhibidores de la Migración de Macrófagos/genética , Ratones , Ratones Noqueados , Mitocondrias/enzimología , Mutación , Neuronas/enzimología , Conformación de Ácido Nucleico , Estrés Oxidativo , Accidente Cerebrovascular/enzimología , Accidente Cerebrovascular/genética
15.
Endocrinology ; 146(7): 2976-84, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15831567

RESUMEN

Kisspeptins are products of the Kiss1 gene, which bind to GPR54, a G protein-coupled receptor. Kisspeptins and GPR54 have been implicated in the neuroendocrine regulation of GnRH secretion. To test the hypothesis that testosterone regulates Kiss1 gene expression, we compared the expression of KiSS-1 mRNA among groups of intact, castrated, and castrated/testosterone (T)-treated male mice. In the arcuate nucleus (Arc), castration resulted in a significant increase in KiSS-1 mRNA, which was completely reversed with T replacement, whereas in the anteroventral periventricular nucleus, the results were the opposite, i.e. castration decreased and T increased KiSS-1 mRNA expression. In the Arc, the effects of T on KiSS-1 mRNA were completely mimicked by estrogen but only partially mimicked by dihydrotestosterone, a nonaromatizable androgen, suggesting that both estrogen receptor (ER) and androgen receptor (AR) play a role in T-mediated regulation of KiSS-1. Studies of the effects of T on KiSS-1 expression in mice with either a deletion of the ERalpha or a hypomorphic allele to the AR revealed that the effects of T are mediated by both ERalpha and AR pathways, which was confirmed by the presence of either ERalpha or AR coexpression in most KiSS-1 neurons in the Arc. These observations suggest that KiSS-1 neurons in the Arc, whose transcriptional activity is inhibited by T, are targets for the negative feedback regulation of GnRH secretion, whereas KiSS-1 neurons in the anteroventral periventricular nucleus, whose activity is stimulated by T, may mediate other T-dependent processes.


Asunto(s)
Encéfalo/metabolismo , Proteínas/genética , ARN Mensajero/metabolismo , Testosterona/fisiología , Animales , Peso Corporal , Dihidrotestosterona/farmacología , Estradiol/farmacología , Receptor alfa de Estrógeno/deficiencia , Receptor alfa de Estrógeno/genética , Hormonas/sangre , Hibridación in Situ , Kisspeptinas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Orquiectomía , Prosencéfalo/efectos de los fármacos , Prosencéfalo/metabolismo , Receptores Androgénicos/deficiencia , Receptores Androgénicos/genética , Testosterona/farmacología , Distribución Tisular
16.
Mol Endocrinol ; 28(7): 1055-72, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24825397

RESUMEN

Sertoli cell tight junctions (SCTJs) of the seminiferous epithelium create a specialized microenvironment in the testis to aid differentiation of spermatocytes and spermatids from spermatogonial stem cells. SCTJs must be chronically broken and rebuilt with high fidelity to allow the transmigration of preleptotene spermatocytes from the basal to adluminal epithelial compartment. Impairment of androgen signaling in Sertoli cells perturbs SCTJ remodeling. Claudin (CLDN) 3, a tight junction component under androgen regulation, localizes to newly forming SCTJs and is absent in Sertoli cell androgen receptor knockout (SCARKO) mice. We show here that Cldn3-null mice do not phenocopy SCARKO mice: Cldn3(-/-) mice are fertile, show uninterrupted spermatogenesis, and exhibit fully functional SCTJs based on imaging and small molecule tracer analyses, suggesting that other androgen-regulated genes must contribute to the SCARKO phenotype. To further investigate the SCTJ phenotype observed in SCARKO mutants, we generated a new SCARKO model and extensively analyzed the expression of other tight junction components. In addition to Cldn3, we identified altered expression of several other SCTJ molecules, including down-regulation of Cldn13 and a noncanonical tight junction protein 2 isoform (Tjp2iso3). Chromatin immunoprecipitation was used to demonstrate direct androgen receptor binding to regions of these target genes. Furthermore, we demonstrated that CLDN13 is a constituent of SCTJs and that TJP2iso3 colocalizes with tricellulin, a constituent of tricellular junctions, underscoring the importance of androgen signaling in the regulation of both bicellular and tricellular Sertoli cell tight junctions.


Asunto(s)
Claudina-3/genética , Claudinas/genética , Epitelio Seminífero/citología , Uniones Estrechas/fisiología , Proteína de la Zonula Occludens-2/metabolismo , Andrógenos/metabolismo , Animales , Barrera Hematotesticular/metabolismo , Diferenciación Celular , Movimiento Celular , Claudinas/biosíntesis , Epidídimo/citología , Perfilación de la Expresión Génica , Genotipo , Proteína 2 con Dominio MARVEL/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Androgénicos/metabolismo , Células de Sertoli/citología , Recuento de Espermatozoides , Espermátides/citología , Espermatocitos/citología , Espermatogénesis/genética , Espermatogonias/citología , Proteína de la Zonula Occludens-2/biosíntesis , Proteína de la Zonula Occludens-2/genética
17.
Front Cell Neurosci ; 7: 136, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23986658

RESUMEN

MicroRNAs (miRNAs) are small 19-23 nucleotide regulatory RNAs that function by modulating mRNA translation and/or turnover in a sequence-specific fashion. In the nervous system, miRNAs regulate the production of numerous proteins involved in synaptic transmission. In turn, neuronal activity can regulate the production and turnover of miRNA through a variety of mechanisms. In this way, miRNAs and neuronal activity are in a reciprocal homeostatic relationship that balances neuronal function. The miRNA function is critical in pathological states related to overexcitation such as epilepsy and stroke, suggesting miRNA's potential as a therapeutic target. We review the current literature relating the interplay of miRNA and neuronal activity and provide future directions for defining miRNA's role in disease.

18.
PLoS One ; 6(10): e25068, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21984899

RESUMEN

Neuronal activity regulates a broad range of processes in the hippocampus, including the precise regulation of translation. Disruptions in proper translational control in the nervous system are associated with a variety of disorders that fall in the autistic spectrum. MicroRNA (miRNA) represent a relatively recently discovered player in the regulation of translation in the nervous system. We have conducted an in depth analysis of how neuronal activity regulates miRNA expression in the hippocampus. Using deep sequencing we exhaustively identify all miRNAs, including 15 novel miRNAs, expressed in hippocampus of the adult mouse. We identified 119 miRNAs documented in miRBase but less than half of these miRNA were expressed at a level greater than 0.1% of total miRNA. Expression profiling following induction of neuronal activity by electroconvulsive shock demonstrates that most miRNA show a biphasic pattern of expression: rapid induction of specific mature miRNA expression followed by a decline in expression. These results have important implications into how miRNAs influence activity-dependent translational control.


Asunto(s)
Regulación de la Expresión Génica , Hipocampo/metabolismo , MicroARNs/genética , Neuronas/metabolismo , Animales , Secuencia de Bases , Electrochoque , Perfilación de la Expresión Génica , Biblioteca de Genes , Genes Inmediatos-Precoces/genética , Sitios Genéticos/genética , Genoma/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Datos de Secuencia Molecular , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
J Clin Invest ; 120(7): 2446-56, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20516644

RESUMEN

Identification of the signaling pathways that mediate neuronal survival signaling could lead to new therapeutic targets for neurologic disorders and stroke. Sublethal doses of NMDA can induce robust endogenous protective mechanisms in neurons. Through differential analysis of primary library expression and microarray analyses, here we have shown that nuclear factor I, subtype A (NFI-A), a member of the NFI/CAAT-box transcription factor family, is induced in mouse neurons by NMDA receptor activation in a NOS- and ERK-dependent manner. Knockdown of NFI-A induction using siRNA substantially reduced the neuroprotective effects of sublethal doses of NMDA. Further analysis indicated that NFI-A transcriptional activity was required for the neuroprotective effects of NMDA receptor activation. Additional evidence of the neuroprotective effects of NFI-A was provided by the observations that Nfia(-/-) neurons were highly sensitive to NMDA-induced excitotoxicity and were more susceptible to developmental cell death than wild-type neurons and that Nfia(+/-) mice were more sensitive to NMDA-induced intrastriatal lesions than were wild-type animals. These results identify NFI-A as what we believe to be a novel neuroprotective transcription factor with implications in neuroprotection and neuronal plasticity following NMDA receptor activation.


Asunto(s)
N-Metilaspartato/farmacología , Neuronas , Fármacos Neuroprotectores/farmacología , Animales , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Ratones , Ratones Noqueados , N-Metilaspartato/metabolismo , Factores de Transcripción NFI/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Fármacos Neuroprotectores/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA