Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 17(2): e0263632, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35192674

RESUMO

Adults of many species will care for young offspring that are not their own, a phenomenon called alloparenting. However, in many cases, nonparental adults must be sensitized by repeated or extended exposures to newborns before they will robustly display parental-like behaviors. To capture neurogenomic events underlying the transition to active parental caring behaviors, we analyzed brain gene expression and chromatin profiles of virgin female mice co-housed with pregnant dams during pregnancy and after birth. After an initial display of antagonistic behaviors and a surge of defense-related gene expression, we observed a dramatic shift in the chromatin landscape specifically in amygdala of the pup-exposed virgin females compared to females co-housed with mother before birth, accompanied by a dampening of anxiety-related gene expression. This epigenetic shift coincided with hypothalamic expression of the oxytocin gene and the emergence of behaviors and gene expression patterns classically associated with maternal care. The results outline a neurogenomic program associated with dramatic behavioral changes and suggest molecular networks relevant to human postpartum mental health.


Assuntos
Tonsila do Cerebelo/metabolismo , Comportamento Animal/fisiologia , Epigênese Genética , Comportamento Materno/fisiologia , Proteínas do Tecido Nervoso/genética , Ocitocina/genética , Animais , Animais Recém-Nascidos , Ansiedade/psicologia , Cromatina/química , Cromatina/metabolismo , Feminino , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Hipotálamo/metabolismo , Comportamento Materno/psicologia , Camundongos , Proteínas do Tecido Nervoso/classificação , Proteínas do Tecido Nervoso/metabolismo , Ocitocina/metabolismo , Gravidez , Abstinência Sexual
2.
G3 (Bethesda) ; 9(11): 3891-3906, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31554716

RESUMO

AUTS2 was originally discovered as the gene disrupted by a translocation in human twins with Autism spectrum disorder, intellectual disability, and epilepsy. Since that initial finding, AUTS2-linked mutations and variants have been associated with a very broad array of neuropsychiatric disorders, sugg esting that AUTS2 is required for fundamental steps of neurodevelopment. However, genotype-phenotype correlations in this region are complicated, because most mutations could also involve neighboring genes. Of particular interest is the nearest downstream neighbor of AUTS2, GALNT17, which encodes a brain-expressed N-acetylgalactosaminyltransferase of unknown brain function. Here we describe a mouse (Mus musculus) mutation, T(5G2;8A1)GSO (abbreviated 16Gso), a reciprocal translocation that breaks between Auts2 and Galnt17 and dysregulates both genes. Despite this complex regulatory effect, 16Gso homozygotes model certain human AUTS2-linked phenotypes very well. In addition to abnormalities in growth, craniofacial structure, learning and memory, and behavior, 16Gso homozygotes display distinct pathologies of the cerebellum and hippocampus that are similar to those associated with autism and other types of AUTS2-linked neurological disease. Analyzing mutant cerebellar and hippocampal transcriptomes to explain this pathology, we identified disturbances in pathways related to neuron and synapse maturation, neurotransmitter signaling, and cellular stress, suggesting possible cellular mechanisms. These pathways, coupled with the translocation's selective effects on Auts2 isoforms and coordinated dysregulation of Galnt17, suggest novel hypotheses regarding the etiology of the human "AUTS2 syndrome" and the wide array of neurodevelopmental disorders linked to variance in this genomic region.


Assuntos
Proteínas do Citoesqueleto/genética , N-Acetilgalactosaminiltransferases/genética , Fatores de Transcrição/genética , Animais , Comportamento Animal , Cerebelo/metabolismo , Cerebelo/patologia , Proteínas do Citoesqueleto/metabolismo , Feminino , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/patologia , Fenótipo , Crânio/anatomia & histologia , Síndrome , Fatores de Transcrição/metabolismo
3.
Genes Brain Behav ; 18(1): e12502, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29968347

RESUMO

Social challenges like territorial intrusions evoke behavioral responses in widely diverging species. Recent work has showed that evolutionary "toolkits"-genes and modules with lineage-specific variations but deep conservation of function-participate in the behavioral response to social challenge. Here, we develop a multispecies computational-experimental approach to characterize such a toolkit at a systems level. Brain transcriptomic responses to social challenge was probed via RNA-seq profiling in three diverged species-honey bees, mice and three-spined stickleback fish-following a common methodology, allowing fair comparisons across species. Data were collected from multiple brain regions and multiple time points after social challenge exposure, achieving anatomical and temporal resolution substantially greater than previous work. We developed statistically rigorous analyses equipped to find homologous functional groups among these species at the levels of individual genes, functional and coexpressed gene modules, and transcription factor subnetworks. We identified six orthogroups involved in response to social challenge, including groups represented by mouse genes Npas4 and Nr4a1, as well as common modulation of systems such as transcriptional regulators, ion channels, G-protein-coupled receptors and synaptic proteins. We also identified conserved coexpression modules enriched for mitochondrial fatty acid metabolism and heat shock that constitute the shared neurogenomic response. Our analysis suggests a toolkit wherein nuclear receptors, interacting with chaperones, induce transcriptional changes in mitochondrial activity, neural cytoarchitecture and synaptic transmission after social challenge. It shows systems-level mechanisms that have been repeatedly co-opted during evolution of analogous behaviors, thus advancing the genetic toolkit concept beyond individual genes.


Assuntos
Evolução Molecular , Genética Comportamental/métodos , Genômica/métodos , Comportamento Social , Análise de Sistemas , Animais , Abelhas , Encéfalo/metabolismo , Encéfalo/fisiologia , Feminino , Redes Reguladoras de Genes , Genoma , Masculino , Camundongos , Smegmamorpha , Transcriptoma
4.
Genome Res ; 27(6): 959-972, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28356321

RESUMO

Agonistic encounters are powerful effectors of future behavior, and the ability to learn from this type of social challenge is an essential adaptive trait. We recently identified a conserved transcriptional program defining the response to social challenge across animal species, highly enriched in transcription factor (TF), energy metabolism, and developmental signaling genes. To understand the trajectory of this program and to uncover the most important regulatory influences controlling this response, we integrated gene expression data with the chromatin landscape in the hypothalamus, frontal cortex, and amygdala of socially challenged mice over time. The expression data revealed a complex spatiotemporal patterning of events starting with neural signaling molecules in the frontal cortex and ending in the modulation of developmental factors in the amygdala and hypothalamus, underpinned by a systems-wide shift in expression of energy metabolism-related genes. The transcriptional signals were correlated with significant shifts in chromatin accessibility and a network of challenge-associated TFs. Among these, the conserved metabolic and developmental regulator ESRRA was highlighted for an especially early and important regulatory role. Cell-type deconvolution analysis attributed the differential metabolic and developmental signals in this social context primarily to oligodendrocytes and neurons, respectively, and we show that ESRRA is expressed in both cell types. Localizing ESRRA binding sites in cortical chromatin, we show that this nuclear receptor binds both differentially expressed energy-related and neurodevelopmental TF genes. These data link metabolic and neurodevelopmental signaling to social challenge, and identify key regulatory drivers of this process with unprecedented tissue and temporal resolution.


Assuntos
Cromatina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Neurônios/metabolismo , Receptores de Estrogênio/genética , Estresse Psicológico/genética , Fatores de Transcrição/genética , Comportamento Agonístico , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/fisiopatologia , Animais , Cromatina/ultraestrutura , Metabolismo Energético/genética , Lobo Frontal/metabolismo , Lobo Frontal/fisiopatologia , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Hipotálamo/metabolismo , Hipotálamo/fisiopatologia , Masculino , Camundongos , Neurônios/citologia , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Ligação Proteica , Receptores de Estrogênio/metabolismo , Transdução de Sinais , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia , Fatores de Transcrição/metabolismo , Transcrição Gênica
5.
Oncotarget ; 7(45): 72571-72592, 2016 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-27732952

RESUMO

Mammalian genomes contain hundreds of genes transcribed by RNA Polymerase III (Pol III), encoding noncoding RNAs and especially the tRNAs specialized to carry specific amino acids to the ribosome for protein synthesis. In addition to this well-known function, tRNAs and their genes (tDNAs) serve a variety of other critical cellular functions. For example, tRNAs and other Pol III transcripts can be cleaved to yield small RNAs with potent regulatory activities. Furthermore, from yeast to mammals, active tDNAs and related "extra-TFIIIC" (ETC) loci provide the DNA scaffolds for the most ancient known mechanism of three-dimensional chromatin architecture. Here we identify the ZSCAN5 TF family - including mammalian ZSCAN5B and its primate-specific paralogs - as proteins that occupy mammalian Pol III promoters and ETC sites. We show that ZSCAN5B binds with high specificity to a conserved subset of Pol III genes in human and mouse. Furthermore, primate-specific ZSCAN5A and ZSCAN5D also bind Pol III genes, although ZSCAN5D preferentially localizes to MIR SINE- and LINE2-associated ETC sites. ZSCAN5 genes are expressed in proliferating cell populations and are cell-cycle regulated, and siRNA knockdown experiments suggested a cooperative role in regulation of mitotic progression. Consistent with this prediction, ZSCAN5A knockdown led to increasing numbers of cells in mitosis and the appearance of cells. Together, these data implicate the role of ZSCAN5 genes in regulation of Pol III genes and nearby Pol II loci, ultimately influencing cell cycle progression and differentiation in a variety of tissues.


Assuntos
Cromatina/metabolismo , RNA Polimerase III/genética , Fatores de Transcrição TFIII/genética , Animais , Ciclo Celular/fisiologia , Progressão da Doença , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mitose/fisiologia , RNA Polimerase III/metabolismo , Fatores de Transcrição TFIII/metabolismo , Transcrição Gênica , Transfecção
6.
PLoS One ; 11(4): e0154413, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27120339

RESUMO

The T-box transcription factor TBX18 is essential to mesenchymal cell differentiation in several tissues and Tbx18 loss-of-function results in dramatic organ malformations and perinatal lethality. Here we demonstrate for the first time that Tbx18 is required for the normal development of periductal smooth muscle stromal cells in prostate, particularly in the anterior lobe, with a clear impact on prostate health in adult mice. Prostate abnormalities are only subtly apparent in Tbx18 mutants at birth; to examine postnatal prostate development we utilized a relatively long-lived hypomorphic mutant and a novel conditional Tbx18 allele. Similar to the ureter, cells that fail to express Tbx18 do not condense normally into smooth muscle cells of the periductal prostatic stroma. However, in contrast to ureter, the periductal stromal cells in mutant prostate assume a hypertrophic, myofibroblastic state and the adjacent epithelium becomes grossly disorganized. To identify molecular events preceding the onset of this pathology, we compared gene expression in the urogenital sinus (UGS), from which the prostate develops, in Tbx18-null and wild type littermates at two embryonic stages. Genes that regulate cell proliferation, smooth muscle differentiation, prostate epithelium development, and inflammatory response were significantly dysregulated in the mutant urogenital sinus around the time that Tbx18 is first expressed in the wild type UGS, suggesting a direct role in regulating those genes. Together, these results argue that Tbx18 is essential to the differentiation and maintenance of the prostate periurethral mesenchyme and that it indirectly regulates epithelial differentiation through control of stromal-epithelial signaling.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Músculo Liso/metabolismo , Miócitos de Músculo Liso/metabolismo , Próstata/metabolismo , Células Estromais/metabolismo , Proteínas com Domínio T/genética , Alelos , Animais , Comunicação Celular , Diferenciação Celular , Proliferação de Células , Ductos Ejaculatórios/crescimento & desenvolvimento , Ductos Ejaculatórios/metabolismo , Ductos Ejaculatórios/patologia , Embrião de Mamíferos , Perfilação da Expressão Gênica , Masculino , Camundongos , Músculo Liso/crescimento & desenvolvimento , Músculo Liso/patologia , Miócitos de Músculo Liso/patologia , Organogênese/genética , Próstata/crescimento & desenvolvimento , Próstata/patologia , Transdução de Sinais , Células Estromais/patologia , Proteínas com Domínio T/deficiência , Ureter/crescimento & desenvolvimento , Ureter/metabolismo , Ureter/patologia
7.
Proc Natl Acad Sci U S A ; 111(50): 17929-34, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25453090

RESUMO

Certain complex phenotypes appear repeatedly across diverse species due to processes of evolutionary conservation and convergence. In some contexts like developmental body patterning, there is increased appreciation that common molecular mechanisms underlie common phenotypes; these molecular mechanisms include highly conserved genes and networks that may be modified by lineage-specific mutations. However, the existence of deeply conserved mechanisms for social behaviors has not yet been demonstrated. We used a comparative genomics approach to determine whether shared neuromolecular mechanisms could underlie behavioral response to territory intrusion across species spanning a broad phylogenetic range: house mouse (Mus musculus), stickleback fish (Gasterosteus aculeatus), and honey bee (Apis mellifera). Territory intrusion modulated similar brain functional processes in each species, including those associated with hormone-mediated signal transduction and neurodevelopment. Changes in chromosome organization and energy metabolism appear to be core, conserved processes involved in the response to territory intrusion. We also found that several homologous transcription factors that are typically associated with neural development were modulated across all three species, suggesting that shared neuronal effects may involve transcriptional cascades of evolutionarily conserved genes. Furthermore, immunohistochemical analyses of a subset of these transcription factors in mouse again implicated modulation of energy metabolism in the behavioral response. These results provide support for conserved genetic "toolkits" that are used in independent evolutions of the response to social challenge in diverse taxa.


Assuntos
Abelhas/fisiologia , Evolução Biológica , Encéfalo/fisiologia , Smegmamorpha/fisiologia , Comportamento Social , Territorialidade , Animais , Sequência de Bases , Abelhas/genética , Primers do DNA/genética , Metabolismo Energético/fisiologia , Genômica/métodos , Imuno-Histoquímica , Camundongos , Microscopia de Fluorescência , Anotação de Sequência Molecular , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Análise de Sequência de RNA , Transdução de Sinais/fisiologia , Smegmamorpha/genética , Especificidade da Espécie , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...