Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
1.
J Cell Biol ; 223(5)2024 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-38506728

RESUMO

The development of multicellular organisms depends on spatiotemporally controlled differentiation of numerous cell types and their maintenance. To generate such diversity based on the invariant genetic information stored in DNA, epigenetic mechanisms, which are heritable changes in gene function that do not involve alterations to the underlying DNA sequence, are required to establish and maintain unique gene expression programs. Polycomb repressive complexes represent a paradigm of epigenetic regulation of developmentally regulated genes, and the roles of these complexes as well as the epigenetic marks they deposit, namely H3K27me3 and H2AK119ub, have been extensively studied. However, an emerging theme from recent studies is that not only the autonomous functions of the Polycomb repressive system, but also crosstalks of Polycomb with other epigenetic modifications, are important for gene regulation. In this review, we summarize how these crosstalk mechanisms have improved our understanding of Polycomb biology and how such knowledge could help with the design of cancer treatments that target the dysregulated epigenome.


Assuntos
Repressão Epigenética , Genes Controladores do Desenvolvimento , Proteínas do Grupo Polycomb , Diferenciação Celular , Proteínas de Drosophila , Epigênese Genética , Proteínas do Grupo Polycomb/genética , Proteínas do Grupo Polycomb/metabolismo , Humanos , Animais
2.
Cancer Res Commun ; 4(2): 279-292, 2024 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-38240752

RESUMO

Gastric cancer metastasis is a major cause of mortality worldwide. Inhibition of RUNX3 in gastric cancer cell lines reduced migration, invasion, and anchorage-independent growth in vitro. Following splenic inoculation, CRISPR-mediated RUNX3-knockout HGC-27 cells show suppression of xenograft growth and liver metastasis. We interrogated the potential of RUNX3 as a metastasis driver in gastric cancer by profiling its target genes. Transcriptomic analysis revealed strong involvement of RUNX3 in the regulation of multiple developmental pathways, consistent with the notion that Runt domain transcription factor (RUNX) family genes are master regulators of development. RUNX3 promoted "cell migration" and "extracellular matrix" programs, which are necessary for metastasis. Of note, we found pro-metastatic genes WNT5A, CD44, and VIM among the top differentially expressed genes in RUNX3 knockout versus control cells. Chromatin immunoprecipitation sequencing and HiChIP analyses revealed that RUNX3 bound to the enhancers and promoters of these genes, suggesting that they are under direct transcriptional control by RUNX3. We show that RUNX3 promoted metastasis in part through its upregulation of WNT5A to promote migration, invasion, and anchorage-independent growth in various malignancies. Our study therefore reveals the RUNX3-WNT5A axis as a key targetable mechanism for gastric cancer metastasis. SIGNIFICANCE: Subversion of RUNX3 developmental gene targets to metastasis program indicates the oncogenic nature of inappropriate RUNX3 regulation in gastric cancer.


Assuntos
Neoplasias Gástricas , Humanos , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Genes Controladores do Desenvolvimento , Neoplasias Gástricas/genética , Regulação para Cima/genética
3.
Mol Cell ; 83(22): 3972-3999, 2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-37922911

RESUMO

The elongation stage of transcription by RNA polymerase II (RNA Pol II) is central to the regulation of gene expression in response to developmental and environmental cues in metazoan. Dysregulated transcriptional elongation has been associated with developmental defects as well as disease and aging processes. Decades of genetic and biochemical studies have painstakingly identified and characterized an ensemble of factors that regulate RNA Pol II elongation. This review summarizes recent findings taking advantage of genetic engineering techniques that probe functions of elongation factors in vivo. We propose a revised model of elongation control in this accelerating field by reconciling contradictory results from the earlier biochemical evidence and the recent in vivo studies. We discuss how elongation factors regulate promoter-proximal RNA Pol II pause release, transcriptional elongation rate and processivity, RNA Pol II stability and RNA processing, and how perturbation of these processes is associated with developmental disorders, neurodegenerative disease, cancer, and aging.


Assuntos
Doenças Neurodegenerativas , RNA Polimerase II , Animais , Humanos , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Fatores de Elongação da Transcrição/genética , Doenças Neurodegenerativas/genética , Transcrição Gênica , Regulação da Expressão Gênica , Envelhecimento/genética , Genes Controladores do Desenvolvimento
5.
Cell Rep ; 41(11): 111791, 2022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-36516776

RESUMO

Transposable elements (TEs) are the major sources of lineage-specific genomic innovation and comprise nearly half of the human genome, but most of their functions remain unclear. Here, we identify that a series of endogenous retroviruses (ERVs), a TE subclass, regulate the transcriptome at the definitive endoderm stage with in vitro differentiation model from human embryonic stem cell. Notably, these ERVs perform as enhancers containing binding sites for critical transcription factors for endoderm lineage specification. Genome-wide methylation analysis shows most of these ERVs are derepressed by TET1-mediated DNA demethylation. LTR6B, a representative definitive endoderm activating ERV, contains binding sites for FOXA2 and GATA4 and governs the primate-specific expression of its neighboring developmental genes such as ERBB4 in definitive endoderm. Together, our study proposes evidence that recently evolved ERVs represent potent de novo developmental regulatory elements, which, in turn, fine-tune species-specific transcriptomes during endoderm and embryonic development.


Assuntos
Retrovirus Endógenos , Animais , Humanos , Retrovirus Endógenos/genética , Endoderma , Ativação Transcricional , Primatas , Genes Controladores do Desenvolvimento , Desmetilação , Oxigenases de Função Mista/genética , Proteínas Proto-Oncogênicas/genética
6.
PLoS One ; 17(5): e0257936, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35551312

RESUMO

Tomato yellow leaf curl virus (TYLCV), a monopartite begomovirus in the family Geminiviridae, is efficiently transmitted by the whitefly, Bemisia tabaci, and causes serious economic losses to tomato crops around the world. TYLCV-infected tomato plants develop distinctive symptoms of yellowing and leaf upward cupping. In recent years, excellent progress has been made in the characterization of TYLCV C4 protein function as a pathogenicity determinant in experimental plants, including Nicotiana benthamiana and Arabidopsis thaliana. However, the molecular mechanism leading to disease symptom development in the natural host plant, tomato, has yet to be characterized. The aim of the current study was to generate transgenic tomato plants expressing the TYLCV C4 gene and evaluate differential gene expression through comparative transcriptome analysis between the transgenic C4 plants and the transgenic green fluorescent protein (Gfp) gene control plants. Transgenic tomato plants expressing TYLCV C4 developed phenotypes, including leaf upward cupping and yellowing, that are similar to the disease symptoms expressed on tomato plants infected with TYLCV. In a total of 241 differentially expressed genes identified in the transcriptome analysis, a series of plant development-related genes, including transcription factors, glutaredoxins, protein kinases, R-genes and microRNA target genes, were significantly altered. These results provide further evidence to support the important function of the C4 protein in begomovirus pathogenicity. These transgenic tomato plants could serve as basic genetic materials for further characterization of plant receptors that are interacting with the TYLCV C4.


Assuntos
Begomovirus , Hemípteros , Solanum lycopersicum , Animais , Begomovirus/fisiologia , Genes Controladores do Desenvolvimento , Hemípteros/genética , Solanum lycopersicum/genética , Fenótipo , Doenças das Plantas/genética , Plantas Geneticamente Modificadas/genética
7.
Oncogene ; 41(20): 2873-2884, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35422476

RESUMO

Sarcomas are difficult to treat and the therapy, even when effective, is associated with long-term and life-threatening side effects. In addition, the treatment regimens for many sarcomas, including Ewing sarcoma, rhabdomyosarcoma, and osteosarcoma, are relatively unchanged over the past two decades, indicating a critical lack of progress. Although differentiation-based therapies are used for the treatment of some cancers, the application of this approach to sarcomas has proven challenging. Here, using a CRISPR-mediated gene knockout approach, we show that Inhibitor of DNA Binding 2 (ID2) is a critical regulator of developmental-related genes and tumor growth in vitro and in vivo in Ewing sarcoma tumors. We also identified that homoharringtonine, which is an inhibitor of protein translation and FDA-approved for the treatment of leukemia, decreases the level of the ID2 protein and significantly reduces tumor growth and prolongs mouse survival in an Ewing sarcoma xenograft model. Furthermore, in addition to targeting ID2, homoharringtonine also reduces the protein levels of ID1 and ID3, which are additional members of the ID family of proteins with well-described roles in tumorigenesis, in multiple types of cancer. Overall, these results provide insight into developmental regulation in Ewing sarcoma tumors and identify a novel, therapeutic approach to target the ID family of proteins using an FDA-approved drug.


Assuntos
Proteína 2 Inibidora de Diferenciação , Sarcoma de Ewing , Animais , Carcinogênese/genética , Transformação Celular Neoplásica/genética , Genes Controladores do Desenvolvimento , Mepesuccinato de Omacetaxina , Humanos , Proteína 2 Inibidora de Diferenciação/genética , Camundongos , Proteínas/genética , Sarcoma de Ewing/tratamento farmacológico , Sarcoma de Ewing/genética , Sarcoma de Ewing/metabolismo
8.
Hum Mol Genet ; 31(13): 2164-2184, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35094088

RESUMO

Sotos syndrome (SS), the most common overgrowth with intellectual disability (OGID) disorder, is caused by inactivating germline mutations of NSD1, which encodes a histone H3 lysine 36 methyltransferase. To understand how NSD1 inactivation deregulates transcription and DNA methylation (DNAm), and to explore how these abnormalities affect human development, we profiled transcription and DNAm in SS patients and healthy control individuals. We identified a transcriptional signature that distinguishes individuals with SS from controls and was also deregulated in NSD1-mutated cancers. Most abnormally expressed genes displayed reduced expression in SS; these downregulated genes consisted mostly of bivalent genes and were enriched for regulators of development and neural synapse function. DNA hypomethylation was strongly enriched within promoters of transcriptionally deregulated genes: overexpressed genes displayed hypomethylation at their transcription start sites while underexpressed genes featured hypomethylation at polycomb binding sites within their promoter CpG island shores. SS patients featured accelerated molecular aging at the levels of both transcription and DNAm. Overall, these findings indicate that NSD1-deposited H3K36 methylation regulates transcription by directing promoter DNA methylation, partially by repressing polycomb repressive complex 2 (PRC2) activity. These findings could explain the phenotypic similarity of SS to OGID disorders that are caused by mutations in PRC2 complex-encoding genes.


Assuntos
Síndrome de Sotos , Metilação de DNA/genética , Genes Controladores do Desenvolvimento , Histona Metiltransferases/genética , Histona-Lisina N-Metiltransferase/genética , Humanos , Mutação , Síndrome de Sotos/genética
9.
J Cell Sci ; 135(2)2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34908116

RESUMO

The histone acetyltransferase Gcn5 is critical for gene expression and development. In Drosophila, Gcn5 is part of four complexes (SAGA, ATAC, CHAT and ADA) that are essential for fly viability and have key roles in regulating gene expression. Here, we show that although the SAGA, ADA and CHAT complexes play redundant roles in embryonic gene expression, the insect-specific CHAT complex uniquely regulates expression of a subset of developmental genes. We also identify a substantial decrease in histone acetylation in chiffon mutant embryos that exceeds that observed in Ada2b, suggesting broader roles for Chiffon in regulating histone acetylation outside of the Gcn5 complexes. The chiffon gene encodes two independent polypeptides that nucleate formation of either the CHAT or Dbf4-dependent kinase (DDK) complexes. DDK includes the cell cycle kinase Cdc7, which is necessary for maternally driven DNA replication in the embryo. We identify a temporal switch between the expression of these chiffon gene products during a short window during the early nuclear cycles in embryos that correlates with the onset of zygotic genome activation, suggesting a potential role for CHAT in this process. This article has an associated First Person interview with the first author of the paper.


Assuntos
Proteínas de Drosophila , Proteínas de Saccharomyces cerevisiae , Acetilação , Animais , Proteínas de Ciclo Celular , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Genes Controladores do Desenvolvimento , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Proteínas Serina-Treonina Quinases
10.
Genome Res ; 31(6): 1106-1119, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33832989

RESUMO

Steps of mRNA maturation are important gene regulatory events that occur in distinct cellular locations. However, transcriptomic analyses often lose information on the subcellular distribution of processed and unprocessed transcripts. We generated extensive RNA-seq data sets to track mRNA maturation across subcellular locations in mouse embryonic stem cells, neuronal progenitor cells, and postmitotic neurons. We find disparate patterns of RNA enrichment between the cytoplasmic, nucleoplasmic, and chromatin fractions, with some genes maintaining more polyadenylated RNA in chromatin than in the cytoplasm. We bioinformatically defined four regulatory groups for intron retention, including complete cotranscriptional splicing, complete intron retention in the cytoplasmic RNA, and two intron groups present in nuclear and chromatin transcripts but fully excised in cytoplasm. We found that introns switch their regulatory group between cell types, including neuronally excised introns repressed by polypyrimidine track binding protein 1 (PTBP1). Transcripts for the neuronal gamma-aminobutyric acid (GABA) B receptor, 1 (Gabbr1) are highly expressed in mESCs but are absent from the cytoplasm. Instead, incompletely spliced Gabbr1 RNA remains sequestered on chromatin, where it is bound by PTBP1, similar to certain long noncoding RNAs. Upon neuronal differentiation, Gabbr1 RNA becomes fully processed and exported for translation. Thus, splicing repression and chromatin anchoring of RNA combine to allow posttranscriptional regulation of Gabbr1 over development. For this and other genes, polyadenylated RNA abundance does not indicate functional gene expression. Our data sets provide a rich resource for analyzing many other aspects of mRNA maturation in subcellular locations and across development.


Assuntos
Precursores de RNA , Splicing de RNA , Animais , Núcleo Celular/genética , Núcleo Celular/metabolismo , Genes Controladores do Desenvolvimento , Íntrons/genética , Camundongos , Precursores de RNA/genética , Precursores de RNA/metabolismo
11.
Dis Model Mech ; 14(5)2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33737325

RESUMO

Heterozygous mutations in HNF1B cause the complex syndrome renal cysts and diabetes (RCAD), characterized by developmental abnormalities of the kidneys, genital tracts and pancreas, and a variety of renal, pancreas and liver dysfunctions. The pathogenesis underlying this syndrome remains unclear as mice with heterozygous null mutations have no phenotype, while constitutive/conditional Hnf1b ablation leads to more severe phenotypes. We generated a novel mouse model carrying an identified human mutation at the intron-2 splice donor site. Unlike heterozygous mice previously characterized, mice heterozygous for the splicing mutation exhibited decreased HNF1B protein levels and bilateral renal cysts from embryonic day 15, originated from glomeruli, early proximal tubules (PTs) and intermediate nephron segments, concurrently with delayed PT differentiation, hydronephrosis and rare genital tract anomalies. Consistently, mRNA sequencing showed that most downregulated genes in embryonic kidneys were primarily expressed in early PTs and the loop of Henle and involved in ion/drug transport, organic acid and lipid metabolic processes, while the expression of previously identified targets upon Hnf1b ablation, including cystic disease genes, was weakly or not affected. Postnatal analyses revealed renal abnormalities, ranging from glomerular cysts to hydronephrosis and, rarely, multicystic dysplasia. Urinary proteomics uncovered a particular profile predictive of progressive decline in kidney function and fibrosis, and displayed common features with a recently reported urine proteome in an RCAD pediatric cohort. Altogether, our results show that reduced HNF1B levels lead to developmental disease phenotypes associated with the deregulation of a subset of HNF1B targets. They further suggest that this model represents a unique clinical/pathological viable model of the RCAD disease.


Assuntos
Doenças do Sistema Nervoso Central/genética , Esmalte Dentário/anormalidades , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 2/genética , Genes Controladores do Desenvolvimento , Haploinsuficiência/genética , Fator 1-beta Nuclear de Hepatócito/genética , Doenças Renais Císticas/genética , Animais , Animais Recém-Nascidos , Polaridade Celular , Doenças do Sistema Nervoso Central/patologia , Cílios/patologia , Esmalte Dentário/patologia , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Embrião de Mamíferos/patologia , Dosagem de Genes , Perfilação da Expressão Gênica , Heterozigoto , Humanos , Hidronefrose/complicações , Doenças Renais Císticas/patologia , Glomérulos Renais/patologia , Túbulos Renais/patologia , Camundongos Endogâmicos C57BL , Mutação/genética , Néfrons/patologia , Splicing de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Índice de Gravidade de Doença
12.
Development ; 148(1)2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33318148

RESUMO

Androgens/androgen receptor (AR)-mediated signaling pathways are essential for prostate development, morphogenesis and regeneration. Specifically, stromal AR signaling has been shown to be essential for prostatic initiation. However, the molecular mechanisms underlying AR-initiated mesenchymal-epithelial interactions in prostate development remain unclear. Here, using a newly generated mouse model, we have directly addressed the fate and role of genetically marked AR-expressing cells during embryonic prostate development. Androgen signaling-initiated signaling pathways were identified in mesenchymal niche populations at single-cell transcriptomic resolution. The dynamic cell-signaling networks regulated by stromal AR were additionally characterized in relation to prostatic epithelial bud formation. Pseudotime analyses further revealed the differentiation trajectory and fate of AR-expressing cells in both prostatic mesenchymal and epithelial cell populations. Specifically, the cellular properties of Zeb1-expressing progenitors were assessed. Selective deletion of AR signaling in a subpopulation of mesenchymal rather than epithelial cells dysregulated the expression of the master regulators and significantly impaired prostatic bud formation. These data provide novel, high-resolution evidence demonstrating the important role of mesenchymal androgen signaling in the cellular niche controlling prostate early development by initiating dynamic mesenchyme-epithelia cell interactions.


Assuntos
Androgênios/farmacologia , Comunicação Celular , Linhagem da Célula , Próstata/citologia , Análise de Célula Única , Animais , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/genética , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Linhagem da Célula/genética , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genes Controladores do Desenvolvimento , Masculino , Mesoderma/citologia , Camundongos , Próstata/efeitos dos fármacos , RNA-Seq , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo
13.
Nucleic Acids Res ; 48(3): 1206-1224, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31799607

RESUMO

The histone demethylase LSD1 is a key chromatin regulator that is often deregulated in cancer. Its ortholog, dLsd1 plays a crucial role in Drosophila oogenesis; however, our knowledge of dLsd1 function is insufficient to explain its role in the ovary. Here, we have performed genome-wide analysis of dLsd1 binding in the ovary, and we document that dLsd1 is preferentially associated to the transcription start site of developmental genes. We uncovered an unanticipated interplay between dLsd1 and the GATA transcription factor Serpent and we report an unexpected role for Serpent in oogenesis. Besides, our transcriptomic data show that reducing dLsd1 levels results in ectopic transposable elements (TE) expression correlated with changes in H3K4me2 and H3K9me2 at TE loci. In addition, our results suggest that dLsd1 is required for Piwi dependent TE silencing. Hence, we propose that dLsd1 plays crucial roles in establishing specific gene expression programs and in repressing transposons during oogenesis.


Assuntos
Elementos de DNA Transponíveis/genética , Proteínas de Drosophila/genética , Fatores de Transcrição GATA/genética , Oogênese/genética , Oxirredutases N-Desmetilantes/genética , Animais , Proteínas Argonautas/genética , Cromatina/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Genes Controladores do Desenvolvimento/genética , Histonas/genética , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Sítio de Iniciação de Transcrição
15.
Development ; 146(14)2019 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-31221640

RESUMO

Bone protrusions provide stable anchoring sites for ligaments and tendons and define the unique morphology of each long bone. Despite their importance, the mechanism by which superstructures are patterned is unknown. Here, we identify components of the genetic program that control the patterning of Sox9+/Scx+ superstructure progenitors in mouse and show that this program includes both global and regional regulatory modules. Using light-sheet fluorescence microscopy combined with genetic lineage labeling, we mapped the broad contribution of the Sox9+/Scx+ progenitors to the formation of bone superstructures. Then, by combining literature-based evidence, comparative transcriptomic analysis and genetic mouse models, we identified Gli3 as a global regulator of superstructure patterning, whereas Pbx1, Pbx2, Hoxa11 and Hoxd11 act as proximal and distal regulators, respectively. Moreover, by demonstrating a dose-dependent pattern regulation in Gli3 and Pbx1 compound mutations, we show that the global and regional regulatory modules work in a coordinated manner. Collectively, our results provide strong evidence for genetic regulation of superstructure patterning, which further supports the notion that long bone development is a modular process.This article has an associated 'The people behind the papers' interview.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Osso e Ossos/anatomia & histologia , Osso e Ossos/embriologia , Genes Controladores do Desenvolvimento , Proteínas de Homeodomínio/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Desenvolvimento Ósseo/genética , Osso e Ossos/metabolismo , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genes Controladores do Desenvolvimento/genética , Proteínas de Homeodomínio/metabolismo , Ligamentos/anatomia & histologia , Ligamentos/embriologia , Ligamentos/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos/genética , Fator de Transcrição 1 de Leucemia de Células Pré-B/genética , Fator de Transcrição 1 de Leucemia de Células Pré-B/metabolismo , Gravidez , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Tendões/anatomia & histologia , Tendões/embriologia , Tendões/metabolismo
16.
Fish Physiol Biochem ; 45(1): 167-176, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30143929

RESUMO

The insulin-like growth factor (IGF) system plays important roles in fish reproduction, but the expression pattern and cellular location of IGF-I and IGF-II during gonadal maturation are uncertain. The present study reports a stage-specific assessment of gonadal expression levels and immunolocalisation of IGF-I and IGF-II in Astyanax fasciatus, a characid fish from South America. Adult fish in different maturity stages were caught in the Furnas Reservoir, Grande River, Brazil. Gonad samples were processed for histology, immunohistochemistry, and ELISA for IGF-I and IGF-II. Ovarian levels of IGF-I were low during ripening and ripe stages, higher in totally spent, and then decreased in resting. Levels of IGF-II increased during ovarian maturation, reaching significantly higher values at stage totally spent. In males, IGF-I levels followed gonadal maturation, with higher values in ripening and ripe stages, whereas IGF-II levels showed higher values in stage ripening and partially spent. A positive correlation was found between IGF-I and gonadosomatic index (GSI) for males (r = 0.59), while females showed a negative correlation (r = - 0.43), but IGF-II showed no correlation to GSI. IGF-I was expressed mainly in oogonia nests whereas IGF-II stained the follicular cells in the perinucleolar follicles, cortical vesicles in the previtellogenic follicles, and oogonia nests. In males, IGF-I was evident in spermatogonia and spermatocytes while IGF-II stained Sertoli cells surrounding spermatids cysts and spermatogonia in late stages. Together, these findings support a hypothesis that the balance between IGF-I and IGF-II levels is important in the regulation of gonad maturation in Astyanax fasciatus.


Assuntos
Characidae/fisiologia , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Ovário/metabolismo , Testículo/metabolismo , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes Controladores do Desenvolvimento , Imuno-Histoquímica/veterinária , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like II/genética , Masculino , Ovário/crescimento & desenvolvimento , Testículo/crescimento & desenvolvimento
17.
J Vet Med Sci ; 80(10): 1534-1539, 2018 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-30175754

RESUMO

Stable reference genes are important for gene expression analyses such as quantitative PCR. The stability of 15 candidate reference genes that can be used to developing mouse gonads was thoroughly verified using combinations of multiple algorithms. The expression of these genes fluctuated greatly depending on the analysis period and/or gender. Peptidylprolyl isomerase A (Ppia) and polymerase (RNA) II (DNA directed) polypeptide A (Polr2a) were the reference genes that were used stably for a wide analysis period in developing mouse gonads. Furthermore, the stable reference genes corresponding to the analysis period and/or gender were ranked. These results are useful for the selection of the optimal reference gene required for high-precision measurements.


Assuntos
Genes Controladores do Desenvolvimento , Gônadas/crescimento & desenvolvimento , Reação em Cadeia da Polimerase em Tempo Real , Algoritmos , Animais , Feminino , Genes Reporter , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Padrões de Referência
18.
Cell ; 174(1): 117-130.e14, 2018 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-29909981

RESUMO

Heterogeneity is a hallmark feature of the adaptive immune system in vertebrates. Following infection, naive T cells differentiate into various subsets of effector and memory T cells, which help to eliminate pathogens and maintain long-term immunity. The current model suggests there is a single lineage of naive T cells that give rise to different populations of effector and memory T cells depending on the type and amounts of stimulation they encounter during infection. Here, we have discovered that multiple sub-populations of cells exist in the naive CD8+ T cell pool that are distinguished by their developmental origin, unique transcriptional profiles, distinct chromatin landscapes, and different kinetics and phenotypes after microbial challenge. These data demonstrate that the naive CD8+ T cell pool is not as homogeneous as previously thought and offers a new framework for explaining the remarkable heterogeneity in the effector and memory T cell subsets that arise after infection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Genes Controladores do Desenvolvimento , Listeria monocytogenes/patogenicidade , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Cromatina/metabolismo , Citocinas/farmacologia , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/metabolismo , Memória Imunológica , Interferon gama/metabolismo , Células Matadoras Naturais/citologia , Células Matadoras Naturais/metabolismo , Listeria monocytogenes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Análise de Componente Principal , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Timo/transplante , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma
19.
Stem Cells ; 36(8): 1179-1197, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29575325

RESUMO

During aging-one the most potent risk factors for Parkinson's disease (PD)-both astrocytes and microglia undergo functional changes that ultimately hamper homoeostasis, defense, and repair of substantia nigra pars compacta (SNpc) midbrain dopaminergic (mDA) neurons. We tested the possibility of rejuvenating the host microenvironment and boosting SNpc DA neuronal plasticity via the unilateral transplantation of syngeneic neural stem/progenitor cells (NSCs) in the SNpc of aged mice with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced experimental PD. Transplanted NSCs within the aged SNpc engrafted and migrated in large proportions to the tegmental aqueduct mDA niche, with 30% acquiring an astroglial phenotype. Both graft-derived exogenous (ex-Astro) and endogenous astrocytes (en-Astro) expressed Wnt1. Both ex-Astro and en-Astro were key triggers of Wnt/ß-catenin signaling in SNpc-mDA neurons and microglia, which was associated with mDA neurorescue and immunomodulation. At the aqueduct-ventral tegmental area level, NSC grafts recapitulated a genetic Wnt1-dependent mDA developmental program, inciting the acquisition of a mature Nurr1+ TH+ neuronal phenotype. Wnt/ß-catenin signaling antagonism abolished mDA neurorestoration and immune modulatory effects of NSC grafts. Our work implicates an unprecedented therapeutic potential for somatic NSC grafts in the restoration of mDA neuronal function in the aged Parkinsonian brain. Stem Cells 2018;36:1179-1197.


Assuntos
Envelhecimento/patologia , Astrócitos/patologia , Encéfalo/patologia , Células-Tronco Neurais/transplante , Doença de Parkinson/patologia , Doença de Parkinson/terapia , Via de Sinalização Wnt , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Animais , Astrócitos/metabolismo , Morte Celular , Diferenciação Celular/genética , Linhagem da Célula , Proliferação de Células , Sobrevivência Celular , Modelos Animais de Doenças , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Regulação para Baixo/genética , Genes Controladores do Desenvolvimento , Inflamação/genética , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Microglia/patologia , Células-Tronco Neurais/citologia , Estresse Oxidativo/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Substância Negra/patologia , Sinaptossomos/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Via de Sinalização Wnt/genética
20.
Cell Mol Life Sci ; 75(3): 385-402, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28821904

RESUMO

Malignant gliomas are the most common, infiltrative, and lethal primary brain tumors affecting the adult population. The grim prognosis for this disease is due to a combination of the presence of highly invasive tumor cells that escape surgical resection and the presence of a population of therapy-resistant cancer stem cells found within these tumors. Several studies suggest that glioma cells have cleverly hijacked the normal developmental program of neural progenitor cells, including their transcriptional programs, to enhance gliomagenesis. In this review, we summarize the role of developmentally regulated signaling pathways that have been found to facilitate glioma growth and invasion. Furthermore, we discuss how the microenvironment and treatment-induced perturbations of these highly interconnected signaling networks can trigger a shift in cellular phenotype and tumor subtype.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Genes Controladores do Desenvolvimento , Glioma/genética , Glioma/patologia , Adulto , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Invasividade Neoplásica , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neurais/metabolismo , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA