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








Base de dados
Intervalo de ano de publicação
1.
Cytogenet Genome Res ; 164(1): 23-32, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38754392

RESUMO

INTRODUCTION: X chromosome inactivation (XCI) is an essential mechanism for dosage compensation between females and males in mammals. In females, XCI is controlled by a complex, conserved locus termed the X inactivation center (Xic), in which the lncRNA Xist is the key regulator. However, little is known about the Xic in species with unusual sex chromosomes. The genus Tokudaia includes three rodent species endemic to Japan. Tokudaia osimensis and Tokudaia tokunoshimensis lost the Y chromosome (XO/XO), while Tokudaia muenninki (TMU) acquired a neo-X region by fusion of the X chromosome and an autosome (XX/XY). We compared the gene location and structure in the Xic among Tokudaia species. METHODS: Gene structure of nine genes in Xic was predicted, and the gene location and genome sequences of Xic were compared between mouse and Tokudaia species. The expression level of the gene was confirmed by transcripts per million calculation using RNA-seq data. RESULTS: Compared to mouse, the Xic gene order and location were conserved in Tokudaia species. However, remarkable structure changes were observed in lncRNA genes, Xist and Tsix, in the XO/XO species. In Xist, important functional repeats, B-, C-, D-, and E-repeats, were partially or completely lost due to deletions in these species. RNA-seq data showed that female-specific expression patterns of Xist and Tsix were confirmed in TMU, however, not in the XO/XO species. Additionally, three deletions and one inversion were confirmed in the intergenic region between Jpx and Ftx in the XO/XO species. CONCLUSION: Our findings indicate that even if the Xist and Tsix lncRNAs are expressed, they are incapable of producing a successful and lasting XCI in the XO/XO species. We hypothesized that the significant structure change in the intergenic region of Jpx-Ftx resulted in the inability to perform the XCI, and, as a result, a lack of Xist expression. Our results collectively suggest that structural changes in the Xic occurred in the ancestral lineage of XO/XO species, likely due to the loss of one X chromosome and the Y chromosome as a consequence of the degradation of the XCI system.


Assuntos
RNA Longo não Codificante , Inativação do Cromossomo X , Cromossomo X , Cromossomo Y , Animais , Inativação do Cromossomo X/genética , Feminino , Cromossomo X/genética , Masculino , Cromossomo Y/genética , RNA Longo não Codificante/genética , Camundongos , Murinae/genética
2.
J Biochem Mol Toxicol ; 37(8): e23384, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37155292

RESUMO

Long noncoding RNA (lncRNA)-X-inactive-specific transcript (TSIX) expression is upregulated in spinal cord tissues following spinal cord injury (SCI). However, the role of lncRNA-TSIX in SCI remains elusive. SCI animal model was established using C57BL/6 mice. LncRNA TSIX and miR-532-3p expression were determined using quantitative reverse transcription polymerase chain reaction (qRT-PCR). Apoptosis, cell proliferation, and migration were evaluated by transferase dUTP nick end labeling staining, CCK-8, and Transwell assays, respectively. The interaction of miR-532-3p with lncRNA TSIX and DDOST was explored via a dual-luciferase reporter system. Hematoxylin-eosin staining and the Basso, Beattie, and Bresnahan locomotor rating (BBB) scale were performed to investigate SCI progression. The expression of the lncRNA TSIX was found to be significantly upregulated in the serum of SCI patients and spinal cord tissues of SCI mice. The overexpression of lncRNA TSIX enhanced spinal cord neural stem cell (SC-NSC) proliferation and migration in vitro while inhibiting apoptosis and inflammatory cell infiltration in vivo. Moreover, lncRNA TSIX acted as a molecular sponge for miR-532-3p, and the knockdown of miR-532-3p promoted proliferation and migration and inhibited apoptosis of SC-NSCs. Moreover, DDOST was found to be the downstream target of miR-532-3p, and DDOST overexpression showed a similar effect as miR-532-3p silencing on the proliferation, migration, and apoptosis of SC-NSCs. Furthermore, we found that lncRNA TSIX overexpression promoted the activation of the PI3K/AKT signaling pathway. LncRNA TSIX aggravates SCI by regulating the PI3K/AKT pathway via the miR-532-3p/DDOST axis, indicating potential applications for targeted therapy of SCI regeneration.


Assuntos
MicroRNAs , RNA Longo não Codificante , Traumatismos da Medula Espinal , Camundongos , Animais , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Camundongos Endogâmicos C57BL , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/metabolismo , Apoptose
3.
Biotechnol Genet Eng Rev ; : 1-23, 2023 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-37013868

RESUMO

Spinal cord injury (SCI) is a serious injury to the central nervous system. Previous studies have discovered that the development of SCI is associated with gene expression. The purpose of this study was to explore the significance of lncRNA TSIX in SCI and its underlying mechanism involved. An in vivo SCI mice model and an in vitro hypoxia-treated HT22 cells model were applied in this study. TSIX and SOCS3 expression in SCI tissues was measured by qRT-PCR, western blot and FISH assay. LV-sh-TSIX was injected into SCI mice intrathecally or subjected to HT22 cells to access the consequent alteration in inflammation response, cell apoptosis and functional recovery through ELISA, immunohistochemistry, TUNEL, flow cytometry assays and BMS scores. Then, the underlying mechanism of TSIX was analyzed by bioinformatics analysis and then confirmed by RIP, RNA pull-down and dual-luciferase reporter assay. It was identified that TSIX was up-regulated in HT22 cells under hypoxia operation and spinal cord tissues of SCI mice. TSIX knockdown improved the lesion size and BMS score and inhibited inflammation and cell apoptosis. MiR-30a was identified as a target for TSIX and SOCS3, and TSIX binds to miR-30a by competing with SOCS3, thereby counteracting miR-30a-mediated SOCS3 inhibition. In addition, LV-sh-TSIX effects were significantly overturned by miR-30a inhibition or SOCS3 over-expression. Knockdown of TSIX improved functional recovery and attenuated the inflammation response and cell apoptosis via miR-30a/SOCS3 axis. These results may provide a potential novel insight for SCI treatment.

4.
Bioengineered ; 2021 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-34784842

RESUMO

The identification of innovative gene biomarkers with clinical efficacy is warranted for the treatment of acute myocardial infarction (AMI). The current study sought to screen potential target genes in AMI via bioinformatic analysis and analyze their effects on cardiomyocyte apoptosis. The differentially expressed long non-coding RNAs (lncRNAs) of AMI were screened, and the downstream microRNAs (miRNAs) and mRNAs of lncRNA antisense for X-inactive-specific transcript (lncRNA TSIX) were predicted accordingly. The diagnostic relationship between the 12 differentially expressed lncRNAs and AMI was analyzed by receiver operating characteristic (ROC). Next, the expressions of 12 lncRNAs, including miR-34a-5p and retinol binding protein 2 (RBP2) were all detected. The targeting relationships of miR-34a-5p with lncRNA TSIX and RBP2 were verified. AMI model was established and treated with Ad-TSIX and/or agomiR-34a-5p to evaluate the cardiac function and cardiomyocyte apoptosis of AMI mice. LncRNA TSIX was identified as the most differentially expressed lncRNA in AMI. Our findings revealed that LncRNA TSIX could function as an AMI diagnostic marker. LncRNA TSIX could target miR-34a-5p and miR-34a-5p could target RBP2. Upregulation of lncRNA TSIX could ameliorate cardiac injury inflicted by AMI and mitigate cardiomyocyte apoptosis. Upregulation of miR-34a-5p reversed the effect of lncRNA TSIX overexpression to ameliorate cardiomyocyte apoptosis in AMI mice. Overall, the overexpression of lncRNA TSIX inhibits cardiomyocyte apoptosis by competing with RBP2 to bind to miR-34a-5p and promoting RBP2.

5.
EMBO J ; 40(24): e105862, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34786738

RESUMO

The onset of random X chromosome inactivation in mouse requires the switch from a symmetric to an asymmetric state, where the identities of the future inactive and active X chromosomes are assigned. This process is known as X chromosome choice. Here, we show that RIF1 and KAP1 are two fundamental factors for the definition of this transcriptional asymmetry. We found that at the onset of differentiation of mouse embryonic stem cells (mESCs), biallelic up-regulation of the long non-coding RNA Tsix weakens the symmetric association of RIF1 with the Xist promoter. The Xist allele maintaining the association with RIF1 goes on to up-regulate Xist RNA expression in a RIF1-dependent manner. Conversely, the promoter that loses RIF1 gains binding of KAP1, and KAP1 is required for the increase in Tsix levels preceding the choice. We propose that the mutual exclusion of Tsix and RIF1, and of RIF1 and KAP1, at the Xist promoters establish a self-sustaining loop that transforms an initially stochastic event into a stably inherited asymmetric X-chromosome state.


Assuntos
Células-Tronco Embrionárias Murinas/citologia , RNA Longo não Codificante/genética , Proteínas de Ligação a Telômeros/metabolismo , Proteína 28 com Motivo Tripartido/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Feminino , Camundongos , Regiões Promotoras Genéticas , Processos Estocásticos , Regulação para Cima , Inativação do Cromossomo X
6.
Genes Dev ; 35(13-14): 1035-1054, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34168040

RESUMO

G9a is a histone methyltransferase responsible for the dimethylation of histone H3 at lysine 9 (H3K9me2). G9a plays key roles in transcriptional silencing of developmentally regulated genes, but its role in X-chromosome inactivation (XCI) has been under debate. Here, we uncover a female-specific function of G9a and demonstrate that deleting G9a has a disproportionate impact on the X chromosome relative to the rest of the genome. G9a deficiency causes a failure of XCI and female-specific hypersensitivity to drug inhibition of H3K9me2. We show that G9a interacts with Tsix and Xist RNAs, and that competitive inhibition of the G9a-RNA interaction recapitulates the XCI defect. During XCI, Xist recruits G9a to silence X-linked genes on the future inactive X. In parallel on the future Xa, Tsix recruits G9a to silence Xist in cis Thus, RNA tethers G9a for allele-specific targeting of the H3K9me2 modification and the G9a-RNA interaction is essential for XCI.


Assuntos
Cromossomos Humanos X , Antígenos de Histocompatibilidade/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Metiltransferases , RNA Longo não Codificante , Feminino , Histonas/metabolismo , Humanos , Metiltransferases/genética , RNA Longo não Codificante/genética , Inativação do Cromossomo X/genética
7.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34161282

RESUMO

Mammalian cells equalize X-linked dosages between the male (XY) and female (XX) sexes by silencing one X chromosome in the female sex. This process, known as "X chromosome inactivation" (XCI), requires a master switch within the X inactivation center (Xic). The Xic spans several hundred kilobases in the mouse and includes a number of regulatory noncoding genes that produce functional transcripts. Over three decades, transgenic and deletional analyses have demonstrated both the necessity and sufficiency of the Xic to induce XCI, including the steps of X chromosome counting, choice, and initiation of whole-chromosome silencing. One recent study, however, reported that deleting the noncoding sequences of the Xic surprisingly had no effect for XCI and attributed a sufficiency to drive counting to the coding gene, Rnf12/Rlim Here, we revisit the question by creating independent Xic deletion cell lines. Multiple independent clones carrying heterozygous deletions of the Xic display an inability to up-regulate Xist expression, consistent with a counting defect. This defect is rescued by a second site mutation in Tsix occurring in trans, bypassing the defect in counting. These findings reaffirm the essential nature of noncoding Xic elements for the initiation of XCI.


Assuntos
Deleção de Sequência , Inativação do Cromossomo X/genética , Alelos , Animais , Morte Celular , Linhagem Celular , Feminino , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Deleção de Sequência/genética
8.
Cell Rep ; 34(13): 108912, 2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33789104

RESUMO

The fine-scale dynamics from euchromatin (EC) to facultative heterochromatin (fHC) has remained largely unclear. Here, we focus on Xist and its silencing initiator Tsix as a paradigm of transcription-mediated conversion from EC to fHC. In mouse epiblast stem cells, induction of Tsix recapitulates the conversion at the Xist promoter. Investigating the dynamics reveals that the conversion proceeds in a stepwise manner. Initially, a transient opened chromatin structure is observed. In the second step, gene silencing is initiated and dependent on Tsix, which is reversible and accompanied by simultaneous changes in multiple histone modifications. At the last step, maintenance of silencing becomes independent of Tsix and irreversible, which correlates with occupation of the -1 position of the transcription start site by a nucleosome and initiation of DNA methylation introduction. This study highlights the hierarchy of multiple chromatin events upon stepwise gene silencing establishment.


Assuntos
Eucromatina/metabolismo , Heterocromatina/metabolismo , Regiões Promotoras Genéticas , RNA Longo não Codificante/genética , Transcrição Gênica , Animais , Fator de Ligação a CCCTC/metabolismo , Metilação de DNA/genética , Epigênese Genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Inativação Gênica , Camadas Germinativas/citologia , Histonas/metabolismo , Camundongos , Nucleossomos/metabolismo , Processamento de Proteína Pós-Traducional , RNA Longo não Codificante/metabolismo , Células-Tronco/metabolismo , Fator de Transcrição YY1/metabolismo
9.
Cell Rep ; 27(2): 442-454.e5, 2019 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-30970248

RESUMO

Neural tube defects (NTDs) are common birth defects in humans and show an unexplained female bias. Female mice lacking the tumor suppressor p53 display NTDs with incomplete penetrance. We found that the combined loss of pro-apoptotic BIM and p53 caused 100% penetrant, female-exclusive NTDs, which allowed us to investigate the female-specific functions of p53. We report that female p53-/- embryonic neural tube samples show fewer cells with inactive X chromosome markers Xist and H3K27me3 and a concomitant increase in biallelic expression of the X-linked genes, Huwe1 and Usp9x. Decreased Xist and increased X-linked gene expression was confirmed by RNA sequencing. Moreover, we found that p53 directly bound response elements in the X chromosome inactivation center (XIC). Together, these findings suggest p53 directly activates XIC genes, without which there is stochastic failure in X chromosome inactivation, and that X chromosome inactivation failure may underlie the female bias in neural tube closure defects.


Assuntos
Defeitos do Tubo Neural/genética , Proteína Supressora de Tumor p53/deficiência , Animais , Células-Tronco Embrionárias/patologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Defeitos do Tubo Neural/patologia , Gravidez , Processos Estocásticos , Proteína Supressora de Tumor p53/genética , Inativação do Cromossomo X
10.
Front Oncol ; 9: 1459, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31998636

RESUMO

Escaping antitumor immunity is a hallmark in cancer progression. Programmed cell death protein 1 (PD-1) is an immune checkpoint receptor responsible for the maintenance of immune tolerance; PD-1 ligand (PD-L1) is overexpressed in tumor cells, simplifying their escape from the immune system through T-cell function suppression. Notwithstanding that cancer antigen (CA)125, carcinoembryonic antigen (CEA), CA15-3, and alpha-fetoprotein (AFP) are among conventional breast cancer diagnostic biomarkers, their lack of sensitivity and specificity resides among their major limitations. Furthermore, human epidermal growth factor receptor (HER)2 and interleukin (IL)-6-demonstrated as breast cancer immune biomarkers-still possess limitations, for instance, technical detection problems and stability problems, which necessitate the discovery of novel, stable non-invasive cancer immune biomarkers. XIST and TSIX are two long non-coding (lnc)RNAs possessing a role in X chromosome inactivation (XCI) as well as in breast cancer (BC). In the present study, they were investigated as stable non-invasive breast cancer immune biomarkers. The study demonstrated that PD-L1 was overexpressed in the different molecular subtypes of breast cancer patients as well as in MDA-MB-231 cells. Furthermore, lncRNAs XIST and TSIX were markedly increased in the tissues, lymph nodes, and different body fluids of breast cancer patients compared to controls. In addition, XIST and TSIX were differentially expressed in subtypes of BC patients, and their levels were correlated to PD-L1 expression level. In conclusion, this correlative study has shed light on the role of both lncRNAs XIST and TSIX as potential non-invasive BC immune biomarkers reflecting the evaded immune system of the patient and overcoming the instability problem of common BC biomarkers.

11.
Proc Natl Acad Sci U S A ; 115(32): 8185-8190, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30038001

RESUMO

Rett syndrome (RTT) is a severe neurodevelopmental disorder caused by a mutation in the X-linked methyl-CpG-binding protein 2 (MECP2). There is currently no disease-specific treatment, but MECP2 restoration through reactivation of the inactive X (Xi) has been of considerable interest. Progress toward an Xi-reactivation therapy has been hampered by a lack of suitable female mouse models. Because of cellular mosaicism due to random X-chromosome inactivation (XCI), Mecp2+/- heterozygous females develop only mild RTT. Here, we create an improved female mouse model by introducing a mutation in Tsix, the antisense regulator of XCI allelic choice. Tsix-Mecp2 mice show reduced MECP2 mosaicism and closely phenocopy the severely affected Mecp2-null males. Tsix-Mecp2 females demonstrate shortened lifespan, motor weakness, tremors, and gait disturbance. Intriguingly, they also exhibit repetitive behaviors, as is often seen in human RTT, including excessive grooming and biting that result in self-injury. With a Tsix allelic series, we vary MECP2 levels in brain and demonstrate a direct, but nonlinear correlation between MECP2 levels and phenotypic improvement. As little as 5-10% MECP2 restoration improves neuromotor function and extends lifespan five- to eightfold. Our study thus guides future pharmacological strategies and suggests that partial MECP2 restoration could have disproportionate therapeutic benefit.


Assuntos
Modelos Animais de Doenças , Genes Ligados ao Cromossomo X/genética , Longevidade/genética , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , RNA Longo não Codificante/genética , Síndrome de Rett/genética , Alelos , Animais , Comportamento Animal , Encéfalo/patologia , Feminino , Heterozigoto , Humanos , Masculino , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mosaicismo , Atividade Motora/genética , Mutação , Fenótipo , RNA Longo não Codificante/metabolismo , Síndrome de Rett/mortalidade , Síndrome de Rett/patologia , Inativação do Cromossomo X
12.
Connect Tissue Res ; 59(6): 534-541, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29260905

RESUMO

OBJECTIVE: This study aims to investigate the role of TSIX/miR-30a-5p axis in particle-induced osteolysis (PIO). METHOD: PIO mouse model was established by the implantation of Co-Cr-Mo metal particles (CoPs). MC3T3-E1 cells received CoPs stimulation. Bone mineral density (BMD) in the skull was detected to evaluate PIO development. The expression of TSIX and miR-30a-5p was detected by using qRT-PCR. Osteoblast apoptosis was measured using flow cytometry. RNA pull-down was used to verify the regulatory relationship between TSIX and miR-30a-5p. RESULT: The results showed that BMD of the skull in PIO mice was significantly decreased compared with control mice, which indicated that the PIO model was established successfully. Moreover, CoPs could up-regulate TSIX level, down-regulate miR-30a-5p expression, and promote osteoblast apoptosis in vivo and in vitro. The results also found that TSIX negatively regulated miR-30a-5p expression, and knockdown of TSIX inhibited Runx2 expression. As expected, miR-30a-5p inhibitor could reverse the inhibition of si-TSIX on osteoblast apoptosis. CONCLUSION: TSIX played a pivotal role in PIO development by negatively regulating miR-30a-5p.


Assuntos
Apoptose/efeitos dos fármacos , MicroRNAs/metabolismo , Osteoblastos/metabolismo , Osteólise , RNA Longo não Codificante/metabolismo , Vitálio/toxicidade , Animais , Densidade Óssea/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , Osteoblastos/patologia , Osteólise/induzido quimicamente , Osteólise/genética , Osteólise/metabolismo , Osteólise/patologia , RNA Longo não Codificante/genética , Crânio/metabolismo , Crânio/patologia
13.
Reprod Sci ; 25(2): 246-255, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28587571

RESUMO

The objective of this study was to identify the expression profile of long noncoding RNAs (lncRNAs) with concurrent assessment of microRNA (miRNA) and messenger RNA (mRNA) profiles in leiomyomas and paired myometrium using next-generation RNA sequencing and assembly of RNA transcripts. Total RNA was isolated from leiomyoma and paired myometrium (N = 8) and samples from 3 pairs were subjected to RNA sequencing. Normalized assembly of over 48 000 lncRNAs resulted in identification of 45 936 lncRNAs. Of these lncRNAs, 22 148 representing overlapping, intergenic, intronic, and antisense subtypes were expressed in all paired tissues, with 5941 (2813 up- and 3128 downregulated at ≥1.5 fold) differentially expressed in leiomyomas. Concurrent RNA sequencing revealed the expression of 2588 miRNAs and 21 814 mRNAs, of which 392 miRNAs and 16 559 mRNAs were expressed in all paired tissues. Of these transcripts, 56 and 92 miRNAs and 2030 and 1825 mRNAs were up- or downregulated at ≥1.5 fold, respectively, in leiomyoma as compared to myometrium. Using quantitative reverse transcription-polymerase chain reaction (QRT-PCR), we confirmed the expression of hepatocellular carcinoma upregulated (HULC), lnc-maternally expressed 3 (MEG3), long intergenic ncRNA 890 (LINC00890), TSIX, long intergenic ncRNA 473 (LINC00473), lnc-KLF9-1, and lnc-POTEM-3 (lncRNA-ATB) in leiomyoma and matched myometrium (N = 8). Collectively, the results presented here provide a comprehensive expression profile of lncRNAs in leiomyomas with concurrent integrated expression of miRNAs and mRNAs and implicate potential regulatory functions of lncRNAs through interactions with specific miRNAs and mRNAs which are known to be critical in the pathogenesis of leiomyoma.


Assuntos
Leiomioma/genética , MicroRNAs/genética , RNA Longo não Codificante/genética , Neoplasias Uterinas/genética , Adulto , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Leiomioma/metabolismo , Leiomioma/patologia , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Miométrio/metabolismo , Miométrio/patologia , RNA Longo não Codificante/metabolismo , RNA Mensageiro/metabolismo , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patologia
14.
Dev Cell ; 43(3): 359-371.e6, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-29107559

RESUMO

X-chromosome inactivation (XCI) silences one X chromosome in the female mammal and is essential to peri-implantation development. XCI is thought to be cell autonomous, with all factors required being produced within each cell. Nevertheless, external cues may exist. Here, we search for such developmental signals by combining bioinformatic, biochemical, and genetic approaches. Using ex vivo and in vivo models, we identify the Hedgehog (HH) paracrine system as a candidate signaling cascade. HH signaling keeps XCI in check in pluripotent cells and is transduced by GLI transcription factors to binding sites in Tsix, the antisense repressor of XCI. GLI potentiates Tsix expression and impedes XCI. In vivo, mutating Indian Hedgehog results in a sex ratio bias against females, and the female lethality is rescued by a second-site mutation in Tsix. These data demonstrate a genetic and functional intersection between HH and XCI and support a role for intercellular signaling during XCI.


Assuntos
Proteínas Hedgehog/metabolismo , Regiões Promotoras Genéticas/genética , RNA Longo não Codificante/genética , Inativação do Cromossomo X/genética , Animais , Diferenciação Celular/fisiologia , Feminino , Camundongos Knockout , Fatores de Transcrição/metabolismo , Transcrição Gênica/genética
15.
Mol Cytogenet ; 9: 56, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27504142

RESUMO

BACKGROUND: Chromosome counting is a process in which cells determine somehow their intrinsic chromosome number(s). The best-studied cellular mechanism that involves chromosome counting is 'chromosome-kissing' and X-chromosome inactivation (XCI) mechanism. It is necessary for the well-known dosage compensation between the genders in mammals to balance the number of active X-chromosomes (Xa) with regard to diploid set of autosomes. At the onset of XCI, two X-chromosomes are coming in close proximity and pair physically by a specific segment denominated X-pairing region (Xpr) that involves the SLC16A2 gene. RESULTS: An Ensembl BLAST search for human and mouse SLC16A2/Slc16a2 homologues revealed, that highly similar sequences can be found at almost each chromosome in the corresponding genomes. Additionally, a BLAST search for SLC16A2/TSIX/XIST (genes responsible for XCI) reveled that "SLC16A2/TSIX/XIST like sequences" cover equally all chromosomes, too. With respect to this we provide following hypotheses. HYPOTHESES: If a single genomic region containing the SLC16A2 gene on X-chromosome is responsible for maintaining "balanced" active copy numbers, it is possible that similar sequences or gene/s have the same function on other chromosomes (autosomes). SLC16A2 like sequences on autosomes could encompass evolutionary older, but functionally active key regions for chromosome counting in early embryogenesis. Also SLC16A2 like sequence on autosomes could be involved in inappropriate chromosomes pairing and, thereby be involved in aneuploidy formation during embryogenesis and cancer development. Also, "SLC16A2/TSIX/XIST gene like sequence combinations" covering the whole genome, could be important for the determination of X:autosome ratio in cells and chromosome counting. CONCLUSIONS: SLC16A2 and/or SLC16A2/TSIX/XIST like sequence dispersed across autosomes and X-chromosome(s) could serve as bases for a counting mechanism to determine X:autosome ratio and could potentially be a mechanism by which a cell also counts its autosomes. It could also be that such specific genomic regions have the same function for each specific autosome. As errors during the obviously existing process of chromosome counting are one if not the major origin of germline/somatic aneuploidy the here presented hypotheses should further elaborated and experimentally tested.

16.
Semin Cell Dev Biol ; 56: 3-8, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27223409

RESUMO

In early female embryos X-chromosome inactivation occurs concomitant with up regulation of the non-coding RNA, Xist, on the future inactive X-chromosome. Up regulation of Xist and coating of the future inactive X is sufficient to induce silencing. Therefore unlocking the mechanisms of X-chromosome inactivation requires thorough understanding of the transcriptional regulators, both activators and repressors, which control Xist. Mouse pluripotent embryonic stem cells, which have two active X chromosomes, provide a tractable ex vivo model system for studying X-chromosome inactivation, since this process is triggered by differentiation signals in these cultured cells. Yet there are significant discrepancies found between ex vivo analyses in mouse embryonic stem cells and in vivo studies of early embryos. In this review we elaborate on potential models of how Xist is up regulated on a single X chromosome in female cells and how ex vivo and in vivo analyses enlighten our understanding of the activators and repressors that control this non-coding RNA gene.


Assuntos
Inativação do Cromossomo X/genética , Animais , Humanos , Modelos Genéticos , Células-Tronco Pluripotentes/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética
17.
Proc Natl Acad Sci U S A ; 113(3): E309-18, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26739568

RESUMO

X-inactive specific transcript (Xist) long noncoding RNA (lncRNA) is thought to catalyze silencing of X-linked genes in cis during X-chromosome inactivation, which equalizes X-linked gene dosage between male and female mammals. To test the impact of Xist RNA on X-linked gene silencing, we ectopically induced endogenous Xist by ablating the antisense repressor Tsix in mice. We find that ectopic Xist RNA induction and subsequent X-linked gene silencing is sex specific in embryos and in differentiating embryonic stem cells (ESCs) and epiblast stem cells (EpiSCs). A higher frequency of X(ΔTsix)Y male cells displayed ectopic Xist RNA coating compared with X(ΔTsix)X female cells. This increase reflected the inability of X(ΔTsix)Y cells to efficiently silence X-linked genes compared with X(ΔTsix)X cells, despite equivalent Xist RNA induction and coating. Silencing of genes on both Xs resulted in significantly reduced proliferation and increased cell death in X(ΔTsix)X female cells relative to X(ΔTsix)Y male cells. Thus, whereas Xist RNA can inactivate the X chromosome in females it may not do so in males. We further found comparable silencing in differentiating X(ΔTsix)Y and 39,X(ΔTsix) (X(ΔTsix)O) ESCs, excluding the Y chromosome and instead implicating the X-chromosome dose as the source of the sex-specific differences. Because X(ΔTsix)X female embryonic epiblast cells and EpiSCs harbor an inactivated X chromosome prior to ectopic inactivation of the active X(ΔTsix) X chromosome, we propose that the increased expression of one or more X-inactivation escapees activates Xist and, separately, helps trigger X-linked gene silencing.


Assuntos
Inativação Gênica , Genes Ligados ao Cromossomo X , RNA Longo não Codificante/genética , Caracteres Sexuais , Animais , Diferenciação Celular/genética , Implantação do Embrião , Embrião de Mamíferos/metabolismo , Feminino , Camadas Germinativas/citologia , Masculino , Camundongos , Modelos Biológicos , Células-Tronco Embrionárias Murinas/metabolismo , RNA Longo não Codificante/metabolismo , Inativação do Cromossomo X/genética , Cromossomo Y/genética
18.
Adv Exp Med Biol ; 886: 33-49, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26659486

RESUMO

Genetic sex determination in mammals relies on dimorphic sex chromosomes that confer phenotypic/physiologic differences between males and females. In this heterogametic system, X and Y chromosomes diverged from an ancestral pair of autosomes, creating a genetic disequilibrium between XX females and XY males. Dosage compensation mechanisms alleviate intrinsic gene dosage imbalance, leading to equal expression levels of most X-linked genes in the two sexes. In therian mammals, this is achieved through inactivation of one of the two X chromosomes in females. Failure to undergo X-chromosome inactivation (XCI) results in developmental arrest and death. Although fundamental for survival, a surprising loose conservation in the mechanisms to achieve XCI during development in therian lineage has been, and continues, to be uncovered. XCI involves the concerted action of non-coding RNAs (ncRNAs), including the well-known Xist RNA, and has thus become a classical paradigm to study the mode of action of this particular class of transcripts. In this chapter, we will describe the processes coping with sex chromosome genetic imbalance and how ncRNAs underlie dosage compensation mechanisms and influence male-female differences in mammals. Moreover, we will discuss how ncRNAs have been tinkered with during therian evolution to adapt XCI mechanistic to species-specific constraints.


Assuntos
Cromossomos Humanos X/metabolismo , Cromossomos Humanos Y/metabolismo , Mecanismo Genético de Compensação de Dose/fisiologia , RNA Longo não Codificante/metabolismo , Processos de Determinação Sexual/fisiologia , Animais , Cromossomos Humanos X/genética , Cromossomos Humanos Y/genética , Feminino , Humanos , Masculino , RNA Longo não Codificante/genética
19.
RNA Biol ; 11(7): 798-807, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25137047

RESUMO

X-chromosome inactivation (XCI) in female mammals is a dramatic example of epigenetic gene regulation, which entails the silencing of an entire chromosome through a wide range of mechanisms involving noncoding RNAs, chromatin-modifications, and DNA-methylation. While XCI is associated with the differentiated cell state, it is reversed by X-chromosome reactivation (XCR) ex vivo in pluripotent stem cells and in vivo in the early mouse embryo and the germline. Critical in the regulation of XCI vs. XCR is the X-inactivation center, a multigene locus on the X-chromosome harboring several long noncoding RNA genes including, most prominently, Xist and Tsix. These genes, which sit at the top of the XCI hierarchy, are by themselves controlled by pluripotency factors, coupling XCR with the naïve pluripotent stem cell state. In this point-of-view article we review the latest findings regarding this intricate relationship between cell differentiation state and epigenetic control of the X-chromosome. In particular, we discuss the emerging picture of complex multifactorial regulatory mechanisms, ensuring both a fine-tuned and robust X-reactivation process.


Assuntos
Células-Tronco Pluripotentes/metabolismo , RNA Longo não Codificante/metabolismo , Fatores de Transcrição/metabolismo , Inativação do Cromossomo X , Animais , Diferenciação Celular , Feminino , Humanos , Camundongos , Cromossomo X/genética
20.
Transcription ; 1(1): 46-56, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21327163

RESUMO

X-chromosome inactivation balances X-chromosome dosages in male and female mammals by transcriptionally repressing one X in the female sex. Proper counting and the mutually exclusive choice of active X and inactive X have been hypothesized to involve X-chromosome crosstalk via homologous chromosome pairing. Transient pairing of two female Xs requires noncoding Tsix and Xite. A recent study suggested a new pairing element (Xpr), located ~200 kb upstream of Xist, in the Xpct region. Xpr is proposed to induce pairing and activate Xist expression. Here, we further characterize Xpr and find that the Xpr sequence is unstable when introduced as transgenes into male ES cells. Xpr transgenes show an unusual tendency to disperse throughout the nucleus. However, we observe neither pairing between Xpr alleles nor ectopic Xist expression. In the absence of Tsix, Xpr does not induce inter-allelic Xic interactions. Female ES cells carrying Xpr transgenes are more stable. Nonetheless, pairing also does not seem to occur in female cells. We conclude that, while Xpr contains unusual properties, it most likely does not serve as a pairing or counting element. Differences in statistical methods and controls may explain some of the discrepancies.


Assuntos
Proteínas de Membrana Transportadoras/genética , RNA não Traduzido/genética , Inativação do Cromossomo X , Animais , Linhagem Celular , Células-Tronco Embrionárias , Feminino , Masculino , Camundongos , Transportadores de Ácidos Monocarboxílicos , RNA Longo não Codificante , Simportadores , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA