Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Mol Cell Biol ; 33(24): 5005-20, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24144980

ABSTRACT

X chromosome inactivation is a remarkable example of chromosome-wide gene silencing and facultative heterochromatin formation. Numerous histone posttranslational modifications, including H3K9me2 and H3K27me3, accompany this process, although our understanding of the enzymes that lay down these marks and the factors that bind to them is still incomplete. Here we identify Cdyl, a chromodomain-containing transcriptional corepressor, as a new chromatin-associated protein partner of the inactive X chromosome (Xi). Using mouse embryonic stem cell lines with mutated histone methyltransferase activities, we show that Cdyl relies on H3K9me2 for its general association with chromatin in vivo. For its association with Xi, Cdyl requires the process of differentiation and the presence of H3K9me2 and H3K27me3, which both become chromosomally enriched following Xist RNA coating. We further show that the removal of the PRC2 component Eed and subsequent loss of H3K27me3 lead to a reduction of both Cdyl and H3K9me2 enrichment on inactive Xi. Finally, we show that Cdyl associates with the H3K9 histone methyltransferase G9a and the MGA protein, both of which are also found on Xi. We propose that the combination of H3K9me2 and H3K27me3 recruits Cdyl to Xi, and this, in turn, may facilitate propagation of the H3K9me2 mark by anchoring G9a.


Subject(s)
Embryonic Stem Cells/physiology , Histones/metabolism , Proteins/metabolism , X Chromosome Inactivation , X Chromosome/metabolism , Animals , Cell Differentiation , Cell Nucleus/metabolism , Co-Repressor Proteins , Epigenesis, Genetic , Female , Gene Expression Regulation, Developmental , HEK293 Cells , Histone Acetyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Humans , Hydro-Lyases , Methylation , Mice , Polycomb Repressive Complex 1/metabolism , Protein Isoforms/metabolism , Protein Processing, Post-Translational , Protein Stability , Tretinoin/physiology , X Chromosome/genetics
2.
Article in English | MEDLINE | ID: mdl-21447823

ABSTRACT

Early development of female mammals is accompanied by transcriptional inactivation of one of their two X chromosomes. This leads to monoallelic expression of most of the X chromosome and ensures dosage compensation with respect to males (XY). One of the most surprising aspects of this phenomenon is that the two X homologs are treated differently even though they are present within the same nucleus. In eutherian mammals, such as humans and mice, either the maternal or the paternal X is inactivated during early embryogenesis. Once set up, the silent state is epigenetically transmitted as cells divide, so that adult females are mosaics of clonal cell populations, which express either of their two X chromosomes. The past years have been marked by the discovery of several molecular events that accompany chromosome-wide silencing.


Subject(s)
Chromatin/chemistry , X Chromosome Inactivation/genetics , Animals , Gene Silencing , Humans , Nuclear Proteins/metabolism , RNA, Long Noncoding , RNA, Untranslated , Transcription, Genetic
3.
IEEE Trans Image Process ; 17(4): 493-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18390358

ABSTRACT

We present an intensity-based nonrigid registration approach for the normalization of 3-D multichannel microscopy images of cell nuclei. A main problem with cell nuclei images is that the intensity structure of different nuclei differs very much; thus, an intensity-based registration scheme cannot be used directly. Instead, we first perform a segmentation of the images from the cell nucleus channel, smooth the resulting images by a Gaussian filter, and then apply an intensity-based registration algorithm. The obtained transformation is applied to the images from the nucleus channel as well as to the images from the other channels. To improve the convergence rate of the algorithm, we propose an adaptive step length optimization scheme and also employ a multiresolution scheme. Our approach has been successfully applied using 2-D cell-like synthetic images, 3-D phantom images as well as 3-D multichannel microscopy images representing different chromosome territories and gene regions. We also describe an extension of our approach, which is applied for the registration of 3D + t (4-D) image series of moving cell nuclei.


Subject(s)
Cell Nucleus/ultrastructure , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Microscopy/methods , Pattern Recognition, Automated/methods , Subtraction Technique , Algorithms , Numerical Analysis, Computer-Assisted , Reproducibility of Results , Sensitivity and Specificity , Signal Processing, Computer-Assisted
4.
Science ; 318(5856): 1632-6, 2007 Dec 07.
Article in English | MEDLINE | ID: mdl-18063799

ABSTRACT

Mammalian dosage compensation involves silencing of one of the two X chromosomes in females and is controlled by the X-inactivation center (Xic). The Xic, which includes Xist and its antisense transcription unit Tsix/Xite, somehow senses the number of X chromosomes and triggers Xist up-regulation from one of the two X chromosomes in females. We found that a segment of the mouse Xic lying several hundred kilobases upstream of Xist brings the two Xics together before the onset of X inactivation. This region can autonomously drive Xic trans-interactions even as an ectopic single-copy transgene. Its introduction into male embryonic stem cells is strongly selected against, consistent with a possible role in trans-activating Xist. We propose that homologous associations driven by this novel X-pairing region (Xpr) of the Xic enable a cell to sense that more than one X chromosome is present and coordinate reciprocal Xist/Tsix expression.


Subject(s)
Chromosome Pairing , X Chromosome Inactivation , X Chromosome/genetics , Alleles , Animals , Cell Differentiation , Cell Line , Chromosomes, Artificial, Bacterial , Down-Regulation , Embryonic Stem Cells , Female , Mice , Mice, Transgenic , RNA, Long Noncoding , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , S Phase , Transfection , Transgenes , Up-Regulation , X Chromosome/physiology
5.
Cytogenet Genome Res ; 113(1-4): 318-24, 2006.
Article in English | MEDLINE | ID: mdl-16575196

ABSTRACT

In the mouse, there are two forms of X chromosome inactivation (XCI), random XCI in the fetus and imprinted paternal XCI, which is limited to the extraembryonic tissues. While the mechanism of random XCI has been studied extensively using the in vitro XX ES cell differentiation system, imprinted XCI during early embryonic development has been less well characterized. Recent studies of early embryos have reported unexpected findings for the paternal X chromosome (Xp). Imprinted XCI may not be linked to meiotic silencing in the male germ line but rather to the imprinted status of the Xist gene. Furthermore, the Xp becomes inactivated in all cells of cleavage-stage embryos and then reactivated in the cells of the inner cell mass (ICM) that form the epiblast, where random XCI ensues.


Subject(s)
Blastula/physiology , Gene Silencing , X Chromosome , Animals , Female , Gene Dosage , Gene Expression Regulation , Kinetics , Male , Mice , Transcription, Genetic
6.
Article in English | MEDLINE | ID: mdl-17381324

ABSTRACT

In female mammals, one of the two X chromosomes is converted from the active euchromatic state into inactive heterochromatin during early embryonic development. This process, known as X-chromosome inactivation, results in the transcriptional silencing of over a thousand genes and ensures dosage compensation between the sexes. Here, we discuss the possible mechanisms of action of the Xist transcript, a remarkable noncoding RNA that triggers the X-inactivation process and also seems to participate in setting up the epigenetic marks that provide the cellular memory of the inactive state. So far, no functional protein partners have been identified for Xist RNA, but different lines of evidence suggest that it may act at multiple levels, including nuclear compartmentalization, chromatin modulation, and recruitment of Polycomb group proteins.


Subject(s)
Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , X Chromosome Inactivation/genetics , X Chromosome Inactivation/physiology , Animals , Cell Compartmentation , Cell Nucleus/metabolism , Female , Gene Expression Regulation, Developmental , Mice , Models, Biological , Polycomb-Group Proteins , RNA, Long Noncoding , RNA, Untranslated/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sex Chromatin/genetics , Sex Chromatin/metabolism
8.
Cytogenet Genome Res ; 99(1-4): 75-84, 2002.
Article in English | MEDLINE | ID: mdl-12900548

ABSTRACT

Inactivation of the X chromosome during early female development and the subsequent maintenance of this transcriptionally inert state through countless cell divisions remain a paradigm for epigenetic regulation in mammals. Nevertheless, the exact mechanisms underlying this chromosome-wide silencing process remain unclear. Using differentiating female embryonic stem (ES) cells as a model system, we recently found that histone H3 tail modifications are among the earliest known chromatin changes in the X inactivation process, appearing as soon as Xist RNA accumulates on the X chromosome, but prior to transcriptional silencing of X-linked genes (Heard et al., 2001). In this report we present an integrated analysis of the sequence of early events and chromatin modifications underlying X inactivation in differentiating female ES cells. We have extended our previous analysis concerning changes in histone tail modification states. We find that the hypomethylation of Arg-17 and that of Lys-36 on histone H3 also characterize the inactive X chromosome, and that these profiles show a similarly early onset during the initiation of X inactivation. In addition, we have investigated the kinetics of the shift in replication timing of the X chromosome undergoing inactivation. This event occurs slightly later than Xist RNA coating and the chromatin modifications. Finally, from an early stage in the X inactivation process, characteristic histone modification patterns can be found on the X chromosome at mitosis, suggesting that they represent true epigenetic marks of the inactive state.


Subject(s)
Dosage Compensation, Genetic , Embryo, Mammalian/metabolism , Stem Cells/metabolism , X Chromosome/genetics , Acetylation , Animals , Cell Differentiation/genetics , Cell Line , Embryo, Mammalian/cytology , Female , Histones/metabolism , In Situ Hybridization, Fluorescence , Interphase/genetics , Kinetics , Mice , Mitosis/genetics , RNA, Long Noncoding , RNA, Untranslated/genetics , Stem Cells/cytology , Time Factors , X Chromosome/metabolism
9.
Cell ; 107(6): 727-38, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11747809

ABSTRACT

Coating of the X chromosome by Xist RNA is an essential trigger for X inactivation. However, little is known about the early chromatin remodeling events that transform this signal into transcriptional silencing. Here we report that methylation of histone H3 lysine 9 on the inactive X chromosome occurs immediately after Xist RNA coating and before transcriptional inactivation of X-linked genes. X-chromosomal H3 Lys-9 methylation occurs during the same window of time as H3 Lys-9 hypoacetylation and H3 Lys-4 hypomethylation. Histone H3 modifications thus represent the earliest known chromatin changes during X inactivation. We also identify a unique "hotspot" of H3 Lys-9 methylation 5' to Xist, and we propose that this acts as a nucleation center for Xist RNA-dependent spread of inactivation along the X chromosome via H3 Lys-9 methylation.


Subject(s)
Cell Differentiation/physiology , Chromatin/metabolism , Chromosomal Proteins, Non-Histone , Dosage Compensation, Genetic , Histones/metabolism , Proto-Oncogene Proteins , Repressor Proteins , X Chromosome/metabolism , A Kinase Anchor Proteins , Acetylation , Adaptor Proteins, Signal Transducing , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27 , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/metabolism , Female , Fibroblasts/physiology , Gene Silencing , In Situ Hybridization, Fluorescence , Male , Methyl-CpG-Binding Protein 2 , Methylation , Mice , Minor Histocompatibility Antigens , Models, Biological , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , RNA/metabolism , RNA, Long Noncoding , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Stem Cells/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
10.
Arch Otolaryngol Head Neck Surg ; 127(8): 970-4, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11493208

ABSTRACT

OBJECTIVE: To evaluate the feasibility and predictive ability of the sentinel node localization technique for patients with squamous cell carcinoma of the oral cavity or oropharynx and clinically negative necks. DESIGN: Prospective, efficacy study comparing the histopathologic status of the sentinel node with that of the remaining neck dissection specimen. SETTING: Tertiary referral center. PATIENTS: Patients with T1 or T2 disease and clinically negative necks were eligible for the study. Nine previously untreated patients with oral cavity or oropharyngeal squamous cell carcinoma were enrolled in the study. INTERVENTIONS: Unfiltered technetium Tc 99m sulfur colloid injections of the primary tumor and lymphoscintigraphy were performed on the day before surgery. Intraoperatively, the sentinel node(s) was localized with a gamma probe and removed after tumor resection and before neck dissection. MAIN OUTCOME MEASURES: The primary outcome was the negative predictive value of the histopathologic status of the sentinel node for predicting cervical metastases. RESULTS: Sentinel nodes were identified in 9 previously untreated patients. In 5 patients, there were no positive nodes. In 4 patients, the sentinel nodes were the only histopathologically positive nodes. In previously untreated patients, the sentinel node technique had a negative predictive value of 100% for cervical metastasis. CONCLUSIONS: Our preliminary investigation shows that sentinel node localization is technically feasible in head and neck surgery and is predictive of cervical metastasis. The sentinel node technique has the potential to decrease the number of neck dissections performed in clinically negative necks, thus reducing the associated morbidity for patients in this group.


Subject(s)
Carcinoma, Squamous Cell/secondary , Lymph Nodes/diagnostic imaging , Mouth Neoplasms/pathology , Oropharyngeal Neoplasms/pathology , Sentinel Lymph Node Biopsy , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/surgery , Feasibility Studies , Female , Humans , Lymph Node Excision , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Middle Aged , Mouth Neoplasms/surgery , Neck , Oropharyngeal Neoplasms/surgery , Predictive Value of Tests , Prospective Studies , Radionuclide Imaging , Radiopharmaceuticals , Technetium Tc 99m Sulfur Colloid
11.
Cytogenet Cell Genet ; 92(1-2): 59-62, 2001.
Article in English | MEDLINE | ID: mdl-11306797

ABSTRACT

Image restoration approaches, such as digital deconvolution, are becoming widely used for improving the quality of microscopic images. However, no quantification of the gain in resolution of fluorescence images is available. We show that, after iterative constrained deconvolution, fluorescent cosmid signals appear to be 25% smaller, and 1.2-kb fragment signals on combed molecules faithfully display the expected length.


Subject(s)
DNA, Fungal/metabolism , In Situ Hybridization, Fluorescence/methods , Chromosomes, Artificial, Yeast/genetics , Chromosomes, Artificial, Yeast/metabolism , Cosmids/genetics , DNA Probes/genetics , DNA, Fungal/genetics , Fibroblasts , Fluorescence , Humans , Sensitivity and Specificity , Yeasts/cytology , Yeasts/genetics
12.
Nat Rev Genet ; 2(1): 59-67, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11253071

ABSTRACT

In many sexually dimorphic species, a mechanism is required to ensure equivalent levels of gene expression from the sex chromosomes. In mammals, such dosage compensation is achieved by X-chromosome inactivation, a process that presents a unique medley of biological puzzles: how to silence one but not the other X chromosome in the same nucleus; how to count the number of X's and keep only one active; how to choose which X chromosome is inactivated; and how to establish this silent state rapidly and efficiently during early development. The key to most of these puzzles lies in a unique locus, the X-inactivation centre and a remarkable RNA--Xist--that it encodes.


Subject(s)
Dosage Compensation, Genetic , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , X Chromosome , Animals , Chromosome Mapping , Drosophila melanogaster/genetics , Female , Male , RNA/genetics , RNA, Long Noncoding
14.
Mol Cell Biol ; 19(12): 8513-25, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10567576

ABSTRACT

X inactivation in female mammals is controlled by a key locus on the X chromosome, the X-inactivation center (Xic). The Xic controls the initiation and propagation of inactivation in cis. It also ensures that the correct number of X chromosomes undergo inactivation (counting) and determines which X chromosome becomes inactivated (choice). The Xist gene maps to the Xic region and is essential for the initiation of X inactivation in cis. Regulatory elements of X inactivation have been proposed to lie 3' to Xist. One such element, lying 15 kb downstream of Xist, is the DXPas34 locus, which was first identified as a result of its hypermethylation on the active X chromosome and the correlation of its methylation level with allelism at the X-controlling element (Xce), a locus known to affect choice. In this study, we have tested the potential function of the DXPas34 locus in Xist regulation and X-inactivation initiation by deleting it in the context of large Xist-containing yeast artificial chromosome transgenes. Deletion of DXPas34 eliminates both Xist expression and antisense transcription present in this region in undifferentiated ES cells. It also leads to nonrandom inactivation of the deleted transgene upon differentiation. DXPas34 thus appears to be a critical regulator of Xist activity and X inactivation. The expression pattern of DXPas34 during early embryonic development, which we report here, further suggests that it could be implicated in the regulation of imprinted Xist expression.


Subject(s)
Gene Expression Regulation , RNA, Untranslated , Regulatory Sequences, Nucleic Acid , Transcription Factors/genetics , X Chromosome , Animals , Cell Differentiation , Cell Line , Chromosomes, Artificial, Yeast , CpG Islands , Embryonic and Fetal Development , Female , In Situ Hybridization, Fluorescence , Male , Mice , Microsatellite Repeats , RNA, Long Noncoding , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transgenes
15.
Hosp Mater Manage Q ; 20(4): 15-23, 1999 May.
Article in English | MEDLINE | ID: mdl-10387776

ABSTRACT

Continuous improvement is an attractive idea, but it is typically more myth than reality. SCK is no myth. It delivers dramatic improvements in traditional measures quickly. SCK accomplishes this via kaizens: rapid, repeated, time-compressed changes for the better in bite-sized chunks of the business.


Subject(s)
Total Quality Management/methods , Efficiency, Organizational , Humans , Industry/organization & administration , Industry/standards , Materials Management, Hospital/organization & administration , Organizational Innovation , Time Management , United States
16.
Proc Natl Acad Sci U S A ; 96(12): 6841-6, 1999 Jun 08.
Article in English | MEDLINE | ID: mdl-10359800

ABSTRACT

Initiation of X chromosome inactivation requires the presence, in cis, of the X inactivation center (XIC). The Xist gene, which lies within the XIC region in both human and mouse and has the unique property of being expressed only from the inactive X chromosome in female somatic cells, is known to be essential for X inactivation based on targeted deletions in the mouse. Although our understanding of the developmental regulation and function of the mouse Xist gene has progressed rapidly, less is known about its human homolog. To address this and to assess the cross-species conservation of X inactivation, a 480-kb yeast artificial chromosome containing the human XIST gene was introduced into mouse embryonic stem (ES) cells. The human XIST transcript was expressed and could coat the mouse autosome from which it was transcribed, indicating that the factors required for cis association are conserved in mouse ES cells. Cis inactivation as a result of human XIST expression was found in only a proportion of differentiated cells, suggesting that the events downstream of XIST RNA coating that culminate in stable inactivation may require species-specific factors. Human XIST RNA appears to coat mouse autosomes in ES cells before in vitro differentiation, in contrast to the behavior of the mouse Xist gene in undifferentiated ES cells, where an unstable transcript and no chromosome coating are found. This may not only reflect important species differences in Xist regulation but also provides evidence that factors implicated in Xist RNA chromosome coating may already be present in undifferentiated ES cells.


Subject(s)
Chromosomes, Artificial, Yeast , Dosage Compensation, Genetic , RNA, Untranslated , Stem Cells/physiology , Transcription Factors/genetics , Animals , Female , Gene Transfer Techniques , Humans , Mice , RNA, Long Noncoding
17.
EMBO J ; 18(10): 2897-907, 1999 May 17.
Article in English | MEDLINE | ID: mdl-10329635

ABSTRACT

We have investigated the role of histone acetylation in X chromosome inactivation, focusing on its possible involvement in the regulation of Xist, an essential gene expressed only from the inactive X (Xi). We have identified a region of H4 hyperacetylation extending up to 120 kb upstream from the Xist somatic promoter P1. This domain includes the promoter P0, which gives rise to the unstable Xist transcript in undifferentiated cells. The hyperacetylated domain was not seen in male cells or in female XT67E1 cells, a mutant cell line heterozygous for a partially deleted Xist allele and in which an increased number of cells fail to undergo X inactivation. The hyperacetylation upstream of Xist was lost by day 7 of differentiation, when X inactivation was essentially complete. Wild-type cells differentiated in the presence of the histone deacetylase inhibitor Trichostatin A were prevented from forming a normally inactivated X, as judged by the frequency of underacetylated X chromosomes detected by immunofluorescence microscopy. Mutant XT67E1 cells, lacking hyperacetylation upstream of Xist, were less affected. We propose that (i) hyperacetylation of chromatin upstream of Xist facilitates the promoter switch that leads to stabilization of the Xist transcript and (ii) that the subsequent deacetylation of this region is essential for the further progression of X inactivation.


Subject(s)
Dosage Compensation, Genetic , Histones/metabolism , RNA, Untranslated , Transcription Factors/genetics , X Chromosome/genetics , Acetylation , Animals , Cell Differentiation , Cell Division , Cell Line , Chromatin/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Developmental , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Mice , Mutation , Promoter Regions, Genetic , RNA, Long Noncoding , Stem Cells , Time Factors
19.
Mol Cell Biol ; 19(4): 3156-66, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10082582

ABSTRACT

X-chromosome inactivation in female mammals is controlled by the X-inactivation center (Xic). This locus is required for inactivation in cis and is thought to be involved in the counting process which ensures that only a single X chromosome remains active per diploid cell. The Xist gene maps to the Xic region and has been shown to be essential for inactivation in cis. Transgenesis represents a stringent test for defining the minimal region that can carry out the functions attributed to the Xic. Although YAC and cosmid Xist-containing transgenes have previously been reported to be capable of cis inactivation and counting, the transgenes were all present as multicopy arrays and it was unclear to what extent individual copies are functional. Using two different yeast artificial chromosomes (YACs), we have found that single-copy transgenes, unlike multicopy arrays, can induce neither inactivation in cis nor counting. These results demonstrate that despite their large size and the presence of Xist, the YACs that we have tested lack sequences critical for autonomous function with respect to X inactivation.


Subject(s)
Dosage Compensation, Genetic , Gene Dosage , RNA, Untranslated , Stem Cells/cytology , Transcription Factors/genetics , X Chromosome/genetics , Animals , Cell Differentiation , Cell Line , Chromosomes, Artificial, Yeast , Female , In Situ Hybridization, Fluorescence , Mice , Mice, Inbred C3H , RNA, Long Noncoding , Transgenes
20.
Genomics ; 48(3): 296-303, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9545634

ABSTRACT

The overall organization of the X-inactivation center (XIC/Xic) candidate region seems poorly conserved between human and mouse. The orientation of a region containing the X-inactive-specific transcript (Xist/ XIST) gene and three genes located 3' of Xist/XIST has been shown to be inverted between the two species, although the actual extent of this rearrangement is unknown. We have cloned and mapped the mouse homolog of the human XPCT (X-linked PEST-containing transporter) gene, which encodes a putative transmembrane transporter. Human XPCT is located about 200 kb outside of the XIC candidate region and 600 kb 5' of or telomeric to the XIST gene. The mouse Xpct gene, which lies approximately 300 kb 5' of and centromeric to Xist, displays 85% identity at the nucleotide level with the human gene, and the overall protein structure is conserved. The transcriptional orientation of mouse Xpct with respect to Xist is the opposite of that in human. Consequently, the evolution of the region between human and mouse appears to be highly complex, with structural rearrangements involving a region of up to 600 kb or more around the Xist gene.


Subject(s)
Carrier Proteins/genetics , Evolution, Molecular , Gene Rearrangement , Membrane Proteins/genetics , Membrane Transport Proteins , Monocarboxylic Acid Transporters , X Chromosome , Amino Acid Sequence , Animals , Blotting, Northern , Chromosome Mapping , Cloning, Molecular , DNA, Complementary , Humans , Mice , Molecular Sequence Data , Restriction Mapping , Sequence Alignment , Sequence Homology, Amino Acid , Symporters
SELECTION OF CITATIONS
SEARCH DETAIL
...