1,396 results on '"X Chromosome Inactivation"'
Search Results
2. Preface.
- Author
-
Heard E and Brockdorff N
- Subjects
- Animals, History, 20th Century, History, 21st Century, Humans, Mammals growth & development, Mice, United Kingdom, Genetics history, Mammals genetics, X Chromosome Inactivation
- Published
- 2017
- Full Text
- View/download PDF
3. [Mary Lyon and the hypothesis on X-chromosome inactivation].
- Author
-
Ørstavik KH
- Subjects
- England, Female, History, 20th Century, Humans, Women, Genetics history, X Chromosome Inactivation
- Published
- 2015
- Full Text
- View/download PDF
4. [The legacy of Mary F. Lyon (1925-2014)].
- Author
-
Guénet JL, Panthier JJ, Avner P, Heard E, and Montagutelli X
- Subjects
- Animals, England, History, 20th Century, History, 21st Century, Humans, Mice, X Chromosome genetics, X Chromosome Inactivation, Genetics history
- Published
- 2015
- Full Text
- View/download PDF
5. Mary Frances Lyon.
- Author
-
Watts G
- Subjects
- England, History, 20th Century, History, 21st Century, Genetics history, X Chromosome Inactivation
- Published
- 2015
- Full Text
- View/download PDF
6. Mary Frances Lyon (1925–2014).
- Author
-
Fisher EM and Peters J
- Subjects
- Animals, Epigenomics, History, 20th Century, Human Genome Project, United Kingdom, Genetics history, X Chromosome Inactivation
- Published
- 2015
- Full Text
- View/download PDF
7. Mammalian developmental genetics in the twentieth century.
- Author
-
Artzt K
- Subjects
- Animals, Cloning, Molecular, Drosophila genetics, History, 20th Century, Mammals growth & development, Mice, Mice, Knockout, Mutation, X Chromosome Inactivation, Genetics history, Growth and Development genetics, Mammals genetics
- Abstract
This Perspectives is a review of the breathtaking history of mammalian genetics in the past century and, in particular, of the ways in which genetic thinking has illuminated aspects of mouse development. To illustrate the power of that thinking, selected hypothesis-driven experiments and technical advances are discussed. Also included in this account are the beginnings of mouse genetics at the Bussey Institute, Columbia University, and The Jackson Laboratory and a retrospective discussion of one of the classic problems in developmental genetics, the T/t complex and its genetic enigmas.
- Published
- 2012
- Full Text
- View/download PDF
8. The demoiselle of X-inactivation: 50 years old and as trendy and mesmerising as ever.
- Author
-
Morey C and Avner P
- Subjects
- Animals, Genetic Diseases, X-Linked genetics, Genetics trends, History, 20th Century, History, 21st Century, Humans, RNA, Long Noncoding, Genetics history, RNA, Untranslated genetics, Sex Chromatin genetics, X Chromosome Inactivation
- Abstract
In humans, sexual dimorphism is associated with the presence of two X chromosomes in the female, whereas males possess only one X and a small and largely degenerate Y chromosome. How do men cope with having only a single X chromosome given that virtually all other chromosomal monosomies are lethal? Ironically, or even typically many might say, women and more generally female mammals contribute most to the job by shutting down one of their two X chromosomes at random. This phenomenon, called X-inactivation, was originally described some 50 years ago by Mary Lyon and has captivated an increasing number of scientists ever since. The fascination arose in part from the realisation that the inactive X corresponded to a dense heterochromatin mass called the "Barr body" whose number varied with the number of Xs within the nucleus and from the many intellectual questions that this raised: How does the cell count the X chromosomes in the nucleus and inactivate all Xs except one? What kind of molecular mechanisms are able to trigger such a profound, chromosome-wide metamorphosis? When is X-inactivation initiated? How is it transmitted to daughter cells and how is it reset during gametogenesis? This review retraces some of the crucial findings, which have led to our current understanding of a biological process that was initially considered as an exception completely distinct from conventional regulatory systems but is now viewed as a paradigm "par excellence" for epigenetic regulation., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2011
- Full Text
- View/download PDF
9. The exception that proves the rule: an interview with Jenny Graves. Interview by Jane Gitschier.
- Author
-
Graves J
- Subjects
- Animals, Humans, Sex Chromosomes genetics, X Chromosome Inactivation, Genetics, Marsupialia genetics
- Published
- 2008
- Full Text
- View/download PDF
10. Dissection of protein and RNA regions required for SPEN binding to XIST A-repeat RNA
- Author
-
Button, Aileen C, Hall, Simone D, Ashley, Ethan L, and McHugh, Colleen Adare
- Subjects
Biochemistry and Cell Biology ,Bioinformatics and Computational Biology ,Biological Sciences ,Genetics ,2.1 Biological and endogenous factors ,Generic health relevance ,Female ,Humans ,Chromatin ,DNA-Binding Proteins ,Gene Silencing ,RNA ,Long Noncoding ,RNA ,Untranslated ,RNA-Binding Proteins ,X Chromosome ,X Chromosome Inactivation ,RNA recognition motif ,RNA-protein interactions ,SPEN ,SHARP ,X chromosome silencing ,XIST noncoding RNA ,RNA–protein interactions ,Developmental Biology ,Biochemistry and cell biology - Abstract
XIST noncoding RNA promotes the initiation of X chromosome silencing by recruiting the protein SPEN to one X chromosome in female mammals. The SPEN protein is also called SHARP (SMRT and HDAC-associated repressor protein) and MINT (Msx-2 interacting nuclear target) in humans. SPEN recruits N-CoR2 and HDAC3 to initiate histone deacetylation on the X chromosome, leading to the formation of repressive chromatin marks and silencing gene expression. We dissected the contributions of different RNA and protein regions to the formation of a human XIST-SPEN complex in vitro and identified novel sequence and structure determinants that may contribute to X chromosome silencing initiation. Binding of SPEN to XIST RNA requires RRM 4 of the protein, in contrast to the requirement of RRM 3 and RRM 4 for specific binding to SRA RNA. Measurements of SPEN binding to full-length, dimeric, trimeric, or other truncated versions of the A-repeat region revealed that high-affinity binding of XIST to SPEN in vitro requires a minimum of four A-repeat segments. SPEN binding to XIST A-repeat RNA changes the accessibility of the RNA at specific nucleotide sequences, as indicated by changes in RNA reactivity through chemical structure probing. Based on computational modeling, we found that inter-repeat duplexes formed by multiple A-repeats can present an unpaired adenosine in the context of a double-stranded region of RNA. The presence of this specific combination of sequence and structural motifs correlates with high-affinity SPEN binding in vitro. These data provide new information on the molecular basis of the XIST and SPEN interaction.
- Published
- 2024
11. Inferring clonal somatic mutations directed by X chromosome inactivation status in single cells
- Author
-
Ilke Demirci, Anton J. M. Larsson, Xinsong Chen, Johan Hartman, Rickard Sandberg, and Jonas Frisén
- Subjects
Mutations ,Clonality ,X chromosome inactivation ,Hematopoiesis ,Biology (General) ,QH301-705.5 ,Genetics ,QH426-470 - Abstract
Abstract Analysis of clonal dynamics in human tissues is enabled by somatic genetic variation. Here, we show that analysis of mitochondrial mutations in single cells is dramatically improved in females when using X chromosome inactivation to select informative clonal mutations. Applying this strategy to human peripheral mononuclear blood cells reveals clonal structures within T cells that otherwise are blurred by non-informative mutations, including the separation of gamma-delta T cells, suggesting this approach can be used to decipher clonal dynamics of cells in human tissues.
- Published
- 2024
- Full Text
- View/download PDF
12. Identification of the RSX interactome in a marsupial shows functional coherence with the Xist interactome during X inactivation
- Author
-
Kim L. McIntyre, Shafagh A. Waters, Ling Zhong, Gene Hart-Smith, Mark Raftery, Zahra A. Chew, Hardip R. Patel, Jennifer A. Marshall Graves, and Paul D. Waters
- Subjects
X chromosome inactivation ,Convergent evolution ,RSX protein interactome ,Biology (General) ,QH301-705.5 ,Genetics ,QH426-470 - Abstract
Abstract The marsupial specific RSX lncRNA is the functional analogue of the eutherian specific XIST, which coordinates X chromosome inactivation. We characterized the RSX interactome in a marsupial representative (the opossum Monodelphis domestica), identifying 135 proteins, of which 54 had orthologues in the XIST interactome. Both interactomes were enriched for biological pathways related to RNA processing, regulation of translation, and epigenetic transcriptional silencing. This represents a remarkable example showcasing the functional coherence of independently evolved lncRNAs in distantly related mammalian lineages.
- Published
- 2024
- Full Text
- View/download PDF
13. X-chromosome inactivation in human iPSCs provides insight into X-regulated gene expression in autosomes
- Author
-
Hande Topa, Clara Benoit-Pilven, Taru Tukiainen, and Olli Pietiläinen
- Subjects
X chromosome inactivation ,hiPSC ,Sex differences ,RNA-seq ,Allele-specific expression ,XIST-bound autosomal genes ,Biology (General) ,QH301-705.5 ,Genetics ,QH426-470 - Abstract
Abstract Background Variation in X chromosome inactivation (XCI) in human-induced pluripotent stem cells (hiPSCs) can impact their ability to model biological sex biases. The gene-wise landscape of X chromosome gene dosage remains unresolved in female hiPSCs. To characterize patterns of de-repression and escape from inactivation, we performed a systematic survey of allele specific expression in 165 female hiPSC lines. Results XCI erosion is non-random and primarily affects genes that escape XCI in human tissues. Individual genes and cell lines vary in the frequency and degree of de-repression. Bi-allelic expression increases gradually after modest decrease of XIST in cultures, whose loss is commonly used to mark lines with eroded XCI. We identify three clusters of female lines at different stages of XCI. Increased XCI erosion amplifies female-biased expression at hypomethylated sites and regions normally occupied by repressive histone marks, lowering male-biased differences in the X chromosome. In autosomes, erosion modifies sex differences in a dose-dependent way. Male-biased genes are enriched for hypermethylated regions, and de-repression of XIST-bound autosomal genes in female lines attenuates normal male-biased gene expression in eroded lines. XCI erosion can compensate for a dominant loss of function effect in several disease genes. Conclusions We present a comprehensive view of X chromosome gene dosage in hiPSCs and implicate a direct mechanism for XCI erosion in regulating autosomal gene expression in trans. The uncommon and variable reactivation of X chromosome genes in female hiPSCs can provide insight into X chromosome’s role in regulating gene expression and sex differences in humans.
- Published
- 2024
- Full Text
- View/download PDF
14. A lifelong duty: how Xist maintains the inactive X chromosome
- Author
-
Jacobson, Elsie C, Pandya-Jones, Amy, and Plath, Kathrin
- Subjects
Biochemistry and Cell Biology ,Genetics ,Biological Sciences ,Human Genome ,Women's Health ,1.1 Normal biological development and functioning ,Generic health relevance ,Chromatin ,Female ,Gene Silencing ,Humans ,RNA ,Long Noncoding ,X Chromosome ,X Chromosome Inactivation ,Developmental Biology ,Biochemistry and cell biology - Abstract
Female eutherians transcriptionally silence one X chromosome to balance gene dosage between the sexes. X-chromosome inactivation (XCI) is initiated by the lncRNA Xist, which assembles many proteins within the inactive X chromosome (Xi) to trigger gene silencing and heterochromatin formation. It is well established that gene silencing on the Xi is maintained through repressive epigenetic processes, including histone deacetylation and DNA methylation. Recent studies revealed a new mechanism where RNA-binding proteins that interact directly with the RNA contribute to the maintenance of Xist localization and gene silencing. In addition, a surprising plasticity of the Xi was uncovered with many genes becoming upregulated upon experimental deletion of Xist. Intriguingly, immune cells normally lose Xist from the Xi, suggesting that thisXist dependence is utilized in vivo to dynamically regulate gene expression from the Xi. These new studies expose fundamental regulatory mechanisms for the chromatin association of RNAs, highlight the need for studying the maintenance of XCI and Xist localization in a gene- and cell-type-specific manner, and are likely to have clinical impact.
- Published
- 2022
15. X Chromosome Inactivation Timing is Not eXACT: Implications for Autism Spectrum Disorders
- Author
-
LaSalle, Janine M
- Subjects
Biological Sciences ,Genetics ,Stem Cell Research ,Mental Health ,Autism ,Pediatric ,Intellectual and Developmental Disabilities (IDD) ,Brain Disorders ,Human Genome ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,DNA methylation ,X chromosome inactivation ,peri-implantation development ,autism ,neurodevelomental disorders ,Clinical Sciences ,Law - Abstract
The etiology of autism spectrum disorders (ASD) is complex, involving different combinations of genetic and environmental factors. My lab's approach has been to investigate DNA methylation as a tractable genome-wide modification at the interface of these complex interactions, reflecting past and future events in the molecular pathogenesis of ASD. Since X-linked genes were enriched in DNA methylation differences discovered from cord blood from newborns later diagnosed with ASD, this has prompted me to review and revisit the recent advancements in the field of X chromosome inactivation (XCI), particularly in humans and other primates. In this Perspective, I compare XCI mechanisms in different mammalian species, including the finding of the noncoding transcript XACT associated with X chromosome erosion in human pluripotent stem cells and recent findings from non-human primate post-implantation embryos. I focus on the experimentally challenging peri- and post-implantation stages of human development when the timing of XCI is prolonged and imprecise in humans. Collectively, this research has raised some important unanswered questions involving biased sex ratios in human births and the male bias in the incidence of ASD.
- Published
- 2022
16. Xist nucleates local protein gradients to propagate silencing across the X chromosome
- Author
-
Markaki, Yolanda, Gan Chong, Johnny, Wang, Yuying, Jacobson, Elsie C, Luong, Christy, Tan, Shawn YX, Jachowicz, Joanna W, Strehle, Mackenzie, Maestrini, Davide, Banerjee, Abhik K, Mistry, Bhaven A, Dror, Iris, Dossin, Francois, Schöneberg, Johannes, Heard, Edith, Guttman, Mitchell, Chou, Tom, and Plath, Kathrin
- Subjects
Biochemistry and Cell Biology ,Bioinformatics and Computational Biology ,Biological Sciences ,Genetics ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Generic health relevance ,Animals ,Apoptosis Regulatory Proteins ,Cell Line ,Embryonic Stem Cells ,Fibroblasts ,Gene Silencing ,Humans ,Mice ,Mitochondrial Proteins ,Protein Binding ,RNA ,Long Noncoding ,X Chromosome ,X Chromosome Inactivation ,RNA-binding proteins ,X chromosome inactivation ,Xist RNA ,biomolecular condensates ,chromatin organization ,heterochromatin ,macromolecular dynamics ,quantitative imaging ,super-resolution microscopy ,supramolecular complexes ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
The lncRNA Xist forms ∼50 diffraction-limited foci to transcriptionally silence one X chromosome. How this small number of RNA foci and interacting proteins regulate a much larger number of X-linked genes is unknown. We show that Xist foci are locally confined, contain ∼2 RNA molecules, and nucleate supramolecular complexes (SMACs) that include many copies of the critical silencing protein SPEN. Aggregation and exchange of SMAC proteins generate local protein gradients that regulate broad, proximal chromatin regions. Partitioning of numerous SPEN molecules into SMACs is mediated by their intrinsically disordered regions and essential for transcriptional repression. Polycomb deposition via SMACs induces chromatin compaction and the increase in SMACs density around genes, which propagates silencing across the X chromosome. Our findings introduce a mechanism for functional nuclear compartmentalization whereby crowding of transcriptional and architectural regulators enables the silencing of many target genes by few RNA molecules.
- Published
- 2021
17. The human Y and inactive X chromosomes similarly modulate autosomal gene expression
- Author
-
Adrianna K. San Roman, Helen Skaletsky, Alexander K. Godfrey, Neha V. Bokil, Levi Teitz, Isani Singh, Laura V. Blanton, Daniel W. Bellott, Tatyana Pyntikova, Julian Lange, Natalia Koutseva, Jennifer F. Hughes, Laura Brown, Sidaly Phou, Ashley Buscetta, Paul Kruszka, Nicole Banks, Amalia Dutra, Evgenia Pak, Patricia C. Lasutschinkow, Colleen Keen, Shanlee M. Davis, Angela E. Lin, Nicole R. Tartaglia, Carole Samango-Sprouse, Maximilian Muenke, and David C. Page
- Subjects
sex chromosomes ,sex differences ,X chromosome inactivation ,aneuploidy ,Turner syndrome ,Klinefelter syndrome ,Genetics ,QH426-470 ,Internal medicine ,RC31-1245 - Abstract
Summary: Somatic cells of human males and females have 45 chromosomes in common, including the “active” X chromosome. In males the 46th chromosome is a Y; in females it is an “inactive” X (Xi). Through linear modeling of autosomal gene expression in cells from individuals with zero to three Xi and zero to four Y chromosomes, we found that Xi and Y impact autosomal expression broadly and with remarkably similar effects. Studying sex chromosome structural anomalies, promoters of Xi- and Y-responsive genes, and CRISPR inhibition, we traced part of this shared effect to homologous transcription factors—ZFX and ZFY—encoded by Chr X and Y. This demonstrates sex-shared mechanisms by which Xi and Y modulate autosomal expression. Combined with earlier analyses of sex-linked gene expression, our studies show that 21% of all genes expressed in lymphoblastoid cells or fibroblasts change expression significantly in response to Xi or Y chromosomes.
- Published
- 2024
- Full Text
- View/download PDF
18. A small-molecule screen reveals novel modulators of MeCP2 and X-chromosome inactivation maintenance
- Author
-
Lee, Hyeong-Min, Kuijer, M Bram, Ruiz Blanes, Nerea, Clark, Ellen P, Aita, Megumi, Galiano Arjona, Lorena, Kokot, Agnieszka, Sciaky, Noah, Simon, Jeremy M, Bhatnagar, Sanchita, Philpot, Benjamin D, and Cerase, Andrea
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Genetics ,Neurodegenerative ,Brain Disorders ,Pediatric ,Rett Syndrome ,Orphan Drug ,Rare Diseases ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Congenital ,Animals ,Chromosomes ,Female ,Male ,Methyl-CpG-Binding Protein 2 ,Mice ,Mutation ,X Chromosome Inactivation ,AG490 ,JAK/STAT ,Janus Kinase ,Janus Kinase inhibitors ,MeCP2 ,PI3K/ATK pathways ,Rett syndrome ,X-chromosome inactivation ,Psychology - Abstract
BackgroundRett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in the X-linked methyl-CpG binding protein 2 (MeCP2) gene. While MeCP2 mutations are lethal in most males, females survive birth but show severe neurological defects. Because X-chromosome inactivation (XCI) is a random process, approximately 50% of the cells silence the wild-type (WT) copy of the MeCP2 gene. Thus, reactivating the silent WT copy of MeCP2 could provide therapeutic intervention for RTT.MethodsToward this goal, we screened ~ 28,000 small-molecule compounds from several libraries using a MeCP2-luciferase reporter cell line and cortical neurons from a MeCP2-EGFP mouse model. We used gain/increase of luminescence or fluorescence as a readout of MeCP2 reactivation and tested the efficacy of these drugs under different drug regimens, conditions, and cellular contexts.ResultsWe identified inhibitors of the JAK/STAT pathway as XCI-reactivating agents, both by in vitro and ex vivo assays. In particular, we show that AG-490, a Janus Kinase 2 (JAK2) kinase inhibitor, and Jaki, a pan JAK/STAT inhibitor, are capable of reactivating MeCP2 from the inactive X chromosome, in different cellular contexts.ConclusionsOur results suggest that inhibition of the JAK/STAT pathway is a new potential pathway to reinstate MeCP2 gene expression as an efficient RTT treatment.
- Published
- 2020
19. Female human primordial germ cells display X-chromosome dosage compensation despite the absence of X-inactivation.
- Author
-
Chitiashvili, Tsotne, Dror, Iris, Kim, Rachel, Hsu, Fei-Man, Chaudhari, Rohan, Pandolfi, Erica, Chen, Di, Liebscher, Simone, Schenke-Layland, Katja, Plath, Kathrin, and Clark, Amander
- Subjects
Germ Cells ,Cells ,Cultured ,Chromosomes ,Human ,X ,Blastocyst ,Humans ,In Situ Hybridization ,Fluorescence ,Gene Expression Profiling ,Gene Expression Regulation ,Developmental ,Embryonic Development ,Dosage Compensation ,Genetic ,Female ,Male ,X Chromosome Inactivation ,RNA ,Long Noncoding ,Stem Cell Research ,Genetics ,1.1 Normal biological development and functioning ,Generic health relevance ,Developmental Biology ,Biological Sciences ,Medical and Health Sciences - Abstract
X-chromosome dosage compensation in female placental mammals is achieved by X-chromosome inactivation (XCI). Human pre-implantation embryos are an exception, in which dosage compensation occurs by X-chromosome dampening (XCD). Here, we examined whether XCD extends to human prenatal germ cells given their similarities to naive pluripotent cells. We found that female human primordial germ cells (hPGCs) display reduced X-linked gene expression before entering meiosis. Moreover, in hPGCs, both X chromosomes are active and express the long non-coding RNAs X active coating transcript (XACT) and X inactive specific transcript (XIST)-the master regulator of XCI-which are silenced after entry into meiosis. We find that XACT is a hPGC marker, describe XCD associated with XIST expression in hPGCs and suggest that XCD evolved in humans to regulate X-linked genes in pre-implantation embryos and PGCs. Furthermore, we found a unique mechanism of X-chromosome regulation in human primordial oocytes. Therefore, future studies of human germline development must consider the sexually dimorphic X-chromosome dosage compensation mechanisms in the prenatal germline.
- Published
- 2020
20. A protein assembly mediates Xist localization and gene silencing
- Author
-
Pandya-Jones, Amy, Markaki, Yolanda, Serizay, Jacques, Chitiashvili, Tsotne, Mancia Leon, Walter R, Damianov, Andrey, Chronis, Constantinos, Papp, Bernadett, Chen, Chun-Kan, McKee, Robin, Wang, Xiao-Jun, Chau, Anthony, Sabri, Shan, Leonhardt, Heinrich, Zheng, Sika, Guttman, Mitchell, Black, Douglas L, and Plath, Kathrin
- Subjects
Biochemistry and Cell Biology ,Bioinformatics and Computational Biology ,Biological Sciences ,Genetics ,1.1 Normal biological development and functioning ,Generic health relevance ,Animals ,CELF1 Protein ,Cell Line ,DNA-Binding Proteins ,Female ,Gene Silencing ,Heterogeneous-Nuclear Ribonucleoproteins ,Humans ,In Situ Hybridization ,Fluorescence ,Male ,Mice ,Nuclear Matrix-Associated Proteins ,Polypyrimidine Tract-Binding Protein ,RNA ,Long Noncoding ,RNA-Binding Proteins ,X Chromosome Inactivation ,General Science & Technology - Abstract
Nuclear compartments have diverse roles in regulating gene expression, yet the molecular forces and components that drive compartment formation remain largely unclear1. The long non-coding RNA Xist establishes an intra-chromosomal compartment by localizing at a high concentration in a territory spatially close to its transcription locus2 and binding diverse proteins3-5 to achieve X-chromosome inactivation (XCI)6,7. The XCI process therefore serves as a paradigm for understanding how RNA-mediated recruitment of various proteins induces a functional compartment. The properties of the inactive X (Xi)-compartment are known to change over time, because after initial Xist spreading and transcriptional shutoff a state is reached in which gene silencing remains stable even if Xist is turned off8. Here we show that the Xist RNA-binding proteins PTBP19, MATR310, TDP-4311 and CELF112 assemble on the multivalent E-repeat element of Xist7 and, via self-aggregation and heterotypic protein-protein interactions, form a condensate1 in the Xi. This condensate is required for gene silencing and for the anchoring of Xist to the Xi territory, and can be sustained in the absence of Xist. Notably, these E-repeat-binding proteins become essential coincident with transition to the Xist-independent XCI phase8, indicating that the condensate seeded by the E-repeat underlies the developmental switch from Xist-dependence to Xist-independence. Taken together, our data show that Xist forms the Xi compartment by seeding a heteromeric condensate that consists of ubiquitous RNA-binding proteins, revealing an unanticipated mechanism for heritable gene silencing.
- Published
- 2020
21. Artificial escape from XCI by DNA methylation editing of the CDKL5 gene
- Author
-
Halmai, Julian ANM, Deng, Peter, Gonzalez, Casiana E, Coggins, Nicole B, Cameron, David, Carter, Jasmine L, Buchanan, Fiona KB, Waldo, Jennifer J, Lock, Samantha R, Anderson, Johnathon D, O’Geen, Henriette, Segal, David J, Nolta, Jan, and Fink, Kyle D
- Subjects
Biological Sciences ,Genetics ,Human Genome ,Brain Disorders ,2.1 Biological and endogenous factors ,Alleles ,CRISPR-Associated Protein 9 ,Catalytic Domain ,Cell Line ,Tumor ,Chromosomes ,Human ,X ,CpG Islands ,Epigenesis ,Genetic ,Gene Editing ,Gene Silencing ,Humans ,Mixed Function Oxygenases ,Neurons ,Promoter Regions ,Genetic ,Protein Serine-Threonine Kinases ,Proto-Oncogene Proteins ,RNA ,Messenger ,Recombinant Fusion Proteins ,Trans-Activators ,X Chromosome Inactivation ,RNA ,Guide ,CRISPR-Cas Systems ,Environmental Sciences ,Information and Computing Sciences ,Developmental Biology ,Biological sciences ,Chemical sciences ,Environmental sciences - Abstract
A significant number of X-linked genes escape from X chromosome inactivation and are associated with a distinct epigenetic signature. One epigenetic modification that strongly correlates with X-escape is reduced DNA methylation in promoter regions. Here, we created an artificial escape by editing DNA methylation on the promoter of CDKL5, a gene causative for an infantile epilepsy, from the silenced X-chromosomal allele in human neuronal-like cells. We identify that a fusion of the catalytic domain of TET1 to dCas9 targeted to the CDKL5 promoter using three guide RNAs causes significant reactivation of the inactive allele in combination with removal of methyl groups from CpG dinucleotides. Strikingly, we demonstrate that co-expression of TET1 and a VP64 transactivator have a synergistic effect on the reactivation of the inactive allele to levels >60% of the active allele. We further used a multi-omics assessment to determine potential off-targets on the transcriptome and methylome. We find that synergistic delivery of dCas9 effectors is highly selective for the target site. Our findings further elucidate a causal role for reduced DNA methylation associated with escape from X chromosome inactivation. Understanding the epigenetics associated with escape from X chromosome inactivation has potential for those suffering from X-linked disorders.
- Published
- 2020
22. A novel truncating variant in ring finger protein 113A (RNF113A) confirms the association of this gene with X‐linked trichothiodystrophy
- Author
-
Mendelsohn, Bryce A, Beleford, Daniah T, Abu‐El‐Haija, Aya, Alsaleh, Norah S, Rahbeeni, Zuhair, Martin, Pierre‐Marie, Rego, Shannon, Huang, Alyssa, Capodanno, Gina, Shieh, Joseph T, Van Ziffle, Jessica, Risch, Neil, Alkuraya, Fowzan S, and Slavotinek, Anne M
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Clinical Research ,Congenital Structural Anomalies ,Brain Disorders ,Pediatric ,Rare Diseases ,2.1 Biological and endogenous factors ,Child ,Child ,Preschool ,DNA-Binding Proteins ,Female ,Genetic Association Studies ,Genetic Diseases ,X-Linked ,Genetic Predisposition to Disease ,Humans ,Male ,Mutation ,Trichothiodystrophy Syndromes ,X Chromosome Inactivation ,RNF113A ,spliceosome ,trichothiodystrophy ,zinc finger ,Clinical Sciences ,Clinical sciences - Abstract
We describe an 11-year old boy with severe global developmental delays, failure to thrive and growth retardation, refractory seizures with recurrent status epilepticus, hypogammaglobulinemia, hypergonadotropic hypogonadism, and duodenal strictures. He had facial and skin findings compatible with trichothiodystrophy, including sparse and brittle hair, thin eyebrows, and dry skin. Exome sequencing showed a hemizygous, truncating variant in RNF113A, c.903_910delGCAGACCA, predicting p.(Gln302fs*12), that was inherited from his mother. Although his clinical features overlap closely with features described in the two previously reported male first cousins with RNF113A loss of function mutations, the duodenal strictures seen in this patient have not been reported. Interestingly, the patient's mother had short stature and 100% skewed X-inactivation as seen in other obligate female carriers. A second male with developmental delays, microcephaly, seizures, ambiguous genitalia, and facial anomalies that included sparse and brittle hair, thin eyebrows and dry skin was recently reported to have c.897_898delTG, predicting p.(Cys299*) in RNF113A and we provide additional clinical details for this patient. This report further supports deleterious variants in RNF113A as a cause of a novel trichothiodystrophy syndrome.
- Published
- 2020
23. Long noncoding RNA XIST: Mechanisms for X chromosome inactivation, roles in sex-biased diseases, and therapeutic opportunities
- Author
-
Jianjian Li, Zhe Ming, Liuyi Yang, Tingxuan Wang, Gaowen Liu, and Qing Ma
- Subjects
Epigenetic regulation ,Long noncoding RNA ,Sex-biased diseases ,X chromosome inactivation ,XIST ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Sexual dimorphism has been reported in various human diseases including autoimmune diseases, neurological diseases, pulmonary arterial hypertension, and some types of cancers, although the underlying mechanisms remain poorly understood. The long noncoding RNA (lncRNA) X-inactive specific transcript (XIST) is involved in X chromosome inactivation (XCI) in female placental mammals, a process that ensures the balanced expression dosage of X-linked genes between sexes. XIST is abnormally expressed in many sex-biased diseases. In addition, escape from XIST-mediated XCI and skewed XCI also contribute to sex-biased diseases. Therefore, its expression or modification can be regarded as a biomarker for the diagnosis and prognosis of many sex-biased diseases. Genetic manipulation of XIST expression can inhibit the progression of some of these diseases in animal models, and therefore XIST has been proposed as a potential therapeutic target. In this manuscript, we summarize the current knowledge about the mechanisms for XIST-mediated XCI and the roles of XIST in sex-biased diseases, and discuss potential therapeutic strategies targeting XIST.
- Published
- 2022
- Full Text
- View/download PDF
24. Identification and functional analysis of a novel de novo missense mutation located in the initiation codon of LAMP2 associated with early onset female Danon disease
- Author
-
Yongxiang Wang, Ming Bai, Piyi Zhang, Yu Peng, Zixian Chen, Zhiyu He, Jin Xu, Youqi Zhu, Dongdong Yan, Runqing Wang, and Zheng Zhang
- Subjects
autophagy ,Danon disease ,haploinsufficiency ,initiation codon mutation ,lysosome‐associated membrane glycoprotein 2 ,X chromosome inactivation ,Genetics ,QH426-470 - Abstract
Abstract Background Danon disease is characterized by the failure of lysosomal biogenesis, maturation, and function due to a deficiency of lysosomal membrane structural protein (LAMP2). Methods The current report describes a female patient with a sudden syncope and hypertrophic cardiomyopathy phenotype. We identified the pathogenic mutations in patients by whole‐exon sequencing, followed by a series of molecular biology and genetic approaches to identify and functional analysis of the mutations. Results Suggestive findings by cardiac magnetic resonance (CMR), electrocardiogram (ECG), and laboratory examination suggested Danon disease which was confirmed by genetic testing. The patient carried a novel de novo mutation, LAMP2 c.2T>C located at the initiation codon. The quantitative polymerase chain reaction (qPCR) and Western blot (WB) analysis of peripheral blood leukocytes from the patients revealed evidence of LAMP2 haploinsufficiency. Labeling of the new initiation codon predicted by the software with green fluorescent protein followed by fluorescence microscopy and Western blotting showed that the first ATG downstream from the original initiation codon became the new translational initiation codon. The three‐dimensional structure of the mutated protein predicted by alphafold2 revealed that it consisted of only six amino acids and failed to form a functional polypeptide or protein. Overexpression of the mutated LAMP2 c.2T>C showed a loss of function of the protein, as assessed by the dual‐fluorescence autophagy indicator system. The mutation was confirmed to be null, AR experiments and sequencing results confirmed that 28% of the mutant X chromosome remained active. Conclusion We propose possible mechanisms of mutations associated with haploinsufficiency of LAMP2: (1) The inactivation X chromosome carrying the mutation was not significantly skewed. However, it decreased in the mRNA level and the expression ratio of the mutant transcripts; (2) The identified mutation is null, and the active mutant transcript fails to translate into the normal LAMP2 proteins. The presence of haploinsufficiency in LAMP2 and the X chromosome inactivation pattern were crucial factors contributing to the early onset of Danon disease in this female patient.
- Published
- 2023
- Full Text
- View/download PDF
25. Orchestrating Asymmetric Expression: Mechanisms behind Xist Regulation
- Author
-
Samuel Jesus Luchsinger-Morcelle, Joost Gribnau, and Hegias Mira-Bontenbal
- Subjects
X chromosome inactivation ,Xist ,epigenetics ,lncRNAs ,asymmetric expression ,Genetics ,QH426-470 ,Biotechnology ,TP248.13-248.65 - Abstract
Compensation for the gene dosage disequilibrium between sex chromosomes in mammals is achieved in female cells by repressing one of its X chromosomes through a process called X chromosome inactivation (XCI), exemplifying the control of gene expression by epigenetic mechanisms. A critical player in this mechanism is Xist, a long, non-coding RNA upregulated from a single X chromosome during early embryonic development in female cells. Over the past few decades, many factors involved at different levels in the regulation of Xist have been discovered. In this review, we hierarchically describe and analyze the different layers of Xist regulation operating concurrently and intricately interacting with each other to achieve asymmetric and monoallelic upregulation of Xist in murine female cells. We categorize these into five different classes: DNA elements, transcription factors, other regulatory proteins, long non-coding RNAs, and the chromatin and topological landscape surrounding Xist.
- Published
- 2024
- Full Text
- View/download PDF
26. Association of Human iPSC Gene Signatures and X Chromosome Dosage with Two Distinct Cardiac Differentiation Trajectories
- Author
-
D'Antonio-Chronowska, Agnieszka, Donovan, Margaret KR, Greenwald, William W Young, Nguyen, Jennifer Phuong, Fujita, Kyohei, Hashem, Sherin, Matsui, Hiroko, Soncin, Francesca, Parast, Mana, Ward, Michelle C, Coulet, Florence, Smith, Erin N, Adler, Eric, D'Antonio, Matteo, and Frazer, Kelly A
- Subjects
Biological Sciences ,Genetics ,Human Genome ,Stem Cell Research - Induced Pluripotent Stem Cell ,Cardiovascular ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research ,Regenerative Medicine ,Heart Disease ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Cell Differentiation ,Cells ,Cultured ,Chromosomes ,Human ,X ,Female ,Humans ,Induced Pluripotent Stem Cells ,Male ,Myocytes ,Cardiac ,Pericardium ,Transcriptome ,X Chromosome Inactivation ,X chromosome erosion ,X chromosome inactivation ,iPSC ,iPSC differentiation ,iPSC-derived cardiomyocytes ,iPSC-derived cardiovascular progenitor cells ,iPSC-derived epicardium ,scRNA-seq ,single-cell transcriptomics ,Biochemistry and Cell Biology ,Clinical Sciences ,Biochemistry and cell biology - Abstract
Despite the importance of understanding how variability across induced pluripotent stem cell (iPSC) lines due to non-genetic factors (clone and passage) influences their differentiation outcome, large-scale studies capable of addressing this question have not yet been conducted. Here, we differentiated 191 iPSC lines to generate iPSC-derived cardiovascular progenitor cells (iPSC-CVPCs). We observed cellular heterogeneity across the iPSC-CVPC samples due to varying fractions of two cell types: cardiomyocytes (CMs) and epicardium-derived cells (EPDCs). Comparing the transcriptomes of CM-fated and EPDC-fated iPSCs, we discovered that 91 signature genes and X chromosome dosage differences are associated with these two distinct cardiac developmental trajectories. In an independent set of 39 iPSCs differentiated into CMs, we confirmed that sex and transcriptional differences affect cardiac-fate outcome. Our study provides novel insights into how iPSC transcriptional and X chromosome gene dosage differences influence their response to differentiation stimuli and, hence, cardiac cell fate.
- Published
- 2019
27. NAA10 polyadenylation signal variants cause syndromic microphthalmia
- Author
-
Johnston, Jennifer J, Williamson, Kathleen A, Chou, Christopher M, Sapp, Julie C, Ansari, Morad, Chapman, Heather M, Cooper, David N, Dabir, Tabib, Dudley, Jeffrey N, Holt, Richard J, Ragge, Nicola K, Schäffer, Alejandro A, Sen, Shurjo K, Slavotinek, Anne M, FitzPatrick, David R, Glaser, Thomas M, Stewart, Fiona, Black, Graeme Cm, and Biesecker, Leslie G
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Rare Diseases ,Human Genome ,Pediatric ,Clinical Research ,2.1 Biological and endogenous factors ,Good Health and Well Being ,3' Untranslated Regions ,Alleles ,Anophthalmos ,Female ,Genes ,X-Linked ,Genetic Association Studies ,Genetic Predisposition to Disease ,Genetic Variation ,Genotype ,Humans ,Lod Score ,Male ,Microphthalmos ,N-Terminal Acetyltransferase A ,N-Terminal Acetyltransferase E ,Pedigree ,Poly A ,Sequence Analysis ,DNA ,X Chromosome Inactivation ,Naa10 ,polyadenylation signal ,Medical and Health Sciences ,Genetics & Heredity ,Clinical sciences - Abstract
BackgroundA single variant in NAA10 (c.471+2T>A), the gene encoding N-acetyltransferase 10, has been associated with Lenz microphthalmia syndrome. In this study, we aimed to identify causative variants in families with syndromic X-linked microphthalmia.MethodsThree families, including 15 affected individuals with syndromic X-linked microphthalmia, underwent analyses including linkage analysis, exome sequencing and targeted gene sequencing. The consequences of two identified variants in NAA10 were evaluated using quantitative PCR and RNAseq.ResultsGenetic linkage analysis in family 1 supported a candidate region on Xq27-q28, which included NAA10. Exome sequencing identified a hemizygous NAA10 polyadenylation signal (PAS) variant, chrX:153,195,397T>C, c.*43A>G, which segregated with the disease. Targeted sequencing of affected males from families 2 and 3 identified distinct NAA10 PAS variants, chrX:g.153,195,401T>C, c.*39A>G and chrX:g.153,195,400T>C, c.*40A>G. All three variants were absent from gnomAD. Quantitative PCR and RNAseq showed reduced NAA10 mRNA levels and abnormal 3' UTRs in affected individuals. Targeted sequencing of NAA10 in 376 additional affected individuals failed to identify variants in the PAS.ConclusionThese data show that PAS variants are the most common variant type in NAA10-associated syndromic microphthalmia, suggesting reduced RNA is the molecular mechanism by which these alterations cause microphthalmia/anophthalmia. We reviewed recognised variants in PAS associated with Mendelian disorders and identified only 23 others, indicating that NAA10 harbours more than 10% of all known PAS variants. We hypothesise that PAS in other genes harbour unrecognised pathogenic variants associated with Mendelian disorders. The systematic interrogation of PAS could improve genetic testing yields.
- Published
- 2019
28. IndiSPENsable for X Chromosome Inactivation and Gene Silencing
- Author
-
Corinne Kaufmann and Anton Wutz
- Subjects
SPEN ,X chromosome inactivation ,RRM ,SPOC domain ,intrinsic disorder ,gene regulation ,Genetics ,QH426-470 ,Biotechnology ,TP248.13-248.65 - Abstract
For about 30 years, SPEN has been the subject of research in many different fields due to its variety of functions and its conservation throughout a wide spectrum of species, like worms, arthropods, and vertebrates. To date, 216 orthologues have been documented. SPEN had been studied for its role in gene regulation in the context of cell signaling, including the NOTCH or nuclear hormone receptor signaling pathways. More recently, SPEN has been identified as a major regulator of initiation of chromosome-wide gene silencing during X chromosome inactivation (XCI) in mammals, where its function remains to be fully understood. Dependent on the biological context, SPEN functions via mechanisms which include different domains. While some domains of SPEN are highly conserved in sequence and secondary structure, species-to-species differences exist that might lead to mechanistic differences. Initiation of XCI appears to be different between humans and mice, which raises additional questions about the extent of generalization of SPEN’s function in XCI. In this review, we dissect the mechanism of SPEN in XCI. We discuss its subregions and domains, focusing on its role as a major regulator. We further highlight species-related research, specifically of mouse and human SPEN, with the aim to reveal and clarify potential species-to-species differences in SPEN’s function.
- Published
- 2023
- Full Text
- View/download PDF
29. The human inactive X chromosome modulates expression of the active X chromosome
- Author
-
Adrianna K. San Roman, Alexander K. Godfrey, Helen Skaletsky, Daniel W. Bellott, Abigail F. Groff, Hannah L. Harris, Laura V. Blanton, Jennifer F. Hughes, Laura Brown, Sidaly Phou, Ashley Buscetta, Paul Kruszka, Nicole Banks, Amalia Dutra, Evgenia Pak, Patricia C. Lasutschinkow, Colleen Keen, Shanlee M. Davis, Nicole R. Tartaglia, Carole Samango-Sprouse, Maximilian Muenke, and David C. Page
- Subjects
sex chromosomes ,X chromosome inactivation ,gene expression ,sex differences ,aneuploidy ,turner syndrome ,Genetics ,QH426-470 ,Internal medicine ,RC31-1245 - Abstract
Summary: The “inactive” X chromosome (Xi) has been assumed to have little impact, in trans, on the “active” X (Xa). To test this, we quantified Xi and Xa gene expression in individuals with one Xa and zero to three Xis. Our linear modeling revealed modular Xi and Xa transcriptomes and significant Xi-driven expression changes for 38% (162/423) of expressed X chromosome genes. By integrating allele-specific analyses, we found that modulation of Xa transcript levels by Xi contributes to many of these Xi-driven changes (≥121 genes). By incorporating metrics of evolutionary constraint, we identified 10 X chromosome genes most likely to drive sex differences in common disease and sex chromosome aneuploidy syndromes. We conclude that human X chromosomes are regulated both in cis, through Xi-wide transcriptional attenuation, and in trans, through positive or negative modulation of individual Xa genes by Xi. The sum of these cis and trans effects differs widely among genes.
- Published
- 2023
- Full Text
- View/download PDF
30. Nance–Horan syndrome pedigree due to a novel microdeletion and skewed X chromosome inactivation
- Author
-
Yazhou Huang, Linya Ma, Zhaoxia Zhang, Shujuan Nie, Yuan Zhou, Jibo Zhang, Chao Wang, Xingxin Fang, Yingting Quan, Ting He, Anhui Liu, and Dan Peng
- Subjects
copy number variation sequencing ,dense congenital cataracts ,genetic counseling ,Nance–Horan syndrome ,X chromosome inactivation ,Genetics ,QH426-470 - Abstract
Abstract Background Nance–Horan syndrome (NHS) is a rare and often overlooked X‐linked dominant disorder characterized by dense congenital cataracts, dental abnormalities, and mental retardation. The majority of NHS variations include frameshift mutations, nonsense mutations, microdeletions, and insertions. Methods Copy number variation sequencing was performed to determine the microdeletion. The expression of NHS was detected by RT‐PCR. Four family members were tested for X chromosome inactivation. Results In this study, all members were examined for systemic examinations and genetic testing of four members and two affected subjects are observed. We identified a heterozygous microdeletion of −0.52 Mb at Xp22.13 in a female proband presenting NHS phenotypically. The microdeletion contains the REPS2 and NHS genes and was inherited from a phenotypically normal mother. Of interest, the expression NHS of proband was reduced and the skewed X chromosome inactivation rate reached more than 85% compared with her mother and the control. It was concluded that the haploinsufficiency of the NHS gene may account for the majority of clinical symptoms in the affected subjects. The variability among female carriers presumably results from nonrandom X chromosome inactivation. Conclusion Our findings broaden the spectrum of NHS mutations and provide molecular insight into NHS clinical prenatal genetic diagnosis.
- Published
- 2023
- Full Text
- View/download PDF
31. The Role of Xist in X-Chromosome Dosage Compensation
- Author
-
Sahakyan, Anna, Yang, Yihao, and Plath, Kathrin
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Human Genome ,Women's Health ,Pediatric ,Stem Cell Research ,Genetics ,1.1 Normal biological development and functioning ,Generic health relevance ,Animals ,Humans ,Mice ,RNA ,Long Noncoding ,X Chromosome ,X Chromosome Inactivation ,X inactivation ,Xist ,dosage compensation ,embryonic stem cells ,lncRNAs ,Medical and Health Sciences ,Developmental Biology ,Biochemistry and cell biology - Abstract
In each somatic cell of a female mammal one X chromosome is transcriptionally silenced via X-chromosome inactivation (XCI), initiating early in development. Although XCI events are conserved in mouse and human postimplantation development, regulation of X-chromosome dosage in preimplantation development occurs differently. In preimplantation development, mouse embryos undergo imprinted form of XCI, yet humans lack imprinted XCI and instead regulate gene expression of both X chromosomes by dampening transcription. The long non-coding RNA Xist/XIST is expressed in mouse and human preimplantation and postimplantation development to orchestrate XCI, but its role in dampening is unclear. In this review, we discuss recent advances in our understanding of the role of Xist in X chromosome dosage compensation in mouse and human.
- Published
- 2018
32. Sex differences in obesity, lipid metabolism, and inflammation—A role for the sex chromosomes?
- Author
-
Zore, Temeka, Palafox, Maria, and Reue, Karen
- Subjects
Biological Sciences ,Genetics ,Nutrition ,Obesity ,Aetiology ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,Animals ,Humans ,Inflammation ,Lipid Metabolism ,Sex Characteristics ,Sex Chromosomes ,X Chromosome Inactivation ,Gonadal hormones ,Adipose tissue ,Sex chromosome anomalies ,X chromosome inactivation ,Mouse models ,Biochemistry and Cell Biology ,Physiology ,Biochemistry and cell biology - Abstract
BackgroundSex differences in obesity and related diseases are well established. Gonadal hormones are a major determinant of these sex differences. However, sex differences in body size and composition are evident prior to exposure to gonadal hormones, providing evidence for gonadal-independent contributions attributable to the XX or XY sex chromosome complement. Large-scale genetic studies have revealed male/female differences in the genetic architecture of adipose tissue amount and anatomical distribution. However, these studies have typically neglected the X and Y chromosomes.Scope of the reviewHere we discuss how the sex chromosome complement may influence obesity, lipid levels, and inflammation. Human sex chromosome anomalies such as Klinefelter syndrome (XXY), as well as mouse models with engineered alterations in sex chromosome complement, support an important role for sex chromosomes in obesity and metabolism. In particular, the Four Core Genotypes mouse model-consisting of XX mice with either ovaries or testes, and XY mice with either ovaries or testes-has revealed an effect of X chromosome dosage on adiposity, hyperlipidemia, and inflammation irrespective of male or female gonads. Mechanisms may include enhanced expression of genes that escape X chromosome inactivation.Major conclusionsAlthough less well studied than effects of gonadal hormones, sex chromosomes exert independent and interactive effects on adiposity, lipid metabolism, and inflammation. In particular, the presence of two X chromosomes has been associated with increased adiposity and dyslipidemia in mouse models and in XXY men. The enhanced expression of genes that escape X chromosome inactivation may contribute, but more work is required.
- Published
- 2018
33. X Chromosome Dosage Influences DNA Methylation Dynamics during Reprogramming to Mouse iPSCs
- Author
-
Pasque, Vincent, Karnik, Rahul, Chronis, Constantinos, Petrella, Paula, Langerman, Justin, Bonora, Giancarlo, Song, Juan, Vanheer, Lotte, Dimashkie, Anupama Sadhu, Meissner, Alexander, and Plath, Kathrin
- Subjects
Biological Sciences ,Genetics ,Stem Cell Research ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Regenerative Medicine ,Stem Cell Research - Embryonic - Human ,Women's Health ,Stem Cell Research - Embryonic - Non-Human ,Human Genome ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research - Induced Pluripotent Stem Cell - Non-Human ,1.1 Normal biological development and functioning ,Generic health relevance ,Animals ,Binding Sites ,Cellular Reprogramming ,Chromosomes ,Mammalian ,CpG Islands ,DNA Methylation ,Embryonic Stem Cells ,Enhancer Elements ,Genetic ,Female ,Genome ,Genomic Imprinting ,Induced Pluripotent Stem Cells ,Male ,Mice ,Transcription Factors ,X Chromosome ,DNA methylation ,ESC ,X chromosome inactivation ,embryonic stem cells ,epigenetics ,iPSC ,induced pluripotency ,pluripotency ,reprogramming ,Biochemistry and Cell Biology ,Clinical Sciences ,Biochemistry and cell biology - Abstract
A dramatic difference in global DNA methylation between male and female cells characterizes mouse embryonic stem cells (ESCs), unlike somatic cells. We analyzed DNA methylation changes during reprogramming of male and female somatic cells and in resulting induced pluripotent stem cells (iPSCs). At an intermediate reprogramming stage, somatic and pluripotency enhancers are targeted for partial methylation and demethylation. Demethylation within pluripotency enhancers often occurs at ESC binding sites of pluripotency transcription factors. Late in reprogramming, global hypomethylation is induced in a female-specific manner. Genome-wide hypomethylation in female cells affects many genomic landmarks, including enhancers and imprint control regions, and accompanies the reactivation of the inactive X chromosome. The loss of one of the two X chromosomes in propagating female iPSCs is associated with genome-wide methylation gain. Collectively, our findings highlight the dynamic regulation of DNA methylation at enhancers during reprogramming and reveal that X chromosome dosage dictates global DNA methylation levels in iPSCs.
- Published
- 2018
34. Evaluation of X chromosome inactivation in endemic Tunisian pemphigus foliaceus
- Author
-
Olfa Abida, Nesrine Elloumi, Emna Bahloul, Hend Hachicha, Khadija Sellami, Raouia Fakhfakh, Sameh Marzouk, Ikhlas Ben Ayed, Nadia Mahfoudh, Hamida Turki, and Hatem Masmoudi
- Subjects
autoimmune disease ,pemphigus foliaceus ,X chromosome inactivation ,Genetics ,QH426-470 - Abstract
Abstract Background Almost 5% of the world's population develops an autoimmune disease (AID), it is considered the fourth leading cause of disability for women, who represent 78% of cases. The sex ratio when it comes to the most prevalent AID varies from 9:1 in systemic lupus erythematosus (SLE) to 13:1 in endemic Tunisian pemphigus foliaceus (PF). Methods To test the potential involvement of skewed x‐inactivation in the pathogenesis of Tunisian PF, we analyzed the methylation status of a highly polymorphic CAG repeat in the androgen receptor gene and evaluated the x chromosome inactivation (XCI) patterns in peripheral blood‐leukocyte‐derived DNA samples of female patients with PF (n = 98) compared to healthy control (HC) subjects (n = 150), as well as female patients with SLE (n = 98) were enrolled as a reference group. Results XCI status was informative for 50 of the 98 PF patients (51%) and 70 of the 150 HC women (47%). Extremely skewed XCI patterns were more frequent in PF and SLEwomen than HC, but the difference was statistically significant only in women with SLE. No statistical difference was observed in XCI patterns between PF and SLE patients. PF phenotype‐XCI correlation analysis revealed that (i) skewed XCI patterns may be involved in the disease's subtype and (ii) it was more pronounced in the endemic group than the sporadic one. Furthermore, preferential XCI showed an increase in heterozygote genotypes of PF's susceptibility polymorphisms in immunity‐related X genes (FOXP3, AR, and TLR7) in PF patients compared to HC. Conclusion Our results suggest that skewed XCI could lead to hemizygosity of X‐linked alleles that might unmask X‐linked deleterious alleles.
- Published
- 2022
- Full Text
- View/download PDF
35. Regulation of X-chromosome dosage compensation in human: mechanisms and model systems
- Author
-
Sahakyan, Anna, Plath, Kathrin, and Rougeulle, Claire
- Subjects
Biochemistry and Cell Biology ,Reproductive Medicine ,Biomedical and Clinical Sciences ,Biological Sciences ,Human Genome ,Genetics ,1.1 Normal biological development and functioning ,Generic health relevance ,Reproductive health and childbirth ,Animals ,Chromosomes ,Human ,X ,Dosage Compensation ,Genetic ,Embryonic Development ,Female ,Humans ,Male ,Mice ,X Chromosome ,X Chromosome Inactivation ,X-chromosome inactivation ,pluripotent stem cells ,X-chromosome dampening ,Xist ,Xact ,Medical and Health Sciences ,Evolutionary Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
The human blastocyst forms 5 days after one of the smallest human cells (the sperm) fertilizes one of the largest human cells (the egg). Depending on the sex-chromosome contribution from the sperm, the resulting embryo will either be female, with two X chromosomes (XX), or male, with an X and a Y chromosome (XY). In early development, one of the major differences between XX female and XY male embryos is the conserved process of X-chromosome inactivation (XCI), which compensates gene expression of the two female X chromosomes to match the dosage of the single X chromosome of males. Most of our understanding of the pre-XCI state and XCI establishment is based on mouse studies, but recent evidence from human pre-implantation embryo research suggests that many of the molecular steps defined in the mouse are not conserved in human. Here, we will discuss recent advances in understanding the control of X-chromosome dosage compensation in early human embryonic development and compare it to that of the mouse.This article is part of the themed issue 'X-chromosome inactivation: a tribute to Mary Lyon'.
- Published
- 2017
36. Landscape of X chromosome inactivation across human tissues
- Author
-
Aguet, François, Ardlie, Kristin G, Cummings, Beryl B, Gelfand, Ellen T, Getz, Gad, Hadley, Kane, Handsaker, Robert E, Huang, Katherine H, Kashin, Seva, Karczewski, Konrad J, Lek, Monkol, Li, Xiao, MacArthur, Daniel G, Nedzel, Jared L, Nguyen, Duyen T, Noble, Michael S, Segrè, Ayellet V, Trowbridge, Casandra A, Tukiainen, Taru, Abell, Nathan S, Balliu, Brunilda, Barshir, Ruth, Basha, Omer, Battle, Alexis, Bogu, Gireesh K, Brown, Andrew, Brown, Christopher D, Castel, Stephane E, Chen, Lin S, Chiang, Colby, Conrad, Donald F, Cox, Nancy J, Damani, Farhan N, Davis, Joe R, Delaneau, Olivier, Dermitzakis, Emmanouil T, Engelhardt, Barbara E, Eskin, Eleazar, Ferreira, Pedro G, Frésard, Laure, Gamazon, Eric R, Garrido-Martín, Diego, Gewirtz, Ariel DH, Gliner, Genna, Gloudemans, Michael J, Guigo, Roderic, Hall, Ira M, Han, Buhm, He, Yuan, Hormozdiari, Farhad, Howald, Cedric, Kyung Im, Hae, Jo, Brian, Yong Kang, Eun, Kim, Yungil, Kim-Hellmuth, Sarah, Lappalainen, Tuuli, Li, Gen, Li, Xin, Liu, Boxiang, Mangul, Serghei, McCarthy, Mark I, McDowell, Ian C, Mohammadi, Pejman, Monlong, Jean, Montgomery, Stephen B, Muñoz-Aguirre, Manuel, Ndungu, Anne W, Nicolae, Dan L, Nobel, Andrew B, Oliva, Meritxell, Ongen, Halit, Palowitch, John J, Panousis, Nikolaos, Papasaikas, Panagiotis, Park, YoSon, Parsana, Princy, Payne, Anthony J, Peterson, Christine B, Quan, Jie, Reverter, Ferran, Sabatti, Chiara, Saha, Ashis, Sammeth, Michael, Scott, Alexandra J, Shabalin, Andrey A, Sodaei, Reza, Stephens, Matthew, Stranger, Barbara E, Strober, Benjamin J, Sul, Jae Hoon, Tsang, Emily K, Urbut, Sarah, van de Bunt, Martijn, Wang, Gao, Wen, Xiaoquan, Wright, Fred A, Xi, Hualin S, Yeger-Lotem, Esti, and Zappala, Zachary
- Subjects
Genetics ,Clinical Research ,Human Genome ,Generic health relevance ,Good Health and Well Being ,Chromosomes ,Human ,X ,Female ,Genes ,X-Linked ,Genome ,Human ,Genomics ,Humans ,Male ,Organ Specificity ,Phenotype ,Sequence Analysis ,RNA ,Single-Cell Analysis ,Transcriptome ,X Chromosome Inactivation ,GTEx Consortium ,Laboratory ,Data Analysis &Coordinating Center (LDACC)—Analysis Working Group ,Statistical Methods groups—Analysis Working Group ,Enhancing GTEx (eGTEx) groups ,NIH Common Fund ,NIH/NCI ,NIH/NHGRI ,NIH/NIMH ,NIH/NIDA ,Biospecimen Collection Source Site—NDRI ,Biospecimen Collection Source Site—RPCI ,Biospecimen Core Resource—VARI ,Brain Bank Repository—University of Miami Brain Endowment Bank ,Leidos Biomedical—Project Management ,ELSI Study ,Genome Browser Data Integration &Visualization—EBI ,Genome Browser Data Integration &Visualization—UCSC Genomics Institute ,University of California Santa Cruz ,General Science & Technology - Abstract
X chromosome inactivation (XCI) silences transcription from one of the two X chromosomes in female mammalian cells to balance expression dosage between XX females and XY males. XCI is, however, incomplete in humans: up to one-third of X-chromosomal genes are expressed from both the active and inactive X chromosomes (Xa and Xi, respectively) in female cells, with the degree of 'escape' from inactivation varying between genes and individuals. The extent to which XCI is shared between cells and tissues remains poorly characterized, as does the degree to which incomplete XCI manifests as detectable sex differences in gene expression and phenotypic traits. Here we describe a systematic survey of XCI, integrating over 5,500 transcriptomes from 449 individuals spanning 29 tissues from GTEx (v6p release) and 940 single-cell transcriptomes, combined with genomic sequence data. We show that XCI at 683 X-chromosomal genes is generally uniform across human tissues, but identify examples of heterogeneity between tissues, individuals and cells. We show that incomplete XCI affects at least 23% of X-chromosomal genes, identify seven genes that escape XCI with support from multiple lines of evidence and demonstrate that escape from XCI results in sex biases in gene expression, establishing incomplete XCI as a mechanism that is likely to introduce phenotypic diversity. Overall, this updated catalogue of XCI across human tissues helps to increase our understanding of the extent and impact of the incompleteness in the maintenance of XCI.
- Published
- 2017
37. Allele-specific non-CG DNA methylation marks domains of active chromatin in female mouse brain
- Author
-
Keown, Christopher L, Berletch, Joel B, Castanon, Rosa, Nery, Joseph R, Disteche, Christine M, Ecker, Joseph R, and Mukamel, Eran A
- Subjects
Genetics ,Human Genome ,Stem Cell Research ,1.1 Normal biological development and functioning ,Underpinning research ,Generic health relevance ,Alleles ,Animals ,Brain ,Chromatin ,DNA Methylation ,Epigenesis ,Genetic ,Female ,Genomic Imprinting ,Male ,Mice ,Inbred C57BL ,Mice ,Mutant Strains ,Polymorphism ,Single Nucleotide ,RNA ,Long Noncoding ,X Chromosome ,X Chromosome Inactivation ,DNA methylation ,X-chromosome inactivation ,imprinting ,Non-CG ,Bcor - Abstract
DNA methylation at gene promoters in a CG context is associated with transcriptional repression, including at genes silenced on the inactive X chromosome in females. Non-CG methylation (mCH) is a distinct feature of the neuronal epigenome that is differentially distributed between males and females on the X chromosome. However, little is known about differences in mCH on the active (Xa) and inactive (Xi) X chromosomes because stochastic X-chromosome inactivation (XCI) confounds allele-specific epigenomic profiling. We used whole-genome bisulfite sequencing in a mouse model with nonrandom XCI to examine allele-specific DNA methylation in frontal cortex. Xi was largely devoid of mCH, whereas Xa contained abundant mCH similar to the male X chromosome and the autosomes. In contrast to the repressive association of DNA methylation at CG dinucleotides (mCG), mCH accumulates on Xi in domains with transcriptional activity, including the bodies of most genes that escape XCI and at the X-inactivation center, validating this epigenetic mark as a signature of transcriptional activity. Escape genes showing CH hypermethylation were the only genes with CG-hypomethylated promoters on Xi, a well-known mark of active transcription. Finally, we found extensive allele-specific mCH and mCG at autosomal imprinted regions, some with a negative correlation between methylation in the two contexts, further supporting their distinct functions. Our findings show that neuronal mCH functions independently of mCG and is a highly dynamic epigenomic correlate of allele-specific gene regulation.
- Published
- 2017
38. Emerging X-linked genes associated with neurodevelopmental disorders in females.
- Author
-
Lukin, Jeronimo, Smith, Corinne M., and De Rubeis, Silvia
- Subjects
- *
X chromosome , *AUTISM spectrum disorders , *INTELLECTUAL disabilities , *GENETICS , *NEURAL development - Abstract
A significant source of risk for neurodevelopmental disorders (NDDs), including intellectual disability (ID) and autism spectrum disorder (ASD), lies in genes located on the X chromosome. Males can be particularly vulnerable to X-linked variation because of hemizygosity, and male-specific segregation in pedigrees has guided earlier gene discovery for X-linked recessive conditions. More recently, X-linked disorders disproportionally affecting females, with complex inheritance patterns and/or presenting with sex differences, have surfaced. Here, we discuss the genetics and neurobiology of X-linked genes that are paradigmatic to understand NDDs in females. Integrating genetic, clinical, and functional data will be key to understand how X-linked variation contributes to the risk architecture of NDDs. • Chromosome X is enriched for genes associated with neurodevelopmental disorders. • X chromosome inactivation evolved to harmonize gene dosage across sexes. • Genes can escape inactivation, contributing to sex differences in neurodevelopment. • X-linked neurodevelopmental disorders more prevalent in females have emerged. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
39. Human Naive Pluripotent Stem Cells Model X Chromosome Dampening and X Inactivation
- Author
-
Sahakyan, Anna, Kim, Rachel, Chronis, Constantinos, Sabri, Shan, Bonora, Giancarlo, Theunissen, Thorold W, Kuoy, Edward, Langerman, Justin, Clark, Amander T, Jaenisch, Rudolf, and Plath, Kathrin
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Stem Cell Research - Embryonic - Human ,Regenerative Medicine ,Stem Cell Research ,Genetics ,Human Genome ,1.1 Normal biological development and functioning ,Generic health relevance ,Base Sequence ,Blastocyst ,Cell Differentiation ,Cells ,Cultured ,Chromosomes ,Human ,X ,DNA Methylation ,Female ,Histones ,Human Embryonic Stem Cells ,Humans ,Lysine ,Methylation ,Pluripotent Stem Cells ,RNA ,Long Noncoding ,X Chromosome Inactivation ,X chromosome ,X chromosome dampening ,X chromosome inactivation ,XIST ,embryonic stem cells ,human development ,human stem cells ,lncRNA ,naive pluripotency ,pluripotent stem cells ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Naive human embryonic stem cells (hESCs) can be derived from primed hESCs or directly from blastocysts, but their X chromosome state has remained unresolved. Here, we show that the inactive X chromosome (Xi) of primed hESCs was reactivated in naive culture conditions. Like cells of the blastocyst, the resulting naive cells contained two active X chromosomes with XIST expression and chromosome-wide transcriptional dampening and initiated XIST-mediated X inactivation upon differentiation. Both establishment of and exit from the naive state (differentiation) happened via an XIST-negative XaXa intermediate. Together, these findings identify a cell culture system for functionally exploring the two X chromosome dosage compensation processes in early human development: X dampening and X inactivation. However, remaining differences between naive hESCs and embryonic cells related to mono-allelic XIST expression and non-random X inactivation highlight the need for further culture improvement. As the naive state resets Xi abnormalities seen in primed hESCs, it may provide cells better suited for downstream applications.
- Published
- 2017
40. Human Embryonic Stem Cells Do Not Change Their X Inactivation Status during Differentiation
- Author
-
Patel, Sanjeet, Bonora, Giancarlo, Sahakyan, Anna, Kim, Rachel, Chronis, Constantinos, Langerman, Justin, Fitz-Gibbon, Sorel, Rubbi, Liudmilla, Skelton, Rhys JP, Ardehali, Reza, Pellegrini, Matteo, Lowry, William E, Clark, Amander T, and Plath, Kathrin
- Subjects
Biological Sciences ,Genetics ,Human Genome ,Stem Cell Research - Embryonic - Human ,Stem Cell Research ,Regenerative Medicine ,Stem Cell Research - Embryonic - Non-Human ,Cell Differentiation ,Cell Line ,DNA Methylation ,Female ,Gene Expression Regulation ,Gene Silencing ,Human Embryonic Stem Cells ,Humans ,In Situ Hybridization ,Fluorescence ,Induced Pluripotent Stem Cells ,Male ,Sequence Analysis ,RNA ,Tretinoin ,X Chromosome Inactivation ,DNA methylation ,X-chromosome dosage compensation ,X-inactivation ,Xi-erosion ,Xist ,human embryonic stem cells ,human induced pluripotent stem cells ,inactive X chromosome ,Biochemistry and Cell Biology ,Medical Physiology ,Biological sciences - Abstract
Applications of embryonic stem cells (ESCs) require faithful chromatin changes during differentiation, but the fate of the X chromosome state in differentiating ESCs is unclear. Female human ESC lines either carry two active X chromosomes (XaXa), an Xa and inactive X chromosome with or without XIST RNA coating (XiXIST+Xa;XiXa), or an Xa and an eroded Xi (XeXa) where the Xi no longer expresses XIST RNA and has partially reactivated. Here, we established XiXa, XeXa, and XaXa ESC lines and followed their X chromosome state during differentiation. Surprisingly, we found that the X state pre-existing in primed ESCs is maintained in differentiated cells. Consequently, differentiated XeXa and XaXa cells lacked XIST, did not induce X inactivation, and displayed higher X-linked gene expression than XiXa cells. These results demonstrate that X chromosome dosage compensation is not required for ESC differentiation. Our data imply that XiXIST+Xa ESCs are most suited for downstream applications and show that all other X states are abnormal byproducts of our ESC derivation and propagation method.
- Published
- 2017
41. A self-enhanced transport mechanism through long noncoding RNAs for X chromosome inactivation.
- Author
-
Li, Chunhe, Hong, Tian, Webb, Chiu-Ho, Karner, Heather, Sun, Sha, and Nie, Qing
- Subjects
Animals ,Mice ,Signal Transduction ,Biological Transport ,Cell Lineage ,Female ,Male ,X Chromosome Inactivation ,Embryonic Stem Cells ,RNA ,Long Noncoding ,Genetics ,Human Genome ,1.1 Normal biological development and functioning ,Generic Health Relevance ,RNA ,Long Noncoding ,Biochemistry and Cell Biology ,Other Physical Sciences - Abstract
X-chromosome inactivation (XCI) is the mammalian dosage compensation strategy for balancing sex chromosome content between females and males. While works exist on initiation of symmetric breaking, the underlying allelic choice mechanisms and dynamic regulation responsible for the asymmetric fate determination of XCI remain elusive. Here we combine mathematical modeling and experimental data to examine the mechanism of XCI fate decision by analyzing the signaling regulatory circuit associated with long noncoding RNAs (lncRNAs) involved in XCI. We describe three plausible gene network models that incorporate features of lncRNAs in their localized actions and rapid transcriptional turnovers. In particular, we show experimentally that Jpx (a lncRNA) is transcribed biallelically, escapes XCI, and is asymmetrically dispersed between two X's. Subjecting Jpx to our test of model predictions against previous experimental observations, we identify that a self-enhanced transport feedback mechanism is critical to XCI fate decision. In addition, the analysis indicates that an ultrasensitive response of Jpx signal on CTCF is important in this mechanism. Overall, our combined modeling and experimental data suggest that the self-enhanced transport regulation based on allele-specific nature of lncRNAs and their temporal dynamics provides a robust and novel mechanism for bi-directional fate decisions in critical developmental processes.
- Published
- 2016
42. Plasticity in the Meiotic Epigenetic Landscape of Sex Chromosomes in Caenorhabditis Species
- Author
-
Larson, Braden J, Van, Mike V, Nakayama, Taylor, and Engebrecht, JoAnne
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Contraception/Reproduction ,Genetics ,Human Genome ,1.1 Normal biological development and functioning ,Generic health relevance ,Animals ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,Chromatin Assembly and Disassembly ,Epigenomics ,Evolution ,Molecular ,Germ Cells ,Histone Demethylases ,Histone-Lysine N-Methyltransferase ,Meiosis ,Mutation ,Sex Chromosomes ,X Chromosome Inactivation ,genetics of sex ,histone methyltransferases ,meiosis ,MSCI ,MSUC ,sex chromosomes ,Developmental Biology ,Biochemistry and cell biology - Abstract
During meiosis in the heterogametic sex in some species, sex chromosomes undergo meiotic sex chromosome inactivation (MSCI), which results in acquisition of repressive chromatin and transcriptional silencing. In Caenorhabditis elegans, MSCI is mediated by MET-2 methyltransferase deposition of histone H3 lysine 9 dimethylation. Here we examined the meiotic chromatin landscape in germ lines of four Caenorhabditis species; C. remanei and C. brenneri represent ancestral gonochorism, while C. briggsae and C. elegans are two lineages that independently evolved hermaphroditism. While MSCI is conserved across all four species, repressive chromatin modifications are distinct and do not correlate with reproductive mode. In contrast to C. elegans and C. remanei germ cells where X chromosomes are enriched for histone H3 lysine 9 dimethylation, X chromosomes in C. briggsae and C. brenneri germ cells are enriched for histone H3 lysine 9 trimethylation. Inactivation of C. briggsae MET-2 resulted in germ-line X chromosome transcription and checkpoint activation. Further, both histone H3 lysine 9 di- and trimethylation were reduced in Cbr-met-2 mutant germ lines, suggesting that in contrast to C. elegans, H3 lysine 9 di- and trimethylation are interdependent. C. briggsae H3 lysine 9 trimethylation was redistributed in the presence of asynapsed chromosomes in a sex-specific manner in the related process of meiotic silencing of unsynapsed chromatin. However, these repressive marks did not influence X chromosome replication timing. Examination of additional Caenorhabditis species revealed diverse H3 lysine 9 methylation patterns on the X, suggesting that the sex chromosome epigenome evolves rapidly.
- Published
- 2016
43. The “lnc” between 3D chromatin structure and X chromosome inactivation
- Author
-
Pandya-Jones, Amy and Plath, Kathrin
- Subjects
Biological Sciences ,Genetics ,Human Genome ,Generic health relevance ,Animals ,Chromatin ,Humans ,Models ,Genetic ,Nuclear Proteins ,RNA ,Long Noncoding ,X Chromosome Inactivation ,X chromosome inactivation Xist ,Chromosome conformation ,Non-coding RNA ,Heterochromatin ,Biochemistry and Cell Biology ,Paediatrics and Reproductive Medicine ,Developmental Biology ,Biochemistry and cell biology - Abstract
The long non-coding RNA Xist directs a remarkable instance of developmentally regulated, epigenetic change known as X Chromosome Inactivation (XCI). By spreading in cis across the X chromosome from which it is expressed, Xist RNA facilitates the creation of a heritably silent, heterochromatic nuclear territory that displays a three-dimensional structure distinct from that of the active X chromosome. How Xist RNA attaches to and propagates across a chromosome and its influence over the three-dimensional (3D) structure of the inactive X are aspects of XCI that have remained largely unclear. Here, we discuss studies that have made significant contributions towards answering these open questions.
- Published
- 2016
44. Impaired imprinted X chromosome inactivation is responsible for the skewed sex ratio following in vitro fertilization
- Author
-
Tan, Kun, An, Lei, Miao, Kai, Ren, Likun, Hou, Zhuocheng, Tao, Li, Zhang, Zhenni, Wang, Xiaodong, Xia, Wei, Liu, Jinghao, Wang, Zhuqing, Xi, Guangyin, Gao, Shuai, Sui, Linlin, Zhu, De-Sheng, Wang, Shumin, Wu, Zhonghong, Bach, Ingolf, Chen, Dong-bao, and Tian, Jianhui
- Subjects
Contraception/Reproduction ,Genetics ,Underpinning research ,1.1 Normal biological development and functioning ,Reproductive health and childbirth ,Chromosomes ,Human ,X ,Female ,Fertilization in Vitro ,Genomic Imprinting ,Humans ,Sex Ratio ,X Chromosome Inactivation ,in vitro fertilization ,sex ratio ,X chromosome inactivation ,Xist ,Rnf12 - Abstract
Dynamic epigenetic reprogramming occurs during normal embryonic development at the preimplantation stage. Erroneous epigenetic modifications due to environmental perturbations such as manipulation and culture of embryos during in vitro fertilization (IVF) are linked to various short- or long-term consequences. Among these, the skewed sex ratio, an indicator of reproductive hazards, was reported in bovine and porcine embryos and even human IVF newborns. However, since the first case of sex skewing reported in 1991, the underlying mechanisms remain unclear. We reported herein that sex ratio is skewed in mouse IVF offspring, and this was a result of female-biased peri-implantation developmental defects that were originated from impaired imprinted X chromosome inactivation (iXCI) through reduced ring finger protein 12 (Rnf12)/X-inactive specific transcript (Xist) expression. Compensation of impaired iXCI by overexpression of Rnf12 to up-regulate Xist significantly rescued female-biased developmental defects and corrected sex ratio in IVF offspring. Moreover, supplementation of an epigenetic modulator retinoic acid in embryo culture medium up-regulated Rnf12/Xist expression, improved iXCI, and successfully redeemed the skewed sex ratio to nearly 50% in mouse IVF offspring. Thus, our data show that iXCI is one of the major epigenetic barriers for the developmental competence of female embryos during preimplantation stage, and targeting erroneous epigenetic modifications may provide a potential approach for preventing IVF-associated complications.
- Published
- 2016
45. Stochastic gene expression and chromosome interactions in protecting the human active X from silencing by XIST
- Author
-
Barbara R. Migeon
- Subjects
x chromosome inactivation ,protecting the active x from xist silencing ,stochastic gene expression ,3-d nuclear interactions ,x/autosome interactions ,19p trisomies ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Mammals use X chromosome inactivation to compensate for the sex difference in numbers of X chromosomes. A relatively unexplored question is how the active X is protected from inactivation by its own XIST gene, the long non-coding RNA, which initiates silence of the inactive X. Previous studies of autosomal duplications show that human chromosome 19 plays a critical role in protecting the active X. I proposed that it genetically interacts with the X chromosome to repress XIST function on the future active X. Here, I show that the type of chromosome 19 duplication influences the outcome of the interaction: the presence of three chromosome 19s is tolerated whereas duplications affecting only one chromosome 19 are not. The different outcomes have mechanistic implications for how chromosome 19 interacts with the future active X, pointing to a role for stochastic gene expression and possibly physical interaction.
- Published
- 2021
- Full Text
- View/download PDF
46. X Chromosome Inactivation Timing is Not eXACT: Implications for Autism Spectrum Disorders
- Author
-
Janine M. LaSalle
- Subjects
DNA methylation ,X chromosome inactivation ,peri-implantation development ,autism ,neurodevelomental disorders ,Genetics ,QH426-470 - Abstract
The etiology of autism spectrum disorders (ASD) is complex, involving different combinations of genetic and environmental factors. My lab’s approach has been to investigate DNA methylation as a tractable genome-wide modification at the interface of these complex interactions, reflecting past and future events in the molecular pathogenesis of ASD. Since X-linked genes were enriched in DNA methylation differences discovered from cord blood from newborns later diagnosed with ASD, this has prompted me to review and revisit the recent advancements in the field of X chromosome inactivation (XCI), particularly in humans and other primates. In this Perspective, I compare XCI mechanisms in different mammalian species, including the finding of the noncoding transcript XACT associated with X chromosome erosion in human pluripotent stem cells and recent findings from non-human primate post-implantation embryos. I focus on the experimentally challenging peri- and post-implantation stages of human development when the timing of XCI is prolonged and imprecise in humans. Collectively, this research has raised some important unanswered questions involving biased sex ratios in human births and the male bias in the incidence of ASD.
- Published
- 2022
- Full Text
- View/download PDF
47. Poised chromatin and bivalent domains facilitate the mitosis-to-meiosis transition in the male germline
- Author
-
Sin, Ho-Su, Kartashov, Andrey V, Hasegawa, Kazuteru, Barski, Artem, and Namekawa, Satoshi H
- Subjects
Regenerative Medicine ,Genetics ,Biotechnology ,Stem Cell Research - Nonembryonic - Non-Human ,Contraception/Reproduction ,Stem Cell Research ,Underpinning research ,1.1 Normal biological development and functioning ,Animals ,Cells ,Cultured ,Chromatin ,Epigenesis ,Genetic ,Gene Expression Regulation ,Developmental ,Genes ,X-Linked ,Germ Cells ,Histones ,Male ,Meiosis ,Mice ,Inbred C57BL ,Mitosis ,Sex Chromosomes ,Spermatogenesis ,Transcriptional Activation ,Transcriptome ,X Chromosome Inactivation ,Developmental Biology - Abstract
BackgroundThe male germline transcriptome changes dramatically during the mitosis-to-meiosis transition to activate late spermatogenesis genes and to transiently suppress genes commonly expressed in somatic lineages and spermatogenesis progenitor cells, termed somatic/progenitor genes.ResultsThese changes reflect epigenetic regulation. Induction of late spermatogenesis genes during spermatogenesis is facilitated by poised chromatin established in the stem cell phases of spermatogonia, whereas silencing of somatic/progenitor genes during meiosis and postmeiosis is associated with formation of bivalent domains which also allows the recovery of the somatic/progenitor program after fertilization. Importantly, during spermatogenesis mechanisms of epigenetic regulation on sex chromosomes are different from autosomes: X-linked somatic/progenitor genes are suppressed by meiotic sex chromosome inactivation without deposition of H3K27me3.ConclusionsOur results suggest that bivalent H3K27me3 and H3K4me2/3 domains are not limited to developmental promoters (which maintain bivalent domains that are silent throughout the reproductive cycle), but also underlie reversible silencing of somatic/progenitor genes during the mitosis-to-meiosis transition in late spermatogenesis.
- Published
- 2015
48. X chromosome reactivation in reprogramming and in development
- Author
-
Pasque, Vincent and Plath, Kathrin
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Stem Cell Research ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research - Embryonic - Human ,Regenerative Medicine ,Human Genome ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research - Induced Pluripotent Stem Cell - Non-Human ,Genetics ,Generic health relevance ,Animals ,Cell Differentiation ,Cellular Reprogramming ,Humans ,Models ,Biological ,Pluripotent Stem Cells ,X Chromosome ,X Chromosome Inactivation ,Developmental Biology ,Biochemistry and cell biology - Abstract
Dramatic epigenetic changes take place during mammalian differentiation from the naïve pluripotent state including the silencing of one of the two X chromosomes in female cells through X chromosome inactivation. Conversely, reprogramming of somatic cells to naive pluripotency is coupled to X chromosome reactivation (XCR). Recent studies in the mouse system have shed light on the mechanisms of XCR by uncovering the timing and steps of XCR during reprogramming to induced pluripotent stem cells (iPSCs), allowing the generation of testable hypotheses during embryogenesis. In contrast, analyses of the X chromosome in human iPSCs have revealed important differences between mouse and human reprogramming processes that can partially be explained by the establishment of distinct pluripotent states and impact disease modeling and the application of human pluripotent stem cells. Here, we review recent literature on XCR as a readout and determinant of reprogramming to pluripotency.
- Published
- 2015
49. Clinical characterization of int22h1/int22h2-mediated Xq28 duplication/deletion: new cases and literature review
- Author
-
El-Hattab, Ayman W, Schaaf, Christian P, Fang, Ping, Roeder, Elizabeth, Kimonis, Virginia E, Church, Joseph A, Patel, Ankita, and Cheung, Sau Wai
- Subjects
Brain Disorders ,Behavioral and Social Science ,Pediatric ,Mental Health ,Prevention ,Intellectual and Developmental Disabilities (IDD) ,Basic Behavioral and Social Science ,Genetics ,Aetiology ,2.1 Biological and endogenous factors ,Mental health ,Adolescent ,Adult ,Child ,Child ,Preschool ,Chromosome Deletion ,Chromosome Duplication ,Chromosomes ,Human ,X ,Female ,Humans ,Infant ,Introns ,Male ,Middle Aged ,Pedigree ,Sex Chromosome Disorders ,X Chromosome Inactivation ,Young Adult ,Chromosomal rearrangements ,X-linked intellectual disability ,Chromosomal microarray analysis ,Medical Biochemistry and Metabolomics ,Oncology and Carcinogenesis ,Genetics & Heredity - Abstract
BackgroundInt22h1/int22h2-mediated Xq28 duplication syndrome is caused by ~0.5 Mb chromosomal duplications mediated by nonallelic homologous recombination between intron 22 homologous region 1 (int22h1) and 2 (int22h2), which, in addition to int22h3, are also responsible for inversions disrupting the F8 gene in hemophilia A. This syndrome has recently been described in 9 males with cognitive impairment, behavioral problems, and distinctive facial features; and 6 females with milder phenotypes. The reciprocal deletion was previously reported in a mother and daughter. It was suggested that this deletion may not have phenotypic effects in females because of skewed chromosome X inactivation, but may be embryonic lethal in males.MethodsArray comparative genomic hybridization analyses were performed using oligonucleotide-based chromosomal microarray. Chromosome X inactivation studies were performed at the AR (androgen receptor) and FMR1 (fragile X mental retardation 1) loci.ResultsWe present here 5 males and 6 females with int22h1/int22h2-mediated Xq28 duplication syndrome. The males manifested cognitive impairment, behavioral problems, and distinctive facial features. Two of the six females manifested mild cognitive impairment. This duplication was maternally inherited, and skewed chromosome X inactivation was observed in the majority of females carrying the duplication. We also report the reciprocal deletion in a mother and daughter with overweight, but normal cognition. In addition, we present the first case of a prenatally diagnosed de novo int22h1/int22h2-mediated deletion in a healthy female infant. We reviewed individuals previously reported with similar or overlapping rearrangements and evaluated the potential roles of genes in the rearrangement region.ConclusionsThe similarity of clinical features among individuals with the int22h1/int22h2-mediated Xq28 duplication supports the notion that this duplication causes a recognizable syndrome that affects males with females exhibiting milder phenotypes. It is suggested that the observed cognitive impairment in this syndrome results from increased dosage of RAB39B gene located within the duplicated region. Increased dosage of CLIC2 may also contribute to the phenotype. The reciprocal deletion results in skewed chromosome X inactivation and no clinical phenotype in females. Review of overlapping deletions suggests that hemizygous loss of VBP1 may be the cause for the proposed male lethality associated with this deletion.
- Published
- 2015
50. A high-throughput screen of inactive X chromosome reactivation identifies the enhancement of DNA demethylation by 5-aza-2′-dC upon inhibition of ribonucleotide reductase
- Author
-
Minkovsky, Alissa, Sahakyan, Anna, Bonora, Giancarlo, Damoiseaux, Robert, Dimitrova, Elizabeth, Rubbi, Liudmilla, Pellegrini, Matteo, Radu, Caius G, and Plath, Kathrin
- Subjects
Biological Sciences ,Genetics ,Hematology ,Human Genome ,Rare Diseases ,Cancer ,X chromosome inactivation ,DNA methylation ,5-aza-2 '-dC ,Ribonucleotide reductase ,Hydroxyurea ,5-aza-2′-dC - Abstract
BackgroundDNA methylation is important for the maintenance of the silent state of genes on the inactive X chromosome (Xi). Here, we screened for siRNAs and chemicals that reactivate an Xi-linked reporter in the presence of 5-aza-2'-deoxycytidine (5-aza-2'-dC), an inhibitor of DNA methyltransferase 1, at a concentration that, on its own, is not sufficient for Xi-reactivation.ResultsWe found that inhibition of ribonucleotide reductase (RNR) induced expression of the reporter. RNR inhibition potentiated the effect of 5-aza-2'-dC by enhancing its DNA incorporation, thereby decreasing DNA methylation levels genome-wide. Since both 5-aza-2'-dC and RNR-inhibitors are used in the treatment of hematological malignancies, we treated myeloid leukemia cell lines with 5-aza-2'-dC and the RNR-inhibitor hydroxyurea, and observed synergistic inhibition of cell growth and a decrease in genome-wide DNA methylation.ConclusionsTaken together, our study identifies a drug combination that enhances DNA demethylation by altering nucleotide metabolism. This demonstrates that Xi-reactivation assays can be used to optimize the epigenetic activity of drug combinations.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.