Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2770: 53-62, 2024.
Article in English | MEDLINE | ID: mdl-38351446

ABSTRACT

In the adult mouse testis, germ cells of various developmental cell states co-exist. FACS isolation of cells stained with the DNA dye Hoechst 33342 has been used for many years to sub-divide these cells based on their DNA content. This approach provides an efficient way to obtain broad categories of male germ cells: pre-meiotic spermatogonia, meiotic spermatocytes and post-meiotic spermatids. The addition of a red filter for Hoechst staining enables further sub-division of spermatocytes depending on sub-stages of meiotic prophase. However, separation of different stage spermatids using Hoechst staining alone is not possible. We recently reported a methodology, combining Hoechst staining with a second DNA dye (SYTO16) that enables the further separation of these cells into three sub-populations: round, early elongating, and late elongating spermatids (Gill et al., Cytometry A 101:529-536, 2022). This method makes it possible to obtain rapidly and simply pure fractions of male germ cells from multiple developental stages from the same animal.


Subject(s)
Benzimidazoles , Spermatogenesis , Testis , Mice , Animals , Male , Meiosis , Spermatids , Spermatocytes , Germ Cells , Staining and Labeling , DNA
2.
Genome Res ; 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38129075

ABSTRACT

In mammals, the adult testis is the tissue with the highest diversity in gene expression. Much of that diversity is attributed to germ cells, primarily meiotic spermatocytes and postmeiotic haploid spermatids. Exploiting a newly developed cell purification method, we profiled the transcriptomes of such postmitotic germ cells of mice. We used a de novo transcriptome assembly approach and identified thousands of novel expressed transcripts characterized by features distinct from those of known genes. Novel loci tend to be short in length, monoexonic, and lowly expressed. Most novel genes have arisen recently in evolutionary time and possess low coding potential. Nonetheless, we identify several novel protein-coding genes harboring open reading frames that encode proteins containing matches to conserved protein domains. Analysis of mass-spectrometry data from adult mouse testes confirms protein production from several of these novel genes. We also examine overlap between transcripts and repetitive elements. We find that although distinct families of repeats are expressed with differing temporal dynamics during spermatogenesis, we do not observe a general mode of regulation wherein repeats drive expression of nonrepetitive sequences in a cell type-specific manner. Finally, we observe many fairly long antisense transcripts originating from canonical gene promoters, pointing to pervasive bidirectional promoter activity during spermatogenesis that is distinct and more frequent compared with somatic cells.

3.
Nature ; 605(7911): 621-622, 2022 05.
Article in English | MEDLINE | ID: mdl-35585262
4.
Cytometry A ; 101(6): 529-536, 2022 06.
Article in English | MEDLINE | ID: mdl-35128792

ABSTRACT

During spermatogenesis, mammalian male germ cells undergo multiple developmental processes, including meiosis and post-meiotic differentiation (spermiogenesis). To understand the transitions between different cellular states it is essential to isolate pure populations of cells at different stages of development. Previous approaches enabled the isolation of cells from different stages of meiotic prophase I, but techniques to sub-fractionate unfixed, post-meiotic spermatids have been lacking. Here we report the development of a protocol enabling simultaneous isolation of cells at different stages of meiotic prophase and post-meiotic differentiation from testes of adult mice. This approach builds on existing fluorescence activated cell sorting protocols designed to purify cells in different stages of meiotic prophase I. By utilizing the specific spectral properties that two different DNA dyes (Hoechst 33342 and SYTO 16) exhibit when bound to chromatin of different stage male germ cells, we obtain highly pure populations of cells in relatively large numbers. This FACS protocol will enable immunocytological and molecular characterization studies of fractionated meiotic and haploid germ cells from both wild type and genetically mutant animals.


Subject(s)
Meiosis , Spermatids , Animals , DNA/metabolism , Germ Cells/metabolism , Male , Mammals/genetics , Mice , Spermatogenesis/genetics , Staining and Labeling , Testis
5.
EMBO J ; 40(12): e106818, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33909924

ABSTRACT

Mouse embryonic stem cells (mESCs) are biased toward producing embryonic rather than extraembryonic endoderm fates. Here, we identify the mechanism of this barrier and report that the histone deacetylase Hdac3 and the transcriptional corepressor Dax1 cooperatively limit the lineage repertoire of mESCs by silencing an enhancer of the extraembryonic endoderm-specifying transcription factor Gata6. This restriction is opposed by the pluripotency transcription factors Nr5a2 and Esrrb, which promote cell type conversion. Perturbation of the barrier extends mESC potency and allows formation of 3D spheroids that mimic the spatial segregation of embryonic epiblast and extraembryonic endoderm in early embryos. Overall, this study shows that transcriptional repressors stabilize pluripotency by biasing the equilibrium between embryonic and extraembryonic lineages that is hardwired into the mESC transcriptional network.


Subject(s)
DAX-1 Orphan Nuclear Receptor , Histone Deacetylases , Mouse Embryonic Stem Cells/cytology , Animals , Cell Differentiation , Cells, Cultured , DAX-1 Orphan Nuclear Receptor/genetics , DAX-1 Orphan Nuclear Receptor/metabolism , Female , GATA6 Transcription Factor/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Male , Mice , RNA, Small Interfering/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism
6.
Nature ; 588(7839): 642-647, 2020 12.
Article in English | MEDLINE | ID: mdl-33177713

ABSTRACT

Gene-expression programs define shared and species-specific phenotypes, but their evolution remains largely uncharacterized beyond the transcriptome layer1. Here we report an analysis of the co-evolution of translatomes and transcriptomes using ribosome-profiling and matched RNA-sequencing data for three organs (brain, liver and testis) in five mammals (human, macaque, mouse, opossum and platypus) and a bird (chicken). Our within-species analyses reveal that translational regulation is widespread in the different organs, in particular across the spermatogenic cell types of the testis. The between-species divergence in gene expression is around 20% lower at the translatome layer than at the transcriptome layer owing to extensive buffering between the expression layers, which especially preserved old, essential and housekeeping genes. Translational upregulation specifically counterbalanced global dosage reductions during the evolution of sex chromosomes and the effects of meiotic sex-chromosome inactivation during spermatogenesis. Despite the overall prevalence of buffering, some genes evolved faster at the translatome layer-potentially indicating adaptive changes in expression; testis tissue shows the highest fraction of such genes. Further analyses incorporating mass spectrometry proteomics data establish that the co-evolution of transcriptomes and translatomes is reflected at the proteome layer. Together, our work uncovers co-evolutionary patterns and associated selective forces across the expression layers, and provides a resource for understanding their interplay in mammalian organs.


Subject(s)
Evolution, Molecular , Mammals/genetics , Protein Biosynthesis , Transcriptome/genetics , Animals , Brain/metabolism , Chickens/genetics , Female , Genes, X-Linked/genetics , Humans , Liver/metabolism , Macaca/genetics , Male , Mice , Opossums/genetics , Organ Specificity/genetics , Platypus/genetics , Protein Biosynthesis/genetics , RNA-Seq , Ribosomes/metabolism , Sex Chromosomes/genetics , Species Specificity , Spermatogenesis/genetics , Testis/metabolism , Up-Regulation
7.
Nat Commun ; 11(1): 2807, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32533074

ABSTRACT

The nuclear receptor binding SET domain protein 1 (NSD1) is recurrently mutated in human cancers including acute leukemia. We show that NSD1 knockdown alters erythroid clonogenic growth of human CD34+ hematopoietic cells. Ablation of Nsd1 in the hematopoietic system of mice induces a transplantable erythroleukemia. In vitro differentiation of Nsd1-/- erythroblasts is majorly impaired despite abundant expression of GATA1, the transcriptional master regulator of erythropoiesis, and associated with an impaired activation of GATA1-induced targets. Retroviral expression of wildtype NSD1, but not a catalytically-inactive NSD1N1918Q SET-domain mutant induces terminal maturation of Nsd1-/- erythroblasts. Despite similar GATA1 protein levels, exogenous NSD1 but not NSDN1918Q significantly increases the occupancy of GATA1 at target genes and their expression. Notably, exogenous NSD1 reduces the association of GATA1 with the co-repressor SKI, and knockdown of SKI induces differentiation of Nsd1-/- erythroblasts. Collectively, we identify the NSD1 methyltransferase as a regulator of GATA1-controlled erythroid differentiation and leukemogenesis.


Subject(s)
Cell Differentiation , Erythroid Cells/metabolism , Erythroid Cells/pathology , GATA1 Transcription Factor/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Erythroblastic, Acute/pathology , Adult , Animals , Antigens, CD/metabolism , Antigens, CD34/metabolism , Cell Line, Tumor , Cell Lineage , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Erythroblasts/metabolism , GATA1 Transcription Factor/genetics , Gene Expression Regulation, Leukemic , Gene Knockdown Techniques , Hematopoiesis , Histone-Lysine N-Methyltransferase/genetics , Humans , Kaplan-Meier Estimate , Leukemia, Erythroblastic, Acute/genetics , Male , Mice , Protein Binding , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-kit/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Transferrin/metabolism
8.
Nat Commun ; 11(1): 2680, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32471981

ABSTRACT

DNA methylation is considered a stable epigenetic mark, yet methylation patterns can vary during differentiation and in diseases such as cancer. Local levels of DNA methylation result from opposing enzymatic activities, the rates of which remain largely unknown. Here we developed a theoretical and experimental framework enabling us to infer methylation and demethylation rates at 860,404 CpGs in mouse embryonic stem cells. We find that enzymatic rates can vary as much as two orders of magnitude between CpGs with identical steady-state DNA methylation. Unexpectedly, de novo and maintenance methylation activity is reduced at transcription factor binding sites, while methylation turnover is elevated in transcribed gene bodies. Furthermore, we show that TET activity contributes substantially more than passive demethylation to establishing low methylation levels at distal enhancers. Taken together, our work unveils a genome-scale map of methylation kinetics, revealing highly variable and context-specific activity for the DNA methylation machinery.


Subject(s)
CpG Islands/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Demethylation , DNA Methylation/genetics , DNA-Binding Proteins/metabolism , Mouse Embryonic Stem Cells/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Binding Sites/genetics , Cell Line , Chromosome Mapping , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , DNA-Binding Proteins/genetics , Dioxygenases/genetics , Dioxygenases/metabolism , Epigenesis, Genetic/genetics , Genome/genetics , Histones/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/genetics , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/metabolism , Transcription, Genetic/genetics , DNA Methyltransferase 3B
9.
Haematologica ; 105(7): 1857-1867, 2020 07.
Article in English | MEDLINE | ID: mdl-31558671

ABSTRACT

Inv(11)(p15q23), found in myelodysplastic syndromes and acute myeloid leukemia, leads to expression of a fusion protein consisting of the N-terminal of nucleoporin 98 (NUP98) and the majority of the lysine methyltransferase 2A (KMT2A). To explore the transforming potential of this fusion we established inducible iNUP98-KMT2A transgenic mice. After a median latency of 80 weeks, over 90% of these mice developed signs of disease, with anemia and reduced bone marrow cellularity, increased white blood cell numbers, extramedullary hematopoiesis, and multilineage dysplasia. Additionally, induction of iNUP98-KMT2A led to elevated lineage marker-negative Sca-1+ c-Kit+ cell numbers in the bone marrow, which outcompeted wildtype cells in repopulation assays. Six iNUP98-KMT2A mice developed transplantable acute myeloid leukemia with leukemic blasts infiltrating multiple organs. Notably, as reported for patients, iNUP98-KMT2A leukemic blasts did not express increased levels of the HoxA-B-C gene cluster, and in contrast to KMT2A-AF9 leukemic cells, the cells were resistant to pharmacological targeting of menin and BET family proteins by MI-2-2 or JQ1, respectively. Expression of iNUP98-KMT2A in mouse embryonic fibroblasts led to an accumulation of cells in G1 phase, and abrogated replicative senescence. In bone marrow-derived hematopoietic progenitors, iNUP98-KMT2A expression similarly resulted in increased cell numbers in the G1 phase of the cell cycle, with aberrant gene expression of Sirt1, Tert, Rbl2, Twist1, Vim, and Prkcd, mimicking that seen in mouse embryonic fibroblasts. In summary, we demonstrate that iNUP98-KMT2A has in vivo transforming activity and interferes with cell cycle progression rather than primarily blocking differentiation.


Subject(s)
Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Animals , Fibroblasts , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Myelodysplastic Syndromes/genetics , Myeloid-Lymphoid Leukemia Protein , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics
10.
Mol Cell ; 77(4): 825-839.e7, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31837995

ABSTRACT

In mammals, chromatin organization undergoes drastic reorganization during oocyte development. However, the dynamics of three-dimensional chromatin structure in this process is poorly characterized. Using low-input Hi-C (genome-wide chromatin conformation capture), we found that a unique chromatin organization gradually appears during mouse oocyte growth. Oocytes at late stages show self-interacting, cohesin-independent compartmental domains marked by H3K27me3, therefore termed Polycomb-associating domains (PADs). PADs and inter-PAD (iPAD) regions form compartment-like structures with strong inter-domain interactions among nearby PADs. PADs disassemble upon meiotic resumption from diplotene arrest but briefly reappear on the maternal genome after fertilization. Upon maternal depletion of Eed, PADs are largely intact in oocytes, but their reestablishment after fertilization is compromised. By contrast, depletion of Polycomb repressive complex 1 (PRC1) proteins attenuates PADs in oocytes, which is associated with substantial gene de-repression in PADs. These data reveal a critical role of Polycomb in regulating chromatin architecture during mammalian oocyte growth and early development.


Subject(s)
Chromatin/chemistry , Oocytes/growth & development , Oogenesis/genetics , Polycomb-Group Proteins/physiology , Animals , Blastocyst/chemistry , Cell Cycle Proteins/physiology , Chromosomal Proteins, Non-Histone/physiology , Embryo, Mammalian/chemistry , Gene Silencing , Histone Code , Mice , Oocytes/chemistry , Transcription, Genetic , Cohesins
11.
Cancer Discov ; 9(12): 1736-1753, 2019 12.
Article in English | MEDLINE | ID: mdl-31662298

ABSTRACT

Fusion oncogenes are prevalent in several pediatric cancers, yet little is known about the specific associations between age and phenotype. We observed that fusion oncogenes, such as ETO2-GLIS2, are associated with acute megakaryoblastic or other myeloid leukemia subtypes in an age-dependent manner. Analysis of a novel inducible transgenic mouse model showed that ETO2-GLIS2 expression in fetal hematopoietic stem cells induced rapid megakaryoblastic leukemia whereas expression in adult bone marrow hematopoietic stem cells resulted in a shift toward myeloid transformation with a strikingly delayed in vivo leukemogenic potential. Chromatin accessibility and single-cell transcriptome analyses indicate ontogeny-dependent intrinsic and ETO2-GLIS2-induced differences in the activities of key transcription factors, including ERG, SPI1, GATA1, and CEBPA. Importantly, switching off the fusion oncogene restored terminal differentiation of the leukemic blasts. Together, these data show that aggressiveness and phenotypes in pediatric acute myeloid leukemia result from an ontogeny-related differential susceptibility to transformation by fusion oncogenes. SIGNIFICANCE: This work demonstrates that the clinical phenotype of pediatric acute myeloid leukemia is determined by ontogeny-dependent susceptibility for transformation by oncogenic fusion genes. The phenotype is maintained by potentially reversible alteration of key transcription factors, indicating that targeting of the fusions may overcome the differentiation blockage and revert the leukemic state.See related commentary by Cruz Hernandez and Vyas, p. 1653.This article is highlighted in the In This Issue feature, p. 1631.


Subject(s)
Leukemia, Myeloid, Acute/pathology , Oncogene Proteins, Fusion/genetics , Adolescent , Age Factors , Animals , Child , Child, Preschool , Female , Humans , Infant , Leukemia, Myeloid, Acute/genetics , Mice , Neoplasm Transplantation , Transcription Factors , Tumor Cells, Cultured
12.
Mol Cell ; 75(3): 483-497.e9, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31253574

ABSTRACT

In mammals, ∼100 deubiquitinases act on ∼20,000 intracellular ubiquitination sites. Deubiquitinases are commonly regarded as constitutively active, with limited regulatory and targeting capacity. The BRCA1-A and BRISC complexes serve in DNA double-strand break repair and immune signaling and contain the lysine-63 linkage-specific BRCC36 subunit that is functionalized by scaffold subunits ABRAXAS and ABRO1, respectively. The molecular basis underlying BRCA1-A and BRISC function is currently unknown. Here we show that in the BRCA1-A complex structure, ABRAXAS integrates the DNA repair protein RAP80 and provides a high-affinity binding site that sequesters the tumor suppressor BRCA1 away from the break site. In the BRISC structure, ABRO1 binds SHMT2α, a metabolic enzyme enabling cancer growth in hypoxic environments, which we find prevents BRCC36 from binding and cleaving ubiquitin chains. Our work explains modularity in the BRCC36 DUB family, with different adaptor subunits conferring diversified targeting and regulatory functions.


Subject(s)
BRCA1 Protein/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Deubiquitinating Enzymes/genetics , Histone Chaperones/genetics , Neoplasms/genetics , Binding Sites/genetics , Carrier Proteins/genetics , Cell Nucleus/genetics , Cell Nucleus/immunology , Cytoplasm/genetics , Cytoplasm/immunology , DNA Breaks, Double-Stranded , DNA Repair/immunology , Deubiquitinating Enzymes/immunology , HeLa Cells , Humans , Immunity, Cellular/genetics , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Neoplasms/immunology , Nuclear Matrix-Associated Proteins/genetics , Protein Binding/genetics , Ubiquitin/genetics , Ubiquitin-Specific Proteases/genetics , Ubiquitination/genetics
13.
Development ; 145(23)2018 11 30.
Article in English | MEDLINE | ID: mdl-30504434

ABSTRACT

The mammalian male germline is sustained by a pool of spermatogonial stem cells (SSCs) that can transmit both genetic and epigenetic information to offspring. However, the mechanisms underlying epigenetic transmission remain unclear. The histone methyltransferase Kmt2b is highly expressed in SSCs and is required for the SSC-to-progenitor transition. At the stem-cell stage, Kmt2b catalyzes H3K4me3 at bivalent H3K27me3-marked promoters as well as at promoters of a new class of genes lacking H3K27me3, which we call monovalent. Monovalent genes are mainly activated in late spermatogenesis, whereas most bivalent genes are mainly not expressed until embryonic development. These data suggest that SSCs are epigenetically primed by Kmt2b in two distinguishable ways for the upregulation of gene expression both during the spermatogenic program and through the male germline into the embryo. Because Kmt2b is also the major H3K4 methyltransferase for bivalent promoters in embryonic stem cells, we also propose that Kmt2b has the capacity to prime stem cells epigenetically.


Subject(s)
Embryo, Mammalian/metabolism , Germ Cells/cytology , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Promoter Regions, Genetic , Spermatogonia/cytology , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Survival , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/genetics , Male , Mice , Myeloid-Lymphoid Leukemia Protein/genetics , Polycomb-Group Proteins/metabolism
14.
Science ; 362(6412): 291-292, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30337398

Subject(s)
Ovum , Humans
15.
Mol Cell Oncol ; 5(3): e1241854, 2018.
Article in English | MEDLINE | ID: mdl-30250880

ABSTRACT

We recently showed that cellular origin impacts the aggressiveness and the phenotype of acute myeloid leukemia (AML). Direct induction of the MLL-AF9 fusion in various hematopoietic compartments in vivo using a doxycycline (DOX) regulated mouse model (iMLL-AF9) led to an invasive chemoresistant AML expressing several genes known to be involved in epithelial to mesenchymal transition (EMT) in solid cancers. Many of these genes play important roles in migration and invasion and are significantly associated with poor overall survival in AML patients.

16.
Hemasphere ; 2(4): e51, 2018 Aug.
Article in English | MEDLINE | ID: mdl-31723780

ABSTRACT

Previous retroviral and knock-in approaches to model human t(11;19)+ acute mixed-lineage leukemia in mice resulted in myeloproliferation and acute myeloid leukemia not fully recapitulating the human disease. The authors established a doxycycline (DOX)-inducible transgenic mouse model "iMLL-ENL" in which induction in long-term hematopoietic stem cells, lymphoid primed multipotent progenitor cells, multipotent progenitors (MPP4) but not in more committed myeloid granulocyte-macrophage progenitors led to a fully reversible acute leukemia expressing myeloid and B-cell markers. iMLL-ENL leukemic cells generally expressed lower MLL-ENL mRNA than those obtained after retroviral transduction. Disease induction was associated with iMLL-ENL levels exceeding the endogenous Mll1 at mRNA and protein levels. In leukemic cells from t(11;19)+ leukemia patients, MLL-ENL mRNA also exceeded the endogenous MLL1 levels suggesting a critical threshold for transformation. Expression profiling of iMLL-ENL acute leukemia revealed gene signatures that segregated t(11;19)+ leukemia patients from those without an MLL translocation. Importantly, B220+ iMLL-ENL leukemic cells showed a higher in vivo leukemia initiation potential than coexisting B220- cells. Collectively, characterization of a novel transgenic mouse model indicates that the cell-of-origin and the fusion gene expression levels are both critical determinants for MLL-ENL-driven acute leukemia.

17.
Epigenetics Chromatin ; 10: 11, 2017.
Article in English | MEDLINE | ID: mdl-28293300

ABSTRACT

BACKGROUND: In the nuclei of most mammalian cells, pericentric heterochromatin is characterized by DNA methylation, histone modifications such as H3K9me3 and H4K20me3, and specific binding proteins like heterochromatin-binding protein 1 isoforms (HP1 isoforms). Maintenance of this specialized chromatin structure is of great importance for genome integrity and for the controlled repression of the repetitive elements within the pericentric DNA sequence. Here we have studied histone modifications at pericentric heterochromatin during primordial germ cell (PGC) development using different fixation conditions and fluorescent immunohistochemical and immunocytochemical protocols. RESULTS: We observed that pericentric heterochromatin marks, such as H3K9me3, H4K20me3, and HP1 isoforms, were retained on pericentric heterochromatin throughout PGC development. However, the observed immunostaining patterns varied, depending on the fixation method, explaining previous findings of a general loss of pericentric heterochromatic features in PGCs. Also, in contrast to the general clustering of multiple pericentric regions and associated centromeres in DAPI-dense regions in somatic cells, the pericentric regions of PGCs were more frequently organized as individual entities. We also observed a transient enrichment of the chromatin remodeler ATRX in pericentric regions in embryonic day 11.5 (E11.5) PGCs. At this stage, a similar and low level of major satellite repeat RNA transcription was detected in both PGCs and somatic cells. CONCLUSIONS: These results indicate that in pericentric heterochromatin of mouse PGCs, only minor reductions in levels of some chromatin-associated proteins occur, in association with a transient increase in ATRX, between E11.5 and E13.5. These pericentric heterochromatin regions more frequently contain only a single centromere in PGCs compared to the surrounding soma, indicating a difference in overall organization, but there is no de-repression of major satellite transcription.


Subject(s)
Germ Cells/metabolism , Heterochromatin/metabolism , Animals , Centromere/metabolism , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Embryo, Mammalian/metabolism , Gene Silencing , Germ Cells/cytology , Germ Cells/growth & development , Histones/metabolism , Immunohistochemistry , Mice , Microscopy, Fluorescence , Protein Isoforms/genetics , Protein Isoforms/metabolism
18.
Dev Cell ; 40(3): 289-301.e3, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28132849

ABSTRACT

Somatic X dosage compensation requires two mechanisms: X inactivation balances X gene output between males (XY) and females (XX), while X upregulation, hypothesized by Ohno and documented in vivo, balances X gene with autosomal gene output. Whether X dosage compensation occurs in germ cells is unclear. We show that mouse and human germ cells exhibit non-canonical X dosage states that differ from the soma and between the sexes. Prior to genome-wide reprogramming, X upregulation is present, consistent with Ohno's hypothesis. Subsequently, however, it is erased. In females, erasure follows loss of X inactivation, causing X dosage excess. Conversely, in males, erasure leads to permanent X dosage decompensation. Sex chromosomally abnormal models exhibit a "sex-reversed" X dosage state: XX males, like XX females, develop X dosage excess, while XO females, like XY males, develop X dosage decompensation. Thus, germline X dosage compensation states are determined by X chromosome number, not phenotypic sex. These unexpected differences in X dosage compensation states between germline and soma offer unique perspectives on sex chromosome infertility.


Subject(s)
Chromosomes, Human, X/genetics , Dosage Compensation, Genetic , Germ Cells/metabolism , Sex Characteristics , X Chromosome/genetics , Animals , Cellular Reprogramming/genetics , Databases, Genetic , Female , Gene Expression Profiling , Gene Expression Regulation , Germ Cells/cytology , Gonads/cytology , Gonads/metabolism , Humans , Male , Mice , Models, Genetic , Sequence Analysis, RNA , Up-Regulation/genetics
19.
Cancer Cell ; 30(1): 43-58, 2016 07 11.
Article in English | MEDLINE | ID: mdl-27344946

ABSTRACT

To address the impact of cellular origin on acute myeloid leukemia (AML), we generated an inducible transgenic mouse model for MLL-AF9-driven leukemia. MLL-AF9 expression in long-term hematopoietic stem cells (LT-HSC) in vitro resulted in dispersed clonogenic growth and expression of genes involved in migration and invasion. In vivo, 20% LT-HSC-derived AML were particularly aggressive with extensive tissue infiltration, chemoresistance, and expressed genes related to epithelial-mesenchymal transition (EMT) in solid cancers. Knockdown of the EMT regulator ZEB1 significantly reduced leukemic blast invasion. By classifying mouse and human leukemias according to Evi1/EVI1 and Erg/ERG expression, reflecting aggressiveness and cell of origin, and performing comparative transcriptomics, we identified several EMT-related genes that were significantly associated with poor overall survival of AML patients.


Subject(s)
Epithelial-Mesenchymal Transition , Hematopoietic Stem Cells/cytology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Animals , Drug Resistance, Neoplasm , Gene Expression Profiling/methods , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Transgenic , Neoplasm Invasiveness , Neoplasms, Experimental , Prognosis , Tumor Cells, Cultured , Zinc Finger E-box-Binding Homeobox 1/genetics
20.
Development ; 143(15): 2767-79, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27317807

ABSTRACT

Oocytes develop the competence for meiosis and early embryogenesis during their growth. Setdb1 is a histone H3 lysine 9 (H3K9) methyltransferase required for post-implantation development and has been implicated in the transcriptional silencing of genes and endogenous retroviral elements (ERVs). To address its role in oogenesis and pre-implantation development, we conditionally deleted Setdb1 in growing oocytes. Loss of Setdb1 expression greatly impaired meiosis. It delayed meiotic resumption, altered the dynamics of chromatin condensation, and impaired kinetochore-spindle interactions, bipolar spindle organization and chromosome segregation in more mature oocytes. The observed phenotypes related to changes in abundance of specific transcripts in mutant oocytes. Setdb1 maternally deficient embryos arrested during pre-implantation development and showed comparable defects during cell cycle progression and in chromosome segregation. Finally, transcriptional profiling data indicate that Setdb1 downregulates rather than silences expression of ERVK and ERVL-MaLR retrotransposons and associated chimearic transcripts during oogenesis. Our results identify Setdb1 as a newly discovered meiotic and embryonic competence factor safeguarding genome integrity at the onset of life.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Meiosis/physiology , Mitosis/physiology , Oocytes/metabolism , Animals , Chromosome Segregation/genetics , Chromosome Segregation/physiology , Embryonic Development/genetics , Embryonic Development/physiology , Female , Histone-Lysine N-Methyltransferase/genetics , Kinetochores/metabolism , Male , Meiosis/genetics , Mice , Mitosis/genetics , Oocytes/cytology , Oogenesis/genetics , Oogenesis/physiology , Retroelements/genetics , Retroelements/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...