Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
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.
J Mammary Gland Biol Neoplasia ; 28(1): 26, 2023 12 08.
Article in English | MEDLINE | ID: mdl-38066300

ABSTRACT

Metastasis is the leading cause of cancer-related deaths of breast cancer patients. Some cancer cells in a tumour go through successive steps, referred to as the metastatic cascade, and give rise to metastases at a distant site. We know that the plasticity and heterogeneity of cancer cells play critical roles in metastasis but the precise underlying molecular mechanisms remain elusive. Here we aimed to identify molecular mechanisms of metastasis during colonization, one of the most important yet poorly understood steps of the cascade. We performed single-cell RNA-Seq (scRNA-Seq) on tumours and matched lung macrometastases of patient-derived xenografts of breast cancer. After correcting for confounding factors such as the cell cycle and the percentage of detected genes (PDG), we identified cells in three states in both tumours and metastases. Gene-set enrichment analysis revealed biological processes specific to proliferation and invasion in two states. Our findings suggest that these states are a balance between epithelial-to-mesenchymal (EMT) and mesenchymal-to-epithelial transitions (MET) traits that results in so-called partial EMT phenotypes. Analysis of the top differentially expressed genes (DEGs) between these cell states revealed a common set of partial EMT transcription factors (TFs) controlling gene expression, including ZNF750, OVOL2, TP63, TFAP2C and HEY2. Our data suggest that the TFs related to EMT delineate different cell states in tumours and metastases. The results highlight the marked interpatient heterogeneity of breast cancer but identify common features of single cells from five models of metastatic breast cancer.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Cell Line, Tumor , Transcription Factors , Single-Cell Analysis , Tumor Suppressor Proteins
3.
Nat Commun ; 14(1): 3242, 2023 06 05.
Article in English | MEDLINE | ID: mdl-37277355

ABSTRACT

Enhancer-promoter interactions preferentially occur within boundary-insulated topologically associating domains (TADs), limiting inter-TAD interactions. Enhancer clusters in linear proximity, termed super-enhancers (SEs), ensure high target gene expression levels. Little is known about SE topological regulatory impact during craniofacial development. Here, we identify 2232 genome-wide putative SEs in mouse cranial neural crest cells (CNCCs), 147 of which target genes establishing CNCC positional identity during face formation. In second pharyngeal arch (PA2) CNCCs, a multiple SE-containing region, partitioned into Hoxa Inter-TAD Regulatory Element 1 and 2 (HIRE1 and HIRE2), establishes long-range inter-TAD interactions selectively with Hoxa2, that is required for external and middle ear structures. HIRE2 deletion in a Hoxa2 haploinsufficient background results in microtia. HIRE1 deletion phenocopies the full homeotic Hoxa2 knockout phenotype and induces PA3 and PA4 CNCC abnormalities correlating with Hoxa2 and Hoxa3 transcriptional downregulation. Thus, SEs can overcome TAD insulation and regulate anterior Hoxa gene collinear expression in a CNCC subpopulation-specific manner during craniofacial development.


Subject(s)
Neural Crest , Regulatory Sequences, Nucleic Acid , Mice , Animals , Neural Crest/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Skull/metabolism , Chromatin/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism
4.
Cell Stem Cell ; 30(6): 867-884.e11, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37209681

ABSTRACT

Gastruloids are 3D structures generated from pluripotent stem cells recapitulating fundamental principles of embryonic pattern formation. Using single-cell genomic analysis, we provide a resource mapping cell states and types during gastruloid development and compare them with the in vivo embryo. We developed a high-throughput handling and imaging pipeline to spatially monitor symmetry breaking during gastruloid development and report an early spatial variability in pluripotency determining a binary response to Wnt activation. Although cells in the gastruloid-core revert to pluripotency, peripheral cells become primitive streak-like. These two populations subsequently break radial symmetry and initiate axial elongation. By performing a compound screen, perturbing thousands of gastruloids, we derive a phenotypic landscape and infer networks of genetic interactions. Finally, using a dual Wnt modulation, we improve the formation of anterior structures in the existing gastruloid model. This work provides a resource to understand how gastruloids develop and generate complex patterns in vitro.


Subject(s)
Embryo, Mammalian , Pluripotent Stem Cells , Mice , Animals , Embryo, Mammalian/metabolism , Primitive Streak/metabolism , Embryonic Development
5.
Nature ; 604(7906): 571-577, 2022 04.
Article in English | MEDLINE | ID: mdl-35418676

ABSTRACT

Chromosome structure in mammals is thought to regulate transcription by modulating three-dimensional interactions between enhancers and promoters, notably through CTCF-mediated loops and topologically associating domains (TADs)1-4. However, how chromosome interactions are actually translated into transcriptional outputs remains unclear. Here, to address this question, we use an assay to position an enhancer at large numbers of densely spaced chromosomal locations relative to a fixed promoter, and measure promoter output and interactions within a genomic region with minimal regulatory and structural complexity. A quantitative analysis of hundreds of cell lines reveals that the transcriptional effect of an enhancer depends on its contact probabilities with the promoter through a nonlinear relationship. Mathematical modelling suggests that nonlinearity might arise from transient enhancer-promoter interactions being translated into slower promoter bursting dynamics in individual cells, therefore uncoupling the temporal dynamics of interactions from those of transcription. This uncovers a potential mechanism of how distal enhancers act from large genomic distances, and of how topologically associating domain boundaries block distal enhancers. Finally, we show that enhancer strength also determines absolute transcription levels as well as the sensitivity of a promoter to CTCF-mediated transcriptional insulation. Our measurements establish general principles for the context-dependent role of chromosome structure in long-range transcriptional regulation.


Subject(s)
Chromosomes , Enhancer Elements, Genetic , Animals , Chromatin/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Genomics , Mammals/genetics , Promoter Regions, Genetic/genetics
6.
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
7.
Nat Cell Biol ; 23(11): 1163-1175, 2021 11.
Article in English | MEDLINE | ID: mdl-34737442

ABSTRACT

The developmental role of histone H3K9 methylation (H3K9me), which typifies heterochromatin, remains unclear. In Caenorhabditis elegans, loss of H3K9me leads to a highly divergent upregulation of genes with tissue and developmental-stage specificity. During development H3K9me is lost from differentiated cell type-specific genes and gained at genes expressed in earlier developmental stages or other tissues. The continuous deposition of H3K9me2 by the SETDB1 homolog MET-2 after terminal differentiation is necessary to maintain repression. In differentiated tissues, H3K9me ensures silencing by restricting the activity of a defined set of transcription factors at promoters and enhancers. Increased chromatin accessibility following the loss of H3K9me is neither sufficient nor necessary to drive transcription. Increased ATAC-seq signal and gene expression correlate at a subset of loci positioned away from the nuclear envelope, while derepressed genes at the nuclear periphery remain poorly accessible despite being transcribed. In conclusion, H3K9me deposition can confer tissue-specific gene expression and maintain the integrity of terminally differentiated muscle by restricting transcription factor activity.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Cell Differentiation , Chromatin Assembly and Disassembly , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Protein Processing, Post-Translational , Transcription, Genetic , Animals , Animals, Genetically Modified , Binding Sites , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Chromatin Immunoprecipitation Sequencing , Gene Expression Profiling , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Methylation , Protein Binding , Time Factors , Transcriptome
8.
Nat Genet ; 53(3): 379-391, 2021 03.
Article in English | MEDLINE | ID: mdl-33603234

ABSTRACT

Rapid cellular responses to environmental stimuli are fundamental for development and maturation. Immediate early genes can be transcriptionally induced within minutes in response to a variety of signals. How their induction levels are regulated and their untimely activation by spurious signals prevented during development is poorly understood. We found that in developing sensory neurons, before perinatal sensory-activity-dependent induction, immediate early genes are embedded into a unique bipartite Polycomb chromatin signature, carrying active H3K27ac on promoters but repressive Ezh2-dependent H3K27me3 on gene bodies. This bipartite signature is widely present in developing cell types, including embryonic stem cells. Polycomb marking of gene bodies inhibits mRNA elongation, dampening productive transcription, while still allowing for fast stimulus-dependent mark removal and bipartite gene induction. We reveal a developmental epigenetic mechanism regulating the rapidity and amplitude of the transcriptional response to relevant stimuli, while preventing inappropriate activation of stimulus-response genes.


Subject(s)
Chromatin/genetics , Gene Expression Regulation, Developmental , Genes, Immediate-Early , Polycomb-Group Proteins/genetics , Animals , Chromatin/metabolism , Embryonic Stem Cells/physiology , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Histones/metabolism , Mice, Transgenic , Mutation , Polycomb-Group Proteins/metabolism , Promoter Regions, Genetic , RNA Polymerase II/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rhombencephalon/drug effects , Rhombencephalon/embryology , Sensory Receptor Cells/physiology
9.
Blood ; 137(21): 2920-2934, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33512466

ABSTRACT

OBF1 is a specific coactivator of the POU family transcription factors OCT1 and OCT2. OBF1 and OCT2 are B cell-specific and indispensable for germinal center (GC) formation, but their mechanism of action is unclear. Here, we show by chromatin immunoprecipitation-sequencing that OBF1 extensively colocalizes with OCT1 and OCT2. We found that these factors also often colocalize with transcription factors of the ETS family. Furthermore, we showed that OBF1, OCT2, and OCT1 bind widely to the promoters or enhancers of genes involved in GC formation in mouse and human GC B cells. Short hairpin RNA knockdown experiments demonstrated that OCT1, OCT2, and OBF1 regulate each other and are essential for proliferation of GC-derived lymphoma cell lines. OBF1 downregulation disrupts the GC transcriptional program: genes involved in GC maintenance, such as BCL6, are downregulated, whereas genes related to exit from the GC program, such as IRF4, are upregulated. Ectopic expression of BCL6 does not restore the proliferation of GC-derived lymphoma cells depleted of OBF1 unless IRF4 is also depleted, indicating that OBF1 controls an essential regulatory node in GC differentiation.


Subject(s)
Germinal Center/metabolism , Octamer Transcription Factor-1/physiology , Octamer Transcription Factor-2/therapeutic use , Trans-Activators/therapeutic use , Transcription, Genetic/genetics , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Gene Ontology , HEK293 Cells , Humans , Lipopolysaccharides/pharmacology , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Octamer Transcription Factor-1/deficiency , Octamer Transcription Factor-1/genetics , Octamer Transcription Factor-2/deficiency , Octamer Transcription Factor-2/genetics , Proto-Oncogene Protein c-ets-1/analysis , RNA Interference , RNA, Small Interfering/genetics , Recombinant Proteins/metabolism , Trans-Activators/deficiency , Trans-Activators/genetics
10.
Cell Rep ; 31(11): 107767, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32553152

ABSTRACT

The mammalian precerebellar pontine nucleus (PN) has a main role in relaying cortical information to the cerebellum. The molecular determinants establishing ordered connectivity patterns between cortical afferents and precerebellar neurons are largely unknown. We show that expression of Hox5 transcription factors is induced in specific subsets of postmitotic PN neurons at migration onset. Hox5 induction is achieved by response to retinoic acid signaling, resulting in Jmjd3-dependent derepression of Polycomb chromatin and 3D conformational changes. Hoxa5 drives neurons to settle posteriorly in the PN, where they are monosynaptically targeted by cortical neuron subsets mainly carrying limb somatosensation. Furthermore, Hoxa5 postmigratory ectopic expression in PN neurons is sufficient to attract cortical somatosensory inputs regardless of position and avoid visual afferents. Transcriptome analysis further suggests that Hoxa5 is involved in circuit formation. Thus, Hoxa5 coordinates postmitotic specification, migration, settling position, and sub-circuit assembly of PN neuron subsets in the cortico-cerebellar pathway.


Subject(s)
Cerebellum/metabolism , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/metabolism , Neurons/metabolism , Transcription Factors/metabolism , Animals , Cell Movement/physiology , Cerebral Cortex/metabolism
11.
Nature ; 567(7749): 540-544, 2019 03.
Article in English | MEDLINE | ID: mdl-30867597

ABSTRACT

Diversity within or between tumours and metastases (known as intra-patient tumour heterogeneity) that develops during disease progression is a serious hurdle for therapy1-3. Metastasis is the fatal hallmark of cancer and the mechanisms of colonization, the most complex step in the metastatic cascade4, remain poorly defined. A clearer understanding of the cellular and molecular processes that underlie both intra-patient tumour heterogeneity and metastasis is crucial for the success of personalized cancer therapy. Here, using transcriptional profiling of tumours and matched metastases in patient-derived xenograft models in mice, we show cancer-site-specific phenotypes and increased glucocorticoid receptor activity in distant metastases. The glucocorticoid receptor mediates the effects of stress hormones, and of synthetic derivatives of these hormones that are used widely in the clinic as anti-inflammatory and immunosuppressive agents. We show that the increase in stress hormones during breast cancer progression results in the activation of the glucocorticoid receptor at distant metastatic sites, increased colonization and reduced survival. Our transcriptomics, proteomics and phospho-proteomics studies implicate the glucocorticoid receptor in the activation of multiple processes in metastasis and in the increased expression of kinase ROR1, both of which correlate with reduced survival. The ablation of ROR1 reduced metastatic outgrowth and prolonged survival in preclinical models. Our results indicate that the activation of the glucocorticoid receptor increases heterogeneity and metastasis, which suggests that caution is needed when using glucocorticoids to treat patients with breast cancer who have developed cancer-related complications.


Subject(s)
Breast Neoplasms/pathology , Glucocorticoids/adverse effects , Glucocorticoids/metabolism , Neoplasm Metastasis/pathology , Animals , Breast Neoplasms/enzymology , Cell Line, Tumor , Dexamethasone/adverse effects , Dexamethasone/metabolism , Disease Progression , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Protein Kinases/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Receptors, Glucocorticoid/metabolism , Signal Transduction/drug effects , Survival Rate
12.
Neuro Oncol ; 20(5): 621-631, 2018 04 09.
Article in English | MEDLINE | ID: mdl-29401256

ABSTRACT

Background: Glioblastoma (GBM) is one of the most aggressive human brain tumors, with a median survival of 15-18 months. There is a desperate need to find novel therapeutic targets. Various receptor protein kinases have been identified as potential targets; however, response rates in clinical studies have been somewhat disappointing. Targeting the spleen tyrosine kinase (SYK), which acts downstream of a range of oncogenic receptors, may therefore show more promising results. Methods: Kinase expression of brain tumor samples including GBM and low-grade tumors were compared with normal brain and normal human astrocytes by microarray analysis. Furthermore, SYK, LYN, SLP76, and PLCG2 protein expressions were analyzed by immunohistochemistry, western blot, and immunofluorescence of additional GBM patient samples, murine glioma samples, and cell lines. SYK was then blocked chemically and genetically in vitro and in vivo in 2 different mouse models. Multiphoton intravital imaging and multicolor flow cytometry were performed in a syngeneic immunocompetent C57BL/6J mouse GL261 glioma model to study the effect of these inhibitors on the tumor microenvironment. Results: SYK, LYN, SLP76, and PLCG2 were found expressed in human and murine glioma samples and cell lines. SYK inhibition blocked proliferation, migration, and colony formation. Flow cytometric and multiphoton imaging imply that targeting SYK in vivo attenuated GBM tumor growth and invasiveness and reduced B and CD11b+ cell mobility and infiltration. Conclusions: Our data suggest that gliomas express a SYK signaling network important in glioma progression, inhibition of which results in reduced invasion with slower tumor progression.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Movement , Cell Proliferation , Disease Models, Animal , Glioblastoma/pathology , Syk Kinase/metabolism , Tumor Microenvironment , Animals , Apoptosis , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Female , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Prognosis , Syk Kinase/genetics , Tumor Cells, Cultured
13.
Elife ; 62017 11 06.
Article in English | MEDLINE | ID: mdl-29106373

ABSTRACT

High-resolution daylight vision is mediated by cone photoreceptors. The molecular program responsible for the formation of their light sensor, the outer segment, is not well understood. We correlated daily changes in ultrastructure and gene expression in postmitotic mouse cones, between birth and eye opening, using serial block-face electron microscopy (EM) and RNA sequencing. Outer segments appeared rapidly at postnatal day six and their appearance coincided with a switch in gene expression. The switch affected over 14% of all expressed genes. Genes that switched off were rich in transcription factors and neurogenic genes. Those that switched on contained genes relevant for cone function. Chromatin rearrangements in enhancer regions occurred before the switch was completed, but not after. We provide a resource comprised of correlated EM, RNAseq, and ATACseq data, showing that the growth of a key compartment of a postmitotic cell involves an extensive switch in gene expression and chromatin accessibility.


Subject(s)
Cell Differentiation , Gene Expression Regulation , Retina/growth & development , Retinal Cone Photoreceptor Cells/physiology , Transcription, Genetic , Animals , Gene Expression Profiling , Mice , Microscopy, Electron , Retinal Cone Photoreceptor Cells/ultrastructure , Sequence Analysis, RNA
14.
Science ; 355(6332)2017 03 31.
Article in English | MEDLINE | ID: mdl-28360266

ABSTRACT

The cranial neural crest cells are multipotent cells that provide head skeletogenic mesenchyme and are crucial for craniofacial patterning. We analyzed the chromatin landscapes of mouse cranial neural crest subpopulations in vivo. Early postmigratory subpopulations contributing to distinct mouse craniofacial structures displayed similar chromatin accessibility patterns yet differed transcriptionally. Accessible promoters and enhancers of differentially silenced genes carried H3K27me3/H3K4me2 bivalent chromatin marks embedded in large enhancer of zeste homolog 2-dependent Polycomb domains, indicating transcriptional poising. These postmigratory bivalent chromatin regions were already present in premigratory progenitors. At Polycomb domains, H3K27me3 antagonized H3K4me2 deposition, which was restricted to accessible sites. Thus, bivalent Polycomb domains provide a chromatin template for the regulation of cranial neural crest cell positional identity in vivo, contributing insights into the epigenetic regulation of face morphogenesis.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Epigenesis, Genetic , Face/embryology , Gene Expression Regulation, Developmental , Neural Crest/embryology , Skull/embryology , Animals , Cell Movement , Cell Plasticity/genetics , Chromatin/metabolism , Enhancer Elements, Genetic , Homeodomain Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Neural Crest/cytology , Promoter Regions, Genetic , Protein Domains
15.
Nature ; 525(7567): 114-8, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26266975

ABSTRACT

The adult mouse mammary epithelium contains self-sustained cell lineages that form the inner luminal and outer basal cell layers, with stem and progenitor cells contributing to its proliferative and regenerative potential. A key issue in breast cancer biology is the effect of genomic lesions in specific mammary cell lineages on tumour heterogeneity and progression. The impact of transforming events on fate conversion in cancer cells of origin and thus their contribution to tumour heterogeneity remains largely elusive. Using in situ genetic lineage tracing and limiting dilution transplantation, we have unravelled the potential of PIK3CA(H1047R), one of the most frequent mutations occurring in human breast cancer, to induce multipotency during tumorigenesis in the mammary gland. Here we show that expression of PIK3CA(H1047R) in lineage-committed basal Lgr5-positive and luminal keratin-8-positive cells of the adult mouse mammary gland evokes cell dedifferentiation into a multipotent stem-like state, suggesting this to be a mechanism involved in the formation of heterogeneous, multi-lineage mammary tumours. Moreover, we show that the tumour cell of origin influences the frequency of malignant mammary tumours. Our results define a key effect of PIK3CA(H1047R) on mammary cell fate in the pre-neoplastic mammary gland and show that the cell of origin of PIK3CA(H1047R) tumours dictates their malignancy, thus revealing a mechanism underlying tumour heterogeneity and aggressiveness.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Lineage/genetics , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Multipotent Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/genetics , Animals , Cell Dedifferentiation/genetics , Cell Transformation, Neoplastic/genetics , Class I Phosphatidylinositol 3-Kinases , Female , Humans , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mice , Multipotent Stem Cells/pathology , Mutation/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Phosphatidylinositol 3-Kinases/metabolism
16.
EMBO Mol Med ; 6(9): 1175-90, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25092770

ABSTRACT

In this report, we describe the development of a modified adeno-associated virus (AAV) capsid and promoter for transduction of retinal ON-bipolar cells. The bipolar cells, which are post-synaptic to the photoreceptors, are important retinal targets for both basic and preclinical research. In particular, a therapeutic strategy under investigation for advanced forms of blindness involves using optogenetic molecules to render ON-bipolar cells light-sensitive. Currently, delivery of adequate levels of gene expression is a limiting step for this approach. The synthetic AAV capsid and promoter described here achieves high level of optogenetic transgene expression in ON-bipolar cells. This evokes high-frequency (~100 Hz) spiking responses in ganglion cells of previously blind, rd1, mice. Our vector is a promising vehicle for further development toward potential clinical use.


Subject(s)
Dependovirus/genetics , Retinal Bipolar Cells/virology , Transduction, Genetic/methods , Animals , Genetic Vectors , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Promoter Regions, Genetic
17.
PLoS One ; 9(4): e92836, 2014.
Article in English | MEDLINE | ID: mdl-24747299

ABSTRACT

We have identified expression of the gene encoding the transcriptional coactivator FOG-1 (Friend of GATA-1; Zfpm1, Zinc finger protein multitype 1) in B lymphocytes. We found that FOG-1 expression is directly or indirectly dependent on the B cell-specific coactivator OBF-1 and that it is modulated during B cell development: expression is observed in early but not in late stages of B cell development. To directly test in vivo the role of FOG-1 in B lymphocytes, we developed a novel embryonic stem cell recombination system. For this, we combined homologous recombination with the FLP recombinase activity to rapidly generate embryonic stem cell lines carrying a Cre-inducible transgene at the Rosa26 locus. Using this system, we successfully generated transgenic mice where FOG-1 is conditionally overexpressed in mature B-cells or in the entire hematopoietic system. While overexpression of FOG-1 in B cells did not significantly affect B cell development or function, we found that enforced expression of FOG-1 throughout all hematopoietic lineages led to a reduction in the number of circulating eosinophils, confirming and extending to mammals the known function of FOG-1 in this lineage.


Subject(s)
B-Lymphocytes/cytology , Eosinophils/cytology , Hematopoiesis , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , B-Lymphocytes/metabolism , Blood Cell Count , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Eosinophils/metabolism , Female , Gene Expression , Genetic Loci/genetics , Humans , Male , Mice , Nuclear Proteins/metabolism , Transcription Factors/metabolism
18.
Nature ; 495(7440): 236-40, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23486062

ABSTRACT

In mammals, sex differentiation of primordial germ cells (PGCs) is determined by extrinsic cues from the environment. In mouse female PGCs, expression of stimulated by retinoic acid gene 8 (Stra8) and meiosis are induced in response to retinoic acid provided from the mesonephroi. Given the widespread role of retinoic acid signalling during development, the molecular mechanisms that enable PGCs to express Stra8 and enter meiosis in a timely manner are unknown. Here we identify gene-dosage-dependent roles in PGC development for Ring1 and Rnf2, two central components of the Polycomb repressive complex 1 (PRC1). Both paralogues are essential for PGC development between days 10.5 and 11.5 of gestation. Rnf2 is subsequently required in female PGCs to maintain high levels of Oct4 (also known as Pou5f1) and Nanog expression, and to prevent premature induction of meiotic gene expression and entry into meiotic prophase. Chemical inhibition of retinoic acid signalling partially suppresses precocious Oct4 downregulation and Stra8 activation in Rnf2-deficient female PGCs. Chromatin immunoprecipitation analyses show that Stra8 is a direct target of PRC1 and PRC2 in PGCs. These data demonstrate the importance of PRC1 gene dosage in PGC development and in coordinating the timing of sex differentiation of female PGCs by antagonizing extrinsic retinoic acid signalling.


Subject(s)
Ovum/cytology , Ovum/metabolism , Polycomb Repressive Complex 1/metabolism , Sex Differentiation/physiology , Adaptor Proteins, Signal Transducing , Animals , Chromatin/genetics , Chromatin/metabolism , Down-Regulation , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Male , Meiosis , Mice , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Polycomb Repressive Complex 1/deficiency , Polycomb Repressive Complex 2/metabolism , Proteins/genetics , Sex Characteristics , Signal Transduction , Time Factors , Transcription, Genetic , Tretinoin/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/metabolism
19.
Development ; 140(1): 117-25, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23154416

ABSTRACT

Tyrosine phosphorylation plays a fundamental role in mammary gland development. However, the role of specific tyrosine phosphatases in controlling mammary cell fate remains ill defined. We have identified protein tyrosine phosphatase 1B (PTP1B) as an essential regulator of alveologenesis and lactogenesis. PTP1B depletion increased the number of luminal mammary progenitors in nulliparous mice, leading to enhanced alveoli formation upon pregnancy. Mechanistically, Ptp1b deletion enhanced the expression of progesterone receptor and phosphorylation of Stat5, two key regulators of alveologenesis. Furthermore, glands from Ptp1b knockout mice exhibited increased expression of milk proteins during pregnancy due to enhanced Stat5 activation. These findings reveal that PTP1B constrains the number of mammary progenitors and thus prevents inappropriate onset of alveologenesis in early pregnancy. Moreover, PTP1B restrains the expression of milk proteins during pregnancy and thus prevents premature lactogenesis. Our work has implications for breast tumorigenesis because Ptp1b deletion has been shown to prevent or delay the onset of mammary tumors.


Subject(s)
Cell Differentiation/physiology , Mammary Glands, Animal/cytology , Mammary Glands, Animal/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology , Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Female , Lactation/genetics , Male , Mammary Glands, Animal/embryology , Mice , Mice, Knockout , Pregnancy , Progesterone/antagonists & inhibitors , Progesterone/biosynthesis , Progesterone/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , STAT5 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/biosynthesis , STAT5 Transcription Factor/physiology , Stem Cells/cytology , Stem Cells/enzymology , Up-Regulation/genetics
20.
Blood ; 119(18): 4291-300, 2012 May 03.
Article in English | MEDLINE | ID: mdl-22422825

ABSTRACT

The DEAH helicase RHAU (alias DHX36, G4R1) is the only helicase shown to have G-quadruplex (G4)-RNA resolvase activity and the major source of G4-DNA resolvase activity. Previous report showed RHAU mRNA expression to be elevated in human lymphoid and CD34(+) BM cells, suggesting a potential role in hematopoiesis. Here, we generated a conditional knockout of the RHAU gene in mice. Germ line deletion of RHAU led to embryonic lethality. We then targeted the RHAU gene specifically in the hematopoiesis system, using a Cre-inducible system in which an optimized variant of Cre recombinase was expressed under the control of the Vav1 promoter. RHAU deletion in hematopoietic system caused hemolytic anemia and differentiation defect at the proerythroblast stage. The partial differentiation block of proerythroblasts was because of a proliferation defect. Transcriptome analysis of RHAU knockout proerythroblasts showed that a statistically significant portion of the deregulated genes contain G4 motifs in their promoters. This suggests that RHAU may play a role in the regulation of gene expression that relies on its G4 resolvase activity.


Subject(s)
DEAD-box RNA Helicases/physiology , Hematopoiesis/genetics , Promoter Regions, Genetic/genetics , Anemia, Hemolytic, Congenital/genetics , Animals , Bone Marrow Transplantation , Cell Cycle , Crosses, Genetic , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Embryonic Development/genetics , Embryonic Development/physiology , Erythroblasts/pathology , Erythropoietin/blood , Genes, Lethal , Genes, Synthetic , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Hematopoiesis/physiology , Leukopenia/congenital , Leukopenia/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Folding , Proto-Oncogene Proteins c-vav/genetics , Radiation Chimera , Recombinases/deficiency , Recombinases/genetics , Recombinases/physiology , Thrombocytopenia/congenital , Thrombocytopenia/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...