Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Viruses ; 15(11)2023 Oct 28.
Article En | MEDLINE | ID: mdl-38005849

Despite remarkable progress, a cure for HIV-1 infection remains elusive. Rebound competent latent and transcriptionally active reservoir cells persevere despite antiretroviral therapy and rekindle infection due to inefficient proviral silencing. We propose a novel "block-lock-stop" approach, entailing long term durable silencing of viral expression towards an irreversible transcriptionally inactive latent provirus to achieve long term antiretroviral free control of the virus. A graded transformation of remnant HIV-1 in PLWH from persistent into silent to permanently defective proviruses is proposed, emulating and accelerating the natural path that human endogenous retroviruses (HERVs) take over millions of years. This hypothesis was based on research into delineating the mechanisms of HIV-1 latency, lessons from latency reversing agents and advances of Tat inhibitors, as well as expertise in the biology of HERVs. Insights from elite controllers and the availability of advanced genome engineering technologies for the direct excision of remnant virus set the stage for a rapid path to an HIV-1 cure.


Endogenous Retroviruses , HIV Infections , HIV Seropositivity , HIV-1 , Humans , HIV-1/genetics , Virus Latency , Proviruses/genetics , HIV Seropositivity/genetics , CD4-Positive T-Lymphocytes
2.
BMC Med Genomics ; 16(1): 68, 2023 04 03.
Article En | MEDLINE | ID: mdl-37013607

BACKGROUND: Human endogenous retroviruses (HERV) are repetitive sequence elements and a substantial part of the human genome. Their role in development has been well documented and there is now mounting evidence that dysregulated HERV expression also contributes to various human diseases. While research on HERV elements has in the past been hampered by their high sequence similarity, advanced sequencing technology and analytical tools have empowered the field. For the first time, we are now able to undertake locus-specific HERV analysis, deciphering expression patterns, regulatory networks and biological functions of these elements. To do so, we inevitable rely on omics datasets available through the public domain. However, technical parameters inevitably differ, making inter-study analysis challenging. We here address the issue of confounding factors for profiling locus-specific HERV transcriptomes using datasets from multiple sources. METHODS: We collected RNAseq datasets of CD4 and CD8 primary T cells and extracted HERV expression profiles for 3220 elements, resembling most intact, near full-length proviruses. Looking at sequencing parameters and batch effects, we compared HERV signatures across datasets and determined permissive features for HERV expression analysis from multiple-source data. RESULTS: We could demonstrate that considering sequencing parameters, sequencing-depth is most influential on HERV signature outcome. Sequencing samples deeper broadens the spectrum of expressed HERV elements. Sequencing mode and read length are secondary parameters. Nevertheless, we find that HERV signatures from smaller RNAseq datasets do reliably reveal most abundantly expressed HERV elements. Overall, HERV signatures between samples and studies overlap substantially, indicating a robust HERV transcript signature in CD4 and CD8 T cells. Moreover, we find that measures of batch effect reduction are critical to uncover genic and HERV expression differences between cell types. After doing so, differences in the HERV transcriptome between ontologically closely related CD4 and CD8 T cells became apparent. CONCLUSION: In our systematic approach to determine sequencing and analysis parameters for detection of locus-specific HERV expression, we provide evidence that analysis of RNAseq datasets from multiple studies can aid confidence of biological findings. When generating de novo HERV expression datasets we recommend increased sequence depth ( > = 100 mio reads) compared to standard genic transcriptome pipelines. Finally, batch effect reduction measures need to be implemented to allow for differential expression analysis.


Endogenous Retroviruses , Humans , Endogenous Retroviruses/genetics , Proviruses , Transcriptome , T-Lymphocytes
3.
PLoS One ; 16(12): e0261269, 2021.
Article En | MEDLINE | ID: mdl-34928979

Adeno-associated viruses (AAV) are considered non-pathogenic in humans, and thus have been developed into powerful vector platforms for in vivo gene therapy. Although the various AAV serotypes display broad tropism, frequently infecting multiple tissues and cell types, vectors for specific and efficient targeting of human CD4+ T lymphocytes are largely missing. In fact, a substantial translational bottleneck exists in the field of therapeutic gene transfer that would require in vivo delivery into peripheral disease-related lymphocytes for subsequent genome editing. To solve this issue, capsid modification for retargeting AAV tropism, and in turn improving vector potency, is considered a promising strategy. Here, we genetically modified the minor AAV2 capsid proteins, VP1 and VP2, with a set of novel nanobodies with high-affinity for the human CD4 receptor. These novel vector variants demonstrated improved targeting of human CD4+ cells, including primary human peripheral blood mononuclear cells (PBMC) and purified human CD4+ T lymphocytes. Thus, the technical approach presented here provides a promising strategy for developing specific gene therapy vectors, particularly targeting disease-related peripheral blood CD4+ leukocytes.


CD4-Positive T-Lymphocytes/metabolism , Capsid Proteins/genetics , Dependovirus/genetics , Genetic Vectors/administration & dosage , Leukocytes, Mononuclear/metabolism , Single-Domain Antibodies/chemistry , Transduction, Genetic , CD4-Positive T-Lymphocytes/immunology , Gene Transfer Techniques , HEK293 Cells , HeLa Cells , Humans , Leukocytes, Mononuclear/immunology
4.
Virus Res ; 293: 198260, 2021 02.
Article En | MEDLINE | ID: mdl-33316352

Chronic infection with human immunodeficiency virus (HIV)-1 is characterized by accumulation of proviral sequences in the genome of target cells. Integration of viral DNA in patients on long-term antiretroviral therapy selectively persists at preferential loci, suggesting site-specific crosstalk of viral sequences and human genes. This crosstalk likely contributes to chronic HIV disease through modulation of host immune pathways and emergence of clonal infected cell populations. To systematically interrogate such effects, we undertook genome engineering to generate Jurkat cell models that replicate integration of HIV-1 long terminal repeat (LTR) sequences at the BTB and CNC Homolog 2 (BACH2) integration locus. This locus is a prominent HIV-1 integration gene in chronic infection, found in 30 % of long-term treated patients with mapped proviral integrations. Using five clonal models carrying an LTR-driven reporter at different BACH2 intergenic regions, we here show that LTR transcriptional activity is repressed in BACH2 regions associated with proviral-DNA integrations in vivo but not in a control region. Our data indicates that this repression is in part epigenetically regulated, particularly through DNA methylation. Importantly, we demonstrate that transcriptional activity of the LTR is independent of BACH2 gene transcription and vice versa in our models. This suggests no transcriptional interference of endogenous and HIV-1 promoters. Taken together, our study provides first insights into how activity of HIV-1 LTR sequences is regulated at the BACH2 locus as prominent example for a recurrently-detected integration gene in chronic infection. Given the importance of integration-site dependent virus/host crosstalk for chronic HIV disease, our findings for the BACH2 locus have potential implications for future therapeutic strategies.


Basic-Leucine Zipper Transcription Factors/genetics , HIV-1 , HIV-1/genetics , Humans , Persistent Infection , Promoter Regions, Genetic , Proviruses/genetics , Virus Integration
5.
Semin Immunopathol ; 42(2): 187-200, 2020 04.
Article En | MEDLINE | ID: mdl-32047948

Human immunodeficiency virus 1 (HIV-1) replicates through the integration of its viral DNA into the genome of human immune target cells. Chronically infected individuals thus carry a genomic burden of virus-derived sequences that persists through antiretroviral therapy. This burden consists of a small fraction of intact, but transcriptionally silenced, i.e. latent, viral genomes and a dominant fraction of defective sequences. Remarkably, all viral-derived sequences are subject to interaction with host cellular physiology at various levels. In this review, we focus on epigenetic aspects of this interaction. We provide a comprehensive overview of how epigenetic mechanisms contribute to establishment and maintenance of HIV-1 gene repression during latency. We furthermore summarize findings indicating that HIV-1 infection leads to changes in the epigenome of target and bystander immune cells. Finally, we discuss how an improved understanding of epigenetic features and mechanisms involved in HIV-1 infection could be exploited for clinical use.


HIV Infections , HIV-1 , CD4-Positive T-Lymphocytes , Epigenesis, Genetic , HIV Infections/genetics , HIV-1/genetics , Humans , Virus Latency
6.
Eur J Immunol ; 49(1): 66-78, 2019 01.
Article En | MEDLINE | ID: mdl-30365177

The interferon-inducible transmembrane (Ifitm/Fragilis) genes encode homologous proteins that are induced by IFNs. Here, we show that IFITM proteins regulate murine CD4+ Th cell differentiation. Ifitm2 and Ifitm3 are expressed in wild-type (WT) CD4+ T cells. On activation, Ifitm3 was downregulated and Ifitm2 was upregulated. Resting Ifitm-family-deficient CD4+ T cells had higher expression of Th1-associated genes than WT and purified naive Ifitm-family-deficient CD4+ T cells differentiated more efficiently to Th1, whereas Th2 differentiation was inhibited. Ifitm-family-deficient mice, but not Ifitm3-deficient mice, were less susceptible than WT to induction of allergic airways disease, with a weaker Th2 response and less severe disease and lower Il4 but higher Ifng expression and IL-27 secretion. Thus, the Ifitm family is important in adaptive immunity, influencing Th1/Th2 polarization, and Th2 immunopathology.


Hypersensitivity/immunology , Inflammation/immunology , Membrane Proteins/metabolism , Respiratory System/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Cell Differentiation/genetics , Cells, Cultured , Interferon-gamma/metabolism , Interleukin-27/metabolism , Interleukin-4/metabolism , Lymphocyte Activation/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Th1-Th2 Balance/genetics
7.
J Vis Exp ; (141)2018 11 14.
Article En | MEDLINE | ID: mdl-30507915

Human immunodeficiency virus (HIV) integrates its proviral DNA non-randomly into the host cell genome at recurrent sites and genomic hotspots. Here we present a detailed protocol for the generation of novel in vitro models for HIV infection with chosen genomic integration sites using CRISPR-Cas9-based genome engineering technology. With this method, a reporter sequence of choice can be integrated into a targeted, chosen genomic locus, reflecting clinically relevant integration sites. In the protocol, the design of an HIV-derived reporter and choosing of a target site and gRNA sequence are described. A targeting vector with homology arms is constructed and transfected into Jurkat T cells. The reporter sequence is targeted to the selected genomic site by homologous recombination facilitated by a Cas9-mediated double-strand break at the target site. Single-cell clones are generated and screened for targeting events by flow cytometry and PCR. Selected clones are then expanded, and correct targeting is verified by PCR, sequencing, and Southern blotting. Potential off-target events of CRISPR-Cas9-mediated genome engineering are analyzed. By using this protocol, novel cell culture systems that model HIV infection at clinically relevant integration sites can be generated. Although the generation of single-cell clones and verification of correct reporter sequence integration is time-consuming, the resulting clonal lines are powerful tools to functionally analyze proviral integration site choice.


CRISPR-Cas Systems , Gene Editing , Genome , HIV Infections/genetics , HIV-1/genetics , Virus Integration , CRISPR-Associated Protein 9/genetics , Humans , Jurkat Cells , RNA, Guide, Kinetoplastida/genetics
8.
Nat Struct Mol Biol ; 25(8): 743, 2018 Aug.
Article En | MEDLINE | ID: mdl-29995840

In this article, the Ponceau staining presented in Fig. 1b (right, bottom) does not follow best practices for figure preparation since itinadvertently included duplications from the Ponceau staining presented in Supplementary Fig. 1b (for which the same preparation ofnucleosomes from HeLa cells had been used). A new Fig. 1b is provided in the Author Correction.

9.
Virus Res ; 249: 69-75, 2018 04 02.
Article En | MEDLINE | ID: mdl-29550509

HIV infection is characterized by accumulation of proviral sequences within the human host genome. Integration of viral-derived DNA occurs at preferential loci, suggesting a site-specific crosstalk between viral sequences and human genes. We here describe a genome engineering workflow to generate models for HIV-1 infection that for the first time recapitulate proviral integration at selected genomic loci and provide unique tools to study effects of HIV proviral integration site choice. Using this workflow, we have derived two BACH2-HIV-1 reporter models that mimic largely latent integration in the clinically relevant BACH2 gene locus, which has been associated with recurrent integration and HIV-reservoir maintenance in chronically infected patients.


HIV Infections/virology , HIV-1/physiology , Proviruses/physiology , Virus Integration , HIV-1/genetics , Humans , Jurkat Cells/virology , Models, Biological , Proviruses/genetics
10.
PLoS One ; 11(6): e0158294, 2016.
Article En | MEDLINE | ID: mdl-27341108

CRISPR/Cas9 technology is currently considered the most advanced tool for targeted genome engineering. Its sequence-dependent specificity has been explored for locus-directed transcriptional modulation. Such modulation, in particular transcriptional activation, has been proposed as key approach to overcome silencing of dormant HIV provirus in latently infected cellular reservoirs. Currently available agents for provirus activation, so-called latency reversing agents (LRAs), act indirectly through cellular pathways to induce viral transcription. However, their clinical performance remains suboptimal, possibly because reservoirs have diverse cellular identities and/or proviral DNA is intractable to the induced pathways. We have explored two CRISPR/Cas9-derived activator systems as targeted approaches to induce dormant HIV-1 proviral DNA. These systems recruit multiple transcriptional activation domains to the HIV 5' long terminal repeat (LTR), for which we have identified an optimal target region within the LTR U3 sequence. Using this target region, we demonstrate transcriptional activation of proviral genomes via the synergistic activation mediator complex in various in culture model systems for HIV latency. Observed levels of induction are comparable or indeed higher than treatment with established LRAs. Importantly, activation is complete, leading to production of infective viral particles. Our data demonstrate that CRISPR/Cas9-derived technologies can be applied to counteract HIV latency and may therefore represent promising novel approaches in the quest for HIV elimination.


CRISPR-Cas Systems , Gene Targeting , HIV Infections/virology , HIV-1/physiology , Proviruses , Virus Latency , Base Sequence , Binding Sites , Gene Editing , HIV Long Terminal Repeat , Humans , Jurkat Cells , Protein Binding , Proviruses/genetics , RNA, Guide, Kinetoplastida , Transcriptional Activation , Virus Latency/genetics , Virus Replication
11.
Nat Biotechnol ; 34(4): 401-9, 2016 Apr.
Article En | MEDLINE | ID: mdl-26900663

Current combination antiretroviral therapies (cART) efficiently suppress HIV-1 reproduction in humans, but the virus persists as integrated proviral reservoirs in small numbers of cells. To generate an antiviral agent capable of eradicating the provirus from infected cells, we employed 145 cycles of substrate-linked directed evolution to evolve a recombinase (Brec1) that site-specifically recognizes a 34-bp sequence present in the long terminal repeats (LTRs) of the majority of the clinically relevant HIV-1 strains and subtypes. Brec1 efficiently, precisely and safely removes the integrated provirus from infected cells and is efficacious on clinical HIV-1 isolates in vitro and in vivo, including in mice humanized with patient-derived cells. Our data suggest that Brec1 has potential for clinical application as a curative HIV-1 therapy.


Antiviral Agents/pharmacology , Directed Molecular Evolution/methods , HIV Infections/virology , HIV-1/drug effects , Proviruses/drug effects , Recombinases/pharmacology , Virus Integration/drug effects , Animals , Antiviral Agents/metabolism , Base Sequence , Cells, Cultured , HIV-1/genetics , Humans , Mice , Molecular Sequence Data , Proviruses/genetics , Recombinases/metabolism , Virus Integration/genetics
12.
Nat Commun ; 4: 2233, 2013.
Article En | MEDLINE | ID: mdl-23903902

To ensure genome stability, pericentromeric regions are compacted in a dense heterochromatic structure through a combination of specific 'epigenetic' factors and modifications. A cascadal pathway is responsible for establishing pericentromeric chromatin involving chromatin modifiers and 'readers', such as H3K9 histone methyltransferases (Suv)39h and heterochromatin protein 1. Here we define how H3K64me3 on the lateral surface of the histone octamer integrates within the heterochromatinization cascade. Our data suggest that enrichment of H3K64me3 at pericentromeric chromatin foci is dependent on H3K9me3 but independent of a number of central factors such as heterochromatin protein 1, DNA methyltransferases and Suv4-20h histone methyltransferases. Our results support a model in which pericentromeric heterochromatin foci are formed along distinct pathways upon H3K9 trimethylation, involving H3K64me3 to potentially stabilize DNA-histone interactions, as well as sequential recruitment of repressive histone tail and DNA modifications. We hence suggest that multiple mechanisms ensure heterochromatin integrity at pericentromeres, with H3K64me3 as an important factor.


Centromere/metabolism , Heterochromatin/metabolism , Histones/metabolism , Lysine/metabolism , Animals , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , DNA Methylation/genetics , Fluorescent Antibody Technique , Histone-Lysine N-Methyltransferase/metabolism , Humans , Mice , NIH 3T3 Cells , Zygote/metabolism
13.
Glia ; 61(5): 679-93, 2013 May.
Article En | MEDLINE | ID: mdl-23382131

Interferon-induced transmembrane protein 3 (IFITM3) iplays a crucial role in the antiviral responses of Type I interferons (IFNs). The role of IFITM3 in the central nervous system (CNS) is, however, largely unknown, despite the fact that its expression is increased in the brains of patients with neurologic and neuropsychiatric diseases. Here, we show the role of IFITM3 in long-lasting neuronal impairments in mice following polyriboinosinic-polyribocytidylic acid (polyI:C, a synthetic double-stranded RNA)-induced immune challenge during the early stages of development. We found that the induction of IFITM3 expression in the brain of mice treated with polyI:C was observed only in astrocytes. Cultured astrocytes were activated by polyI:C treatment, leading to an increase in the mRNA levels of inflammatory cytokines as well as Ifitm3. When cultured neurons were treated with the conditioned medium of polyI:C-treated astrocytes (polyI:C-ACM), neurite development was impaired. These polyI:C-ACM-induced neurodevelopmental abnormalities were alleviated by ifitm3(-/-) astrocyte-conditioned medium. Furthermore, decreases of MAP2 expression, spine density, and dendrite complexity in the frontal cortex as well as memory impairment were evident in polyI:C-treated wild-type mice, but such neuronal impairments were not observed in ifitm3(-) (/) (-) mice. We also found that IFITM3 proteins were localized to the early endosomes of astrocytes following polyI:C treatment and reduced endocytic activity. These findings suggest that the induction of IFITM3 expression in astrocytes by the activation of the innate immune system during the early stages of development has non-cell autonomous effects that affect subsequent neurodevelopment, leading to neuropathological impairments and brain dysfunction, by impairing endocytosis in astrocytes.


Astrocytes/pathology , Membrane Proteins/physiology , Neurons/pathology , Animals , Animals, Newborn , Astrocytes/immunology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cytokines/biosynthesis , Cytokines/genetics , Endocytosis/drug effects , Endocytosis/immunology , Female , Immunity, Innate/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Neurons/immunology , Poly I-C/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/drug effects
14.
Bioessays ; 32(8): 659-68, 2010 Aug.
Article En | MEDLINE | ID: mdl-20658704

During mammalian development, maintenance of cell fate through mitotic divisions require faithful replication not only of the DNA but also of a particular epigenetic state. Germline cells have the capacity of erasing this epigenetic memory at crucial times during development, thereby resetting their epigenome. Certain marks, however, appear to escape this reprogramming, which allows their transmission to the offspring and potentially guarantees transgenerational epigenetic inheritance. Here we discuss the molecular requirements for faithful transmission of epigenetic information and our current knowledge about the transmission of epigenetic information through generations.


Epigenesis, Genetic/genetics , Animals , DNA Methylation/genetics , Histones/genetics , Humans , Mitosis/genetics
15.
Nat Struct Mol Biol ; 16(7): 777-81, 2009 Jul.
Article En | MEDLINE | ID: mdl-19561610

Histone modifications are central to the regulation of all DNA-dependent processes. Lys64 of histone H3 (H3K64) lies within the globular domain at a structurally important position. We identify trimethylation of H3K64 (H3K64me3) as a modification that is enriched at pericentric heterochromatin and associated with repeat sequences and transcriptionally inactive genomic regions. We show that this new mark is dynamic during the two main epigenetic reprogramming events in mammals. In primordial germ cells, H3K64me3 is present at the time of specification, but it disappears transiently during reprogramming. In early mouse embryos, it is inherited exclusively maternally; subsequently, the modification is rapidly removed, suggesting an important role for H3K64me3 turnover in development. Taken together, our findings establish H3K64me3 as a previously uncharacterized histone modification that is preferentially localized to repressive chromatin. We hypothesize that H3K64me3 helps to 'secure' nucleosomes, and perhaps the surrounding chromatin, in an appropriately repressed state during development.


Epigenesis, Genetic , Heterochromatin/metabolism , Histones/metabolism , Lysine/metabolism , Animals , Cell Line , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Methylation , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/metabolism , Heterochromatin/chemistry , Heterochromatin/genetics , Histones/genetics , Humans , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Nucleosomes/chemistry , Nucleosomes/metabolism , Protein Conformation , Repressor Proteins/genetics , Repressor Proteins/metabolism , Xenopus laevis
16.
Mol Cell Biol ; 28(15): 4688-96, 2008 Aug.
Article En | MEDLINE | ID: mdl-18505827

The family of interferon-inducible transmembrane proteins (Ifitm) consists of five highly sequence-related cell surface proteins, which are implicated in diverse cellular processes. Ifitm genes are conserved, widely expressed, and characteristically found in genomic clusters, such as the 67-kb Ifitm family locus on mouse chromosome 7. Recently, Ifitm1 and Ifitm3 have been suggested to mediate migration of early primordial germ cells (PGCs), a process that is little understood. To investigate Ifitm function during germ cell development, we used targeted chromosome engineering to generate mutants which either lack the entire Ifitm locus or carry a disrupted Ifitm3 gene only. Here we show that the mutations have no detectable effects on development of the germ line or on the generation of live young. Hence, contrary to previous reports, Ifitm genes are not essential for PGC migration. The Ifitm family is a striking example of a conserved gene cluster which appears to be functionally redundant during development.


Gene Deletion , Germ Cells/growth & development , Membrane Proteins/genetics , Multigene Family , Animals , Cell Lineage , Cell Movement , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Fertility , Gene Expression Regulation, Developmental , Gene Targeting , Germ Cells/cytology , Gonads/cytology , Gonads/embryology , Green Fluorescent Proteins/metabolism , Homozygote , Mice
17.
Nature ; 452(7189): 877-81, 2008 Apr 17.
Article En | MEDLINE | ID: mdl-18354397

A unique feature of the germ cell lineage is the generation of totipotency. A critical event in this context is DNA demethylation and the erasure of parental imprints in mouse primordial germ cells (PGCs) on embryonic day 11.5 (E11.5) after they enter into the developing gonads. Little is yet known about the mechanism involved, except that it is apparently an active process. We have examined the associated changes in the chromatin to gain further insights into this reprogramming event. Here we show that the chromatin changes occur in two steps. The first changes in nascent PGCs at E8.5 establish a distinctive chromatin signature that is reminiscent of pluripotency. Next, when PGCs are residing in the gonads, major changes occur in nuclear architecture accompanied by an extensive erasure of several histone modifications and exchange of histone variants. Furthermore, the histone chaperones HIRA and NAP-1 (NAP111), which are implicated in histone exchange, accumulate in PGC nuclei undergoing reprogramming. We therefore suggest that the mechanism of histone replacement is critical for these chromatin rearrangements to occur. The marked chromatin changes are intimately linked with genome-wide DNA demethylation. On the basis of the timing of the observed events, we propose that if DNA demethylation entails a DNA repair-based mechanism, the evident histone replacement would represent a repair-induced response event rather than being a prerequisite.


Chromatin Assembly and Disassembly , Chromatin/metabolism , Epigenesis, Genetic , Germ Cells/metabolism , Animals , DNA Methylation , Gonads/cytology , Gonads/metabolism , Histones/metabolism , Mice , Stem Cells/metabolism
18.
Nat Cell Biol ; 8(6): 623-30, 2006 Jun.
Article En | MEDLINE | ID: mdl-16699504

Blimp1, a transcriptional repressor, has a crucial role in the specification of primordial germ cells (PGCs) in mice at embryonic day 7.5 (E7.5). This SET-PR domain protein can form complexes with various chromatin modifiers in a context-dependent manner. Here, we show that Blimp1 has a novel interaction with Prmt5, an arginine-specific histone methyltransferase, which mediates symmetrical dimethylation of arginine 3 on histone H2A and/or H4 tails (H2A/H4R3me2s). Prmt5 has been shown to associate with Tudor, a component of germ plasm in Drosophila melanogaster. Blimp1-Prmt5 colocalization results in high levels of H2A/H4 R3 methylation in PGCs at E8.5. However, at E11.5, Blimp1-Prmt5 translocates from the nucleus to the cytoplasm, resulting in the loss of H2A/H4 R3 methylation at the time of extensive epigenetic reprogramming of germ cells. Subsequently, Dhx38, a putative target of the Blimp1-Prmt5 complex, is upregulated. Interestingly, expression of Dhx38 is also seen in pluripotent embryonic germ cells that are derived from PGCs when Blimp1 expression is lost. Our study demonstrates that Blimp1 is involved in a novel transcriptional regulatory complex in the mouse germ-cell lineage.


Arginine/metabolism , Germ Cells/metabolism , Histones/metabolism , Repressor Proteins/physiology , Transcription Factors/physiology , Active Transport, Cell Nucleus , Adenosine Triphosphatases/genetics , Age Factors , Animals , Embryo, Mammalian , Gene Expression Regulation , Methylation , Mice , Positive Regulatory Domain I-Binding Factor 1 , Protein Binding , Protein Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic
19.
Mol Cell Biol ; 26(9): 3595-609, 2006 May.
Article En | MEDLINE | ID: mdl-16611999

Tumor suppressor of lung cancer 1 (TSLC1), also known as SgIGSF, IGSF4, and SynCAM, is strongly expressed in spermatogenic cells undergoing the early and late phases of spermatogenesis (spermatogonia to zygotene spermatocytes and elongating spermatids to spermiation). Using embryonic stem cell technology to generate a null mutation of Tslc1 in mice, we found that Tslc1 null male mice were infertile. Tslc1 null adult testes showed that spermatogenesis had arrested at the spermatid stage, with degenerating and apoptotic spermatids sloughing off into the lumen. In adult mice, Tslc1 null round spermatids showed evidence of normal differentiation (an acrosomal cap and F-actin polarization indistinguishable from that of wild-type spermatids); however, the surviving spermatozoa were immature, malformed, found at very low levels in the epididymis, and rarely motile. Analysis of the first wave of spermatogenesis in Tslc1 null mice showed a delay in maturation by day 22 and degeneration of round spermatids by day 28. Expression profiling of the testes revealed that Tslc1 null mice showed increases in the expression levels of genes involved in apoptosis, adhesion, and the cytoskeleton. Taken together, these data show that Tslc1 is essential for normal spermatogenesis in mice.


Immunoglobulins/physiology , Infertility, Male/genetics , Membrane Proteins/physiology , Spermatids/growth & development , Spermatogenesis/genetics , Spermatozoa/abnormalities , Tumor Suppressor Proteins/physiology , Animals , Apoptosis , Cell Adhesion/genetics , Cell Adhesion Molecule-1 , Cell Adhesion Molecules , Cell Differentiation , Cytoskeleton/genetics , Embryo, Mammalian/cytology , Epididymis/chemistry , Epididymis/cytology , Gene Expression , Immunoglobulins/analysis , Immunoglobulins/genetics , Infertility, Male/metabolism , Male , Membrane Proteins/analysis , Membrane Proteins/genetics , Mice , Mice, Mutant Strains , Mutation , Oligonucleotide Array Sequence Analysis , Sperm Motility/genetics , Spermatids/cytology , Spermatids/metabolism , Spermatozoa/cytology , Spermatozoa/metabolism , Stem Cells/metabolism , Testis/chemistry , Testis/cytology , Tumor Suppressor Proteins/analysis , Tumor Suppressor Proteins/genetics , Up-Regulation
20.
Philos Trans R Soc Lond B Biol Sci ; 358(1436): 1363-70, 2003 Aug 29.
Article En | MEDLINE | ID: mdl-14511483

An early fundamental event during development is the segregation of germ cells from somatic cells. In many organisms, this is accomplished by the inheritance of preformed germ plasm, which apparently imposes transcriptional repression to prevent somatic cell fate. However, in mammals, pluripotent epiblast cells acquire germ cell fate in response to signalling molecules. We have used single cell analysis to study how epiblast cells acquire germ cell competence and undergo specification. Germ cell competent cells express Fragilis and initially progress towards a somatic mesodermal fate. However, a subset of these cells, the future primordial germ cells (PGCs), then shows rapid upregulation of Fragilis with concomitant transcriptional repression of a number of genes, including Hox and Smad genes. This repression may be a key event associated with germ cell specification. Furthermore, PGCs express Stella and other genes, such as Oct-4 that are associated with pluripotency. While these molecules are also detected in mature oocytes as maternally inherited factors, their early role is to regulate development and maintain pluripotency, and they do not serve the role of classical germline determinants.


Cell Differentiation/physiology , Gene Expression Regulation, Developmental/physiology , Germ Cells/physiology , Pluripotent Stem Cells/physiology , Signal Transduction/physiology , Animals , Cell Differentiation/genetics , Gene Silencing/physiology , Germ Cells/cytology , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Transcription, Genetic/physiology , Up-Regulation/physiology
...