Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Aging Cell ; 23(5): e14105, 2024 May.
Article in English | MEDLINE | ID: mdl-38504487

ABSTRACT

Hutchinson-Gilford Progeria syndrome (HGPS) is a severe premature ageing disorder caused by a 50 amino acid truncated (Δ50AA) and permanently farnesylated lamin A (LA) mutant called progerin. On a cellular level, progerin expression leads to heterochromatin loss, impaired nucleocytoplasmic transport, telomeric DNA damage and a permanent growth arrest called cellular senescence. Although the genetic basis for HGPS has been elucidated 20 years ago, the question whether the Δ50AA or the permanent farnesylation causes cellular defects has not been addressed. Moreover, we currently lack mechanistic insight into how the only FDA-approved progeria drug Lonafarnib, a farnesyltransferase inhibitor (FTI), ameliorates HGPS phenotypes. By expressing a variety of LA mutants using a doxycycline-inducible system, and in conjunction with FTI, we demonstrate that the permanent farnesylation, and not the Δ50AA, is solely responsible for progerin-induced cellular defects, as well as its rapid accumulation and slow clearance. Importantly, FTI does not affect clearance of progerin post-farnesylation and we demonstrate that early, but not late FTI treatment prevents HGPS phenotypes. Collectively, our study unravels the precise contributions of progerin's permanent farnesylation to its turnover and HGPS cellular phenotypes, and how FTI treatment ameliorates these. These findings are applicable to other diseases associated with permanently farnesylated proteins, such as adult-onset autosomal dominant leukodystrophy.


Subject(s)
Lamin Type A , Progeria , Lamin Type A/metabolism , Lamin Type A/genetics , Humans , Progeria/metabolism , Progeria/genetics , Progeria/pathology , Progeria/drug therapy , Farnesyltranstransferase/metabolism , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/genetics , Protein Prenylation , Dibenzocycloheptenes , Piperidines , Pyridines
2.
PLoS One ; 19(2): e0292479, 2024.
Article in English | MEDLINE | ID: mdl-38349923

ABSTRACT

Recombinase enzymes are extremely efficient at integrating very large DNA fragments into target genomes. However, intrinsic sequence specificities curtail their use to DNA sequences with sufficient homology to endogenous target motifs. Extensive engineering is therefore required to broaden applicability and robustness. Here, we describe the directed evolution of novel lambda integrase variants capable of editing exogenous target sequences identified in the diatom Phaeodactylum tricornutum and the algae Nannochloropsis oceanica. These microorganisms hold great promise as conduits for green biomanufacturing and carbon sequestration. The evolved enzyme variants show >1000-fold switch in specificity towards the non-natural target sites when assayed in vitro. A single-copy target motif in the human genome with homology to the Nannochloropsis oceanica site can also be efficiently targeted using an engineered integrase, both in vitro and in human cells. The developed integrase variants represent useful additions to the DNA editing toolbox, with particular application for targeted genomic insertion of large DNA cargos.


Subject(s)
Diatoms , Stramenopiles , Humans , Integrases/genetics , Genome, Human/genetics , DNA , Genomics , Diatoms/genetics , Stramenopiles/genetics , Gene Editing
3.
Front Bioeng Biotechnol ; 11: 1198465, 2023.
Article in English | MEDLINE | ID: mdl-37425360

ABSTRACT

Reliable cell-based platforms to test and/or produce biologics in a sustainable manner are important for the biotech industry. Utilizing enhanced λ integrase, a sequence-specific DNA recombinase, we developed a novel transgenesis platform involving a fully characterized single genomic locus as an artificial landing pad for transgene insertion in human Expi293F cells. Importantly, transgene instability and variation in expression were not observed in the absence of selection pressure, thus enabling reliable long-term biotherapeutics testing or production. The artificial landing pad for λ integrase can be targeted with multi-transgene constructs and offers future modularity involving additional genome manipulation tools to generate sequential or nearly seamless insertions. We demonstrated broad utility with expression constructs for anti PD-1 monoclonal antibodies and showed that the orientation of heavy and light chain transcription units profoundly affected antibody expression levels. In addition, we demonstrated encapsulation of our PD-1 platform cells into bio-compatible mini-bioreactors and the continued secretion of antibodies, thus providing a basis for future cell-based applications for more effective and affordable therapies.

4.
FEBS Lett ; 597(15): 1977-1988, 2023 08.
Article in English | MEDLINE | ID: mdl-37259564

ABSTRACT

The architectural chromatin factor high-mobility group AT-hook 2 (HMGA2) is causally involved in several human malignancies and pathologies. HMGA2 is not expressed in most normal adult somatic cells, which renders the protein an attractive drug target. An established cell-based compound library screen identified the fibroblast growth factor receptor (FGFR) inhibitor PD173074 as an antagonist of HMGA2-mediated transcriptional reporter gene activation. We determined that PD173074 binds the C-terminus of HMGA2 and interferes with functional coordination of the three AT-hook DNA-binding domains mediated by the C-terminus. The HMGA2-antagonistic effect of PD173074 on transcriptional activation may therefore result from an induced altered DNA-binding mode of HMGA2. PD173074 as a novel HMGA2-specific antagonist could trigger the development of derivates with enhanced attributes and clinical potential.


Subject(s)
Neoplasms , Receptors, Fibroblast Growth Factor , Adult , Humans , Transcriptional Activation , Chromatin , DNA/metabolism , HMGA2 Protein/genetics , HMGA2 Protein/metabolism
5.
Nat Commun ; 14(1): 1919, 2023 04 06.
Article in English | MEDLINE | ID: mdl-37024489

ABSTRACT

Alternative lengthening of telomeres (ALT) supports telomere maintenance in 10-15% of cancers, thus representing a compelling target for therapy. By performing anti-cancer compound library screen on isogenic cell lines and using extrachromosomal telomeric C-circles, as a bona fide marker of ALT activity, we identify a receptor tyrosine kinase inhibitor ponatinib that deregulates ALT mechanisms, induces telomeric dysfunction, reduced ALT-associated telomere synthesis, and targets, in vivo, ALT-positive cells. Using RNA-sequencing and quantitative phosphoproteomic analyses, combined with C-circle level assessment, we find an ABL1-JNK-JUN signalling circuit to be inhibited by ponatinib and to have a role in suppressing telomeric C-circles. Furthermore, transcriptome and interactome analyses suggest a role of JUN in DNA damage repair. These results are corroborated by synergistic drug interactions between ponatinib and either DNA synthesis or repair inhibitors, such as triciribine. Taken together, we describe here a signalling pathway impacting ALT which can be targeted by a clinically approved drug.


Subject(s)
Signal Transduction , Telomere , Cell Survival , Signal Transduction/drug effects , Gene Expression Regulation , DNA Repair , DNA Replication , JNK Mitogen-Activated Protein Kinases/metabolism , Humans , Animals , Mice , Cell Line, Tumor
6.
PLoS One ; 17(9): e0270173, 2022.
Article in English | MEDLINE | ID: mdl-36149906

ABSTRACT

Seamless DNA vectors derived from bacterial plasmids are devoid of bacterial genetic elements and represent attractive alternatives for biomedical applications including DNA vaccines. Larger scale production of seamless vectors employs engineered Escherichia coli strains in order to enable tightly regulated expression of site-specific DNA recombinases which precisely delete unwanted sequences from bacterial plasmids. As a novel component of a developing lambda integrase genome editing platform, we describe here strain MG1655-ISC as a means to easily produce different scales of seamless vectors, ranging in size from a few hundred base pairs to more than ten kilo base pairs. Since we employed an engineered lambda integrase that is able to efficiently recombine pairs of DNA crossover sites that differ in sequence, the resulting seamless vectors will be useful for subsequent genome editing in higher eukaryotes to accommodate variations in target site sequences. Future inclusion of single cognate sites for other genome targeting systems could enable modularity. These features, together with the demonstrated simplicity of in vivo seamless vector production, add to their utility in the biomedical space.


Subject(s)
Bacteriophage lambda , Vaccines, DNA , Bacteriophage lambda/genetics , DNA , Escherichia coli/genetics , Escherichia coli/metabolism , Genetic Vectors/genetics , Integrases/genetics , Integrases/metabolism , Plasmids/genetics , Recombination, Genetic , Vaccines, DNA/genetics
7.
Microb Pathog ; 158: 105105, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34311016

ABSTRACT

COVID-19 exhibits a global health threat among the elderly and the population with underlying health conditions. During infection, the host's innate immune response acts as a frontline of defense by releasing cytokines such as type I interferon (IFN α and ß) thereby initiating antiviral activity. However, this particular interferon response is interrupted by factors such as SARS-CoV-2 non-structural proteins, aging, diabetes, and germ-line errors eventually making the host more susceptible to illness. Therefore, enhancing the host's innate immune response by administering type I IFN could be an effective treatment against COVID-19. Here, we highlight the importance of innate immune response and the role of IFN ß monotherapy against COVID-19.


Subject(s)
COVID-19 , Interferon Type I , Aged , Humans , Immunity, Innate , Interferon-beta , SARS-CoV-2
8.
Stem Cell Res Ther ; 11(1): 380, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32883366

ABSTRACT

BACKGROUND: The precise, functional and safe insertion of large DNA payloads into host genomes offers versatility in downstream genetic engineering-associated applications, spanning cell and gene therapies, therapeutic protein production, high-throughput cell-based drug screening and reporter cell lines amongst others. Employing viral- and non-viral-based genome engineering tools to achieve specific insertion of large DNA-despite being successful in E. coli and animal models-still pose challenges in the human system. In this study, we demonstrate the applicability of our lambda integrase-based genome insertion tool for human cell and gene therapy applications that require insertions of large functional genes, as exemplified by the integration of a functional copy of the F8 gene and a Double Homeobox Protein 4 (DUX4)-based reporter cassette for potential hemophilia A gene therapy and facioscapulohumeral muscular dystrophy (FSHD)-based high-throughput drug screening purposes, respectively. Thus, we present a non-viral genome insertion tool for safe and functional delivery of large seamless DNA cargo into the human genome that can enable novel designer cell-based therapies. METHODS: Previously, we have demonstrated the utility of our phage λ-integrase platform to generate seamless vectors and subsequently achieve functional integration of large-sized DNA payloads at defined loci in the human genome. To further explore this tool for therapeutic applications, we used pluripotent human embryonic stem cells (hESCs) to integrate large seamless vectors comprising a 'gene of interest'. Clonal cell populations were screened for the correct integration events and further characterized by southern blotting, gene expression and protein activity assays. In the case of our hemophilia A-related study, clones were differentiated to confirm that the targeted locus is active after differentiation and actively express and secrete Factor VIII. RESULTS: The two independent approaches demonstrated specific and functional insertions of a full-length blood clotting F8 expression cassette of ~ 10 kb and of a DUX4 reporter cassette of ~ 7 kb in hESCs. CONCLUSION: We present a versatile tool for site-specific human genome engineering with large transgenes for cell/gene therapies and other synthetic biology and biomedical applications.


Subject(s)
Escherichia coli , Gene Editing , Animals , Escherichia coli/genetics , Genetic Vectors/genetics , Humans , Integrases/genetics , Transgenes
9.
Genome Res ; 30(7): 1027-1039, 2020 07.
Article in English | MEDLINE | ID: mdl-32699019

ABSTRACT

Joint profiling of transcriptome and chromatin accessibility within single cells allows for the deconstruction of the complex relationship between transcriptional states and upstream regulatory programs determining different cell fates. Here, we developed an automated method with high sensitivity, assay for single-cell transcriptome and accessibility regions (ASTAR-seq), for simultaneous measurement of whole-cell transcriptome and chromatin accessibility within the same single cell. To show the utility of ASTAR-seq, we profiled 384 mESCs under naive and primed pluripotent states as well as a two-cell like state, 424 human cells of various lineage origins (BJ, K562, JK1, and Jurkat), and 480 primary cord blood cells undergoing erythroblast differentiation. With the joint profiles, we configured the transcriptional and chromatin accessibility landscapes of discrete cell states, uncovered linked sets of cis-regulatory elements and target genes unique to each state, and constructed interactome and transcription factor (TF)-centered upstream regulatory networks for various cell states.


Subject(s)
Chromatin/metabolism , Gene Expression Profiling/methods , Gene Regulatory Networks , Single-Cell Analysis/methods , Animals , Cell Differentiation , Cell Line , Cells, Cultured , Embryonic Stem Cells , Epigenesis, Genetic , Erythroblasts/cytology , Erythroblasts/metabolism , Humans , Mice , Regulatory Elements, Transcriptional , Transcription Factors/metabolism , Transcriptome
10.
Int J Mol Sci ; 21(12)2020 Jun 24.
Article in English | MEDLINE | ID: mdl-32599919

ABSTRACT

Key DNA transactions, such as genome replication and transcription, rely on the speedy translocation of specialized protein complexes along a double-stranded, right-handed helical template. Physical tethering of these molecular machines during translocation, in conjunction with their internal architectural features, generates DNA topological strain in the form of template supercoiling. It is known that the build-up of transient excessive supercoiling poses severe threats to genome function and stability and that highly specialized enzymes-the topoisomerases (TOP)-have evolved to mitigate these threats. Furthermore, due to their intracellular abundance and fast supercoil relaxation rates, it is generally assumed that these enzymes are sufficient in coping with genome-wide bursts of excessive supercoiling. However, the recent discoveries of chromatin architectural factors that play important accessory functions have cast reasonable doubts on this concept. Here, we reviewed the background of these new findings and described emerging models of how these accessory factors contribute to supercoil homeostasis. We focused on DNA replication and the generation of positive (+) supercoiling in front of replisomes, where two accessory factors-GapR and HMGA2-from pro- and eukaryotic cells, respectively, appear to play important roles as sinks for excessive (+) supercoiling by employing a combination of supercoil constrainment and activation of topoisomerases. Looking forward, we expect that additional factors will be identified in the future as part of an expanding cellular repertoire to cope with bursts of topological strain. Furthermore, identifying antagonists that target these accessory factors and work synergistically with clinically relevant topoisomerase inhibitors could become an interesting novel strategy, leading to improved treatment outcomes.


Subject(s)
Chromatin/chemistry , Chromatin/genetics , DNA Replication , DNA, Superhelical , Gene Expression Regulation , Transcription, Genetic , Animals , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/metabolism , HMGA2 Protein/metabolism , Humans
11.
Sci Rep ; 10(1): 9397, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32523028

ABSTRACT

Pathogenicity islands (PAIs) represent horizontally acquired chromosomal regions and encode their cognate integrase, which mediates chromosomal integration and excision of the island. These site-specific recombination reactions have to be tightly controlled to maintain genomic stability, and their directionality depends on accessory proteins. The integration host factor (IHF) and the factor for inversion stimulation (Fis) are often involved in recombinogenic complex formation and controlling the directionality of the recombination reaction. We investigated the role of the accessory host factors IHF and Fis in controlling the stability of six PAIs in uropathogenic Escherichia coli strain 536. By comparing the loss of individual PAIs in the presence or absence of IHF or Fis, we showed that IHF specifically stabilized PAI I536 and that in particular the IHFB subunit seems to be important for this function. We employed complex genetic studies to address the role of IHF in PAI I536-encoded integrase (IntI) expression. Based on different YFP-reporter constructs and electrophoretic mobility shift assays we demonstrated that IntI acts a strong repressor of its own synthesis, and that IHF binding to the intI promoter region reduces the probability of intI promoter activation. Our results extend the current knowledge of the role of IHF in controlling directionality of site specific recombination reactions and thus PAI stability.


Subject(s)
Escherichia coli Proteins/genetics , Genomic Islands/genetics , Integrases/genetics , Integration Host Factors/genetics , Promoter Regions, Genetic/genetics , Uropathogenic Escherichia coli/genetics , Uropathogenic Escherichia coli/pathogenicity , Factor For Inversion Stimulation Protein/genetics , Gene Expression Regulation, Bacterial/genetics , Recombination, Genetic/genetics
12.
Angew Chem Int Ed Engl ; 59(32): 13295-13304, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32337801

ABSTRACT

The F1 FO -ATP synthase is required for growth and viability of Mycobacterium tuberculosis and is a validated clinical target. A mycobacterium-specific loop of the enzyme's rotary γ subunit plays a role in the coupling of ATP synthesis within the enzyme complex. We report the discovery of a novel antimycobacterial, termed GaMF1, that targets this γ subunit loop. Biochemical and NMR studies show that GaMF1 inhibits ATP synthase activity by binding to the loop. GaMF1 is bactericidal and is active against multidrug- as well as bedaquiline-resistant strains. Chemistry efforts on the scaffold revealed a dynamic structure activity relationship and delivered analogues with nanomolar potencies. Combining GaMF1 with bedaquiline or novel diarylquinoline analogues showed potentiation without inducing genotoxicity or phenotypic changes in a human embryonic stem cell reporter assay. These results suggest that GaMF1 presents an attractive lead for the discovery of a novel class of anti-tuberculosis F-ATP synthase inhibitors.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Bacterial Proton-Translocating ATPases/antagonists & inhibitors , Diarylquinolines/pharmacology , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/drug effects , Benzamides/chemistry , Benzamides/pharmacology , Benzamides/toxicity , Drug Synergism , Embryonic Stem Cells/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/toxicity , Humans , Microbial Sensitivity Tests , Molecular Structure , Mycobacterium tuberculosis/enzymology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrimidines/toxicity , Structure-Activity Relationship
13.
Mol Oncol ; 13(10): 2062-2078, 2019 10.
Article in English | MEDLINE | ID: mdl-31271486

ABSTRACT

Rapidly dividing cells maintain chromatin supercoiling homeostasis via two specialized classes of enzymes, DNA topoisomerase type 1 and 2 (TOP1/2). Several important anticancer drugs perturb this homeostasis by targeting TOP1/2, thereby generating genotoxic DNA damage. Our recent studies indicated that the oncofetal chromatin structuring high-mobility group AT-hook 2 (HMGA2) protein plays an important role as a DNA replication fork chaperone in coping with DNA topological ramifications that occur during replication stress, both genomewide and at fragile sites such as subtelomeres. Intriguingly, a recent large-scale clinical study identified HMGA2 expression as a sole predicting marker for relapse and poor clinical outcomes in 350 acute myeloid leukemia (AML) patients receiving combinatorial treatments that targeted TOP2 and replicative DNA synthesis. Here, we demonstrate that HMGA2 significantly enhanced the DNA supercoil relaxation activity of the drug target TOP2A and that this activator function is mechanistically linked to HMGA2's known ability to constrain DNA supercoils within highly compacted ternary complexes. Furthermore, we show that HMGA2 significantly reduced genotoxic DNA damage in each tested cancer cell model during treatment with the TOP2A poison etoposide or the catalytic TOP2A inhibitor merbarone. Taken together with the recent clinical data obtained with AML patients targeted with TOP2 poisons, our study suggests a novel mechanism of cancer chemoresistance toward combination therapies administering TOP2 poisons or inhibitors. We therefore strongly argue for the future implementation of trials of HMGA2 expression profiling to stratify patients before finalizing clinical treatment regimes.


Subject(s)
DNA Damage/drug effects , DNA/genetics , Etoposide/toxicity , HMGA2 Protein/metabolism , Thiobarbiturates/toxicity , Topoisomerase II Inhibitors/toxicity , Cell Line, Tumor , DNA/chemistry , DNA/metabolism , DNA Topoisomerases, Type II/metabolism , Humans , Nucleic Acid Conformation/drug effects , Poly-ADP-Ribose Binding Proteins/antagonists & inhibitors , Poly-ADP-Ribose Binding Proteins/metabolism
14.
PLoS One ; 14(5): e0215696, 2019.
Article in English | MEDLINE | ID: mdl-31067275

ABSTRACT

The transient build-up of DNA supercoiling during the translocation of replication forks threatens genome stability and is controlled by DNA topoisomerases (TOPs). This crucial process has been exploited with TOP poisons for cancer chemotherapy. However, pinpointing cellular determinants of the best clinical response to TOP poisons still remains enigmatic. Here, we present an integrated approach and demonstrate that endogenous and exogenous expression of the oncofetal high-mobility group AT-hook 2 (HMGA2) protein exhibited broad protection against the formation of hydroxyurea-induced DNA breaks in various cancer cells, thus corroborating our previously proposed model in which HMGA2 functions as a replication fork chaperone that forms a protective DNA scaffold at or close to stalled replication forks. We now further demonstrate that high levels of HMGA2 also protected cancer cells against DNA breaks triggered by the clinically important TOP1 poison irinotecan. This protection is most likely due to the recently identified DNA supercoil constraining function of HMGA2 in combination with exclusion of TOP1 from binding to supercoiled substrate DNA. In contrast, low to moderate HMGA2 protein levels surprisingly potentiated the formation of irinotecan-induced genotoxic covalent TOP1-DNA cleavage complexes. Our data from cell-based and several in vitro assays indicate that, mechanistically, this potentiating role involves enhanced drug-target interactions mediated by HMGA2 in ternary complexes with supercoiled DNA. Subtelomeric regions were found to be extraordinarily vulnerable to these genotoxic challenges induced by TOP1 poisoning, pointing at strong DNA topological barriers located at human telomeres. These findings were corroborated by an increased irinotecan sensitivity of patient-derived xenografts of colorectal cancers exhibiting low to moderate HMGA2 levels. Collectively, we uncovered a therapeutically important control mechanism of transient changes in chromosomal DNA topology that ultimately leads to enhanced human subtelomere stability.


Subject(s)
Chromatin/metabolism , HMGA2 Protein/metabolism , Telomere/genetics , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Replication/genetics , DNA Topoisomerases, Type I/metabolism , Female , Gene Expression Regulation, Neoplastic , HMGA2 Protein/genetics , Humans , Male
15.
Elife ; 72018 10 25.
Article in English | MEDLINE | ID: mdl-30355447

ABSTRACT

Telomerase RNA (TR) provides the template for DNA repeat synthesis at telomeres and is essential for genome stability in continuously dividing cells. We mapped the RNA interactome of human TR (hTR) and identified a set of non-coding and coding hTR-interacting RNAs, including the histone 1C mRNA (HIST1H1C). Disruption of the hTR-HIST1H1C RNA association resulted in markedly increased telomere elongation without affecting telomerase enzymatic activity. Conversely, over-expression of HIST1H1C led to telomere attrition. By using a combination of mutations to disentangle the effects of histone 1 RNA synthesis, protein expression, and hTR interaction, we show that HIST1H1C RNA negatively regulates telomere length independently of its protein coding potential. Taken together, our data provide important insights into a surprisingly complex hTR-RNA interaction network and define an unexpected non-coding RNA role for HIST1H1C in regulating telomere length homeostasis, thus offering a glimpse into the mostly uncharted, vast space of non-canonical messenger RNA functions.


Subject(s)
Histones/genetics , RNA, Messenger/metabolism , RNA/metabolism , Telomerase/metabolism , Telomere Homeostasis , Cell Line , Humans
16.
Nucleic Acids Res ; 46(16): e99, 2018 09 19.
Article in English | MEDLINE | ID: mdl-29893931

ABSTRACT

Advances in stem cell engineering, gene therapy and molecular medicine often involve genome engineering at a cellular level. However, functionally large or multi transgene cassette insertion into the human genome still remains a challenge. Current practices such as random transgene integration or targeted endonuclease-based genome editing are suboptimal and might pose safety concerns. Taking this into consideration, we previously developed a transgenesis tool derived from phage λ integrase (Int) that precisely recombines large plasmid DNA into an endogenous sequence found in human Long INterspersed Elements-1 (LINE-1). Despite this advancement, biosafety concerns associated with bacterial components of plasmids, enhanced uptake and efficient transgene expression remained problematic. We therefore further improved and herein report a more superior Int-based transgenesis tool. This novel Int platform allows efficient and easy derivation of sufficient amounts of seamless supercoiled transgene vectors from conventional plasmids via intramolecular recombination as well as subsequent intermolecular site-specific genome integration into LINE-1. Furthermore, we identified certain LINE-1 as preferred insertion sites for Int-mediated seamless vector transgenesis, and showed that targeted anti-CD19 chimeric antigen receptor gene integration achieves high-level sustained transgene expression in human embryonic stem cell clones for potential downstream therapeutic applications.


Subject(s)
Bacteriophage lambda/genetics , Genetic Engineering/methods , Genetic Vectors/genetics , Integrases/genetics , Recombinant Fusion Proteins/metabolism , Transgenes/genetics , Bacteriophage lambda/enzymology , Gene Editing/methods , Gene Expression , Genetic Therapy/methods , Humans , Integrases/metabolism , Long Interspersed Nucleotide Elements/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use
17.
Sci Rep ; 8(1): 1664, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29362495

ABSTRACT

A correction to this article has been published and is linked from the HTML version of this paper. The error has been fixed in the paper.

18.
Sci Rep ; 7(1): 14612, 2017 11 06.
Article in English | MEDLINE | ID: mdl-29097685

ABSTRACT

Undifferentiated embryonic cell transcription factor 1 (Utf1) is expressed in pluripotent embryonic stem cells (ESCs) and primordial germ cells (PGCs). Utf1 expression is directly controlled by pluripotency factors Oct4 and Sox2, which form a ternary complex with the Utf1 enhancer. The Utf1 protein plays a role in chromatin organization and epigenetic control of bivalent gene expression in ESCs in vitro, where it promotes effective cell differentiation during exit from pluripotency. The function of Utf1 in PGCs in vivo, however, is not known. Here, we report that proper development of Utf1 null embryos almost entirely depends on the presence of functional Utf1 alleles in the parental germline. This indicates that Utf1's proposed epigenetic role in ESC pluripotency in vitro may be linked to intergenerational epigenetic inheritance in vivo. One component - or at least facilitator - of the relevant epigenetic mark appears to be Utf1 itself, since Utf1-driven tomato reporter and Utf1 are detected in mature germ cells. We also provide initial evidence for a reduced adult testis size in Utf1 null mice. Our findings thus point at unexpected functional links between the core ESC pluripotency factor network and epigenetic inheritance of pluripotency.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Epigenesis, Genetic , Pluripotent Stem Cells/metabolism , Trans-Activators/metabolism , Animals , Chromosomal Proteins, Non-Histone/genetics , Female , Gametogenesis/physiology , Gene Expression , Germ Cells/metabolism , Kidney/growth & development , Kidney/metabolism , Kidney/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Organ Size , Pluripotent Stem Cells/pathology , RNA, Messenger/metabolism , Testis/growth & development , Testis/metabolism , Testis/pathology , Trans-Activators/genetics
19.
Sci Rep ; 7(1): 8440, 2017 08 16.
Article in English | MEDLINE | ID: mdl-28814752

ABSTRACT

HMGA2 belongs to the family of the high mobility group (HMG) proteins. It binds DNA via three AT-hook domains to the minor groove of adenine-thymine (AT) rich DNA. Recently, a new function of HMGA2 as a replication fork chaperone that protects stem and cancer cells from replication fork collapse induced by chemotherapeutic agents was uncovered, suggesting a previously uncharacterized binding at replication forks. In this study, we examined HMGA2 binding to four DNA structures relevant to replication forks, namely ds DNA, ss DNA, forked DNA and supercoiled DNA plectonemes. We detected HMGA2 binding to supercoiled DNA at the lowest concentration and this binding mode transiently stabilizes the supercoiled plectonemes against relaxation by type I topoisomerase. Together, these findings suggest a plausible mechanism how fork regression and collapse are attenuated by HMGA2 during replication stress, i.e. through transient stabilization of positively supercoiled plectonemes in the parental duplex.


Subject(s)
DNA, Single-Stranded/metabolism , DNA, Superhelical/metabolism , DNA/metabolism , HMGA2 Protein/metabolism , DNA/chemistry , DNA Replication , DNA, Single-Stranded/chemistry , DNA, Superhelical/chemistry , Fetus/metabolism , HMGA2 Protein/genetics , Humans , Magnetics/methods , Models, Molecular , Neoplasms/metabolism , Nucleic Acid Conformation , Optical Tweezers , Protein Binding , Recombinant Proteins/metabolism
20.
Nucleic Acids Res ; 44(22): e162, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27587582

ABSTRACT

HMGA2 is an important chromatin factor that interacts with DNA via three AT-hook domains, thereby regulating chromatin architecture and transcription during embryonic and fetal development. The protein is absent from differentiated somatic cells, but aberrantly re-expressed in most aggressive human neoplasias where it is causally linked to cell transformation and metastasis. DNA-binding also enables HMGA2 to protect cancer cells from DNA-damaging agents. HMGA2 therefore is considered to be a prime drug target for many aggressive malignancies. Here, we have developed a broadly applicable cell-based reporter system which can identify HMGA2 antagonists targeting functionally important protein domains, as validated with the known AT-hook competitor netropsin. In addition, high-throughput screening can uncover functional links between HMGA2 and cellular factors important for cell transformation. This is demonstrated with the discovery that HMGA2 potentiates the clinically important topoisomerase I inhibitor irinotecan/SN-38 in trapping the enzyme in covalent DNA-complexes, thereby attenuating transcription.


Subject(s)
Camptothecin/analogs & derivatives , DNA Topoisomerases, Type I/physiology , HMGA2 Protein/physiology , Topoisomerase I Inhibitors/pharmacology , Amino Acid Sequence , Camptothecin/pharmacology , Cell Differentiation/drug effects , Enzyme Stability , Genes, Reporter , HEK293 Cells , HeLa Cells , High-Throughput Screening Assays , Humans , Irinotecan , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Netropsin/pharmacology , Promoter Regions, Genetic , Transcription, Genetic/drug effects , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...