Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Nucleic Acids Res ; 51(4): 1662-1673, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36156096

ABSTRACT

The histone H3 variant, H3.3, is localized at specific regions in the genome, especially promoters and active enhancers, and has been shown to play important roles in development. A lysine to methionine substitution in position 27 (H3.3K27M) is a main cause of Diffuse Intrinsic Pontine Glioma (specifically Diffuse Midline Glioma, K27M-mutant), a lethal type of pediatric cancer. H3.3K27M has a dominant-negative effect by inhibiting the Polycomb Repressor Complex 2 (PRC2) activity. Here, we studied the immediate, genome-wide, consequences of the H3.3K27M mutation independent of PRC2 activity. We developed Doxycycline (Dox)-inducible mouse embryonic stem cells (ESCs) carrying a single extra copy of WT-H3.3, H3.3K27M and H3.3K27L, all fused to HA. We performed RNA-Seq and ChIP-Seq at different times following Dox induction in undifferentiated and differentiated ESCs. We find increased binding of H3.3 around transcription start sites in cells expressing both H3.3K27M and H3.3K27L compared with WT, but not in cells treated with PRC2 inhibitors. Differentiated cells carrying either H3.3K27M or H3.3K27L retain expression of ESC-active genes, in expense of expression of genes related to neuronal differentiation. Taken together, our data suggest that a modifiable H3.3K27 is required for proper histone incorporation and cellular maturation, independent of PRC2 activity.


Subject(s)
Embryonic Stem Cells , Histones , Animals , Mice , Cell Differentiation , Cell Nucleus/metabolism , Gene Expression Regulation , Glioma/genetics , Histones/metabolism , Mutation , Polycomb-Group Proteins/metabolism , Doxycycline/pharmacology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism
2.
Mol Psychiatry ; 26(2): 666-681, 2021 02.
Article in English | MEDLINE | ID: mdl-30953002

ABSTRACT

Mutations in AUTS2 are associated with autism, intellectual disability, and microcephaly. AUTS2 is expressed in the brain and interacts with polycomb proteins, yet it is still unclear how mutations in AUTS2 lead to neurodevelopmental phenotypes. Here we report that when neuronal differentiation is initiated, there is a shift in expression from a long isoform to a short AUTS2 isoform. Yeast two-hybrid screen identified the splicing factor SF3B1 as an interactor of both isoforms, whereas the polycomb group proteins, PCGF3 and PCGF5, were found to interact exclusively with the long AUTS2 isoform. Reporter assays showed that the first exons of the long AUTS2 isoform function as a transcription repressor, but the part that consist of the short isoform acts as a transcriptional activator, both influenced by the cellular context. The expression levels of PCGF3 influenced the ability of the long AUTS2 isoform to activate or repress transcription. Mouse embryonic stem cells (mESCs) with heterozygote mutations in Auts2 had an increase in cell death during in vitro corticogenesis, which was significantly rescued by overexpressing the human AUTS2 transcripts. mESCs with a truncated AUTS2 protein (missing exons 12-20) showed premature neuronal differentiation, whereas cells overexpressing AUTS2, especially the long transcript, showed increase in expression of pluripotency markers and delayed differentiation. Taken together, our data suggest that the precise expression of AUTS2 isoforms is essential for regulating transcription and the timing of neuronal differentiation.


Subject(s)
Cell Differentiation , Cytoskeletal Proteins , Neurons/cytology , Transcription Factors , Animals , Exons , Mice , Phenotype , Protein Isoforms/genetics , Transcription Factors/genetics
3.
Nucleic Acids Res ; 41(12): 6300-15, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23630323

ABSTRACT

The transcriptional landscape in embryonic stem cells (ESCs) and during ESC differentiation has received considerable attention, albeit mostly confined to the polyadenylated fraction of RNA, whereas the non-polyadenylated (NPA) fraction remained largely unexplored. Notwithstanding, the NPA RNA super-family has every potential to participate in the regulation of pluripotency and stem cell fate. We conducted a comprehensive analysis of NPA RNA in ESCs using a combination of whole-genome tiling arrays and deep sequencing technologies. In addition to identifying previously characterized and new non-coding RNA members, we describe a group of novel conserved RNAs (snacRNAs: small NPA conserved), some of which are differentially expressed between ESC and neuronal progenitor cells, providing the first evidence of a novel group of potentially functional NPA RNA involved in the regulation of pluripotency and stem cell fate. We further show that minor spliceosomal small nuclear RNAs, which are NPA, are almost completely absent in ESCs and are upregulated in differentiation. Finally, we show differential processing of the minor intron of the polycomb group gene Eed. Our data suggest that NPA RNA, both known and novel, play important roles in ESCs.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , RNA, Small Untranslated/metabolism , Transcription, Genetic , Animals , Cells, Cultured , Epigenesis, Genetic , Histones/genetics , Male , Mice , Proteins/genetics , RNA Polymerase II/metabolism , RNA Splicing , RNA, Small Untranslated/biosynthesis , RNA, Small Untranslated/physiology , Spliceosomes/metabolism
4.
Mol Ther Methods Clin Dev ; 29: 329-346, 2023 Jun 08.
Article in English | MEDLINE | ID: mdl-37214315

ABSTRACT

Upscaling of kidney epithelial cells is crucial for renal regenerative medicine. Nonetheless, the adult kidney lacks a distinct stem cell hierarchy, limiting the ability to long-term propagate clonal populations of primary cells that retain renal identity. Toward this goal, we tested the paradigm of shifting the balance between differentiation and stemness in the kidney by introducing a single pluripotency factor, OCT4. Here we show that ectopic expression of OCT4 in human adult kidney epithelial cells (hKEpC) induces the cells to dedifferentiate, stably proliferate, and clonally emerge over many generations. Control hKEpC dedifferentiate, assume fibroblastic morphology, and completely lose clonogenic capacity. Analysis of gene expression and histone methylation patterns revealed that OCT4 represses the HNF1B gene module, which is critical for kidney epithelial differentiation, and concomitantly activates stemness-related pathways. OCT4-hKEpC can be long-term expanded in the dedifferentiated state that is primed for renal differentiation. Thus, when expanded OCT4-hKEpC are grown as kidney spheroids (OCT4-kSPH), they reactivate the HNF1B gene signature, redifferentiate, and efficiently generate renal structures in vivo. Hence, changes occurring in the cellular state of hKEpC following OCT4 induction, long-term propagation, and 3D aggregation afford rapid scale-up technology of primary renal tissue-forming cells.

5.
Nat Commun ; 12(1): 6718, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34795250

ABSTRACT

In mammals, imprinted genes are regulated by differentially methylated regions (DMRs) that are inherited from germ cells, leading to monoallelic expression in accordance with parent-of-origin. Yet, it is largely unknown how imprinted DMRs are maintained in human embryos despite global DNA demethylation following fertilization. Here, we explored the mechanisms involved in imprinting regulation by employing human parthenogenetic embryonic stem cells (hpESCs), which lack paternal alleles. We show that although global loss of DNA methylation in hpESCs affects most imprinted DMRs, many paternally-expressed genes (PEGs) remain repressed. To search for factors regulating PEGs, we performed a genome-wide CRISPR/Cas9 screen in haploid hpESCs. This revealed ATF7IP as an essential repressor of a set of PEGs, which we further show is also required for silencing sperm-specific genes. Our study reinforces an important role for histone modifications in regulating imprinted genes and suggests a link between parental imprinting and germ cell identity.


Subject(s)
CRISPR-Cas Systems , Gene Expression Regulation , Genomic Imprinting , Haploidy , Human Embryonic Stem Cells/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cells, Cultured , Chromatin Immunoprecipitation Sequencing/methods , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , Human Embryonic Stem Cells/cytology , Humans , MAP Kinase Signaling System/genetics , Male , Parthenogenesis/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Spermatogenesis/genetics
6.
Genome Biol ; 22(1): 73, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33663567

ABSTRACT

BACKGROUND: Many neurodegenerative diseases develop only later in life, when cells in the nervous system lose their structure or function. In many forms of neurodegenerative diseases, this late-onset phenomenon remains largely unexplained. RESULTS: Analyzing single-cell RNA sequencing from Alzheimer's disease (AD) and Huntington's disease (HD) patients, we find increased transcriptional heterogeneity in disease-state neurons. We hypothesize that transcriptional heterogeneity precedes neurodegenerative disease pathologies. To test this idea experimentally, we use juvenile forms (72Q; 180Q) of HD iPSCs, differentiate them into committed neuronal progenitors, and obtain single-cell expression profiles. We show a global increase in gene expression variability in HD. Autophagy genes become more stable, while energy and actin-related genes become more variable in the mutant cells. Knocking down several differentially variable genes results in increased aggregate formation, a pathology associated with HD. We further validate the increased transcriptional heterogeneity in CHD8+/- cells, a model for autism spectrum disorder. CONCLUSIONS: Overall, our results suggest that although neurodegenerative diseases develop over time, transcriptional regulation imbalance is present already at very early developmental stages. Therefore, an intervention aimed at this early phenotype may be of high diagnostic value.


Subject(s)
Gene Expression Regulation , Genetic Heterogeneity , Genetic Predisposition to Disease , Models, Biological , Neurodegenerative Diseases/etiology , Pluripotent Stem Cells/metabolism , Adult , Gene Expression Profiling , Gene Regulatory Networks , Genetic Background , High-Throughput Nucleotide Sequencing , Humans , Mutation , RNA-Seq , Single-Cell Analysis/methods
7.
J Clin Invest ; 117(3): 683-92, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17290305

ABSTRACT

Aminoglycosides can readthrough premature termination codons (PTCs), permitting translation of full-length proteins. Previously we have found variable efficiency of readthrough in response to the aminoglycoside gentamicin among cystic fibrosis (CF) patients, all carrying the W1282X nonsense mutation. Here we demonstrate that there are patients in whom the level of CF transmembrane conductance regulator (CFTR) nonsense transcripts is markedly reduced, while in others it is significantly higher. Response to gentamicin was found only in patients with the higher level. We further investigated the possibility that the nonsense-mediated mRNA decay (NMD) might vary among cells and hence governs the level of nonsense transcripts available for readthrough. Our results demonstrate differences in NMD efficiency of CFTR transcripts carrying the W1282X mutation among different epithelial cell lines derived from the same tissue. Variability was also found for 5 physiologic NMD substrates, RPL3, SC35 1.6 kb, SC35 1.7 kb, ASNS, and CARS. Importantly, our results demonstrate the existence of cells in which NMD of all transcripts was efficient and others in which the NMD was less efficient. Downregulation of NMD in cells carrying the W1282X mutation increased the level of CFTR nonsense transcripts and enhanced the CFTR chloride channel activity in response to gentamicin. Together our results suggest that the efficiency of NMD might vary and hence have an important role in governing the response to treatments aiming to promote readthrough of PTCs in many genetic diseases.


Subject(s)
Aminoglycosides/therapeutic use , Anti-Bacterial Agents/therapeutic use , Codon, Nonsense/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/drug therapy , Gentamicins/therapeutic use , RNA Stability/genetics , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Drug Resistance/genetics , Humans , Mutation , RNA, Messenger/metabolism , Ribosomal Protein L3 , Transcription, Genetic
8.
Stem Cell Reports ; 15(6): 1275-1286, 2020 12 08.
Article in English | MEDLINE | ID: mdl-32559458

ABSTRACT

Chromatin regulators play fundamental roles in controlling pluripotency and differentiation. We examined the effect of mutations in 703 genes from nearly 70 chromatin-modifying complexes on human embryonic stem cell (ESC) growth. While the vast majority of chromatin-associated complexes are essential for ESC growth, the only complexes that conferred growth advantage upon mutation of their members, were the repressive complexes LSD-CoREST and BHC. Both complexes include the most potent growth-restricting chromatin-related protein, ZMYM2. Interestingly, while ZMYM2 expression is rather low in human blastocysts, its expression peaks in primed ESCs and is again downregulated upon differentiation. ZMYM2-null ESCs overexpress pluripotency genes and show genome-wide promotor-localized histone H3 hyper-acetylation. These mutant cells were also refractory to differentiate in vitro and failed to produce teratomas upon injection into immunodeficient mice. Our results suggest a central role for ZMYM2 in the transcriptional regulation of the undifferentiated state and in the exit-from-pluripotency of human ESCs.


Subject(s)
Cell Proliferation , DNA-Binding Proteins/metabolism , Mutation , Neoplasm Proteins/metabolism , Teratoma/metabolism , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/genetics , Human Embryonic Stem Cells , Humans , Mice , Mice, SCID , Neoplasm Proteins/genetics , Teratoma/genetics , Teratoma/pathology , Transcription Factors/genetics
9.
Stem Cell Reports ; 15(6): 1260-1274, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33296674

ABSTRACT

The multifunctional histone chaperone, SET, is essential for embryonic development in the mouse. Previously, we identified SET as a factor that is rapidly downregulated during embryonic stem cell (ESC) differentiation, suggesting a possible role in the maintenance of pluripotency. Here, we explore SET's function in early differentiation. Using immunoprecipitation coupled with protein quantitation by LC-MS/MS, we uncover factors and complexes, including P53 and ß-catenin, by which SET regulates lineage specification. Knockdown for P53 in SET-knockout (KO) ESCs partially rescues lineage marker misregulation during differentiation. Paradoxically, SET-KO ESCs show increased expression of several Wnt target genes despite reduced levels of active ß-catenin. Further analysis of RNA sequencing datasets hints at a co-regulatory relationship between SET and TCF proteins, terminal effectors of Wnt signaling. Overall, we discover a role for both P53 and ß-catenin in SET-regulated early differentiation and raise a hypothesis for SET function at the ß-catenin-TCF regulatory axis.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Histone Chaperones/metabolism , Mouse Embryonic Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , Histone Chaperones/genetics , Mice , Mouse Embryonic Stem Cells/cytology , Tumor Suppressor Protein p53/genetics , beta Catenin/genetics
10.
Mol Neurobiol ; 57(3): 1778, 2020 03.
Article in English | MEDLINE | ID: mdl-31939145

ABSTRACT

In the original version of the paper, the name of one of the contributing authors, Dr. Mundackal S. Divya (orcid:0000-0002-2869-7191).

11.
Mol Neurobiol ; 57(3): 1768-1777, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31834602

ABSTRACT

Huntington's disease (HD) is a neurodegenerative late-onset genetic disorder caused by CAG expansions in the coding region of the Huntingtin (HTT) gene, resulting in a poly-glutamine (polyQ) expanded HTT protein. Considerable efforts have been devoted for studying HD and other polyQ diseases using animal models and cell culture systems, but no treatment currently exists. Human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) offer an elegant solution for modeling human diseases. However, as embryonic or rejuvenated cells, respectively, these pluripotent stem cells (PSCs) do not recapitulate the late-onset feature of the disease. Here, we applied a robust and rapid differentiation protocol to derive electrophysiologically active striatal GABAergic neurons from human wild-type (WT) and HD ESCs and iPSCs. RNA-seq analyses revealed that HD and WT PSC-derived neurons are highly similar in their gene expression patterns. Interestingly, ectopic expression of Progerin in both WT and HD neurons exacerbated the otherwise non-significant changes in gene expression between these cells, revealing IGF1 and genes involved in neurogenesis and nervous system development as consistently altered in the HD cells. This work provides a useful tool for modeling HD in human PSCs and reveals potential molecular targets altered in HD neurons.


Subject(s)
Huntington Disease/genetics , Induced Pluripotent Stem Cells/metabolism , Lamin Type A/metabolism , Neurons/cytology , Pluripotent Stem Cells/metabolism , Transcription, Genetic , Cell Differentiation/physiology , Embryonic Stem Cells/metabolism , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/metabolism
12.
Nat Commun ; 11(1): 1189, 2020 03 04.
Article in English | MEDLINE | ID: mdl-32132541

ABSTRACT

Changes in potential regulatory elements are thought to be key drivers of phenotypic divergence. However, identifying changes to regulatory elements that underlie human-specific traits has proven very challenging. Here, we use 63 reconstructed and experimentally measured DNA methylation maps of ancient and present-day humans, as well as of six chimpanzees, to detect differentially methylated regions that likely emerged in modern humans after the split from Neanderthals and Denisovans. We show that genes associated with face and vocal tract anatomy went through particularly extensive methylation changes. Specifically, we identify widespread hypermethylation in a network of face- and voice-associated genes (SOX9, ACAN, COL2A1, NFIX and XYLT1). We propose that these repression patterns appeared after the split from Neanderthals and Denisovans, and that they might have played a key role in shaping the modern human face and vocal tract.


Subject(s)
DNA Methylation , DNA, Ancient , Face/anatomy & histology , Phenotype , Phonation/genetics , Adult , Aged , Animals , Cells, Cultured , Child , Chondrocytes , Evolution, Molecular , Female , Gene Regulatory Networks , Genetic Speciation , Humans , Larynx/anatomy & histology , Male , Middle Aged , Neanderthals/genetics , Pan troglodytes/genetics , Primary Cell Culture , Tongue/anatomy & histology , Vocal Cords/anatomy & histology , Vocalization, Animal
13.
J Cell Mol Med ; 13(6): 1059-85, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19210577

ABSTRACT

The nuclear lamina is a proteinaceous structure located underneath the inner nuclear membrane (INM), where it associates with the peripheral chromatin. It contains lamins and lamin-associated proteins, including many integral proteins of the INM, chromatin modifying proteins, transcriptional repressors and structural proteins. A fraction of lamins is also present in the nucleoplasm, where it forms stable complexes and is associated with specific nucleoplasmic proteins. The lamins and their associated proteins are required for most nuclear activities, mitosis and for linking the nucleoplasm to all major cytoskeletal networks in the cytoplasm. Mutations in nuclear lamins and their associated proteins cause about 20 different diseases that are collectively called laminopathies'. This review concentrates mainly on lamins, their structure and their roles in DNA replication, chromatin organization, adult stem cell differentiation, aging, tumorogenesis and the lamin mutations leading to laminopathic diseases.


Subject(s)
Cell Nucleus/metabolism , Lamins/metabolism , Nuclear Envelope/metabolism , Nuclear Lamina/metabolism , Animals , Chromatin/metabolism , Humans , Lamins/genetics , Models, Biological , Mutation , Protein Binding
14.
Lancet ; 372(9640): 719-27, 2008 Aug 30.
Article in English | MEDLINE | ID: mdl-18722008

ABSTRACT

BACKGROUND: In about 10% of patients worldwide and more than 50% of patients in Israel, cystic fibrosis results from nonsense mutations (premature stop codons) in the messenger RNA (mRNA) for the cystic fibrosis transmembrane conductance regulator (CFTR). PTC124 is an orally bioavailable small molecule that is designed to induce ribosomes to selectively read through premature stop codons during mRNA translation, to produce functional CFTR. METHODS: This phase II prospective trial recruited adults with cystic fibrosis who had at least one nonsense mutation in the CFTR gene. Patients were assessed in two 28-day cycles. During the first cycle, patients received PTC124 at 16 mg/kg per day in three doses every day for 14 days, followed by 14 days without treatment; in the second cycle, patients received 40 mg/kg of PTC124 in three doses every day for 14 days, followed by 14 days without treatment. The primary outcome had three components: change in CFTR-mediated total chloride transport; proportion of patients who responded to treatment; and normalisation of chloride transport, as assessed by transepithelial nasal potential difference (PD) at baseline, at the end of each 14-day treatment course, and after 14 days without treatment. The trial was registered with who.int/ictrp, and with clinicaltrials.gov, number NCT00237380. FINDINGS: Transepithelial nasal PD was evaluated in 23 patients in the first cycle and in 21 patients in the second cycle. Mean total chloride transport increased in the first treatment phase, with a change of -7.1 (SD 7.0) mV (p<0.0001), and in the second, with a change of -3.7 (SD 7.3) mV (p=0.032). We recorded a response in total chloride transport (defined as a change in nasal PD of -5 mV or more) in 16 of the 23 patients in the first cycle's treatment phase (p<0.0001) and in eight of the 21 patients in the second cycle (p<0.0001). Total chloride transport entered the normal range for 13 of 23 patients in the first cycle's treatment phase (p=0.0003) and for nine of 21 in the second cycle (p=0.02). Two patients given PTC124 had constipation without intestinal obstruction, and four had mild dysuria. No drug-related serious adverse events were recorded. INTERPRETATION: In patients with cystic fibrosis who have a premature stop codon in the CFTR gene, oral administration of PTC124 to suppress nonsense mutations reduces the epithelial electrophysiological abnormalities caused by CFTR dysfunction.


Subject(s)
Codon, Terminator/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Oxadiazoles/therapeutic use , Adolescent , Adult , Chlorides/metabolism , Codon, Nonsense/drug effects , Codon, Nonsense/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Female , Humans , Male , Middle Aged , Oxadiazoles/adverse effects , Oxadiazoles/pharmacology , Treatment Outcome
15.
Oncogene ; 38(17): 3103-3118, 2019 04.
Article in English | MEDLINE | ID: mdl-30622338

ABSTRACT

The cancer stem cell (CSC) model suggests that a subpopulation of cells within the tumor, the CSCs, is responsible for cancer relapse and metastasis formation. CSCs hold unique characteristics, such as self-renewal, differentiation abilities, and resistance to chemotherapy, raising the need for discovering drugs that target CSCs. Previously we have found that the antihypertensive drug spironolactone impairs DNA damage response in cancer cells. Here we show that spironolactone, apart from inhibiting cancerous cell growth, is also highly toxic to CSCs. Notably, we demonstrate that CSCs have high basal levels of DNA double-strand breaks (DSBs). Mechanistically, we reveal that spironolactone does not damage the DNA but impairs DSB repair and induces apoptosis in cancer cells and CSCs while sparing healthy cells. In vivo, spironolactone treatment reduced the size and CSC content of tumors. Overall, we suggest spironolactone as an anticancer reagent, toxic to both cancer cells and, particularly to, CSCs.


Subject(s)
Antineoplastic Agents/administration & dosage , DNA Repair/drug effects , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Spironolactone/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Drug Repositioning , HeLa Cells , Humans , Mice , Neoplasms/genetics , Spironolactone/pharmacology , Xenograft Model Antitumor Assays
16.
Trends Genet ; 21(9): 480-3, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16039004

ABSTRACT

Disease severity correlates with the level of correctly spliced RNA transcribed from genes carrying splicing mutations and with the ratio of alternatively spliced isoforms. Hence, a role for splicing regulation as a genetic modifier has been suggested. Here we discuss recent experiments that provide direct evidence that changes in the level of splicing factors modulate the splicing pattern of disease-associated genes. Importantly, modulation of the splicing pattern led to regulation of the protein function and modification of disease severity.


Subject(s)
Alternative Splicing , Genetic Diseases, Inborn/genetics , Transcription Factors/metabolism , Animals , Genetic Predisposition to Disease , Humans , Mice , Mutation , Severity of Illness Index , Transcription Factors/genetics
17.
J Cell Biol ; 217(2): 473-481, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29284668

ABSTRACT

Neuronal stimulation leads to immediate early gene (IEG) expression through calcium-dependent mechanisms. In recent years, considerable attention has been devoted to the transcriptional responses after neuronal stimulation, but relatively little is known about the changes in chromatin dynamics that follow neuronal activation. Here, we use fluorescence recovery after photobleaching, biochemical fractionations, and chromatin immunoprecipitation to show that KCl-induced depolarization in primary cultured cortical neurons causes a rapid release of the linker histone H1 from chromatin, concomitant with IEG expression. H1 release is repressed by PARP inhibition, PARP1 deletion, a non-PARylatable H1, as well as phosphorylation inhibitions and a nonphosphorylatable H1, leading to hindered IEG expression. Further, H1 is replaced by PARP1 on IEG promoters after neuronal stimulation, and PARP inhibition blocks this reciprocal binding response. Our results demonstrate the relationship between neuronal excitation and chromatin plasticity by identifying the roles of polyadenosine diphosphate ribosylation and phosphorylation of H1 in regulating H1 chromatin eviction and IEG expression in stimulated neurons.


Subject(s)
Gene Expression Regulation , Histones/metabolism , Neurons/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Animals , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Mice , Mice, Knockout , Neurons/drug effects , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/genetics , Potassium Chloride/pharmacology
18.
Trends Genet ; 18(3): 123-7, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11858835

ABSTRACT

Inherited diseases are associated with profound phenotypic variability, which is affected strongly by genetic modifiers. The splicing machinery could be one such modifying system, through a mechanism involving splicing motifs and their interaction with a complex repertoire of splicing factors. Mutations in splicing motifs and changes in levels of splicing factors can result in different splicing patterns. Changes in the level of normal transcripts or in the relative pattern of different mRNA isoforms affect disease expression, leading to phenotypic variability. Here, we discuss the splicing machinery in terms of its significance in disease severity and its potential role as a genetic modifier.


Subject(s)
Alternative Splicing/genetics , Genetic Variation , RNA Splicing/genetics , Animals , Base Sequence , Cell Nucleus/metabolism , Gene Expression Regulation , Humans , Models, Genetic , Molecular Sequence Data , Transcription, Genetic
19.
Stem Cell Reports ; 9(4): 1291-1303, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28966118

ABSTRACT

Embryonic stem cells (ESCs) are regulated by pluripotency-related transcription factors in concert with chromatin regulators. To identify additional stem cell regulators, we screened a library of endogenously labeled fluorescent fusion proteins in mouse ESCs for fluorescence loss during differentiation. We identified SET, which displayed a rapid isoform shift during early differentiation from the predominant isoform in ESCs, SETα, to the primary isoform in differentiated cells, SETß, through alternative promoters. SETα is selectively bound and regulated by pluripotency factors. SET depletion causes proliferation slowdown and perturbed neuronal differentiation in vitro and developmental arrest in vivo, and photobleaching methods demonstrate SET's role in maintaining a dynamic chromatin state in ESCs. This work identifies an important regulator of pluripotency and early differentiation, which is controlled by alternative promoter usage.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Histone Acetyltransferases/genetics , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Adaptor Proteins, Signal Transducing , Animals , Cell Proliferation , Cell Survival/genetics , Chromatin Assembly and Disassembly , Histones/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Neural Plate/cytology , Octamer Transcription Factor-3/metabolism , Protein Isoforms
20.
Prog Mol Subcell Biol ; 44: 233-54, 2006.
Article in English | MEDLINE | ID: mdl-17076271

ABSTRACT

A significant fraction of CF-causing mutations affects pre-mRNA splicing. These mutations can generate both aberrant and correct transcripts, the level of which varies among different patients. An inverse correlation was found between this level and disease severity, suggesting a role for splicing regulation as a genetic modifier. Subsequent studies showed that overexpression of splicing factors modulated the level of correctly spliced RNA, transcribed from minigenes carrying CF-causing splicing mutations. Overexpression of splicing factors also modulated the level of normal CFTR transcripts, transcribed from the endogenous CFTR allele carrying splicing mutations, in CF-derived epithelial cells. Several of the factors promoted higher level of correct CFTR transcripts. The increased level of normal transcripts led to activation of the CFTR channel and restoration of its function. Restoration was also obtained by sodium butyrate, a histone deacetylase inhibitor, known to up-regulate the expression of splicing factors. These results highlight the role of the splicing machinery as a modifier of disease severity in patients carrying splicing mutations and shed a new light on the therapeutic potential of splicing modulation for genetic diseases caused by splicing mutations.


Subject(s)
Alternative Splicing/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Animals , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Humans , Mutation/genetics , Oligonucleotides, Antisense/metabolism , RNA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL