Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Nucleic Acids Res ; 49(7): 3907-3918, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33751106

ABSTRACT

Somatic expansion of the CAG repeat tract that causes Huntington's disease (HD) is thought to contribute to the rate of disease pathogenesis. Therefore, factors influencing repeat expansion are potential therapeutic targets. Genes in the DNA mismatch repair pathway are critical drivers of somatic expansion in HD mouse models. Here, we have tested, using genetic and pharmacological approaches, the role of the endonuclease domain of the mismatch repair protein MLH3 in somatic CAG expansion in HD mice and patient cells. A point mutation in the MLH3 endonuclease domain completely eliminated CAG expansion in the brain and peripheral tissues of a HD knock-in mouse model (HttQ111). To test whether the MLH3 endonuclease could be manipulated pharmacologically, we delivered splice switching oligonucleotides in mice to redirect Mlh3 splicing to exclude the endonuclease domain. Splice redirection to an isoform lacking the endonuclease domain was associated with reduced CAG expansion. Finally, CAG expansion in HD patient-derived primary fibroblasts was also significantly reduced by redirecting MLH3 splicing to the endogenous endonuclease domain-lacking isoform. These data indicate the potential of targeting the MLH3 endonuclease domain to slow somatic CAG repeat expansion in HD, a therapeutic strategy that may be applicable across multiple repeat expansion disorders.


Subject(s)
DNA Repair , Endonucleases , Huntington Disease/genetics , MutL Proteins , Protein Splicing , Trinucleotide Repeat Expansion , Animals , Cells, Cultured , Endonucleases/physiology , Female , Fibroblasts , Gene Knock-In Techniques , Genomic Instability , Humans , Male , Mice , Mice, Inbred C57BL , MutL Proteins/physiology , Oligonucleotides
2.
Nucleic Acids Res ; 46(8): 4022-4032, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29529236

ABSTRACT

DNA repeat expansion underlies dozens of progressive neurodegenerative disorders. While the mechanisms driving repeat expansion are not fully understood, increasing evidence suggests a central role for DNA mismatch repair. The mismatch repair recognition complex MutSß (MSH2-MSH3) that binds mismatched bases and/or insertion/deletion loops has previously been implicated in GAA•TTC, CAG•CTG and CGG•CCG repeat expansion, suggesting a shared mechanism. MutSß has been studied in a number of models, but the contribution of subsequent steps mediated by the MutL endonuclease in this pathway is less clear. Here we show that MutLγ (MLH1-MLH3) is the MutL complex responsible for GAA•TTC repeat expansion. Lentiviral expression of shRNA targeting MutL nuclease components MLH1, PMS2, and MLH3 revealed that reduced expression of MLH1 or MLH3 reduced the repeat expansion rate in a human Friedreich ataxia cell model, while targeting PMS2 did not. Using splice-switching oligonucleotides we show that MLH3 isoform 1 is active in GAA•TTC repeat expansion while the nuclease-deficient MLH3 isoform 2 is not. MLH3 isoform switching slowed repeat expansion in both model cells and FRDA patient fibroblasts. Our work indicates a specific and active role for MutLγ in the expansion process and reveals plausible targets for disease-modifying therapies.


Subject(s)
DNA Repeat Expansion , MutL Protein Homolog 1/metabolism , MutL Proteins/metabolism , Cells, Cultured , Endonucleases/chemistry , Exons , Friedreich Ataxia/genetics , Gene Knockdown Techniques , HEK293 Cells , Humans , MutL Proteins/chemistry , MutL Proteins/genetics , Protein Domains , Protein Isoforms/metabolism
3.
J Biol Chem ; 287(35): 29958-67, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22787155

ABSTRACT

While DNA repair has been implicated in CAG·CTG repeat expansion, its role in the GAA·TTC expansion of Friedreich ataxia (FRDA) is less clear. We have developed a human cellular model that recapitulates the DNA repeat expansion found in FRDA patient tissues. In this model, GAA·TTC repeats expand incrementally and continuously. We have previously shown that the expansion rate is linked to transcription within the repeats. Our working hypothesis is that structures formed within the GAA·TTC repeat during transcription attract DNA repair enzymes that then facilitate the expansion process. MutSß, a heterodimer of MSH2 and MSH3, is known to have a role in CAG·CTG repeat expansion. We now show that shRNA knockdown of either MSH2 or MSH3 slowed GAA·TTC expansion in our system. We further characterized the role of MutSß in GAA·TTC expansion using a functional assay in primary FRDA patient-derived fibroblasts. These fibroblasts have no known propensity for instability in their native state. Ectopic expression of MSH2 and MSH3 induced GAA·TTC repeat expansion in the native FXN gene. MSH2 is central to mismatch repair and its absence or reduction causes a predisposition to cancer. Thus, despite its essential role in GAA·TTC expansion, MSH2 is not an attractive therapeutic target. The absence or reduction of MSH3 is not strongly associated with cancer predisposition. Accordingly, MSH3 has been suggested as a therapeutic target for CAG·CTG repeat expansion disorders. Our results suggest that MSH3 may also serve as a therapeutic target to slow the expansion of GAA·TTC repeats in the future.


Subject(s)
DNA Mismatch Repair , DNA-Binding Proteins/metabolism , Multienzyme Complexes/metabolism , MutS DNA Mismatch-Binding Protein/metabolism , MutS Homolog 2 Protein/metabolism , Trinucleotide Repeat Expansion , DNA-Binding Proteins/genetics , Fibroblasts/enzymology , Fibroblasts/pathology , Friedreich Ataxia/enzymology , Friedreich Ataxia/genetics , Friedreich Ataxia/pathology , Genetic Predisposition to Disease , HEK293 Cells , Humans , Models, Biological , Multienzyme Complexes/genetics , MutS DNA Mismatch-Binding Protein/genetics , MutS Homolog 2 Protein/genetics , MutS Homolog 3 Protein , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology
4.
PLoS One ; 5(6): e11121, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20559546

ABSTRACT

BACKGROUND: Expansion of an unstable GAA*TTC repeat in the first intron of the FXN gene causes Friedreich ataxia by reducing frataxin expression. Structure formation by the repeat has been implicated in both frataxin repression and GAA*TTC instability. The GAA*TTC sequence is capable of adopting multiple non-B DNA structures including Y*R*Y and R*R*Y triplexes. Lower pH promotes the formation of Y*R*Y triplexes by GAA*TTC. Here we used the bacterial transposon Tn7 as an in vitro tool to probe whether GAA*TTC repeats can attract a well-characterized recombinase. METHODOLOGY/PRINCIPAL FINDINGS: Tn7 showed a pH-dependent preference for insertion into uninterrupted regions of a Friedreich ataxia patient-derived repeat, inserting 48, 39 and 14 percent of the time at pH 7, pH 8 and pH 9, respectively. Moreover, Tn7 also showed orientation and region specific insertion within the repeat at pH 7 and pH 8, but not at pH 9. In contrast, transposon Tn5 showed no strong preference for or against the repeat during in vitro transposition at any pH tested. Y*R*Y triplex formation was reduced in predictable ways by transposon interruption of the GAA*TTC repeat. However, transposon interruptions in the GAA*TTC repeats did not increase the in vitro transcription efficiency of the templates. CONCLUSIONS/SIGNIFICANCE: We have demonstrated that transposon Tn7 will recognize structures that form spontaneously in GAA*TTC repeats and insert in a specific orientation within the repeat. The conditions used for in vitro transposition span the physiologically relevant range suggesting that long GAA*TTC repeats can form triplex structures in vivo, attracting enzymes involved in DNA repair, recombination and chromatin modification.


Subject(s)
DNA Transposable Elements , DNA/genetics , Repetitive Sequences, Nucleic Acid , Base Sequence , Blotting, Northern , Cloning, Molecular , Electrophoretic Mobility Shift Assay , Hydrogen-Ion Concentration , Molecular Sequence Data , Polymerase Chain Reaction
5.
PLoS Genet ; 5(10): e1000704, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19876374

ABSTRACT

Trinucleotide repeat expansion is the genetic basis for a sizeable group of inherited neurological and neuromuscular disorders. Friedreich ataxia (FRDA) is a relentlessly progressive neurodegenerative disorder caused by GAA.TTC repeat expansion in the first intron of the FXN gene. The expanded repeat reduces FXN mRNA expression and the length of the repeat tract is proportional to disease severity. Somatic expansion of the GAA.TTC repeat sequence in disease-relevant tissues is thought to contribute to the progression of disease severity during patient aging. Previous models of GAA.TTC instability have not been able to produce substantial levels of expansion within an experimentally useful time frame, which has limited our understanding of the molecular basis for this expansion. Here, we present a novel model for studying GAA.TTC expansion in human cells. In our model system, uninterrupted GAA.TTC repeat sequences display high levels of genomic instability, with an overall tendency towards progressive expansion. Using this model, we characterize the relationship between repeat length and expansion. We identify the interval between 88 and 176 repeats as being an important length threshold where expansion rates dramatically increase. We show that expansion levels are affected by both the purity and orientation of the repeat tract within the genomic context. We further demonstrate that GAA.TTC expansion in our model is independent of cell division. Using unique reporter constructs, we identify transcription through the repeat tract as a major contributor to GAA.TTC expansion. Our findings provide novel insight into the mechanisms responsible for GAA.TTC expansion in human cells.


Subject(s)
Genomic Instability , Trinucleotide Repeat Expansion , Cell Line , Gene Expression Regulation , Humans , Models, Genetic , Transcription, Genetic
6.
Anal Biochem ; 395(2): 237-43, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19733147

ABSTRACT

Splicing and nuclear export are vital components of eukaryotic gene expression. Defects in splicing due to cis mutations are known to cause a number of human diseases. Here we present a dual reporter system that can be used to look at splicing or export deficiencies resulting from an insufficiency in components of the cotranscriptional machinery. The constructs use a bidirectional promoter to coexpress a test reporter and a control reporter. In the splicing construct, maximal expression of the test reporter is dependent on efficient splicing and splicing-related nuclear export, whereas the control reporter is an intronless complementary DNA expression cassette. The dual reporters allow a robust ratiometric output that is independent of cell number or transfection efficiency. Therefore, our construct is internally controlled and amenable to high-throughput analysis. As a counterscreen, we have a nonsplicing control construct in which neither reporter bears an intron. We demonstrate the sensitivity of our construct to defects in nuclear export by depleting UAP56 and NXF1, essential components of the cotranscriptional machinery.


Subject(s)
RNA Splicing , RNA, Messenger/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Base Sequence , Blotting, Northern , Cell Line , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Genes, Reporter , High-Throughput Screening Assays , Humans , Molecular Sequence Data , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , RNA Interference , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
7.
PLoS One ; 4(7): e6193, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19587781

ABSTRACT

BACKGROUND: Making the correct choice between transcription elongation and transcription termination is essential to the function of RNA polymerase II, and fundamental to gene expression. This choice can be influenced by factors modifying the transcription complex, factors modifying chromatin, or signals mediated by the template or transcript. To aid in the study of transcription elongation and termination we have developed a transcription elongation reporter system that consists of tandem luciferase reporters flanking a test sequence of interest. The ratio of expression from the reporters provides a measure of the relative rates of successful elongation through the intervening sequence. METHODOLOGY/PRINCIPAL FINDINGS: Size matched fragments containing the polyadenylation signal of the human beta-actin gene (ACTB) and the human beta-globin gene (HBB) were evaluated for transcription termination using this new ratiometric tandem reporter assay. Constructs bearing just 200 base pairs on either side of the consensus poly(A) addition site terminated 98% and 86% of transcription for ACTB and HBB sequences, respectively. The nearly 10-fold difference in read-through transcription between the two short poly(A) regions was eclipsed when additional downstream poly(A) sequence was included for each gene. Both poly(A) regions proved very effective at termination when 1100 base pairs were included, stopping 99.6% of transcription. To determine if part of the increased termination was simply due to the increased template length, we inserted several kilobases of heterologous coding sequence downstream of each poly(A) region test fragment. Unexpectedly, the additional length reduced the effectiveness of termination of HBB sequences 2-fold and of ACTB sequences 3- to 5-fold. CONCLUSIONS/SIGNIFICANCE: The tandem construct provides a sensitive measure of transcription termination in human cells. Decreased Xrn2 or Senataxin levels produced only a modest release from termination. Our data support overlap in allosteric and torpedo mechanisms of transcription termination and suggest that efficient termination is ensured by redundancy.


Subject(s)
Genes, Reporter , RNA Polymerase II/metabolism , Terminator Regions, Genetic , Transcription, Genetic , Actins/genetics , Humans , beta-Globins/genetics
8.
J Biol Chem ; 283(8): 4578-87, 2008 Feb 22.
Article in English | MEDLINE | ID: mdl-18160403

ABSTRACT

Ferritin plays an important role in the storage and release of iron, an element utilized in cellular processes such as respiration, gene regulation, and DNA replication and repair. Ferritin in animals is composed of 24 ferritin L (FTL) and ferritin H (FTH) subunits in ratios that vary in different cell types. Because the subunits are not functionally interchangeable, both L and H units are critical for maintaining iron homeostasis and protecting against iron overload. FTL and FTH are regulated primarily at a post-transcriptional level in response to cellular iron concentrations. Individual regulation of FTL and FTH is of much interest, and although transcriptional differences between FTL and FTH have been shown, differences in their post-transcriptional regulation have not been evaluated. We report here that FTL and FTH are differentially regulated in 1% oxygen on a post-transcriptional level. We have designed a quantitative assay system sensitive enough to detect differences between FTL and FTH iron regulatory elements (IREs) that a standard electrophoretic mobility shift assay does not. The FTL IRE is the primary responder in the presence of an iron donor in hypoxic conditions, and this response is reflected in endogenous FTL protein levels. These results provide evidence that FTL and FTH subunits respond independently to cellular iron concentrations and underscore the importance of evaluating FTL and FTH IREs separately.


Subject(s)
Apoferritins/biosynthesis , Ferritins/biosynthesis , Gene Expression Regulation/physiology , Homeostasis/physiology , Iron/metabolism , Oxygen/metabolism , Apoferritins/genetics , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Line , DNA Replication/drug effects , DNA Replication/physiology , Ferritins/genetics , Gene Expression Regulation/drug effects , Homeostasis/drug effects , Humans , Iron/pharmacology , Iron Overload/genetics , Iron Overload/metabolism , Oxygen/pharmacology , Oxygen Consumption/drug effects , Oxygen Consumption/physiology , Response Elements/physiology
9.
Nucleic Acids Res ; 35(16): 5351-9, 2007.
Article in English | MEDLINE | ID: mdl-17693431

ABSTRACT

Expansion of an unstable GAA.TTC repeat in the first intron of the FXN gene causes Friedreich ataxia by reducing frataxin expression. Deficiency of frataxin, an essential mitochondrial protein, leads to progressive neurodegeneration and cardiomyopathy. The degree of frataxin reduction correlates with GAA.TTC tract length, but the mechanism of reduction remains controversial. Here we show that transcription causes extensive RNA.DNA hybrid formation on GAA.TTC templates in bacteria as well as in defined transcription reactions using T7 RNA polymerase in vitro. RNA.DNA hybrids can also form to a lesser extent on smaller, so-called 'pre-mutation' size GAA.TTC repeats, that do not cause disease, but are prone to expansion. During in vitro transcription of longer repeats, T7 RNA polymerase arrests in the promoter distal end of the GAA.TTC tract and an extensive RNA.DNA hybrid is tightly linked to this arrest. RNA.DNA hybrid formation appears to be an intrinsic property of transcription through long GAA.TTC tracts. RNA.DNA hybrids have a potential role in GAA.TTC tract instability and in the mechanism underlying reduced frataxin mRNA levels in Friedreich Ataxia.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , DNA/biosynthesis , Iron-Binding Proteins/genetics , RNA/biosynthesis , Transcription, Genetic , Trinucleotide Repeats , Viral Proteins/metabolism , Bacteria/genetics , Templates, Genetic , Frataxin
10.
Gene ; 395(1-2): 125-34, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17442505

ABSTRACT

Carriers of FMR1 alleles with 55-200 repeats in the 5' UTR are at risk for Fragile X associated tremor and ataxia syndrome. The cause of the neuropathology is unknown but is thought to be RNA-mediated. Maternally transmitted premutation alleles are also at risk of expansion of the repeat tract into the "full mutation" range (>200 repeats). The mechanism responsible for expansion is unknown. Full mutation alleles produce reduced amounts of the FMR1 gene product, FMRP, which leads to Fragile X mental retardation syndrome. We have developed a murine model for Fragile X premutation carriers that recapitulates key features seen in humans including a direct relationship between repeat number and Fmr1 mRNA levels, an inverse relationship with FMRP levels and Purkinje cell dropout that have not been seen in a previously described knock-in mouse model. In addition, these mice also show a differential deficit of FMRP in different parts of the brain that might account for symptoms of the full mutation that are seen in premutation carriers. As in humans, repeat instability is high with expansions predominating and, for the first time in a mouse model, large expansions into the full mutation range are seen that occur within a single generation. Thus, contrary to what was previously thought, mice may be good models not only for the symptoms seen in human carriers of FMR1 premutation alleles but also for understanding the mechanism responsible for repeat expansion, a phenomenon that is responsible for a number of neurological and neurodevelopmental disorders.


Subject(s)
DNA Repeat Expansion , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , 5' Untranslated Regions , Alleles , Animals , Base Sequence , Brain/metabolism , DNA Methylation , Disease Models, Animal , Female , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Fragile X Syndrome/physiopathology , Gene Targeting , Genomic Instability , Humans , Male , Mice , Mice, Mutant Strains , Mutation , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
Anal Biochem ; 346(2): 210-6, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16212928

ABSTRACT

Coordinated control of protein expression is highly desirable for functional genomics. Here we show a widely applicable approach to construction and use of custom bidirectional promoters capable of reproducible coexpression of two proteins. The use of a bidirectional promoter system overcomes many of the limitations of current coexpression systems such as unpredictable upstream and downstream expression ratios mediated by an internal ribosome entry site. We present examples of tetracycline-inducible, bidirectional promoter systems that produce simultaneous and rapid coinduction of two separate reporters to predictable levels and ratios. Steric blocking of transcription initiation by simple tetracycline repressors, rather than the use of transcriptional activator/repressor domain fusions, makes the system described here superior for investigating downstream transcriptional consequences of protein expression.


Subject(s)
Gene Expression Regulation/drug effects , Gene Order/genetics , Plasmids/genetics , Promoter Regions, Genetic , Tetracyclines/pharmacology , Cell Line , Genes, Reporter , Genetic Vectors , Humans , Repressor Proteins/genetics , Transfection
12.
Eur J Neurosci ; 2(10): 822-827, 1990.
Article in English | MEDLINE | ID: mdl-12106089

ABSTRACT

GAP-43 is a gene expressed only in the nervous system. The protein product is believed to be important to neuronal growth and plasticity. Most, and likely all, neurons express high levels of GAP-43 during periods of neurite elongation. To initiate studies of GAP-43 gene regulation we have cloned the rat gene encoding GAP-43. The GAP-43 gene includes three exons. The first exon encodes only the amino terminal 10 amino acids, which corresponds to the membrane targeting domain of GAP-43. The second exon encodes a putative calmodulin binding domain and a protein kinase C phosphorylation site. The 5'-flanking sequence is unusual in that it lacks CAAT or TATA elements, and directs RNA transcription initiation from several sites. Some of the transcription start sites are used to a different degree in the central and peripheral nervous systems.

SELECTION OF CITATIONS
SEARCH DETAIL