Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Med ; 25(7): 1131-1142, 2019 07.
Article in English | MEDLINE | ID: mdl-31263285

ABSTRACT

Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder caused by a CAG trinucleotide expansion in the huntingtin gene (HTT), which codes for the pathologic mutant HTT (mHTT) protein. Since normal HTT is thought to be important for brain function, we engineered zinc finger protein transcription factors (ZFP-TFs) to target the pathogenic CAG repeat and selectively lower mHTT as a therapeutic strategy. Using patient-derived fibroblasts and neurons, we demonstrate that ZFP-TFs selectively repress >99% of HD-causing alleles over a wide dose range while preserving expression of >86% of normal alleles. Other CAG-containing genes are minimally affected, and virally delivered ZFP-TFs are active and well tolerated in HD neurons beyond 100 days in culture and for at least nine months in the mouse brain. Using three HD mouse models, we demonstrate improvements in a range of molecular, histopathological, electrophysiological and functional endpoints. Our findings support the continued development of an allele-selective ZFP-TF for the treatment of HD.


Subject(s)
Alleles , Huntingtin Protein/genetics , Huntington Disease/therapy , Mutation , Transcription, Genetic , Zinc Fingers , Animals , Cells, Cultured , Disease Models, Animal , Female , Humans , Huntington Disease/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Neuroprotection , Trinucleotide Repeats
2.
Cell Syst ; 8(2): 122-135.e7, 2019 02 27.
Article in English | MEDLINE | ID: mdl-30772379

ABSTRACT

Transcriptional regulatory changes in the developing and adult brain are prominent features of brain diseases, but the involvement of specific transcription factors (TFs) remains poorly understood. We integrated brain-specific DNase footprinting and TF-gene co-expression to reconstruct a transcriptional regulatory network (TRN) model for the human brain. We identified key regulator TFs whose predicted target genes were enriched for differentially expressed genes in the prefrontal cortex of individuals with psychiatric and neurodegenerative diseases. Many of these TFs were further implicated in the same diseases through disruption of their binding sites by disease-associated SNPs and associations of TF loci with disease risk. Using primary human neural stem cells, we validated network predictions that link the TF POU3F2 to schizophrenia and bipolar disorder via both cis- and trans-acting mechanisms. Our models of brain-specific TF binding sites and target genes provide a resource for network analysis of brain diseases.


Subject(s)
Gene Regulatory Networks/genetics , Genomics/methods , Neurodegenerative Diseases/genetics , Psychiatry/methods , Transcription Factors/genetics , Female , Humans , Male
3.
Mol Syst Biol ; 14(3): e7435, 2018 03 26.
Article in English | MEDLINE | ID: mdl-29581148

ABSTRACT

Transcriptional changes occur presymptomatically and throughout Huntington's disease (HD), motivating the study of transcriptional regulatory networks (TRNs) in HD We reconstructed a genome-scale model for the target genes of 718 transcription factors (TFs) in the mouse striatum by integrating a model of genomic binding sites with transcriptome profiling of striatal tissue from HD mouse models. We identified 48 differentially expressed TF-target gene modules associated with age- and CAG repeat length-dependent gene expression changes in Htt CAG knock-in mouse striatum and replicated many of these associations in independent transcriptomic and proteomic datasets. Thirteen of 48 of these predicted TF-target gene modules were also differentially expressed in striatal tissue from human disease. We experimentally validated a specific model prediction that SMAD3 regulates HD-related gene expression changes using chromatin immunoprecipitation and deep sequencing (ChIP-seq) of mouse striatum. We found CAG repeat length-dependent changes in the genomic occupancy of SMAD3 and confirmed our model's prediction that many SMAD3 target genes are downregulated early in HD.


Subject(s)
Gene Expression Profiling/methods , Gene Regulatory Networks , Huntington Disease/genetics , Smad3 Protein/genetics , Animals , Corpus Striatum/metabolism , Disease Models, Animal , Gene Expression Regulation , Humans , Huntington Disease/metabolism , Mice , Protein Interaction Maps , Proteomics , Smad3 Protein/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
4.
J Huntingtons Dis ; 6(3): 237-247, 2017.
Article in English | MEDLINE | ID: mdl-28968243

ABSTRACT

BACKGROUND: Huntington's disease (HD) is a fatal progressive neurodegenerative disease characterized by chorea, cognitive impairment and psychiatric symptoms. Retinal examination of HD patients as well as in HD animal models have shown evidence of retinal dysfunction. However, a detailed retinal study employing clinically available measurement tools has not been reported to date in HD. OBJECTIVE: The goal of this study was to assess retinal responses measured by electroretinogram (ERG) between HD patients and controls and evaluate any correlation between ERG measurements and stage of disease. METHODS: Eighteen patients and 10 controls with inclusion criteria of ages 18-70 years (average age HD subjects: 52.1 yrs and control subjects: 51.9 yrs) were recruited for the study. Subjects with previous history of retinal or ophthalmologic disease were excluded. Retinal function was examined by full-field ERG in both eyes of each subject. Amplitudes and latencies to increasing flash intensities in both light- and dark-adaptation were measured in all subjects. Statistical analyses employed generalized estimating equations, which account for repeated measures per subject. RESULTS: We analyzed the b-wave amplitudes of ERG response in all flash intensities and with 30 Hz flicker stimulation. We found statistically significant increased amplitudes in HD patients compared to controls at light-adapted (photopic) 24.2 and 60.9 cd.sec/m2 intensities, dark-adapted (scotopic, red flash) 0.22 cd.sec/m2 intensity, and a trend toward significance at light-adapted 30 Hz flicker. Furthermore, we found a significant increase in light-adapted ERG response from female compared to male HD patients, but no significant difference between gender amongst controls. We also noted a positive association between number of CAG repeats and ERG response at the smallest light adapted intensity (3.1 cd.sec/m2). CONCLUSIONS: ERG studies revealed significantly altered retinal responses at multiple flash intensities in subjects with an HD expansion allele compared to controls. Significant differences were observed with either light-adapted tests or the dark-adapted red flash which suggests that the enhanced responses in HD patients is specific to the cone photoreceptor pathway.


Subject(s)
Huntington Disease/pathology , Retina/physiopathology , Adaptation, Ocular/physiology , Adolescent , Adult , Aged , Dark Adaptation/physiology , Electroretinography/methods , Female , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Male , Middle Aged , Photic Stimulation , Trinucleotide Repeats/genetics , Young Adult
5.
Hum Mol Genet ; 26(5): 913-922, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28334820

ABSTRACT

Huntington's disease is a dominantly inherited neurodegenerative disease caused by the expansion of a CAG repeat in the HTT gene. In addition to the length of the CAG expansion, factors such as genetic background have been shown to contribute to the age at onset of neurological symptoms. A central challenge in understanding the disease progression that leads from the HD mutation to massive cell death in the striatum is the ability to characterize the subtle and early functional consequences of the CAG expansion longitudinally. We used dense time course sampling between 4 and 20 postnatal weeks to characterize early transcriptomic, molecular and cellular phenotypes in the striatum of six distinct knock-in mouse models of the HD mutation. We studied the effects of the HttQ111 allele on the C57BL/6J, CD-1, FVB/NCr1, and 129S2/SvPasCrl genetic backgrounds, and of two additional alleles, HttQ92 and HttQ50, on the C57BL/6J background. We describe the emergence of a transcriptomic signature in HttQ111/+ mice involving hundreds of differentially expressed genes and changes in diverse molecular pathways. We also show that this time course spanned the onset of mutant huntingtin nuclear localization phenotypes and somatic CAG-length instability in the striatum. Genetic background strongly influenced the magnitude and age at onset of these effects. This work provides a foundation for understanding the earliest transcriptional and molecular changes contributing to HD pathogenesis.


Subject(s)
Corpus Striatum/metabolism , Huntingtin Protein/genetics , Huntington Disease/genetics , Trinucleotide Repeat Expansion/genetics , Animals , Corpus Striatum/pathology , Disease Models, Animal , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Genetic Background , Genomic Instability/genetics , Humans , Huntingtin Protein/biosynthesis , Huntington Disease/pathology , Mice , Mutation/genetics , Neurons/metabolism , Neurons/pathology , Phenotype , Transcriptome/genetics
6.
Nat Biotechnol ; 34(4): 424-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26950749

ABSTRACT

Gene therapy with genetically modified human CD34(+) hematopoietic stem and progenitor cells (HSPCs) may be safer using targeted integration (TI) of transgenes into a genomic 'safe harbor' site rather than random viral integration. We demonstrate that temporally optimized delivery of zinc finger nuclease mRNA via electroporation and adeno-associated virus (AAV) 6 delivery of donor constructs in human HSPCs approaches clinically relevant levels of TI into the AAVS1 safe harbor locus. Up to 58% Venus(+) HSPCs with 6-16% human cell marking were observed following engraftment into mice. In HSPCs from patients with X-linked chronic granulomatous disease (X-CGD), caused by mutations in the gp91phox subunit of the NADPH oxidase, TI of a gp91phox transgene into AAVS1 resulted in ∼15% gp91phox expression and increased NADPH oxidase activity in ex vivo-derived neutrophils. In mice transplanted with corrected HSPCs, 4-11% of human cells in the bone marrow expressed gp91phox. This method for TI into AAVS1 may be broadly applicable to correction of other monogenic diseases.


Subject(s)
Antigens, CD34/chemistry , Genetic Therapy/methods , Granulomatous Disease, Chronic/therapy , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Animals , Cells, Cultured , Humans , Mice , Mice, Transgenic
7.
Nature ; 500(7462): 296-300, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23863942

ABSTRACT

Down's syndrome is a common disorder with enormous medical and social costs, caused by trisomy for chromosome 21. We tested the concept that gene imbalance across an extra chromosome can be de facto corrected by manipulating a single gene, XIST (the X-inactivation gene). Using genome editing with zinc finger nucleases, we inserted a large, inducible XIST transgene into the DYRK1A locus on chromosome 21, in Down's syndrome pluripotent stem cells. The XIST non-coding RNA coats chromosome 21 and triggers stable heterochromatin modifications, chromosome-wide transcriptional silencing and DNA methylation to form a 'chromosome 21 Barr body'. This provides a model to study human chromosome inactivation and creates a system to investigate genomic expression changes and cellular pathologies of trisomy 21, free from genetic and epigenetic noise. Notably, deficits in proliferation and neural rosette formation are rapidly reversed upon silencing one chromosome 21. Successful trisomy silencing in vitro also surmounts the major first step towards potential development of 'chromosome therapy'.


Subject(s)
Chromosomes, Human, Pair 21/genetics , Dosage Compensation, Genetic , Down Syndrome/genetics , RNA, Long Noncoding/metabolism , Animals , Cell Line , Cell Proliferation , DNA Methylation , Down Syndrome/therapy , Gene Silencing , Humans , Induced Pluripotent Stem Cells , Male , Mice , Mutagenesis, Insertional , Neurogenesis , RNA, Long Noncoding/genetics , Sex Chromatin/genetics , X Chromosome Inactivation/genetics
8.
Plant Biotechnol J ; 11(6): 671-80, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23521778

ABSTRACT

Targeted gene regulation via designed transcription factors has great potential for precise phenotypic modification and acceleration of novel crop trait development. To this end, designed transcriptional activators have been constructed by fusing transcriptional activation domains to DNA-binding proteins. In this study, a transcriptional activator from the herpes simplex virus, VP16, was used to identify plant regulatory proteins. Transcriptional activation domains were identified from each protein and fused with zinc finger DNA-binding proteins (ZFPs) to generate designed transcriptional activators. In addition, specific sequences within each transcriptional activation domain were modified to mimic the VP16 contact motif that interacts directly with RNA polymerase II core transcription factors. To evaluate these designed transcriptional activators, test systems were built in yeast and tobacco comprising reporter genes driven by promoters containing ZFP-binding sites upstream of the transcriptional start site. In yeast, transcriptional domains from the plant proteins ERF2 and PTI4 activated MEL1 reporter gene expression to levels similar to VP16 and the modified sequences displayed even greater levels of activation. Following stable transformation of the tobacco reporter system with transcriptional activators derived from ERF2, GUS reporter gene transcript accumulation was equal to or greater than those derived from VP16. Moreover, a modified ERF2 domain displayed significantly enhanced transcriptional activation compared with VP16 and with the unmodified ERF2 sequence. These results demonstrate that plant sequences capable of facilitating transcriptional activation can be found and, when fused to DNA-binding proteins, can enhance gene expression.


Subject(s)
Gene Expression Regulation, Plant , Nicotiana/genetics , Protein Engineering , Transcription Factors/metabolism , Transcriptional Activation/genetics , Amino Acid Motifs , Amino Acid Sequence , Chromosomes, Plant/genetics , Evolution, Molecular , Genes, Reporter , Herpes Simplex Virus Protein Vmw65/metabolism , Molecular Sequence Data , Plant Proteins/chemistry , Protein Structure, Tertiary , Proteome/metabolism , Saccharomyces cerevisiae/metabolism , Sequence Alignment , Transcription, Genetic
9.
Nat Methods ; 9(10): 993-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22922501

ABSTRACT

Malaria afflicts over 200 million people worldwide, and its most lethal etiologic agent, Plasmodium falciparum, is evolving to resist even the latest-generation therapeutics. Efficient tools for genome-directed investigations of P. falciparum-induced pathogenesis, including drug-resistance mechanisms, are clearly required. Here we report rapid and targeted genetic engineering of this parasite using zinc-finger nucleases (ZFNs) that produce a double-strand break in a user-defined locus and trigger homology-directed repair. Targeting an integrated egfp locus, we obtained gene-deletion parasites with unprecedented speed (2 weeks), both with and without direct selection. ZFNs engineered against the parasite gene pfcrt, responsible for escape under chloroquine treatment, rapidly produced parasites that carried either an allelic replacement or a panel of specified point mutations. This method will enable a diverse array of genome-editing approaches to interrogate this human pathogen.


Subject(s)
Endonucleases/physiology , Genome, Protozoan , Plasmodium falciparum/genetics , Protein Engineering/methods , Zinc Fingers/physiology , Alleles , Base Sequence , Chloroquine/pharmacology , Drug Resistance/genetics , Endonucleases/genetics , Molecular Sequence Data , Plasmodium falciparum/drug effects , Zinc Fingers/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...