Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
bioRxiv ; 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38659854

ABSTRACT

The human genome contains millions of retrotransposons, several of which could become active due to somatic mutations having phenotypic consequences, including disease. However, it is not thoroughly understood how nucleotide changes in retrotransposons affect their jumping activity. Here, we developed a novel massively parallel jumping assay (MPJA) that can test the jumping potential of thousands of transposons en masse. We generated nucleotide variant library of selected four Alu retrotransposons containing 165,087 different haplotypes and tested them for their jumping ability using MPJA. We found 66,821 unique jumping haplotypes, allowing us to pinpoint domains and variants vital for transposition. Mapping these variants to the Alu-RNA secondary structure revealed stem-loop features that contribute to jumping potential. Combined, our work provides a novel high-throughput assay that assesses the ability of retrotransposons to jump and identifies nucleotide changes that have the potential to reactivate them in the human genome.

3.
Cells ; 10(12)2021 12 05.
Article in English | MEDLINE | ID: mdl-34943930

ABSTRACT

Experimental models of the central nervous system (CNS) are imperative for developmental and pathophysiological studies of neurological diseases. Among these models, three-dimensional (3D) induced pluripotent stem cell (iPSC)-derived brain organoid models have been successful in mitigating some of the drawbacks of 2D models; however, they are plagued by high organoid-to-organoid variability, making it difficult to compare specific gene regulatory pathways across 3D organoids with those of the native brain. Single-cell RNA sequencing (scRNA-seq) transcriptome datasets have recently emerged as powerful tools to perform integrative analyses and compare variability across organoids. However, transcriptome studies focusing on late-stage neural functionality development have been underexplored. Here, we combine and analyze 8 brain organoid transcriptome databases to study the correlation between differentiation protocols and their resulting cellular functionality across various 3D organoid and exogenous brain models. We utilize dimensionality reduction methods including principal component analysis (PCA) and uniform manifold approximation projection (UMAP) to identify and visualize cellular diversity among 3D models and subsequently use gene set enrichment analysis (GSEA) and developmental trajectory inference to quantify neuronal behaviors such as axon guidance, synapse transmission and action potential. We showed high similarity in cellular composition, cellular differentiation pathways and expression of functional genes in human brain organoids during induction and differentiation phases, i.e., up to 3 months in culture. However, during the maturation phase, i.e., 6-month timepoint, we observed significant developmental deficits and depletion of neuronal and astrocytes functional genes as indicated by our GSEA results. Our results caution against use of organoids to model pathophysiology and drug response at this advanced time point and provide insights to tune in vitro iPSC differentiation protocols to achieve desired neuronal functionality and improve current protocols.


Subject(s)
Brain/metabolism , Cell Differentiation/genetics , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Organoids/metabolism , Transcriptome/genetics , Brain/embryology , Databases, Genetic , Gene Expression Regulation, Developmental , Humans , Neurons/cytology , Neurons/metabolism , Reproduction , Sequence Analysis, RNA , Signal Transduction/genetics , Single-Cell Analysis
4.
Nature ; 599(7883): 131-135, 2021 11.
Article in English | MEDLINE | ID: mdl-34646010

ABSTRACT

Oestrogen depletion in rodents and humans leads to inactivity, fat accumulation and diabetes1,2, underscoring the conserved metabolic benefits of oestrogen that inevitably decrease with age. In rodents, the preovulatory surge in 17ß-oestradiol (E2) temporarily increases energy expenditure to coordinate increased physical activity with peak sexual receptivity. Here we report that a subset of oestrogen-sensitive neurons in the ventrolateral ventromedial hypothalamic nucleus (VMHvl)3-7 projects to arousal centres in the hippocampus and hindbrain, and enables oestrogen to rebalance energy allocation in female mice. Surges in E2 increase melanocortin-4 receptor (MC4R) signalling in these VMHvl neurons by directly recruiting oestrogen receptor-α (ERα) to the Mc4r gene. Sedentary behaviour and obesity in oestrogen-depleted female mice were reversed after chemogenetic stimulation of VMHvl neurons expressing both MC4R and ERα. Similarly, a long-term increase in physical activity is observed after CRISPR-mediated activation of this node. These data extend the effect of MC4R signalling - the most common cause of monogenic human obesity8 - beyond the regulation of food intake and rationalize reported sex differences in melanocortin signalling, including greater disease severity of MC4R insufficiency in women9. This hormone-dependent node illuminates the power of oestrogen during the reproductive cycle in motivating behaviour and maintaining an active lifestyle in women.


Subject(s)
Brain/physiology , Estrogens/metabolism , Physical Exertion/physiology , Receptor, Melanocortin, Type 4/metabolism , Signal Transduction , Animals , CRISPR-Cas Systems , Energy Metabolism , Estrogen Receptor alpha/metabolism , Estrogens/deficiency , Female , Gene Editing , Hippocampus/metabolism , Male , Melanocortins/metabolism , Mice , Neurons/metabolism , Obesity/metabolism , Rhombencephalon/metabolism , Sedentary Behavior , Sex Characteristics , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/physiology
5.
Cells Dev ; 166: 203682, 2021 06.
Article in English | MEDLINE | ID: mdl-33994355

ABSTRACT

Homeotic genes and their genomic organization show remarkable conservation across bilaterians. Consequently, the regulatory mechanisms, which control hox gene expression, are also highly conserved. The crucial presence of conserved GA rich motifs between Hox genes has been previously observed but what factor binds to these is still unknown. Previously we have reported that the vertebrate homologue of Drosophila Trl-GAF preferentially binds to GA rich regions in Evx2-hoxd13 intergenic region of vertebrate HoxD cluster. In this study, we show that the vertebrate-GAF (v-GAF) binds at known cis-regulatory elements in the HoxD complex of zebrafish and mouse. We further used morpholino based knockdown and CRISPR-cas9 knockout technique to deplete the v-GAF in zebrafish. We checked expression of the HoxD genes and found gain of the HoxD4 gene in GAF knockout embryos. Further, we partially rescued the morphological phenotypes in GAF depleted embryos by providing GAF mRNA. Our results show that GAF binds at intergenic regions of the HoxD complex and is important for maintaining the spatial domains of HoxD4 expression during embryonic development.


Subject(s)
Embryonic Development , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Body Patterning/genetics , Embryonic Development/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Zebrafish Proteins/genetics
6.
Nucleic Acids Res ; 49(1): e4, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33211865

ABSTRACT

DNA strand asymmetries can have a major effect on several biological functions, including replication, transcription and transcription factor binding. As such, DNA strand asymmetries and mutational strand bias can provide information about biological function. However, a versatile tool to explore this does not exist. Here, we present Asymmetron, a user-friendly computational tool that performs statistical analysis and visualizations for the evaluation of strand asymmetries. Asymmetron takes as input DNA features provided with strand annotation and outputs strand asymmetries for consecutive occurrences of a single DNA feature or between pairs of features. We illustrate the use of Asymmetron by identifying transcriptional and replicative strand asymmetries of germline structural variant breakpoints. We also show that the orientation of the binding sites of 45% of human transcription factors analyzed have a significant DNA strand bias in transcribed regions, that is also corroborated in ChIP-seq analyses, and is likely associated with transcription. In summary, we provide a novel tool to assess DNA strand asymmetries and show how it can be used to derive new insights across a variety of biological disciplines.


Subject(s)
Computational Biology/methods , DNA Replication/genetics , DNA/genetics , Mutation , Transcription, Genetic/genetics , A549 Cells , Algorithms , Cell Line, Transformed , DNA/chemistry , DNA/metabolism , Hep G2 Cells , Humans , K562 Cells , MCF-7 Cells , Models, Genetic , Protein Binding , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Nat Rev Drug Discov ; 19(11): 757-775, 2020 11.
Article in English | MEDLINE | ID: mdl-33020616

ABSTRACT

Over a thousand diseases are caused by mutations that alter gene expression levels. The potential of nuclease-deficient zinc fingers, TALEs or CRISPR fusion systems to treat these diseases by modulating gene expression has recently emerged. These systems can be applied to modify the activity of gene-regulatory elements - promoters, enhancers, silencers and insulators, subsequently changing their target gene expression levels to achieve therapeutic benefits - an approach termed cis-regulation therapy (CRT). Here, we review emerging CRT technologies and assess their therapeutic potential for treating a wide range of diseases caused by abnormal gene dosage. The challenges facing the translation of CRT into the clinic are discussed.


Subject(s)
Gene Expression Regulation/drug effects , Genetic Diseases, Inborn/drug therapy , Pharmaceutical Preparations/administration & dosage , Animals , CRISPR-Cas Systems/drug effects , CRISPR-Cas Systems/genetics , Gene Expression Regulation/genetics , Genetic Diseases, Inborn/genetics , Humans , Mutation/drug effects , Mutation/genetics
8.
Biofabrication ; 13(1)2020 11 10.
Article in English | MEDLINE | ID: mdl-33059333

ABSTRACT

A crucial step in creating reliablein vitroplatforms for neural development and disorder studies is the reproduction of the multicellular three-dimensional (3D) brain microenvironment and the capturing of cell-cell interactions within the model. The power of self-organization of diverse cell types into brain spheroids could be harnessed to study mechanisms underlying brain development trajectory and diseases. A challenge of current 3D organoid and spheroid models grown in petri-dishes is the lack of control over cellular localization and diversity. To overcome this limitation, neural spheroids can be patterned into customizable 3D structures using microfabrication. We developed a 3D brain-like co-culture construct using embedded 3D bioprinting as a flexible solution for composing heterogenous neural populations with neurospheroids and glia. Specifically, neurospheroid-laden free-standing 3D structures were fabricated in an engineered astrocyte-laden support bath resembling a neural stem cell niche environment. A photo-crosslinkable bioink and a thermal-healing supporting bath were engineered to mimic the mechanical modulus of soft tissue while supporting the formation of self-organizing neurospheroids within elaborate 3D networks. Moreover, bioprinted neurospheroid-laden structures exhibited the capability to differentiate into neuronal cells. These brain-like co-cultures could provide a reproducible platform for modeling neurological diseases, neural regeneration, and drug development and repurposing.


Subject(s)
Bioprinting , Brain , Coculture Techniques , Hydrogels , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds
9.
Nat Metab ; 1(4): 475-484, 2019 04.
Article in English | MEDLINE | ID: mdl-31535083

ABSTRACT

Genome wide association studies (GWAS) in obesity have identified a large number of noncoding loci located near genes expressed in the central nervous system. However, due to the difficulties in isolating and characterizing specific neuronal subpopulations, few obesity-associated SNPs have been functionally characterized. Leptin responsive neurons in the hypothalamus are essential in controlling energy homeostasis and body weight. Here, we combine FACS-sorting of leptin-responsive hypothalamic neuron nuclei with genomic and epigenomic approaches (RNA-seq, ChIP-seq, ATAC-seq) to generate a comprehensive map of leptin-response specific regulatory elements, several of which overlap obesity-associated GWAS variants. We demonstrate the usefulness of our leptin-response neuron regulome, by functionally characterizing a novel enhancer near Socs3, a leptin response-associated transcription factor. We envision our data to serve as a useful resource and a blueprint for functionally characterizing obesity-associated SNPs in the hypothalamus.


Subject(s)
Body Weight/genetics , Epigenomics , Genomics , Leptin/physiology , Animals , Genome-Wide Association Study , Humans , Hypothalamus/metabolism , Hypothalamus/physiology , Mice , Neurons/physiology , Obesity/genetics , Polymorphism, Single Nucleotide , Transcriptome
11.
Science ; 363(6424)2019 01 18.
Article in English | MEDLINE | ID: mdl-30545847

ABSTRACT

A wide range of human diseases result from haploinsufficiency, where the function of one of the two gene copies is lost. Here, we targeted the remaining functional copy of a haploinsufficient gene using CRISPR-mediated activation (CRISPRa) in Sim1 and Mc4r heterozygous mouse models to rescue their obesity phenotype. Transgenic-based CRISPRa targeting of the Sim1 promoter or its distant hypothalamic enhancer up-regulated its expression from the endogenous functional allele in a tissue-specific manner, rescuing the obesity phenotype in Sim1 heterozygous mice. To evaluate the therapeutic potential of CRISPRa, we injected CRISPRa-recombinant adeno-associated virus into the hypothalamus, which led to reversal of the obesity phenotype in Sim1 and Mc4r haploinsufficient mice. Our results suggest that endogenous gene up-regulation could be a potential strategy to treat altered gene dosage diseases.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Enhancer Elements, Genetic , Haploinsufficiency , Obesity/genetics , Promoter Regions, Genetic , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line , Dependovirus , Disease Models, Animal , Female , Gene Expression Regulation , Gene Transfer Techniques , Heterozygote , Hypothalamus , Loss of Function Mutation , Male , Mice , Mice, Transgenic , Obesity/therapy , Phenotype , Receptor, Melanocortin, Type 4/genetics , Repressor Proteins/genetics , Up-Regulation , Weight Gain
12.
Proc Natl Acad Sci U S A ; 115(43): 11018-11023, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30297428

ABSTRACT

Erectile dysfunction affects millions of men worldwide. Twin studies support the role of genetic risk factors underlying erectile dysfunction, but no specific genetic variants have been identified. We conducted a large-scale genome-wide association study of erectile dysfunction in 36,649 men in the multiethnic Kaiser Permanente Northern California Genetic Epidemiology Research in Adult Health and Aging cohort. We also undertook replication analyses in 222,358 men from the UK Biobank. In the discovery cohort, we identified a single locus (rs17185536-T) on chromosome 6 near the single-minded family basic helix-loop-helix transcription factor 1 (SIM1) gene that was significantly associated with the risk of erectile dysfunction (odds ratio = 1.26, P = 3.4 × 10-25). The association replicated in the UK Biobank sample (odds ratio = 1.25, P = 6.8 × 10-14), and the effect is independent of known erectile dysfunction risk factors, including body mass index (BMI). The risk locus resides on the same topologically associating domain as SIM1 and interacts with the SIM1 promoter, and the rs17185536-T risk allele showed differential enhancer activity. SIM1 is part of the leptin-melanocortin system, which has an established role in body weight homeostasis and sexual function. Because the variants associated with erectile dysfunction are not associated with differences in BMI, our findings suggest a mechanism that is specific to sexual function.


Subject(s)
Erectile Dysfunction/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Aged , Alleles , Body Mass Index , Case-Control Studies , Chromosomes, Human, Pair 6/genetics , Cohort Studies , Humans , Leptin/genetics , Male , Melanocortins/genetics , Middle Aged , Promoter Regions, Genetic/genetics
13.
PLoS Genet ; 11(12): e1005640, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26632825

ABSTRACT

The organization and folding of chromatin within the nucleus can determine the outcome of gene expression. Recent technological advancements have enabled us to study chromatin interactions in a genome-wide manner at high resolution. These studies have increased our understanding of the hierarchy and dynamics of chromatin domains that facilitate cognate enhancer-promoter looping, defining the transcriptional program of different cell types. In this review, we focus on vertebrate chromatin long-range interactions as they relate to transcriptional regulation. In addition, we describe how the alteration of boundaries that mark discrete regions in the genome with high interaction frequencies within them, called topological associated domains (TADs), could lead to various phenotypes, including human diseases, which we term as "TADopathies."


Subject(s)
Chromatin Assembly and Disassembly/genetics , Enhancer Elements, Genetic , Promoter Regions, Genetic , Transcription, Genetic , Animals , Chromatin/genetics , Gene Expression Regulation , Humans , Mammals
14.
Genes (Basel) ; 6(3): 790-811, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26340639

ABSTRACT

The spatial organization of metazoan genomes has a direct influence on fundamental nuclear processes that include transcription, replication, and DNA repair. It is imperative to understand the mechanisms that shape the 3D organization of the eukaryotic genomes. Chromatin insulators have emerged as one of the central components of the genome organization tool-kit across species. Recent advancements in chromatin conformation capture technologies have provided important insights into the architectural role of insulators in genomic structuring. Insulators are involved in 3D genome organization at multiple spatial scales and are important for dynamic reorganization of chromatin structure during reprogramming and differentiation. In this review, we will discuss the classical view and our renewed understanding of insulators as global genome organizers. We will also discuss the plasticity of chromatin structure and its re-organization during pluripotency and differentiation and in situations of cellular stress.

15.
Methods Mol Biol ; 1196: 121-31, 2014.
Article in English | MEDLINE | ID: mdl-25151161

ABSTRACT

Hox clusters have served as a favorite system to study the role of cis-regulatory elements at multiple layers of gene regulation. Organization and regulation of Hox genes show remarkable conservation and determine the anterior-posterior body axis across the bilaterians. Identification of a variety of regulatory regions within the complex and around it, embedded primarily in the noncoding part of the corresponding genomic region that can spread 100-150 kb, is a challenging problem. Multiple experimental and computational tools need to be employed to investigate functional features of such elements. Here we discuss parallel approaches to mine the most plausible regulatory information from the noncoding sequences of Hox clusters, among diverse species.


Subject(s)
Computational Biology/methods , Gene Expression Regulation , Homeodomain Proteins/genetics , Multigene Family , Regulatory Sequences, Nucleic Acid , Animals , Conserved Sequence , Data Mining , Epigenesis, Genetic , Gene Order , Mice
16.
Sci Rep ; 3: 3011, 2013 Oct 22.
Article in English | MEDLINE | ID: mdl-24145990

ABSTRACT

Anterior-posterior body axis in all bilaterians is determined by the Hox gene clusters that are activated in a spatio-temporal order. This expression pattern of Hox genes is established and maintained by regulatory mechanisms that involve higher order chromatin structure and Polycomb group (PcG) and trithorax group (trxG) proteins. We identified earlier a Polycomb response element (PRE) in the mouse HoxD complex that is functionally conserved in flies. We analyzed the molecular and genetic interactions of mouse PRE using Drosophila melanogaster and vertebrate cell culture as the model systems. We demonstrate that the repressive activity of this PRE depends on PcG/trxG genes as well as the heterochromatin components. Our findings indicate that a wide range of factors interact with the HoxD PRE that can contribute to establishing the expression pattern of homeotic genes in the complex early during development and maintain that pattern at subsequent stages.


Subject(s)
Heterochromatin/metabolism , Homeodomain Proteins/metabolism , Polycomb-Group Proteins/metabolism , Response Elements , Animals , Animals, Genetically Modified , Cell Line , Chromosome Mapping , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Epigenesis, Genetic , Gene Expression , Gene Order , Genes, Reporter , Genotype , Heterochromatin/genetics , Humans , Male , Mice , Multiprotein Complexes , Mutation , Phenotype , Protein Binding , Temperature , Transcription Factors/metabolism
17.
Subcell Biochem ; 61: 177-207, 2013.
Article in English | MEDLINE | ID: mdl-23150252

ABSTRACT

The organization of eukaryotic genome into chromatin within the nucleus eventually dictates the cell type specific expression pattern of genes. This higher order of chromatin organization is established during development and dynamically maintained throughout the life span. Developmental mechanisms are conserved in bilaterians and hence they have body plan in common, which is achieved by regulatory networks controlling cell type specific gene expression. Homeotic genes are conserved in metazoans and are crucial for animal development as they specify cell type identity along the anterior-posterior body axis. Hox genes are the best studied in the context of epigenetic regulation that has led to significant understanding of the organismal development. Epigenome specific regulation is brought about by conserved chromatin modulating factors like PcG/trxG proteins during development and differentiation. Here we discuss the conserved epigenetic mechanisms relevant to homeotic gene regulation in metazoans.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Developmental , Genes, Homeobox , Animals , Chromatin Assembly and Disassembly , Genotype , Humans , Morphogenesis/genetics , Mutation , Phenotype , Regulatory Elements, Transcriptional , Transcription, Genetic
19.
J Mol Biol ; 400(3): 434-47, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20471984

ABSTRACT

Polycomb group (PcG) and trithorax group (trxG) proteins are chromatin-mediated regulators of a number of developmentally important genes including the homeotic genes. In Drosophila melanogaster, one of the trxG members, Trithorax like (Trl), encodes the essential multifunctional DNA binding protein called GAGA factor (GAF). While most of the PcG and trxG genes are conserved from flies to humans, a Trl-GAF homologue has been conspicuously missing in vertebrates. Here, we report the first identification of c-Krox/Th-POK as the vertebrate homologue of GAF on the basis of sequence similarity and comparative structural analysis. The in silico structural analysis of the zinc finger region showed preferential interaction of vertebrate GAF with GAGA sites similar to that of fly GAF. We also show by cross-immunoreactivity studies that both fly and vertebrate GAFs are highly conserved and share a high degree of structural similarity. Electrophoretic mobility shift assays show that vertebrate GAF binds to GAGA sites in vitro. Finally, in vivo studies by chromatin immunoprecipitation confirmed that vertebrate GAF binds to GAGA-rich DNA sequences present in hox clusters. Identification of vertebrate GAF and the presence of its target sites at various developmentally regulated loci, including hox complexes, highlight the evolutionarily conserved components involved in developmental mechanisms across the evolutionary lineage and answer a long-standing question of the presence of vertebrate GAF.


Subject(s)
Transcription Factors/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , Binding Sites , Cluster Analysis , Computational Biology , Conserved Sequence , Cross Reactions , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Drosophila Proteins/genetics , Drosophila Proteins/immunology , Electrophoretic Mobility Shift Assay , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Binding , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology, Amino Acid , Transcription Factors/immunology , Zebrafish Proteins/immunology , Zinc Fingers/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...