Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Brain ; 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38833182

ABSTRACT

Parkinson's disease (PD) is characterised neuropathologically by the degeneration of dopaminergic neurons in the ventral midbrain, the accumulation of α-synuclein (α-syn) aggregates in neurons, and chronic neuroinflammation. In the past two decades, in vitro, ex vivo and in vivo studies have consistently shown the involvement of inflammatory responses mediated by microglia and astrocytes, which may be elicited by pathological α-syn or signals from affected neurons and other cell types, and are directly linked to neurodegeneration and disease development. Besides the prominent immune alterations seen in the central nervous system (CNS), including the infiltration of T-cells into the brain, more recent studies have demonstrated important changes in the peripheral immune profile within both the innate and adaptive compartments, particularly involving monocytes, CD4+ and CD8+ T-cells. This review aims to integrate the consolidated understanding of immune-related processes underlying the pathogenesis of PD, focusing on both central and peripheral immune cells, neuron-glia crosstalk as well as the central-peripheral immune interaction during the development of PD. Our analysis seeks to provide a comprehensive view of the emerging knowledge of the mechanisms of immunity in PD and the implications of this for better understanding the overall pathogenesis of this disease.

2.
Mol Cell ; 84(5): 822-838.e8, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38157845

ABSTRACT

Chromatin loops between gene pairs have been observed in diverse contexts in both flies and vertebrates. Combining high-resolution Capture-C, DNA fluorescence in situ hybridization, and genetic perturbations, we dissect the functional role of three loops between genes with related function during Drosophila embryogenesis. By mutating the loop anchor (but not the gene) or the gene (but not loop anchor), we disentangle loop formation and gene expression and show that the 3D proximity of paralogous gene loci supports their co-regulation. Breaking the loop leads to either an attenuation or enhancement of expression and perturbs their relative levels of expression and cross-regulation. Although many loops appear constitutive across embryogenesis, their function can change in different developmental contexts. Taken together, our results indicate that chromatin gene-gene loops act as architectural scaffolds that can be used in different ways in different contexts to fine-tune the coordinated expression of genes with related functions and sustain their cross-regulation.


Subject(s)
Chromatin , Chromosomes , Animals , In Situ Hybridization, Fluorescence , Chromatin/genetics , Drosophila/genetics
3.
Nucleic Acids Res ; 45(21): 12285-12300, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29036346

ABSTRACT

A special class of poorly characterized architectural proteins is required for chromatin topology and enhancer-promoter interactions. Here, we identify Opbp as a new Drosophila architectural protein, interacting with CP190 both in vivo and in vitro. Opbp binds to a very restrictive set of genomic regions, through a rare sequence specific motif. These sites are co-bound by CP190 in vivo, and generally located at bidirectional promoters of ribosomal protein genes. We show that Opbp is essential for viability, and loss of opbp function, or destruction of its motif, leads to reduced ribosomal protein gene expression, indicating a functional role in promoter activation. As characteristic of architectural/insulator proteins, the Opbp motif is sufficient for distance-dependent reporter gene activation and enhancer-blocking activity, suggesting an Opbp-mediated enhancer-promoter interaction. Rather than having a constitutive role, Opbp represents a new type of architectural protein with a very restricted, yet essential, function in regulation of housekeeping gene expression.


Subject(s)
Drosophila Proteins/metabolism , Gene Expression Regulation , Genes, rRNA , Transcription Factors/metabolism , Animals , CRISPR-Cas Systems , Chromatin/metabolism , Drosophila/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Enhancer Elements, Genetic , Gene Deletion , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Ribosomal Proteins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/physiology , Transcriptional Activation
4.
Elife ; 62017 08 09.
Article in English | MEDLINE | ID: mdl-28792889

ABSTRACT

Sequence variation within enhancers plays a major role in both evolution and disease, yet its functional impact on transcription factor (TF) occupancy and enhancer activity remains poorly understood. Here, we assayed the binding of five essential TFs over multiple stages of embryogenesis in two distant Drosophila species (with 1.4 substitutions per neutral site), identifying thousands of orthologous enhancers with conserved or diverged combinatorial occupancy. We used these binding signatures to dissect two properties of developmental enhancers: (1) potential TF cooperativity, using signatures of co-associations and co-divergence in TF occupancy. This revealed conserved combinatorial binding despite sequence divergence, suggesting protein-protein interactions sustain conserved collective occupancy. (2) Enhancer in-vivo activity, revealing orthologous enhancers with conserved activity despite divergence in TF occupancy. Taken together, we identify enhancers with diverged motifs yet conserved occupancy and others with diverged occupancy yet conserved activity, emphasising the need to functionally measure the effect of divergence on enhancer activity.


Subject(s)
DNA/metabolism , Enhancer Elements, Genetic , Evolution, Molecular , Transcription Factors/metabolism , Animals , Drosophila/embryology , Drosophila/genetics , Protein Binding
5.
Nature ; 541(7637): 402-406, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28024300

ABSTRACT

Embryonic development is driven by tightly regulated patterns of gene expression, despite extensive genetic variation among individuals. Studies of expression quantitative trait loci (eQTL) indicate that genetic variation frequently alters gene expression in cell-culture models and differentiated tissues. However, the extent and types of genetic variation impacting embryonic gene expression, and their interactions with developmental programs, remain largely unknown. Here we assessed the effect of genetic variation on transcriptional (expression levels) and post-transcriptional (3' RNA processing) regulation across multiple stages of metazoan development, using 80 inbred Drosophila wild isolates, identifying thousands of developmental-stage-specific and shared QTL. Given the small blocks of linkage disequilibrium in Drosophila, we obtain near base-pair resolution, resolving causal mutations in developmental enhancers, validated transcription-factor-binding sites and RNA motifs. This fine-grain mapping uncovered extensive allelic interactions within enhancers that have opposite effects, thereby buffering their impact on enhancer activity. QTL affecting 3' RNA processing identify new functional motifs leading to transcript isoform diversity and changes in the lengths of 3' untranslated regions. These results highlight how developmental stage influences the effects of genetic variation and uncover multiple mechanisms that regulate and buffer expression variation during embryogenesis.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Genetic Variation , 3' Untranslated Regions/genetics , Alleles , Animals , Binding Sites , Enhancer Elements, Genetic , Linkage Disequilibrium , Mutation , Quantitative Trait Loci , RNA 3' End Processing , Transcription Factors/metabolism , Transcription, Genetic
6.
PLoS Comput Biol ; 12(9): e1005073, 2016 09.
Article in English | MEDLINE | ID: mdl-27599298

ABSTRACT

Given the complexity of developmental networks, it is often difficult to predict the effect of genetic perturbations, even within coding genes. Regulatory factors generally have pleiotropic effects, exhibit partially redundant roles, and regulate highly interconnected pathways with ample cross-talk. Here, we delineate a logical model encompassing 48 components and 82 regulatory interactions involved in mesoderm specification during Drosophila development, thereby providing a formal integration of all available genetic information from the literature. The four main tissues derived from mesoderm correspond to alternative stable states. We demonstrate that the model can predict known mutant phenotypes and use it to systematically predict the effects of over 300 new, often non-intuitive, loss- and gain-of-function mutations, and combinations thereof. We further validated several novel predictions experimentally, thereby demonstrating the robustness of model. Logical modelling can thus contribute to formally explain and predict regulatory outcomes underlying cell fate decisions.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Mesoderm/physiology , Models, Biological , Signal Transduction , Animals , Computational Biology , Drosophila/genetics , Drosophila/growth & development , Drosophila/physiology , Mutation , Phenotype , Signal Transduction/genetics , Signal Transduction/physiology
8.
Curr Biol ; 26(1): 38-51, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26687625

ABSTRACT

Embryogenesis is remarkably robust to segregating mutations and environmental variation; under a range of conditions, embryos of a given species develop into stereotypically patterned organisms. Such robustness is thought to be conferred, in part, through elements within regulatory networks that perform similar, redundant tasks. Redundant enhancers (or "shadow" enhancers), for example, can confer precision and robustness to gene expression, at least at individual, well-studied loci. However, the extent to which enhancer redundancy exists and can thereby have a major impact on developmental robustness remains unknown. Here, we systematically assessed this, identifying over 1,000 predicted shadow enhancers during Drosophila mesoderm development. The activity of 23 elements, associated with five genes, was examined in transgenic embryos, while natural structural variation among individuals was used to assess their ability to buffer against genetic variation. Our results reveal three clear properties of enhancer redundancy within developmental systems. First, it is much more pervasive than previously anticipated, with 64% of loci examined having shadow enhancers. Their spatial redundancy is often partial in nature, while the non-overlapping function may explain why these enhancers are maintained within a population. Second, over 70% of loci do not follow the simple situation of having only two shadow enhancers-often there are three (rols), four (CadN and ade5), or five (Traf1), at least one of which can be deleted with no obvious phenotypic effects. Third, although shadow enhancers can buffer variation, patterns of segregating variation suggest that they play a more complex role in development than generally considered.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Animals , Drosophila , Embryonic Development/genetics , Transcription, Genetic
9.
Nature ; 512(7512): 96-100, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25043061

ABSTRACT

Developmental enhancers initiate transcription and are fundamental to our understanding of developmental networks, evolution and disease. Despite their importance, the properties governing enhancer-promoter interactions and their dynamics during embryogenesis remain unclear. At the ß-globin locus, enhancer-promoter interactions appear dynamic and cell-type specific, whereas at the HoxD locus they are stable and ubiquitous, being present in tissues where the target genes are not expressed. The extent to which preformed enhancer-promoter conformations exist at other, more typical, loci and how transcription is eventually triggered is unclear. Here we generated a high-resolution map of enhancer three-dimensional contacts during Drosophila embryogenesis, covering two developmental stages and tissue contexts, at unprecedented resolution. Although local regulatory interactions are common, long-range interactions are highly prevalent within the compact Drosophila genome. Each enhancer contacts multiple enhancers, and promoters with similar expression, suggesting a role in their co-regulation. Notably, most interactions appear unchanged between tissue context and across development, arising before gene activation, and are frequently associated with paused RNA polymerase. Our results indicate that the general topology governing enhancer contacts is conserved from flies to humans and suggest that transcription initiates from preformed enhancer-promoter loops through release of paused polymerase.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Embryonic Development/genetics , Enhancer Elements, Genetic/genetics , Promoter Regions, Genetic/genetics , Animals , Binding Sites , Chromosomes, Insect/genetics , Chromosomes, Insect/metabolism , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental/genetics , Genetic Loci/genetics , Genome, Insect/genetics , Humans , Transcription Initiation, Genetic , Transcriptional Activation
10.
Development ; 141(13): 2633-43, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24961800

ABSTRACT

Molecular models of cell fate specification typically focus on the activation of specific lineage programs. However, the concurrent repression of unwanted transcriptional networks is also essential to stabilize certain cellular identities, as shown in a number of diverse systems and phyla. Here, we demonstrate that this dual requirement also holds true in the context of Drosophila myogenesis. By integrating genetics and genomics, we identified a new role for the pleiotropic transcriptional repressor Tramtrack69 in myoblast specification. Drosophila muscles are formed through the fusion of two discrete cell types: founder cells (FCs) and fusion-competent myoblasts (FCMs). When tramtrack69 is removed, FCMs appear to adopt an alternative muscle FC-like fate. Conversely, ectopic expression of this repressor phenocopies muscle defects seen in loss-of-function lame duck mutants, a transcription factor specific to FCMs. This occurs through Tramtrack69-mediated repression in FCMs, whereas Lame duck activates a largely distinct transcriptional program in the same cells. Lineage-specific factors are therefore not sufficient to maintain FCM identity. Instead, their identity appears more plastic, requiring the combination of instructive repressive and activating programs to stabilize cell fate.


Subject(s)
Cell Differentiation/physiology , Drosophila Proteins/metabolism , Drosophila/embryology , Gene Expression Regulation, Developmental/physiology , Muscle Development/physiology , Myoblasts/physiology , Myogenic Regulatory Factors/metabolism , Repressor Proteins/metabolism , Animals , Chromatin Immunoprecipitation , Drosophila/genetics , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental/genetics , In Situ Hybridization, Fluorescence , Mesoderm/physiology , Myoblasts/metabolism
11.
Genes Dev ; 28(2): 167-81, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24402316

ABSTRACT

The transcription factors of the Snail family are key regulators of epithelial-mesenchymal transitions, cell morphogenesis, and tumor metastasis. Since its discovery in Drosophila ∼25 years ago, Snail has been extensively studied for its role as a transcriptional repressor. Here we demonstrate that Drosophila Snail can positively modulate transcriptional activation. By combining information on in vivo occupancy with expression profiling of hand-selected, staged snail mutant embryos, we identified 106 genes that are potentially directly regulated by Snail during mesoderm development. In addition to the expected Snail-repressed genes, almost 50% of Snail targets showed an unanticipated activation. The majority of "Snail-activated" genes have enhancer elements cobound by Twist and are expressed in the mesoderm at the stages of Snail occupancy. Snail can potentiate Twist-mediated enhancer activation in vitro and is essential for enhancer activity in vivo. Using a machine learning approach, we show that differentially enriched motifs are sufficient to predict Snail's regulatory response. In silico mutagenesis revealed a likely causative motif, which we demonstrate is essential for enhancer activation. Taken together, these data indicate that Snail can potentiate enhancer activation by collaborating with different activators, providing a new mechanism by which Snail regulates development.


Subject(s)
Drosophila/genetics , Drosophila/metabolism , Transcription Factors/metabolism , Amino Acid Motifs , Animals , Drosophila/embryology , Drosophila Proteins/metabolism , Embryo, Nonmammalian , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Mesoderm/metabolism , Protein Binding , Snail Family Transcription Factors , Transcription Factors/genetics , Twist-Related Protein 1/metabolism
12.
PLoS Genet ; 6(7): e1001014, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20617173

ABSTRACT

Understanding how complex patterns of temporal and spatial expression are regulated is central to deciphering genetic programs that drive development. Gene expression is initiated through the action of transcription factors and their cofactors converging on enhancer elements leading to a defined activity. Specific constellations of combinatorial occupancy are therefore often conceptualized as rigid binding codes that give rise to a common output of spatio-temporal expression. Here, we assessed this assumption using the regulatory input of two essential transcription factors within the Drosophila myogenic network. Mutations in either Myocyte enhancing factor 2 (Mef2) or the zinc-finger transcription factor lame duck (lmd) lead to very similar defects in myoblast fusion, yet the underlying molecular mechanism for this shared phenotype is not understood. Using a combination of ChIP-on-chip analysis and expression profiling of loss-of-function mutants, we obtained a global view of the regulatory input of both factors during development. The majority of Lmd-bound enhancers are co-bound by Mef2, representing a subset of Mef2's transcriptional input during these stages of development. Systematic analyses of the regulatory contribution of both factors demonstrate diverse regulatory roles, despite their co-occupancy of shared enhancer elements. These results indicate that Lmd is a tissue-specific modulator of Mef2 activity, acting as both a transcriptional activator and repressor, which has important implications for myogenesis. More generally, this study demonstrates considerable flexibility in the regulatory output of two factors, leading to additive, cooperative, and repressive modes of co-regulation.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Gene Expression Regulation, Developmental , Myogenic Regulatory Factors/metabolism , Animals , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/genetics , Enhancer Elements, Genetic , Female , Male , Muscle Development , Muscles/metabolism , Myogenic Regulatory Factors/genetics , Organ Specificity , Protein Binding
13.
Curr Opin Cell Biol ; 21(6): 754-60, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19896355

ABSTRACT

Understanding developmental networks has recently been enhanced through the identification of a large number of conserved essential regulators. Interspecies comparisons of the transcriptional networks regulated by these factors are still at a rather early stage, with limited global data available. Here we use the accumulating phenotypic information from multiple species to provide initial insights into the wiring and rewiring of developmental networks, with particular emphasis on myogenesis, a highly conserved developmental process. This review highlights the most recent findings on the transcriptional program driving Drosophila myogenesis and compares this with vertebrates, revealing emerging themes that may be applicable to other developmental contexts.


Subject(s)
Drosophila/embryology , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Muscle Development/genetics , Animals , Body Patterning/genetics , Drosophila/genetics , Drosophila/physiology , Drosophila Proteins/genetics , Genes, Insect , Genome, Insect , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...