Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 13(7)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38607052

RESUMEN

Transcription factors (TFs) regulate gene expression by recognizing specific target enhancers in the genome. The DNA-binding and regulatory activity of TFs depend on the presence of additional protein partners, leading to the formation of versatile and dynamic multimeric protein complexes. Visualizing these protein-protein interactions (PPIs) in the nucleus is key for decrypting the molecular cues underlying TF specificity in vivo. Over the last few years, Bimolecular Fluorescence Complementation (BiFC) has been developed in several model systems and applied in the analysis of different types of PPIs. In particular, BiFC has been applied when analyzing PPIs with hundreds of TFs in the nucleus of live Drosophila embryos. However, the visualization of PPIs at the level of specific target enhancers or genomic regions of interest awaits the advent of DNA-labelling methods that can be coupled with BiFC. Here, we present a novel experimental strategy that we have called BiFOR and that is based on the coupling of BiFC with the bacterial ANCHOR DNA-labelling system. We demonstrate that BiFOR enables the precise quantification of the enrichment of specific dimeric protein complexes on target enhancers in Drosophila salivary gland nuclei. Given its versatility and sensitivity, BiFOR could be applied more widely to other tissues during Drosophila development. Our work sets up the experimental basis for future applications of this strategy.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/metabolismo , Microscopía Fluorescente/métodos , Factores de Transcripción/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , ADN/metabolismo
2.
J Mol Evol ; 91(5): 616-627, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37341745

RESUMEN

Hox genes encode Homeodomain-containing transcription factors, which specify segmental identities along the anterior-posterior axis. Functional changes in Hox genes have been directly implicated in the evolution of body plans across the metazoan lineage. The Hox protein Ultrabithorax (Ubx) is expressed and required in developing third thoracic (T3) segments in holometabolous insects studied so far, particularly, of the order Coleoptera, Lepidoptera and Diptera. Ubx function is key to specify differential development of the second (T2) and T3 thoracic segments in these insects. While Ubx is expressed in the third thoracic segment in developing larvae of Hymenopteran Apis mellifera, the morphological differences between T2 and T3 are subtle. To identify evolutionary changes that are behind the differential function of Ubx in Drosophila and Apis, which are diverged for more than 350 million years, we performed comparative analyses of genome wide Ubx-binding sites between these two insects. Our studies reveal that a motif with a TAAAT core is a preferred binding site for Ubx in Drosophila, but not in Apis. Biochemical and transgenic assays suggest that in Drosophila, the TAAAT core sequence in the Ubx binding sites is required for Ubx-mediated regulation of two of its target genes studied here; CG13222, a gene that is normally upregulated by Ubx and vestigial (vg), whose expression is repressed by Ubx in T3. Interestingly, changing the TAAT site to a TAAAT site was sufficient to bring an otherwise unresponsive enhancer of the vg gene from Apis under the control of Ubx in a Drosophila transgenic assay. Taken together, our results suggest an evolutionary mechanism by which critical wing patterning genes might have come under the regulation of Ubx in the Dipteran lineage.

3.
Cells ; 12(1)2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36611993

RESUMEN

Biological pathways rely on the formation of intricate protein interaction networks called interactomes. Getting a comprehensive map of interactomes implies the development of tools that allow one to capture transient and low-affinity protein-protein interactions (PPIs) in live conditions. Here we presented an experimental strategy: the Cell-PCA (cell-based protein complementation assay), which was based on bimolecular fluorescence complementation (BiFC) for ORFeome-wide screening of proteins that interact with different bait proteins in the same live cell context, by combining high-throughput sequencing method. The specificity and sensitivity of the Cell-PCA was established by using a wild-type and a single-amino-acid-mutated HOXA9 protein, and the approach was subsequently applied to seven additional human HOX proteins. These proof-of-concept experiments revealed novel molecular properties of HOX interactomes and led to the identification of a novel cofactor of HOXB13 that promoted its proliferative activity in a cancer cell context. Taken together, our work demonstrated that the Cell-PCA was pertinent for revealing and, importantly, comparing the interactomes of different or highly related bait proteins in the same cell context.


Asunto(s)
Mapas de Interacción de Proteínas , Humanos , Microscopía Fluorescente/métodos
4.
Blood ; 141(10): 1209-1220, 2023 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-36375119

RESUMEN

Understanding the functional role of mutated genes in cancer is required to translate the findings of cancer genomics into therapeutic improvement. BTG1 is recurrently mutated in the MCD/C5 subtype of diffuse large B-cell lymphoma (DLBCL), which is associated with extranodal dissemination. Here, we provide evidence that Btg1 knock out accelerates the development of a lethal lymphoproliferative disease driven by Bcl2 overexpression. Furthermore, we show that the scaffolding protein BCAR1 is a BTG1 partner. Moreover, after BTG1 deletion or expression of BTG1 mutations observed in patients with DLBCL, the overactivation of the BCAR1-RAC1 pathway confers increased migration ability in vitro and in vivo. These modifications are targetable with the SRC inhibitor dasatinib, which opens novel therapeutic opportunities in BTG1 mutated DLBCL.


Asunto(s)
Linfoma de Células B Grandes Difuso , Humanos , Linfoma de Células B Grandes Difuso/patología , Mutación , Genes cdc , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína Sustrato Asociada a CrK/genética , Proteína Sustrato Asociada a CrK/metabolismo
5.
Wiley Interdiscip Rev RNA ; 14(2): e1752, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-35899407

RESUMEN

Transcription factors (TFs) are present in all life forms and conserved across great evolutionary distances in eukaryotes. From yeast to complex multicellular organisms, they are pivotal players of cell fate decision by orchestrating gene expression at diverse molecular layers. Notably, TFs fine-tune gene expression by coordinating RNA fate at both the expression and splicing levels. They regulate alternative splicing, an essential mechanism for cell plasticity, allowing the production of many mRNA and protein isoforms in precise cell and tissue contexts. Despite this apparent role in splicing, how TFs integrate transcription and splicing to ultimately orchestrate diverse cell functions and cell fate decisions remains puzzling. We depict substantial studies in various model organisms underlining the key role of TFs in alternative splicing for promoting tissue-specific functions and cell fate. Furthermore, we emphasize recent advances describing the molecular link between the transcriptional and splicing activities of TFs. As TFs can bind both DNA and/or RNA to regulate transcription and splicing, we further discuss their flexibility and compatibility for DNA and RNA substrates. Finally, we propose several models integrating transcription and splicing activities of TFs in the coordination and diversification of cell and tissue identities. This article is categorized under: RNA Processing > Splicing Regulation/Alternative Splicing RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications RNA Processing > Splicing Mechanisms.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Empalme del ARN , Factores de Transcripción , Transcripción Genética , Diferenciación Celular/genética , Empalme del ARN/genética , Factores de Transcripción/metabolismo , Linaje de la Célula/genética , Análisis Espacio-Temporal , ADN Polimerasa II/química , ADN Polimerasa II/metabolismo , ADN/metabolismo , ARN/metabolismo , Humanos , Animales
6.
J Dev Biol ; 10(3)2022 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-35997398

RESUMEN

In this Special Issue on "Hox genes in development: new paradigms", we present a compilation of articles and reviews tackling various aspects of the Hox biology field [...].

7.
Cells ; 10(11)2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34831206

RESUMEN

It is recognized that a large proportion of eukaryotic RNAs and proteins is not produced from conventional genes but from short and alternative (alt) open reading frames (ORFs) that are not captured by gene prediction programs. Here we present an in silico prediction of altORFs by applying several selecting filters based on evolutionary conservation and annotations of previously characterized altORF peptides. Our work was performed in the Bithorax-complex (BX-C), which was one of the first genomic regions described to contain long non-coding RNAs in Drosophila. We showed that several altORFs could be predicted from coding and non-coding sequences of BX-C. In addition, the selected altORFs encode for proteins that contain several interesting molecular features, such as the presence of transmembrane helices or a general propensity to be rich in short interaction motifs. Of particular interest, one altORF encodes for a protein that contains a peptide sequence found in specific isoforms of two Drosophila Hox proteins. Our work thus suggests that several altORF proteins could be produced from a particular genomic region known for its critical role during Drosophila embryonic development. The molecular signatures of these altORF proteins further suggests that several of them could make numerous protein-protein interactions and be of functional importance in vivo.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Anotación de Secuencia Molecular , Sistemas de Lectura Abierta/genética , Péptidos/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Proteínas de Drosophila/química
8.
Front Cell Dev Biol ; 9: 713282, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34368162

RESUMEN

Developmental processes have to be robust but also flexible enough to respond to genetic and environmental variations. Different mechanisms have been described to explain the apparent antagonistic nature of developmental robustness and plasticity. Here, we present a "self-sufficient" molecular model to explain the development of a particular flight organ that is under the control of the Hox gene Ultrabithorax (Ubx) in the fruit fly Drosophila melanogaster. Our model is based on a candidate RNAi screen and additional genetic analyses that all converge to an autonomous and cofactor-independent mode of action for Ubx. We postulate that this self-sufficient molecular mechanism is possible due to an unusually high expression level of the Hox protein. We propose that high dosage could constitute a so far poorly investigated molecular strategy for allowing Hox proteins to both innovate and stabilize new forms during evolution.

9.
Methods Mol Biol ; 2350: 173-190, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34331286

RESUMEN

Deciphering protein-protein interactions (PPIs) in vivo is crucial to understand protein function. Bimolecular fluorescence complementation (BiFC) makes applicable the analysis of PPIs in many different native contexts, including human live cells. It relies on the property of monomeric fluorescent proteins to be reconstituted from two separate subfragments upon spatial proximity. Candidate partners fused to such complementary subfragments can form a fluorescent protein complex upon interaction, allowing visualization of weak and transient PPIs. It can also be applied for investigation of distinct PPIs at the same time using a multicolor setup. In this chapter, we provide a detailed protocol for analyzing PPIs by doing BiFC in cultured cells. Proof-of-principle experiments rely on the complementation property between the N-terminal fragment of mVenus (designated VN173) and the C-terminal fragment of mCerulean (designated CC155) and the partnership between HOXA7 and PBX1 proteins. This protocol is compatible with any other fluorescent complementation pair fragments and any type of candidate interacting proteins.


Asunto(s)
Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Imagen Molecular/métodos , Mapeo de Interacción de Proteínas/métodos , Línea Celular , Rastreo Celular , Análisis de Datos , Expresión Génica , Orden Génico , Vectores Genéticos , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Espectrofotometría , Transfección
10.
Nat Commun ; 12(1): 2892, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-34001903

RESUMEN

Flying insects have invaded all the aerial space on Earth and this astonishing radiation could not have been possible without a remarkable morphological diversification of their flight appendages. Here, we show that characteristic spatial expression profiles and levels of the Hox genes Antennapedia (Antp) and Ultrabithorax (Ubx) underlie the formation of two different flight organs in the fruit fly Drosophila melanogaster. We further demonstrate that flight appendage morphology is dependent on specific Hox doses. Interestingly, we find that wing morphology from evolutionary distant four-winged insect species is also associated with a differential expression of Antp and Ubx. We propose that variation in the spatial expression profile and dosage of Hox proteins is a major determinant of flight appendage diversification in Drosophila and possibly in other insect species during evolution.


Asunto(s)
Proteína con Homeodominio Antennapedia/genética , Proteínas de Drosophila/genética , Vuelo Animal , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Animales , Proteína con Homeodominio Antennapedia/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Dosificación de Gen , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Alas de Animales/anatomía & histología , Alas de Animales/metabolismo
11.
J Exp Med ; 218(3)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33606008

RESUMEN

Juvenile idiopathic arthritis is the most common chronic rheumatic disease in children, and its etiology remains poorly understood. Here, we explored four families with early-onset arthritis carrying homozygous loss-of-expression mutations in LACC1. To understand the link between LACC1 and inflammation, we performed a functional study of LACC1 in human immune cells. We showed that LACC1 was primarily expressed in macrophages upon mTOR signaling. We found that LACC1 deficiency had no obvious impact on inflammasome activation, type I interferon response, or NF-κB regulation. Using bimolecular fluorescence complementation and biochemical assays, we showed that autophagy-inducing proteins, RACK1 and AMPK, interacted with LACC1. Autophagy blockade in macrophages was associated with LACC1 cleavage and degradation. Moreover, LACC1 deficiency reduced autophagy flux in primary macrophages. This was associated with a defect in the accumulation of lipid droplets and mitochondrial respiration, suggesting that LACC1-dependent autophagy fuels macrophage bioenergetics metabolism. Altogether, LACC1 deficiency defines a novel form of genetically inherited juvenile arthritis associated with impaired autophagy in macrophages.


Asunto(s)
Artritis Juvenil/metabolismo , Artritis Juvenil/patología , Autofagia , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Macrófagos/metabolismo , Adenilato Quinasa/metabolismo , Adolescente , Secuencia de Aminoácidos , Apoptosis/efectos de los fármacos , Artritis Juvenil/genética , Autofagia/efectos de los fármacos , Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Bacterias/metabolismo , Diferenciación Celular/efectos de los fármacos , Niño , Exoma/genética , Femenino , Homocigoto , Humanos , Inflamasomas/metabolismo , Inflamación/complicaciones , Inflamación/patología , Interferones/metabolismo , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/metabolismo , Mutación con Pérdida de Función/genética , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/efectos de los fármacos , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Monocitos/efectos de los fármacos , Monocitos/patología , FN-kappa B/metabolismo , Linaje , Proteómica , Receptores de Cinasa C Activada/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Adulto Joven
12.
J Cell Sci ; 133(18)2020 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-32878938

RESUMEN

Hox proteins are major regulators of embryonic development, acting in the nucleus to regulate the expression of their numerous downstream target genes. By analyzing deletion forms of the Drosophila Hox protein Ultrabithorax (Ubx), we identified the presence of an unconventional nuclear export signal (NES) that overlaps with a highly conserved motif originally described as mediating the interaction with the PBC proteins, a generic and crucial class of Hox transcriptional cofactors that act in development and cancer. We show that this unconventional NES is involved in the interaction with the major exportin protein CRM1 (also known as Embargoed in flies) in vivo and in vitro We find that this interaction is tightly regulated in the Drosophila fat body to control the autophagy-repressive activity of Ubx during larval development. The role of the PBC interaction motif as part of an unconventional NES was also uncovered in other Drosophila and human Hox proteins, highlighting the evolutionary conservation of this novel function. Together, our results reveal the extreme molecular versatility of a unique short peptide motif for controlling the context-dependent activity of Hox proteins both at transcriptional and non-transcriptional levels.


Asunto(s)
Proteínas de Drosophila , Drosophila , Transporte Activo de Núcleo Celular , Animales , Autofagia/genética , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Cuerpo Adiposo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Péptidos , Factores de Transcripción/metabolismo
13.
Dev Biol ; 454(2): 145-155, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31251896

RESUMEN

The specification and morphogenesis of an organ requires the coordinate deployment and integration of regulatory information, including sex specific information when the organ is sex specific. Only a few gene networks controlling size and pattern development have been deciphered, which limits the emergence of principles, general or not, underlying the organ-specifying gene networks. Here we elucidate the genetic and molecular network determining the control of size in the Drosophila abdominal A9 primordium, contributing to the female genitalia. This network requires axial regulatory information provided by the Hox protein Abdominal-BR (Abd-BR), the Hox cofactors Extradenticle (Exd) and Homothorax (Hth), and the sex specific transcription factor Doublesex Female (DsxF). These factors synergize to control size in the female A9 by the coordinate regulation of the Decapentaplegic (Dpp) growth pathway. Molecular dissection of the dpp regulatory region and in vivo protein interaction experiments suggest that Abd-BR, Exd, Hth and DsxF coordinately regulate a short dpp enhancer to repress dpp expression and restrict female A9 size. The same regulators can also suppress dpp expression in the A8, but this requires the absence of the Abd-BM isoform, which specifies A8. These results delineate the network controlling female A9 growth in Drosophila.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Genitales Femeninos/crecimiento & desarrollo , Animales , Proteínas de Unión al ADN/metabolismo , Drosophila/genética , Drosophila/crecimiento & desarrollo , Desarrollo Embrionario/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Genes Homeobox/genética , Genes de Insecto/genética , Proteínas de Homeodominio/metabolismo , Morfogénesis/genética , Proteínas Nucleares/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Factores de Transcripción/metabolismo
14.
Sci Rep ; 9(1): 5664, 2019 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-30952900

RESUMEN

HOX proteins interact with PBX and MEIS cofactors, which belong to the TALE-class of homeodomain (HD)-containing transcription factors. Although the formation of HOX-PBX complexes depends on a unique conserved HOX motif called hexapeptide (HX), the additional presence of MEIS induces a remodeling of the interaction, leading to a global dispensability of the HX motif for trimeric complex formation in the large majority of HOX proteins. In addition, it was shown that the anterior HOXB3 and central HOXA7 and HOXC8 proteins could use different alternative TALE interaction motifs, with or without the HX motif, depending on the DNA-binding site and cell context. Here we dissected the molecular interaction properties of the human posterior HOXA9 protein with its TALE cofactors, PBX1 and MEIS1. Analysis was performed on different DNA-binding sites in vitro and by doing Bimolecular Fluorescence Complementation (BiFC) in different cell lines. Notably, we observed that the HOXA9-TALE interaction relies consistently on the redundant activity of the HX motif and two paralog-specific residues of the HOXA9 HD. Together with previous work, our results show that HOX proteins interact with their generic TALE cofactors through various modalities, ranging from unique and context-independent to versatile and context-dependent TALE binding interfaces.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Sitios de Unión/fisiología , Línea Celular , Línea Celular Tumoral , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Células HeLa , Humanos , Células MCF-7 , Proteínas de Neoplasias/metabolismo , Unión Proteica/fisiología
15.
Elife ; 72018 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-30247122

RESUMEN

Transcription factors achieve specificity by establishing intricate interaction networks that will change depending on the cell context. Capturing these interactions in live condition is however a challenging issue that requires sensitive and non-invasive methods.We present a set of fly lines, called 'multicolor BiFC library', which covers most of the Drosophila transcription factors for performing Bimolecular Fluorescence Complementation (BiFC). The multicolor BiFC library can be used to probe two different binary interactions simultaneously and is compatible for large-scale interaction screens. The library can also be coupled with established Drosophila genetic resources to analyze interactions in the developmentally relevant expression domain of each protein partner. We provide proof of principle experiments of these various applications, using Hox proteins in the live Drosophila embryo as a case study. Overall this novel collection of ready-to-use fly lines constitutes an unprecedented genetic toolbox for the identification and analysis of protein-protein interactions in vivo.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/genética , Biblioteca de Genes , Mapeo de Interacción de Proteínas/métodos , Factores de Transcripción/genética , Animales , Animales Modificados Genéticamente , Color , Drosophila/embriología , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Fluorescencia , Regulación del Desarrollo de la Expresión Génica , Proteínas Luminiscentes/química , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Fluorescente/métodos , Unión Proteica , Factores de Transcripción/metabolismo
16.
Cell Rep ; 22(11): 3058-3071, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29539431

RESUMEN

HOX proteins achieve numerous functions by interacting with the TALE class PBX and MEIS cofactors. In contrast to this established partnership in development and disease, how HOX proteins could interact with PBX and MEIS remains unclear. Here, we present a systematic analysis of HOX/PBX/MEIS interaction properties, scanning all paralog groups with human and mouse HOX proteins in vitro and in live cells. We demonstrate that a previously characterized HOX protein motif known to be critical for HOX-PBX interactions becomes dispensable in the presence of MEIS in all except the two most anterior paralog groups. We further identify paralog-specific TALE-binding sites that are used in a highly context-dependent manner. One of these binding sites is involved in the proliferative activity of HOXA7 in breast cancer cells. Together these findings reveal an extraordinary level of interaction flexibility between HOX proteins and their major class of developmental cofactors.


Asunto(s)
Genes Homeobox/genética , Proteínas de Homeodominio/metabolismo , Proteínas de Neoplasias/metabolismo , Factores de Transcripción/metabolismo , Humanos
17.
Int J Dev Biol ; 62(11-12): 865-876, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30604856

RESUMEN

HOX and TALE genes encode homeodomain (HD)-containing transcription factors that act in concert in different tissues to coordinate cell fates and morphogenesis throughout embryonic development. These two evolutionary conserved families contain several members that form different types of protein complexes on DNA. Mutations affecting the expression of HOX or TALE genes have been reported in a number of cancers, but whether and how the two gene families could be perturbed together has never been explored systematically. As a consequence, the putative collaborative role between HOX and TALE members for promoting or inhibiting oncogenesis remains to be established in most cancer contexts. Here, we address this issue by considering HOX and TALE expression profiling in normal and cancer adult tissues, using normalized RNA-sequencing expression data deriving from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) research projects. Information was extracted from 28 cancer types originating from 21 different tissues, constituting a unique comparative analysis of HOX and TALE expression profiles between normal and cancer contexts in human. We present the general and specific rules that could be deduced from this large-scale comparative analysis. Overall this work provides a precious annotated support to better understand the role of specific HOX/TALE combinatorial codes in human cancers.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Genes Homeobox , Proteínas de Homeodominio/genética , Neoplasias/genética , Factores de Transcripción/genética , Transformación Celular Neoplásica/genética , Bases de Datos Genéticas , Perfilación de la Expresión Génica , Humanos
18.
Curr Opin Genet Dev ; 43: 1-8, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27768937

RESUMEN

Homeodomain proteins are evolutionary conserved proteins present in the entire eukaryote kingdom. They execute functions that are essential for life, both in developing and adult organisms. Most homeodomain proteins act as transcription factors and bind DNA to control the activity of other genes. In contrast to their similar DNA binding specificity, homeodomain proteins execute highly diverse and context-dependent functions. Several factors, including genome accessibility, DNA shape, combinatorial binding and the ability to interact with many transcriptional partners, diversify the activity of homeodomain proteins and culminate in the activation of highly dynamic, context-specific transcriptional programs. Clarifying how homeodomain transcription factors work is central to our understanding of development, disease and evolution.


Asunto(s)
Proteínas de Unión al ADN/genética , ADN/genética , Proteínas de Homeodominio/genética , Transcripción Genética , Secuencia de Aminoácidos/genética , Sitios de Unión/genética , Eucariontes/genética , Humanos
19.
Trends Genet ; 32(6): 334-347, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27066866

RESUMEN

Hox proteins are key regulatory transcription factors that act in different tissues of the embryo to provide specific spatial and temporal coordinates to each cell. These patterning functions often depend on the presence of the TALE-homeodomain class cofactors, which form cooperative DNA-binding complexes with all Hox proteins. How this family of cofactors contributes to the highly diverse and specific functions of Hox proteins in vivo remains an important unsolved question. We review here the most recent advances in understanding the molecular mechanisms underlying Hox-TALE function. In particular, we discuss the role of DNA shape, DNA-binding affinity, and protein-protein interaction flexibility in dictating Hox-TALE specificity. We propose several models to explain how these mechanisms are integrated with each other in the context of the many distinct functions that Hox and TALE factors carry out in vivo.


Asunto(s)
Proteínas de Unión al ADN/genética , ADN/genética , Proteínas de Homeodominio/genética , Secuencia de Aminoácidos/genética , Animales , Regulación del Desarrollo de la Expresión Génica , Unión Proteica , Mapas de Interacción de Proteínas/genética
20.
Front Genet ; 6: 267, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26347770

RESUMEN

Hox genes are major regulators of embryonic development. One of their most conserved functions is to coordinate the formation of specific body structures along the anterior-posterior (AP) axis in Bilateria. This architectural role was at the basis of several morphological innovations across bilaterian evolution. In this review, we traced the origin of the Hox patterning system by considering the partnership with PBC and Meis proteins. PBC and Meis belong to the TALE-class of homeodomain-containing transcription factors and act as generic cofactors of Hox proteins for AP axis patterning in Bilateria. Recent data indicate that Hox proteins acquired the ability to interact with their TALE partners in the last common ancestor of Bilateria and Cnidaria. These interactions relied initially on a short peptide motif called hexapeptide (HX), which is present in Hox and non-Hox protein families. Remarkably, Hox proteins can also recruit the TALE cofactors by using specific PBC Interaction Motifs (SPIMs). We describe how a functional Hox/TALE patterning system emerged in eumetazoans through the acquisition of SPIMs. We anticipate that interaction flexibility could be found in other patterning systems, being at the heart of the astonishing morphological diversity observed in the animal kingdom.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA