Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Genes (Basel) ; 14(1)2023 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-36672963

RESUMEN

The SOX transcription factor family is pivotal in controlling aspects of development. To identify genotype-phenotype relationships of SOX proteins, we performed a non-biased study of SOX using 1890 open-reading frame and 6667 amino acid sequences in combination with structural dynamics to interpret 3999 gnomAD, 485 ClinVar, 1174 Geno2MP, and 4313 COSMIC human variants. We identified, within the HMG (High Mobility Group)- box, twenty-seven amino acids with changes in multiple SOX proteins annotated to clinical pathologies. These sites were screened through Geno2MP medical phenotypes, revealing novel SOX15 R104G associated with musculature abnormality and SOX8 R159G with intellectual disability. Within gnomAD, SOX18 E137K (rs201931544), found within the HMG box of ~0.8% of Latinx individuals, is associated with seizures and neurological complications, potentially through blood-brain barrier alterations. A total of 56 highly conserved variants were found at sites outside the HMG-box, including several within the SOX2 HMG-box-flanking region with neurological associations, several in the SOX9 dimerization region associated with Campomelic Dysplasia, SOX14 K88R (rs199932938) flanking the HMG box associated with cardiovascular complications within European populations, and SOX7 A379V (rs143587868) within an SOXF conserved far C-terminal domain heterozygous in 0.716% of African individuals with associated eye phenotypes. This SOX data compilation builds a robust genotype-to-phenotype association for a gene family through more robust ortholog data integration.


Asunto(s)
Proteínas del Grupo de Alta Movilidad , Factores de Transcripción SOX , Humanos , Proteínas del Grupo de Alta Movilidad/química , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Factores de Transcripción SOX/genética , Secuencia de Aminoácidos , Dimerización , Genotipo , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo , Factores de Transcripción SOXB2/genética , Factores de Transcripción SOXB2/metabolismo , Factores de Transcripción SOXE/genética
3.
EMBO J ; 42(5): e109032, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36715213

RESUMEN

Despite a growing catalog of secreted factors critical for lymphatic network assembly, little is known about the mechanisms that modulate the expression level of these molecular cues in blood vascular endothelial cells (BECs). Here, we show that a BEC-specific transcription factor, SOX7, plays a crucial role in a non-cell-autonomous manner by modulating the transcription of angiocrine signals to pattern lymphatic vessels. While SOX7 is not expressed in lymphatic endothelial cells (LECs), the conditional loss of SOX7 function in mouse embryos causes a dysmorphic dermal lymphatic phenotype. We identify novel distant regulatory regions in mice and humans that contribute to directly repressing the transcription of a major lymphangiogenic growth factor (Vegfc) in a SOX7-dependent manner. Further, we show that SOX7 directly binds HEY1, a canonical repressor of the Notch pathway, suggesting that transcriptional repression may also be modulated by the recruitment of this protein partner at Vegfc genomic regulatory regions. Our work unveils a role for SOX7 in modulating downstream signaling events crucial for lymphatic patterning, at least in part via the transcriptional repression of VEGFC levels in the blood vascular endothelium.


Asunto(s)
Células Endoteliales , Vasos Linfáticos , Humanos , Ratones , Animales , Células Endoteliales/metabolismo , Vasos Linfáticos/metabolismo , Regulación de la Expresión Génica , Endotelio Vascular , Factores de Transcripción/metabolismo , Linfangiogénesis/genética , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo
4.
Nucleic Acids Res ; 49(19): 10931-10955, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34570228

RESUMEN

Few genetically dominant mutations involved in human disease have been fully explained at the molecular level. In cases where the mutant gene encodes a transcription factor, the dominant-negative mode of action of the mutant protein is particularly poorly understood. Here, we studied the genome-wide mechanism underlying a dominant-negative form of the SOX18 transcription factor (SOX18RaOp) responsible for both the classical mouse mutant Ragged Opossum and the human genetic disorder Hypotrichosis-lymphedema-telangiectasia-renal defect syndrome. Combining three single-molecule imaging assays in living cells together with genomics and proteomics analysis, we found that SOX18RaOp disrupts the system through an accumulation of molecular interferences which impair several functional properties of the wild-type SOX18 protein, including its target gene selection process. The dominant-negative effect is further amplified by poisoning the interactome of its wild-type counterpart, which perturbs regulatory nodes such as SOX7 and MEF2C. Our findings explain in unprecedented detail the multi-layered process that underpins the molecular aetiology of dominant-negative transcription factor function.


Asunto(s)
Glomerulonefritis/genética , Hipotricosis/genética , Linfedema/genética , Factores de Transcripción SOXF/genética , Telangiectasia/genética , Transcripción Genética , Animales , Células COS , Chlorocebus aethiops , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Genes Reporteros , Glomerulonefritis/metabolismo , Glomerulonefritis/patología , Células HeLa , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hipotricosis/metabolismo , Hipotricosis/patología , Luciferasas/genética , Luciferasas/metabolismo , Linfedema/metabolismo , Linfedema/patología , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Ratones , Mutación , Factores de Transcripción SOXF/metabolismo , Imagen Individual de Molécula , Telangiectasia/metabolismo , Telangiectasia/patología
5.
Emerg Microbes Infect ; 10(1): 178-195, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33372854

RESUMEN

The genome of SARS-CoV-2 encodes two viral proteases (NSP3/papain-like protease and NSP5/3C-like protease) that are responsible for cleaving viral polyproteins during replication. Here, we discovered new functions of the NSP3 and NSP5 proteases of SARS-CoV-2, demonstrating that they could directly cleave proteins involved in the host innate immune response. We identified 3 proteins that were specifically and selectively cleaved by NSP3 or NSP5: IRF-3, and NLRP12 and TAB1, respectively. Direct cleavage of IRF3 by NSP3 could explain the blunted Type-I IFN response seen during SARS-CoV-2 infections while NSP5 mediated cleavage of NLRP12 and TAB1 point to a molecular mechanism for enhanced production of cytokines and inflammatory responThe genome of SARS-CoV-2 encodes two viral proteases (NSP3/papain-like protease and NSP5/3C-like protease) that are responsible for cleaving viral polyproteins during replication. Here, we discovered new functions of the NSP3 and NSP5 proteases of SARS-CoV-2, demonstrating that they could directly cleave proteins involved in the host innate immune response. We identified 3 proteins that were specifically and selectively cleaved by NSP3 or NSP5: IRF-3, and NLRP12 and TAB1, respectively. Direct cleavage of IRF3 by NSP3 could explain the blunted Type-I IFN response seen during SARS-CoV-2 infections while NSP5 mediated cleavage of NLRP12 and TAB1 point to a molecular mechanism for enhanced production of cytokines and inflammatory response observed in COVID-19 patients. We demonstrate that in the mouse NLRP12 protein, one of the recognition site is not cleaved in our in-vitro assay. We pushed this comparative alignment of IRF-3 and NLRP12 homologs and show that the lack or presence of cognate cleavage motifs in IRF-3 and NLRP12 could contribute to the presentation of disease in cats and tigers, for example. Our findings provide an explanatory framework for indepth studies into the pathophysiology of COVID-19.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteasas 3C de Coronavirus/metabolismo , Proteasas Similares a la Papaína de Coronavirus/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Secuencia de Aminoácidos , Animales , COVID-19/patología , Línea Celular , Quirópteros/virología , Proteasas 3C de Coronavirus/genética , Proteasas Similares a la Papaína de Coronavirus/genética , Células HEK293 , Humanos , Ratones , SARS-CoV-2/enzimología , SARS-CoV-2/genética
6.
Int J Mol Sci ; 21(7)2020 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-32225120

RESUMEN

In the post-genome era, pathologies become associated with specific gene expression profiles and defined molecular lesions can be identified. The traditional therapeutic strategy is to block the identified aberrant biochemical activity. However, an attractive alternative could aim at antagonizing key transcriptional events underlying the pathogenesis, thereby blocking the consequences of a disorder, irrespective of the original biochemical nature. This approach, called transcription therapy, is now rendered possible by major advances in biophysical technologies. In the last two decades, techniques have evolved to become key components of drug discovery platforms, within pharmaceutical companies as well as academic laboratories. This review outlines the current biophysical strategies for transcription manipulation and provides examples of successful applications. It also provides insights into the future development of biophysical methods in drug discovery and personalized medicine.


Asunto(s)
Microscopía por Crioelectrón/métodos , Descubrimiento de Drogas/métodos , Terapia Molecular Dirigida/métodos , Imagen Individual de Molécula/métodos , Factores de Transcripción/antagonistas & inhibidores , Animales , Humanos
7.
Elife ; 82019 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-31358114

RESUMEN

Propranolol is an approved non-selective ß-adrenergic blocker that is first line therapy for infantile hemangioma. Despite the clinical benefit of propranolol therapy in hemangioma, the mechanistic understanding of what drives this outcome is limited. Here, we report successful treatment of pericardial edema with propranolol in a patient with Hypotrichosis-Lymphedema-Telangiectasia and Renal (HLTRS) syndrome, caused by a mutation in SOX18. Using a mouse pre-clinical model of HLTRS, we show that propranolol treatment rescues its corneal neo-vascularisation phenotype. Dissection of the molecular mechanism identified the R(+)-propranolol enantiomer as a small molecule inhibitor of the SOX18 transcription factor, independent of any anti-adrenergic effect. Lastly, in a patient-derived in vitro model of infantile hemangioma and pre-clinical model of HLTRS we demonstrate the therapeutic potential of the R(+) enantiomer. Our work emphasizes the importance of SOX18 etiological role in vascular neoplasms, and suggests R(+)-propranolol repurposing to numerous indications ranging from vascular diseases to metastatic cancer.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Inhibidores Enzimáticos/farmacología , Hemangioma/tratamiento farmacológico , Hipotricosis/tratamiento farmacológico , Linfedema/tratamiento farmacológico , Propranolol/farmacología , Factores de Transcripción SOXF/antagonistas & inhibidores , Telangiectasia/tratamiento farmacológico , Antagonistas Adrenérgicos beta/administración & dosificación , Animales , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/administración & dosificación , Humanos , Ratones , Modelos Teóricos , Propranolol/administración & dosificación
8.
Nucleic Acids Res ; 46(21): 11381-11395, 2018 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-30335167

RESUMEN

During embryogenesis, vascular development relies on a handful of transcription factors that instruct cell fate in a distinct sub-population of the endothelium (1). The SOXF proteins that comprise SOX7, 17 and 18, are molecular switches modulating arterio-venous and lymphatic endothelial differentiation (2,3). Here, we show that, in the SOX-F family, only SOX18 has the ability to switch between a monomeric and a dimeric form. We characterized the SOX18 dimer in binding assays in vitro, and using a split-GFP reporter assay in a zebrafish model system in vivo. We show that SOX18 dimerization is driven by a novel motif located in the vicinity of the C-terminus of the DNA binding region. Insertion of this motif in a SOX7 monomer forced its assembly into a dimer. Genome-wide analysis of SOX18 binding locations on the chromatin revealed enrichment for a SOX dimer binding motif, correlating with genes with a strong endothelial signature. Using a SOX18 small molecule inhibitor that disrupts dimerization, we revealed that dimerization is important for transcription. Overall, we show that dimerization is a specific feature of SOX18 that enables the recruitment of key endothelial transcription factors, and refines the selectivity of the binding to discrete genomic locations assigned to endothelial specific genes.


Asunto(s)
Factores de Transcripción SOXF/química , Secuencias de Aminoácidos , Animales , Técnicas Biosensibles , Proteínas de Unión al ADN/química , Células Endoteliales/metabolismo , Endotelio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/química , Humanos , Ratones , Mutación , Sistemas de Lectura Abierta , Dominios Proteicos , Multimerización de Proteína , Pez Cebra , Proteínas de Pez Cebra/química
9.
MAbs ; 10(4): 596-606, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29648920

RESUMEN

Antibodies are routinely used to study the activity of transcription factors, using various in vitro and in vivo approaches such as electrophoretic mobility shift assay, enzyme-linked immunosorbent assay, genome-wide method analysis coupled with next generation sequencing, or mass spectrometry. More recently, a new application for antibodies has emerged as crystallisation scaffolds for difficult to crystallise proteins, such as transcription factors. Only in a few rare cases, antibodies have been used to modulate the activity of transcription factors, and there is a real gap in our knowledge on how to efficiently design antibodies to interfere with transcription. The molecular function of transcription factors is underpinned by complex networks of protein-protein interaction and in theory, setting aside intra-cellular delivery challenges, developing antibody-based approaches to modulate transcription factor activity appears a viable option. Here, we demonstrate that antibodies or an antibody single-chain variable region fragments are powerful molecular tools to unravel complex protein-DNA and protein-protein binding mechanisms. In this study, we focus on the molecular mode of action of the transcription factor SOX18, a key modulator of endothelial cell fate during development, as well as an attractive target in certain pathophysiological conditions such as solid cancer metastasis. The engineered antibody we designed inhibits SOX18 transcriptional activity, by interfering specifically with an 8-amino-acid motif in the C-terminal region directly adjacent to α-Helix 3 of SOX18 HMG domain, thereby disrupting protein-protein interaction. This new approach establishes a framework to guide the study of transcription factors interactomes using antibodies as molecular handles.


Asunto(s)
Factores de Transcripción SOXF/análisis , Factores de Transcripción SOXF/química , Anticuerpos de Cadena Única , Humanos
10.
Int J Mol Sci ; 18(12)2017 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-29232933

RESUMEN

The use of fluorescently-tagged proteins in microscopy has become routine, and anti-GFP (Green fluorescent protein) affinity matrices are increasingly used in proteomics protocols. However, some protein-protein interactions assays, such as protein complementation assays (PCA), require recloning of each protein as a fusion with the different parts of the complementation system. Here we describe a generic system where the complementation is separated from the proteins and can be directly used with fluorescently-tagged proteins. By using nanobodies and performing tests in cell-free expression systems, we accelerated the development of multiple reporters, detecting heterodimers and homodimers or oligomers tagged with GFP or mCherry. We demonstrate that the system can detect interactions at a broad range of concentrations, from low nanomolar up to micromolar.


Asunto(s)
Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Luciferasas/genética , Proteínas Luminiscentes/metabolismo , Sistema Libre de Células/metabolismo , Ingeniería Genética , Luciferasas/metabolismo , Microscopía Fluorescente , Mapas de Interacción de Proteínas , Proteómica , Proteína Fluorescente Roja
11.
Cell Chem Biol ; 24(3): 346-359, 2017 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-28163017

RESUMEN

Pharmacological modulation of transcription factors (TFs) has only met little success over the past four decades. This is mostly due to standard drug discovery approaches centered on blocking protein/DNA binding or interfering with post-translational modifications. Recent advances in the field of TF biology have revealed a central role of protein-protein interaction in their mode of action. In an attempt to modulate the activity of SOX18 TF, a known regulator of vascular growth in development and disease, we screened a marine extract library for potential small-molecule inhibitors. We identified two compounds, which inspired a series of synthetic SOX18 inhibitors, able to interfere with the SOX18 HMG DNA-binding domain, and to disrupt HMG-dependent protein-protein interaction with RBPJ. These compounds also perturbed SOX18 transcriptional activity in a cell-based reporter gene system. This approach may prove useful in developing a new class of anti-angiogenic compounds based on the inhibition of TF activity.


Asunto(s)
Factores de Transcripción SOXF/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/química , Animales , Sitios de Unión , Productos Biológicos/química , Productos Biológicos/metabolismo , Productos Biológicos/farmacología , Células COS , Chlorocebus aethiops , ADN/química , ADN/metabolismo , Diseño de Fármacos , Genes Reporteros , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/química , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Concentración 50 Inhibidora , Ratones , Conformación de Ácido Nucleico , Unión Proteica , Mapas de Interacción de Proteínas , Estructura Terciaria de Proteína , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo , Ácido Salicílico/química , Ácido Salicílico/metabolismo , Ácido Salicílico/farmacología , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad , Activación Transcripcional/efectos de los fármacos
12.
Elife ; 62017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28137359

RESUMEN

Pharmacological targeting of transcription factors holds great promise for the development of new therapeutics, but strategies based on blockade of DNA binding, nuclear shuttling, or individual protein partner recruitment have yielded limited success to date. Transcription factors typically engage in complex interaction networks, likely masking the effects of specifically inhibiting single protein-protein interactions. Here, we used a combination of genomic, proteomic and biophysical methods to discover a suite of protein-protein interactions involving the SOX18 transcription factor, a known regulator of vascular development and disease. We describe a small-molecule that is able to disrupt a discrete subset of SOX18-dependent interactions. This compound selectively suppressed SOX18 transcriptional outputs in vitro and interfered with vascular development in zebrafish larvae. In a mouse pre-clinical model of breast cancer, treatment with this inhibitor significantly improved survival by reducing tumour vascular density and metastatic spread. Our studies validate an interactome-based molecular strategy to interfere with transcription factor activity, for the development of novel disease therapeutics.


Asunto(s)
Antineoplásicos/metabolismo , Neoplasias de la Mama/prevención & control , Factores de Transcripción SOXF/antagonistas & inhibidores , Transcripción Genética/efectos de los fármacos , Animales , Fenómenos Biofísicos , Vasos Sanguíneos/embriología , Modelos Animales de Enfermedad , Genómica , Ratones , Proteómica , Resultado del Tratamiento , Pez Cebra/embriología , Proteínas de Pez Cebra/antagonistas & inhibidores
13.
Mol Cell Proteomics ; 13(9): 2233-45, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24866125

RESUMEN

Protein dimerization and oligomerization is commonly used by nature to increase the structural and functional complexity of proteins. Regulated protein assembly is essential to transfer information in signaling, transcriptional, and membrane trafficking events. Here we show that a combination of cell-free protein expression, a proximity based interaction assay (AlphaScreen), and single-molecule fluorescence allow rapid mapping of homo- and hetero-oligomerization of proteins. We have applied this approach to the family of BAR domain-containing sorting nexin (SNX-BAR) proteins, which are essential regulators of membrane trafficking and remodeling in all eukaryotes. Dimerization of BAR domains is essential for creating a concave structure capable of sensing and inducing membrane curvature. We have systematically mapped 144 pairwise interactions between the human SNX-BAR proteins and generated an interaction matrix of preferred dimerization partners for each family member. We find that while nine SNX-BAR proteins are able to form homo-dimers, several including the retromer-associated SNX1, SNX2, and SNX5 require heteromeric interactions for dimerization. SNX2, SNX4, SNX6, and SNX8 show a promiscuous ability to bind other SNX-BAR proteins and we also observe a novel interaction with the SNX3 protein which lacks the BAR domain structure.


Asunto(s)
Nexinas de Clasificación/metabolismo , Dimerización , Humanos , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Espectrometría de Fluorescencia/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...