Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
PLoS Comput Biol ; 14(7): e1006279, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-30024886

RESUMO

Cell autonomous cancer dependencies are now routinely identified using CRISPR loss-of-function viability screens. However, a bias exists that makes it difficult to assess the true essentiality of genes located in amplicons, since the entire amplified region can exhibit lethal scores. These false-positive hits can either be discarded from further analysis, which in cancer models can represent a significant number of hits, or methods can be developed to rescue the true-positives within amplified regions. We propose two methods to rescue true positive hits in amplified regions by correcting for this copy number artefact. The Local Drop Out (LDO) method uses the relative lethality scores within genomic regions to assess true essentiality and does not require additional orthogonal data (e.g. copy number value). LDO is meant to be used in screens covering a dense region of the genome (e.g. a whole chromosome or the whole genome). The General Additive Model (GAM) method models the screening data as a function of the known copy number values and removes the systematic effect from the measured lethality. GAM does not require the same density as LDO, but does require prior knowledge of the copy number values. Both methods have been developed with single sample experiments in mind so that the correction can be applied even in smaller screens. Here we demonstrate the efficacy of both methods at removing the copy number effect and rescuing hits from some of the amplified regions. We estimate a 70-80% decrease of false positive hits with either method in regions of high copy number compared to no correction.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Variações do Número de Cópias de DNA/genética , Neoplasias/genética , Artefatos , Astrocitoma/genética , Astrocitoma/patologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Conjuntos de Dados como Assunto , Reações Falso-Positivas , Genômica , Humanos , Modelos Teóricos , Neoplasias/patologia , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia
2.
Bioorg Med Chem Lett ; 29(16): 2316-2319, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31235263

RESUMO

The YAP-TEAD protein-protein interaction is a potential therapeutic target to treat cancers in which the Hippo signaling pathway is deregulated. However, the extremely large surface of interaction between the two proteins presents a formidable challenge for a small molecule interaction disrupter approach. We have accomplished progress towards showing the feasibility of this approach by the identification of a 15-mer peptide able to potently (nanomolar range) disrupt the YAP-TEAD interaction by targeting only one of the two important sites of interaction. This peptide, incorporating non-natural amino acids selected by structure-based design, is derived from the Ω-loop sequence 85-99 of YAP.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Desenho de Fármacos , Peptídeos/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Fatores de Transcrição/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/química , Relação Dose-Resposta a Droga , Humanos , Estrutura Molecular , Peptídeos/síntese química , Peptídeos/química , Ligação Proteica/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade , Fatores de Transcrição/química , Proteínas de Sinalização YAP
3.
PLoS Genet ; 11(8): e1005465, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26295846

RESUMO

YAP1 is a major effector of the Hippo pathway and a well-established oncogene. Elevated YAP1 activity due to mutations in Hippo pathway components or YAP1 amplification is observed in several types of human cancers. Here we investigated its genomic binding landscape in YAP1-activated cancer cells, as well as in non-transformed cells. We demonstrate that TEAD transcription factors mediate YAP1 chromatin-binding genome-wide, further explaining their dominant role as primary mediators of YAP1-transcriptional activity. Moreover, we show that YAP1 largely exerts its transcriptional control via distal enhancers that are marked by H3K27 acetylation and that YAP1 is necessary for this chromatin mark at bound enhancers and the activity of the associated genes. This work establishes YAP1-mediated transcriptional regulation at distal enhancers and provides an expanded set of target genes resulting in a fundamental source to study YAP1 function in a normal and cancer setting.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/fisiologia , Fatores de Transcrição/metabolismo , Acetilação , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Sequência Consenso , Elementos Facilitadores Genéticos , Histonas/metabolismo , Humanos , Ligação Proteica , Processamento de Proteína Pós-Traducional , Fatores de Transcrição de Domínio TEA , Ativação Transcricional , Transcriptoma , Proteínas de Sinalização YAP
4.
Hepatology ; 62(5): 1497-510, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26173433

RESUMO

UNLABELLED: The Yes-associated protein (YAP)/Hippo pathway has been implicated in tissue development, regeneration, and tumorigenesis. However, its role in cholangiocarcinoma (CC) is not established. We show that YAP activation is a common feature in CC patient biopsies and human CC cell lines. Using microarray expression profiling of CC cells with overexpressed or down-regulated YAP, we show that YAP regulates genes involved in proliferation, apoptosis, and angiogenesis. YAP activity promotes CC growth in vitro and in vivo by functionally interacting with TEAD transcription factors (TEADs). YAP activity together with TEADs prevents apoptosis induced by cytotoxic drugs, whereas YAP knockdown sensitizes CC cells to drug-induced apoptosis. We further show that the proangiogenic microfibrillar-associated protein 5 (MFAP5) is a direct transcriptional target of YAP/TEAD in CC cells and that secreted MFAP5 promotes tube formation of human microvascular endothelial cells. High YAP activity in human CC xenografts and clinical samples correlates with increased MFAP5 expression and CD31(+) vasculature. CONCLUSIONS: These findings establish YAP as a key regulator of proliferation and antiapoptotic mechanisms in CC and provide first evidence that YAP promotes angiogenesis by regulating the expression of secreted proangiogenic proteins.


Assuntos
Neoplasias dos Ductos Biliares/patologia , Ductos Biliares Intra-Hepáticos , Colangiocarcinoma/patologia , Proteínas de Ligação a DNA/fisiologia , Resistencia a Medicamentos Antineoplásicos , Neovascularização Patológica/etiologia , Proteínas Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Animais , Apoptose , Neoplasias dos Ductos Biliares/irrigação sanguínea , Neoplasias dos Ductos Biliares/tratamento farmacológico , Proteínas de Ciclo Celular , Proliferação de Células , Colangiocarcinoma/irrigação sanguínea , Colangiocarcinoma/tratamento farmacológico , Proteínas Contráteis/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Glicoproteínas/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Oncogenes , Fatores de Transcrição de Domínio TEA
5.
Chembiochem ; 15(4): 537-42, 2014 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-24504694

RESUMO

The Hippo signaling pathway, which controls organ size in animals, is altered in various human cancers. The TEAD transcription factors, the most downstream elements in this pathway, are regulated by different cofactors, such as the Vgll (vestigial-like) proteins. Having studied the interaction between Vgll1-derived peptides and human TEAD4, we show that, although it lacks a key secondary structure element required for tight binding by two other TEAD cofactors (YAP and TAZ), Vgll1-derived peptides bind to TEAD with nanomolar affinity. We identify a ß-strand:loop:α-helix motif as the minimal Vgll binding site. Finally, we reveal an unexpected difference between mouse and human Vgll1-derived peptides.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas Musculares/química , Fatores de Transcrição/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Fatores de Transcrição de Domínio TEA , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
6.
Nat Cancer ; 5(7): 1102-1120, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38565920

RESUMO

The YAP-TEAD protein-protein interaction mediates YAP oncogenic functions downstream of the Hippo pathway. To date, available YAP-TEAD pharmacologic agents bind into the lipid pocket of TEAD, targeting the interaction indirectly via allosteric changes. However, the consequences of a direct pharmacological disruption of the interface between YAP and TEADs remain largely unexplored. Here, we present IAG933 and its analogs as potent first-in-class and selective disruptors of the YAP-TEAD protein-protein interaction with suitable properties to enter clinical trials. Pharmacologic abrogation of the interaction with all four TEAD paralogs resulted in YAP eviction from chromatin and reduced Hippo-mediated transcription and induction of cell death. In vivo, deep tumor regression was observed in Hippo-driven mesothelioma xenografts at tolerated doses in animal models as well as in Hippo-altered cancer models outside mesothelioma. Importantly this also extended to larger tumor indications, such as lung, pancreatic and colorectal cancer, in combination with RTK, KRAS-mutant selective and MAPK inhibitors, leading to more efficacious and durable responses. Clinical evaluation of IAG933 is underway.


Assuntos
Via de Sinalização Hippo , Proteínas Serina-Treonina Quinases , Fatores de Transcrição , Ensaios Antitumorais Modelo de Xenoenxerto , Humanos , Animais , Fatores de Transcrição/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Camundongos , Linhagem Celular Tumoral , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Sinalização YAP/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição de Domínio TEA , Proteínas ras/metabolismo , Feminino , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico
7.
Chembiochem ; 14(10): 1218-25, 2013 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-23780915

RESUMO

The Hippo pathway controls cell homeostasis, and its deregulation can lead to human diseases. In this pathway, the YAP and TAZ transcriptional cofactors play a key role in stimulating gene transcription through their interaction with the TEAD transcriptional factors. Our study of YAP and TAZ peptides in biochemical and biophysical assays shows that both proteins have essentially the same affinity for TEAD. Molecular modeling and structural biology data suggest that they also bind to the same site on TEAD. However, this apparent similarity hides differences in the ways in which the two proteins interact with TEAD. The secondary structure elements of their TEAD binding site do not contribute equally to the overall affinity, and critical interactions with TEAD are made through different residues. This convergent optimization of the YAP/TAZ TEAD binding site suggests that the similarity in the affinities of binding of YAP to TEAD and of TAZ to TEAD is important for Hippo pathway functionality.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas Musculares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição/metabolismo , Aciltransferases , Sequência de Aminoácidos , Animais , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Via de Sinalização Hippo , Humanos , Imuno-Histoquímica , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Musculares/química , Proteínas Musculares/genética , Mutagênese Sítio-Dirigida , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Domínios e Motivos de Interação entre Proteínas , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Fatores de Transcrição de Domínio TEA , Fatores de Transcrição/química , Fatores de Transcrição/genética
8.
Nat Commun ; 14(1): 3907, 2023 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-37400441

RESUMO

YAP is a key transcriptional co-activator of TEADs, it regulates cell growth and is frequently activated in cancer. In Malignant Pleural Mesothelioma (MPM), YAP is activated by loss-of-function mutations in upstream components of the Hippo pathway, while, in Uveal Melanoma (UM), YAP is activated in a Hippo-independent manner. To date, it is unclear if and how the different oncogenic lesions activating YAP impact its oncogenic program, which is particularly relevant for designing selective anti-cancer therapies. Here we show that, despite YAP being essential in both MPM and UM, its interaction with TEAD is unexpectedly dispensable in UM, limiting the applicability of TEAD inhibitors in this cancer type. Systematic functional interrogation of YAP regulatory elements in both cancer types reveals convergent regulation of broad oncogenic drivers in both MPM and UM, but also strikingly selective programs. Our work reveals unanticipated lineage-specific features of the YAP regulatory network that provide important insights to guide the design of tailored therapeutic strategies to inhibit YAP signaling across different cancer types.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Neoplasias , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Sinalização YAP , Epigenômica , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transdução de Sinais/genética
9.
ChemMedChem ; 18(11): e202300051, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-36988034

RESUMO

The inhibition of the YAP-TEAD protein-protein interaction constitutes a promising therapeutic approach for the treatment of cancers linked to the dysregulation of the Hippo signaling pathway. The identification of a class of small molecules which potently inhibit the YAP-TEAD interaction by binding tightly to the Ω-loop pocket of TEAD has previously been communicated. This report details the further multi-parameter optimization of this class of compounds resulting in advanced analogs combining nanomolar cellular potency with a balanced ADME and off-target profile, and efficacy of these compounds in tumor bearing mice is demonstrated for the first time.


Assuntos
Neoplasias , Fatores de Transcrição , Animais , Camundongos , Fatores de Transcrição/metabolismo , Proteínas de Sinalização YAP
10.
Dev Cell ; 12(2): 221-34, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17276340

RESUMO

The adult, virgin mammary gland is a highly organized tree-like structure formed by ducts with hollowed lumen. Although lumen formation during pubertal development appears to involve apoptosis, the molecular mechanisms that regulate this process are not known. Here, we demonstrate that disruption of the BH3-only proapoptotic factor Bim in mice prevents induction of apoptosis in and clearing of the lumen in terminal end buds during puberty. However, cells that fill the presumptive luminal space are eventually cleared from the adjacent ducts by a caspase-independent death process. Within the filled Bim(-/-) ducts, epithelial cells are deprived of matrix attachment and undergo squamous differentiation prior to clearing. Similarly, we also detect squamous differentiation in vitro when immortalized mammary epithelial cells are detached from the matrix. These data provide important mechanistic information on the processes involved in sculpting the mammary gland and demonstrate that BIM is a critical regulator of apoptosis in vivo.


Assuntos
Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/metabolismo , Apoptose , Glândulas Mamárias Animais/embriologia , Proteínas de Membrana/deficiência , Proteínas de Membrana/metabolismo , Morfogênese , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/metabolismo , Animais , Anoikis , Proteína 11 Semelhante a Bcl-2 , Caspases/metabolismo , Diferenciação Celular , Proliferação de Células , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Feminino , Humanos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Puberdade/fisiologia , Fatores de Tempo
11.
ChemMedChem ; 17(19): e202200303, 2022 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-35950546

RESUMO

Inhibition of the YAP-TEAD protein-protein interaction is an attractive therapeutic concept under intense investigation with the objective to treat cancers associated with a dysregulation of the Hippo pathway. However, owing to the very extended surface of interaction of the two proteins, the identification of small drug-like molecules able to efficiently prevent YAP from binding to TEAD by direct competition has been elusive so far. We disclose here the discovery of the first class of small molecules potently inhibiting the YAP-TEAD interaction by binding at one of the main interaction sites of YAP at the surface of TEAD. These inhibitors, providing a path forward to pharmacological intervention in the Hippo pathway, evolved from a weakly active virtual screening hit advanced to high potency by structure-based design.


Assuntos
Neoplasias , Fatores de Transcrição , Proteínas Adaptadoras de Transdução de Sinal/química , Humanos , Fatores de Transcrição/metabolismo , Proteínas de Sinalização YAP
12.
Nat Commun ; 12(1): 2442, 2021 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-33903593

RESUMO

The transcription factor PAX8 is critical for the development of the thyroid and urogenital system. Comprehensive genomic screens furthermore indicate an additional oncogenic role for PAX8 in renal and ovarian cancers. While a plethora of PAX8-regulated genes in different contexts have been proposed, we still lack a mechanistic understanding of how PAX8 engages molecular complexes to drive disease-relevant oncogenic transcriptional programs. Here we show that protein isoforms originating from the MECOM locus form a complex with PAX8. These include MDS1-EVI1 (also called PRDM3) for which we map its interaction with PAX8 in vitro and in vivo. We show that PAX8 binds a large number of genomic sites and forms transcriptional hubs. At a subset of these, PAX8 together with PRDM3 regulates a specific gene expression module involved in adhesion and extracellular matrix. This gene module correlates with PAX8 and MECOM expression in large scale profiling of cell lines, patient-derived xenografts (PDXs) and clinical cases and stratifies gynecological cancer cases with worse prognosis. PRDM3 is amplified in ovarian cancers and we show that the MECOM locus and PAX8 sustain in vivo tumor growth, further supporting that the identified function of the MECOM locus underlies PAX8-driven oncogenic functions in ovarian cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteína do Locus do Complexo MDS1 e EVI1/genética , Neoplasias Ovarianas/genética , Fator de Transcrição PAX8/genética , Animais , Linhagem Celular Tumoral , Feminino , Células HEK293 , Humanos , Proteína do Locus do Complexo MDS1 e EVI1/metabolismo , Camundongos Nus , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Fator de Transcrição PAX8/metabolismo , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Carga Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
13.
Protein Sci ; 29(2): 509-520, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31697419

RESUMO

The Hippo pathway is a key signaling pathway in the control of organ size and development. The most distal elements of this pathway, the TEAD transcription factors, are regulated by several proteins, such as YAP (Yes-associated protein), TAZ (transcriptional co-activator with PDZ-binding motif) and VGLL1-4 (Vestigial-like members 1-4). In this article, combining structural data and motif searches in protein databases, we identify two new TEAD interactors: FAM181A and FAM181B. Our structural data show that they bind to TEAD via an Ω-loop as YAP/TAZ do, but only FAM181B possesses the LxxLF motif (x any amino acid) found in YAP/TAZ. The affinity of different FAM181A/B fragments for TEAD is in the low micromolar range and full-length FAM181A/B proteins interact with TEAD in cells. These findings, together with a recent report showing that FAM181A/B proteins have a role in nervous system development, suggest a potential new involvement of the TEAD transcription factors in the development of this tissue.


Assuntos
Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Bases de Dados de Proteínas , Células HEK293 , Humanos , Conformação Proteica , Fatores de Transcrição/genética
14.
Sci Rep ; 10(1): 17442, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33060790

RESUMO

The most downstream elements of the Hippo pathway, the TEAD transcription factors, are regulated by several cofactors, such as Vg/VGLL1-3. Earlier findings on human VGLL1 and here on human VGLL3 show that these proteins interact with TEAD via a conserved amino acid motif called the TONDU domain. Surprisingly, our studies reveal that the TEAD-binding domain of Drosophila Vg and of human VGLL2 is more complex and contains an additional structural element, an Ω-loop, that contributes to TEAD binding. To explain this unexpected structural difference between proteins from the same family, we propose that, after the genome-wide duplications at the origin of vertebrates, the Ω-loop present in an ancestral VGLL gene has been lost in some VGLL variants. These findings illustrate how structural and functional constraints can guide the evolution of transcriptional cofactors to preserve their ability to compete with other cofactors for binding to transcription factors.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas Musculares/química , Proteínas Nucleares/química , Fatores de Transcrição/química , Animais , Sítios de Ligação , Drosophila , Células HEK293 , Humanos , Concentração Inibidora 50 , Cinética , Modelos Moleculares , Mutação , Ligação Proteica , Domínios Proteicos , Fatores de Transcrição de Domínio TEA
15.
FEBS J ; 286(12): 2381-2398, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30903741

RESUMO

Four TEAD transcription factors (TEAD1-4) mediate the signalling output of the Hippo pathway that controls organ size in humans. TEAD transcriptional activity is regulated via interactions with the YAP, TAZ and VGLL proteins. A mutation in the TEAD1 gene, Tyr421His, has been identified in patients suffering from Sveinsson's chorioretinal atrophy (SCA), an autosomal dominant eye disease. This mutation prevents the YAP/TAZ:TEAD1 interaction. In this study, we measure the affinity of YAP, TAZ and VGLL1 for the four human TEADs and find that they have a similar affinity for all TEADs. We quantitate the effect of the mutation found in SCA patients and show that it destabilizes the YAP/TAZ:TEAD interaction by about 3 kcal·mol-1 . We determine the structure of YAP in complex with this mutant form of TEAD and show that the histidine residue adopts different conformations at the binding interface. The presence of this flexible residue induces the destabilization of several H-bonds and the loss of van der Waals contacts, which explains why the Tyr421HisTEAD1 mutation has such a large destabilizing effect on the formation of the YAP:TEAD complex. DATABASE: The crystallographic data have been deposited at the RSCB Protein Data Bank (PDB, www.pdb.org) with the access codes: 6HIL (wtYAP :Tyr421HisTEAD1 ), 6HIK (wtYAP :Tyr429HisTEAD4 ).


Assuntos
Proteínas de Ciclo Celular/genética , Distrofias Hereditárias da Córnea/genética , Proteínas de Ligação a DNA/química , Proteínas Nucleares/química , Mapas de Interação de Proteínas/genética , Degeneração Retiniana/genética , Fatores de Transcrição/química , Fatores de Transcrição/genética , Aciltransferases , Proteínas de Ciclo Celular/química , Distrofias Hereditárias da Córnea/patologia , Cristalografia por Raios X , Proteínas de Ligação a DNA/genética , Humanos , Proteínas Intrinsicamente Desordenadas/química , Proteínas Intrinsicamente Desordenadas/genética , Mutação/genética , Proteínas Nucleares/genética , Ligação Proteica/genética , Degeneração Retiniana/patologia , Transdução de Sinais/genética , Fatores de Transcrição de Domínio TEA
17.
Protein Sci ; 27(10): 1810-1820, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30058229

RESUMO

Many interactions between proteins are mediated by intrinsically disordered regions (IDRs). Intrinsically disordered proteins (IDPs) do not adopt a stable three-dimensional structure in their unbound form, but they become more structured upon binding to their partners. In this communication, we study how a bound IDR adapts to mutations, preventing the formation of hydrogen bonds at the binding interface that needs a precise positioning of the interacting residues to be formed. We use as a model the YAP:TEAD interface, where one YAP (IDP) and two TEAD residues form hydrogen bonds via their side chain. Our study shows that the conformational flexibility of bound YAP and the reorganization of water molecules at the interface help to reduce the energetic constraints created by the loss of H-bonds at the interface. The residual flexibility/dynamic of bound IDRs and water might, therefore, be a key for the adaptation of IDPs to different interface landscapes and to mutations occurring at binding interfaces.


Assuntos
Proteínas Intrinsicamente Desordenadas/química , Proteínas Intrinsicamente Desordenadas/genética , Mutação , Ligação Proteica , Conformação Proteica , Termodinâmica , Água/química
18.
Mol Cell Biol ; 22(5): 1329-39, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11839800

RESUMO

The PKC1-associated mitogen-activated protein (MAP) kinase pathway of Saccharomyces cerevisiae regulates cell integrity by controlling the actin cytoskeleton and cell wall synthesis. Activation of PKC1 occurs via the GTPase RHO1 and the kinase pair PKH1 and PKH2. Here we report that YPK1 and YPK2, an essential pair of homologous kinases and proposed downstream effectors of PKH and sphingolipids, are also regulators of the PKC1-controlled MAP kinase cascade. ypk mutants display random distribution of the actin cytoskeleton and severely reduced activation of the MAP kinase MPK1. Upregulation of the RHO1 GTPase switch or the PKC1 effector MAP kinase pathway suppresses the growth and actin defects of ypk cells. ypk lethality is also suppressed by overexpression of an uncharacterized gene termed TUS1. TUS1 is a novel RHO1 exchange factor that contributes to cell wall integrity-mediated modulation of RHO1 activity. Thus, TUS1 and the YPKs add to the growing complexity of RHO1 and PKC1 regulation in the cell integrity signaling pathway. Furthermore, our findings suggest that the YPKs are a missing link between sphingolipid signaling and the cell integrity pathway.


Assuntos
Parede Celular/fisiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Quinases/metabolismo , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo , Ativação Enzimática , Resposta ao Choque Térmico , Sistema de Sinalização das MAP Quinases , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína Quinase C/metabolismo , Saccharomyces cerevisiae/citologia
19.
Mol Cell Biol ; 24(1): 338-51, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14673167

RESUMO

The TOR (target of rapamycin) and RAS/cyclic AMP (cAMP) signaling pathways are the two major pathways controlling cell growth in response to nutrients in yeast. In this study we examine the functional interaction between TOR and the RAS/cAMP pathway. First, activation of the RAS/cAMP signaling pathway confers pronounced resistance to rapamycin. Second, constitutive activation of the RAS/cAMP pathway prevents several rapamycin-induced responses, such as the nuclear translocation of the transcription factor MSN2 and induction of stress genes, the accumulation of glycogen, the induction of autophagy, the down-regulation of ribosome biogenesis (ribosomal protein gene transcription and RNA polymerase I and III activity), and the down-regulation of the glucose transporter HXT1. Third, many of these TOR-mediated responses are independent of the previously described TOR effectors TAP42 and the type 2A-related protein phosphatase SIT4. Conversely, TOR-controlled TAP42/SIT4-dependent events are not affected by the RAS/cAMP pathway. Finally, and importantly, TOR controls the subcellular localization of both the protein kinase A catalytic subunit TPK1 and the RAS/cAMP signaling-related kinase YAK1. Our findings suggest that TOR signals through the RAS/cAMP pathway, independently of TAP42/SIT4. Therefore, the RAS/cAMP pathway may be a novel TOR effector branch.


Assuntos
AMP Cíclico/metabolismo , Proteínas Fúngicas , Fosfatidilinositol 3-Quinases/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas ras/metabolismo , Antifúngicos/farmacologia , Autofagia/efeitos dos fármacos , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA/metabolismo , Proteínas Facilitadoras de Transporte de Glucose , Glicogênio/metabolismo , Proteínas de Transporte de Monossacarídeos/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/deficiência , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Ribossomos/efeitos dos fármacos , Saccharomyces cerevisiae/metabolismo , Sirolimo/farmacologia , Fatores de Transcrição/metabolismo , Transcrição Gênica , Proteínas ras/efeitos dos fármacos
20.
Elife ; 62017 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-28430104

RESUMO

TEAD (TEA/ATTS domain) transcription factors are the most distal effectors of the Hippo pathway. YAP (Yes-associated protein) is a coactivator protein which, upon binding to TEAD proteins, stimulates their transcriptional activity. Since the Hippo pathway is deregulated in various cancers, designing inhibitors of the YAP:TEAD interaction is an attractive therapeutic strategy for oncology. Understanding the molecular events that take place at the YAP:TEAD interface is therefore important not only to devise drug discovery approaches, but also to gain knowledge on TEAD regulation. In this report, combining single site-directed mutagenesis and double mutant analyses, we conduct a detailed analysis on the role of several residues located at the YAP:TEAD interface. Our results provide quantitative understanding of the interactions taking place at the YAP:TEAD interface and give insights into the formation of the YAP:TEAD complex and more particularly on the interaction between TEAD and the Ω-loop found in YAP.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Musculares/metabolismo , Fosfoproteínas/metabolismo , Mapas de Interação de Proteínas , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Análise Mutacional de DNA , Proteínas de Ligação a DNA/genética , Proteínas Musculares/genética , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fosfoproteínas/genética , Ligação Proteica , Fatores de Transcrição de Domínio TEA , Fatores de Transcrição/genética , Proteínas de Sinalização YAP
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA