Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Elife ; 122024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38289221

RESUMO

Eph receptor tyrosine kinases participate in a variety of normal and pathogenic processes during development and throughout adulthood. This versatility is likely facilitated by the ability of Eph receptors to signal through diverse cellular signalling pathways: primarily by controlling cytoskeletal dynamics, but also by regulating cellular growth, proliferation, and survival. Despite many proteins linked to these signalling pathways interacting with Eph receptors, the specific mechanisms behind such links and their coordination remain to be elucidated. In a proteomics screen for novel EPHB2 multi-effector proteins, we identified human MYC binding protein 2 (MYCBP2 or PAM or Phr1). MYCBP2 is a large signalling hub involved in diverse processes such as neuronal connectivity, synaptic growth, cell division, neuronal survival, and protein ubiquitination. Our biochemical experiments demonstrate that the formation of a complex containing EPHB2 and MYCBP2 is facilitated by FBXO45, a protein known to select substrates for MYCBP2 ubiquitin ligase activity. Formation of the MYCBP2-EPHB2 complex does not require EPHB2 tyrosine kinase activity and is destabilised by binding of ephrin-B ligands, suggesting that the MYCBP2-EPHB2 association is a prelude to EPHB2 signalling. Paradoxically, the loss of MYCBP2 results in increased ubiquitination of EPHB2 and a decrease of its protein levels suggesting that MYCBP2 stabilises EPHB2. Commensurate with this effect, our cellular experiments reveal that MYCBP2 is essential for efficient EPHB2 signalling responses in cell lines and primary neurons. Finally, our genetic studies in Caenorhabditis elegans provide in vivo evidence that the ephrin receptor VAB-1 displays genetic interactions with known MYCBP2 binding proteins. Together, our results align with the similarity of neurodevelopmental phenotypes caused by MYCBP2 and EPHB2 loss of function, and couple EPHB2 to a signalling effector that controls diverse cellular functions.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas F-Box , Receptor EphB2 , Ubiquitina-Proteína Ligases , Animais , Humanos , Proteínas Adaptadoras de Transdução de Sinal/genética , Caenorhabditis elegans/genética , Receptor EphB2/genética , Transdução de Sinais , Ubiquitina , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
2.
bioRxiv ; 2023 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-37693478

RESUMO

Eph receptor tyrosine kinases participate in a variety of normal and pathogenic processes during development and throughout adulthood. This versatility is likely facilitated by the ability of Eph receptors to signal through diverse cellular signalling pathways: primarily by controlling cytoskeletal dynamics, but also by regulating cellular growth, proliferation, and survival. Despite many proteins linked to these signalling pathways interacting with Eph receptors, the specific mechanisms behind such links and their coordination remain to be elucidated. In a proteomics screen for novel EPHB2 multi-effector proteins, we identified human MYC binding protein 2 (MYCBP2 or PAM or Phr1). MYCBP2 is a large signalling hub involved in diverse processes such as neuronal connectivity, synaptic growth, cell division, neuronal survival, and protein ubiquitination. Our biochemical experiments demonstrate that the formation of a complex containing EPHB2 and MYCBP2 is facilitated by FBXO45, a protein known to select substrates for MYCBP2 ubiquitin ligase activity. Formation of the MYCBP2-EPHB2 complex does not require EPHB2 tyrosine kinase activity and is destabilised by binding of ephrin-B ligands, suggesting that the MYCBP2-EPHB2 association is a prelude to EPHB2 signalling. Paradoxically, the loss of MYCBP2 results in increased ubiquitination of EPHB2 and a decrease of its protein levels suggesting that MYCBP2 stabilises EPHB2. Commensurate with this effect, our cellular experiments reveal that MYCBP2 is essential for efficient EPHB2 signalling responses in cell lines and primary neurons. Finally, our genetic studies in C. elegans provide in vivo evidence that the ephrin receptor VAB-1 displays genetic interactions with known MYCBP2 binding proteins. Together, our results align with the similarity of neurodevelopmental phenotypes caused by MYCBP2 and EPHB2 loss of function, and couple EPHB2 to a signaling effector that controls diverse cellular functions.

3.
Bioorg Med Chem Lett ; 55: 128441, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34767912

RESUMO

The combination of androgen receptor antagonists with histone deacetylase inhibitors (HDACi) has been shown to be more effective than antiandrogens alone in halting growth of prostate cancer cell lines. Here we have designed, synthesized and assessed a series of antiandrogen/HDACi hybrids by combining structural features of enzalutamide with either SAHA or entinostat. The hybrids are demonstrated to maintain bifunctionality using a fluorometric HDAC assay and a bioluminescence resonance energy transfer (BRET) antiandrogen assay. Antiproliferative assays showed that hybrids bearing o-aminoanilide-based HDACi motifs outperformed hydroxamic acid based HDACi's. The hybrids demonstrated selectivity for epithelial cell lines vs. stromal cell lines, suggesting a potentially useful therapeutic window.


Assuntos
Antagonistas de Androgênios/farmacologia , Antineoplásicos/farmacologia , Benzamidas/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Nitrilas/farmacologia , Feniltioidantoína/farmacologia , Piridinas/farmacologia , Antagonistas de Androgênios/síntese química , Antagonistas de Androgênios/química , Antineoplásicos/síntese química , Antineoplásicos/química , Benzamidas/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Transferência Ressonante de Energia de Fluorescência , Fluorometria , Inibidores de Histona Desacetilases/síntese química , Inibidores de Histona Desacetilases/química , Humanos , Estrutura Molecular , Nitrilas/química , Feniltioidantoína/química , Piridinas/química , Relação Estrutura-Atividade , Células Tumorais Cultivadas
4.
Biochim Biophys Acta Mol Cell Res ; 1868(2): 118908, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33189785

RESUMO

Nur77 (NGFI-B) is a nuclear receptor that belongs to the Nr4a family of orphan nuclear receptors (Nr4a1). This transcription factor has been implicated in the regulation of multiple functions, such as cell cycle regulation, apoptosis, inflammation, glucose and lipid metabolism, and brain function. However, the mechanisms involved in its different regulatory properties remain unclear. In search for regulatory mechanisms of Nur77 function, we identified that Protein Inhibitor of Activated STAT gamma (PIASγ), an E3 SUMO-protein ligase, potently repressed Nur77 transcriptional activity in HEK-293T cells. This PIASγ activity was sensitive to Sentrin SUMO-specific protease 1 (SENP1). Substitution of two putative phylogenetically well-conserved small ubiquitin-like modifier (SUMO) acceptor sites, lysine 102 (K102) and 577 (K577) by arginine residues (R) modulated Nur77 transcriptional activity. In particular, Nur77-K102R and Nur77-K102R/K577R mutants strongly decreased the transcriptional activity of Nur77, whereas single K577R substitution increased transcriptional activity of Nur77. Repression of Nur77 transcriptional activity by SUMO2 and PIASγ was reduced by the K577R mutation, whereas the K102R mutant remained insensitive to SUMO2. Interestingly, the roles of these SUMO acceptor sites in Nur77 are distinct from previously observed activities on its close homolog Nurr1. Thus, the present study identified SUMO2 and PIASγ as important transcriptional co-regulators of Nur77.


Assuntos
Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Proteínas Inibidoras de STAT Ativados/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Transcrição Gênica/genética , Ativação Transcricional , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Plasmídeos/genética , Proteínas de Ligação a Poli-ADP-Ribose/genética , Proteínas Inibidoras de STAT Ativados/genética , Transdução de Sinais/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Transfecção
5.
Bioorg Med Chem ; 26(15): 4428-4440, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-30078609

RESUMO

Hybrid antiestrogen/histone deacetylase (HDAC) inhibitors were designed by appending zinc binding groups to the 4-hydroxystilbene core of 4-hydroxytamoxifen. The resulting hybrids were fully bifunctional, and displayed high nanomolar to low micromolar IC50 values against both the estrogen receptor α (ERα) and HDACs in vitro and in cell-based assays. The hybrids were antiproliferative against ER+ MCF-7 breast cancer cells, with hybrid 28b possessing an improved activity profile compared to either 4-hydroxytamoxifen or SAHA. Hybrid 28b displayed gene expression patterns that reflected both ERα and HDAC inhibition.


Assuntos
Desenho de Fármacos , Antagonistas de Estrogênios/química , Inibidores de Histona Desacetilases/química , Histona Desacetilases/metabolismo , Tamoxifeno/química , Sítios de Ligação , Proliferação de Células/efeitos dos fármacos , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/metabolismo , Histona Desacetilases/química , Humanos , Concentração Inibidora 50 , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Células MCF-7 , Simulação de Acoplamento Molecular , Estrutura Terciária de Proteína , Tamoxifeno/síntese química , Tamoxifeno/farmacologia , Zinco/química
6.
Proc Natl Acad Sci U S A ; 115(11): E2653-E2662, 2018 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-29487210

RESUMO

There is currently an unmet need for versatile techniques to monitor the assembly and dynamics of ternary complexes in live cells. Here we describe bioluminescence resonance energy transfer with fluorescence enhancement by combined transfer (BRETFect), a high-throughput technique that enables robust spectrometric detection of ternary protein complexes based on increased energy transfer from a luciferase to a fluorescent acceptor in the presence of a fluorescent intermediate. Its unique donor-intermediate-acceptor relay system is designed so that the acceptor can receive energy either directly from the donor or indirectly via the intermediate in a combined transfer, taking advantage of the entire luciferase emission spectrum. BRETFect was used to study the ligand-dependent cofactor interaction properties of the estrogen receptors ERα and ERß, which form homo- or heterodimers whose distinctive regulatory properties are difficult to dissect using traditional methods. BRETFect uncovered the relative capacities of hetero- vs. homodimers to recruit receptor-specific cofactors and regulatory proteins, and to interact with common cofactors in the presence of receptor-specific ligands. BRETFect was also used to follow the assembly of ternary complexes between the V2R vasopressin receptor and two different intracellular effectors, illustrating its use for dissection of ternary protein-protein interactions engaged by G protein-coupled receptors. Our results indicate that BRETFect represents a powerful and versatile technique to monitor the dynamics of ternary interactions within multimeric complexes in live cells.


Assuntos
Técnicas Citológicas/métodos , Transferência Ressonante de Energia de Fluorescência/métodos , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Fatores de Complexo Ternário/metabolismo , Células HEK293 , Humanos , Proteínas Luminescentes , Receptores Citoplasmáticos e Nucleares/análise , Receptores Citoplasmáticos e Nucleares/química , Receptores Acoplados a Proteínas G/análise , Receptores Acoplados a Proteínas G/química , Fatores de Complexo Ternário/análise , Fatores de Complexo Ternário/química
7.
Bioorg Med Chem ; 23(24): 7597-606, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26613635

RESUMO

The combination of antiestrogens and histone deacetylase inhibitors (HDACi) has been found to be antiproliferative in breast cancer models. We designed and synthesized hybrid structures which combined structural features of the pure antiestrogen ICI-164,384 and HDACi's SAHA and entinostat in a single bifunctional molecule. The hybrids retained antiestrogenic and HDACi activity and, in the case of benzamide hybrids, were selective for Class I HDAC3 over Class II HDAC6. The hybrids possessed low micromolar to high nanomolar activity against both ER+ MCF-7 and ER- MDA-MB-231 breast cancer cell models.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Moduladores de Receptor Estrogênico/química , Moduladores de Receptor Estrogênico/farmacologia , Inibidores de Histona Desacetilases/química , Inibidores de Histona Desacetilases/farmacologia , Antineoplásicos/síntese química , Benzamidas/síntese química , Benzamidas/química , Benzamidas/farmacologia , Mama/efeitos dos fármacos , Mama/metabolismo , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Estradiol/análogos & derivados , Estradiol/síntese química , Estradiol/química , Estradiol/farmacologia , Moduladores de Receptor Estrogênico/síntese química , Feminino , Inibidores de Histona Desacetilases/síntese química , Histona Desacetilases/metabolismo , Humanos , Simulação de Acoplamento Molecular , Alcamidas Poli-Insaturadas/síntese química , Alcamidas Poli-Insaturadas/química , Alcamidas Poli-Insaturadas/farmacologia , Piridinas/síntese química , Piridinas/química , Piridinas/farmacologia
8.
FASEB J ; 29(10): 4256-67, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26148973

RESUMO

Retinoid X receptors (RXRs) play a role as master regulators because of their capacity to form heterodimers with other nuclear receptors (NRs). Accordingly, retinoid signaling is involved in multiple biologic processes, including development, cell differentiation, metabolism, and cell death. However, the role and function of RXRs in different heterodimer complexes remain unidentified, mainly because most RXR drugs (called rexinoids) are not selective of specific heterodimer complexes. The lack of selectivity strongly limits the use of rexinoids for specific therapeutic approaches. To better characterize rexinoids at specific NR complexes, we have developed and optimized luciferase (Luc) protein complementation(PCA)-based bioluminescence resonance energy transfer (BRET) assays that can directly measure recruitment of a coactivator (CoA) motif fused to yellow fluorescent protein (YFP) by specific NR dimers. To validate the assays, we compared rexinoid modulation of CoA recruitment by the RXR homodimer and by the heterodimers Nur77/RXR and Nurr1/RXR. Results revealed that some rexinoids display selective CoA recruitment activities with homo- or heterodimer complexes. In particular, SR11237 (BMS649) has stronger potency for recruitment of CoA motif and transcriptional activity with the heterodimer Nur77/RXR than other complexes. This technology should be useful in identifying new compounds with specificity for individual dimeric species formed by NRs.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Receptor X Retinoide gama/metabolismo , Alitretinoína , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Benzoatos/farmacologia , Células HEK293 , Humanos , Ligantes , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microscopia de Fluorescência , Modelos Biológicos , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Coativadores de Receptor Nuclear/genética , Coativadores de Receptor Nuclear/metabolismo , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/química , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/química , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Ligação Proteica/efeitos dos fármacos , Multimerização Proteica , Receptor X Retinoide gama/química , Receptor X Retinoide gama/genética , Retinoides/farmacologia , Tretinoína/farmacologia
9.
Mol Cell Endocrinol ; 334(1-2): 76-82, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21056084

RESUMO

Nuclear receptors (NRs) are ligand-dependent transcription factors with important roles in normal development and physiology and in a wide array of pathologies. While identification of natural or synthetic ligands for all human NRs has clarified their physiological roles and led to numerous therapeutic applications, much remains to be understood about the mechanisms by which NRs control transcription of specific networks of target genes. The DNA binding domain, composed of two C4 type zinc fingers, is the most conserved region in NRs. Binding motif selectivity stems from variations in dimerization interfaces in the DNA and ligand binding domains as well as from divergence in a few base-contacting residues in the first zinc finger. However, overlaps in DNA binding patterns suggest competition between different NRs at target sites in vivo, resulting in cross-regulation of subsets of target gene networks. As NRs can regulate transcription even when bound far from the transcription initiation sites of target genes, the considerable expansion in intergenic DNA in genomes throughout evolution likely contributes to the diversity and complexity of tissue-specific gene regulation by NRs. In particular, the presence of NR binding sites in the primate-specific Alu family of short-interspersed elements has been shown to confer transcriptional regulation by NRs to adjacent genes, although the impact on NR regulatory networks at the genome-wide scale remains to be more fully evaluated.


Assuntos
Proteínas de Ligação a DNA/genética , Evolução Molecular , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , DNA/metabolismo , DNA Intergênico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Variação Genética , Genoma , Humanos , Ligantes , Dados de Sequência Molecular , Especificidade de Órgãos , Ligação Proteica/genética , Estrutura Terciária de Proteína , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Dedos de Zinco/genética
10.
Mol Endocrinol ; 21(4): 797-816, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17299137

RESUMO

The basis for the differential repressive effects of antiestrogens on transactivation by estrogen receptor-alpha (ERalpha) remains incompletely understood. Here, we show that the full antiestrogen ICI182,780 and, to a lesser extent, the selective ER modulator raloxifene (Ral), induce accumulation of exogenous ERalpha in a poorly soluble fraction in transiently transfected HepG2 or stably transfected MDA-MB231 cells and of endogenous receptor in MCF7 cells. ERalpha remained nuclear in HepG2 cells treated with either compound. Replacement of selected hydrophobic residues of ERalpha ligand-binding domain helix 12 (H12) enhanced receptor solubility in the presence of ICI182,780 or Ral. These mutations also increased transcriptional activity with Ral or ICI182,780 on reporter genes or on the endogenous estrogen target gene TFF1 in a manner requiring the integrity of the N-terminal AF-1 domain. The antiestrogen-specific effects of single mutations suggest that they affect receptor function by mechanisms other than a simple decrease in hydrophobicity of H12, possibly due to relief from local steric hindrance between these residues and the antiestrogen side chains. Fluorescence anisotropy experiments indicated an enhanced regional stabilization of mutant ligand-binding domains in the presence of antiestrogens. H12 mutations also prevent the increase in bioluminescence resonance energy transfer between ERalpha monomers induced by Ral or ICI182,780 and increase intranuclear receptor mobility in correlation with transcriptional activity in the presence of these antiestrogens. Our data indicate that ICI182,780 and Ral locally alter the ERalpha ligand binding structure via specific hydrophobic residues of H12 and decrease its transcriptional activity through tighter association with an insoluble nuclear structure.


Assuntos
Núcleo Celular/metabolismo , Estradiol/análogos & derivados , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio/efeitos dos fármacos , Leucina/metabolismo , Cloridrato de Raloxifeno/farmacologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos , Aminoácidos/química , Aminoácidos/genética , Aminoácidos/metabolismo , Núcleo Celular/química , Estradiol/farmacologia , Receptor alfa de Estrogênio/análise , Receptor alfa de Estrogênio/metabolismo , Fulvestranto , Humanos , Leucina/química , Leucina/genética , Dados de Sequência Molecular , Mutação , Estrutura Secundária de Proteína , Solubilidade , Transcrição Gênica/efeitos dos fármacos , Fator Trefoil-1 , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA