Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
SLAS Discov ; 25(1): 21-32, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31513463

RESUMO

Focal adhesion kinase (FAK) is a promising cancer drug target due to its massive overexpression in multiple solid tumors and its critical role in the integration of signals that control proliferation, invasion, apoptosis, and metastasis. Previous FAK drug discovery and high-throughput screening have exclusively focused on the identification of inhibitors that target the kinase domain of FAK. Because FAK is both a kinase and scaffolding protein, the development of novel screening assays that detect inhibitors of FAK protein-protein interactions remains a critical need. In this report, we describe the development of a high-throughput fluorescence polarization (FP) screening assay that measures the interactions between FAK and paxillin, a focal adhesion-associated protein. We designed a tetramethylrhodamine (TAMRA)-tagged paxillin peptide based on the paxillin LD2 motif that binds to the focal adhesion targeting (FAT) domain with significant dynamic range, specificity, variability, stability, and a Z'-factor suitable for high-throughput screening. In addition, we performed a pilot screen of 1593 compounds using this FP assay, showing its feasibility for high-throughput drug screening. Finally, we identified three compounds that show dose-dependent competition of FAT-paxillin binding. This assay represents the first described high-throughput screening assay for FAK scaffold inhibitors and can accelerate drug discovery efforts for this promising drug target.


Assuntos
Descoberta de Drogas , Polarização de Fluorescência , Quinase 1 de Adesão Focal/metabolismo , Ensaios de Triagem em Larga Escala , Paxilina/metabolismo , Ligação Proteica/efeitos dos fármacos , Descoberta de Drogas/métodos , Polarização de Fluorescência/métodos , Quinase 1 de Adesão Focal/química , Humanos , Modelos Moleculares , Conformação Molecular , Paxilina/química , Domínios e Motivos de Interação entre Proteínas , Relação Estrutura-Atividade
2.
Biochim Biophys Acta Gen Subj ; 1864(1): 129450, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31676296

RESUMO

BACKGROUND: Leucine rich Aspartate motifs (LD motifs) are molecular recognition motifs on Paxillin that recognize LD-motif binding domains (LDBD) of a number of focal adhesion proteins in order to carry out downstream signaling and actin cytoskeleton remodeling. In this study, we identified structural features within LDBDs that influence their binding affinity with Paxillin LD motifs. METHODS: Various point mutants of focal adhesion targeting (FAT) domain of Focal Adhesion Kinase (FAK) were created by moving a key Lysine residue two and three helical turns in order to match the unique conformations as observed in LDBDs of two other focal adhesion proteins, Vinculin and CCM3. RESULTS: This led to identify a mutant of FAT domain of FAK, named as FAT(NV) (Asn992 of FAT domain was replaced by Val), with remarkable high affinity for LD1 (Kd = 1.5 µM vs no-binding with wild type) and LD2 peptides (Kd = 7.2 µM vs 63 µM with wild type). Consistently, the focal adhesions of MCF7 cells expressing FAK(NV) were highly stable (turnover rate = 1.25 × 10-5 µm2/s) as compared to wild type FAK transfected cells (turnover rate = 1.5 × 10-3 µm2/s). CONCLUSIONS: We observed that the relative disposition of key LD binding amino-acids at LDBD surface, hydrophobic burial of long Leucine side chains of LD-motifs and complementarity of charged surfaces are the key factors determining the binding affinities of LD motifs with LDBDs. GENERAL SIGNIFICANCE: Our study will help in protein engineering of FAT domain of FAK by modulating FAK-LD motif interactions which have implications in cellular focal adhesions and cell migration.


Assuntos
Adesão Celular/genética , Quinase 1 de Adesão Focal/genética , Adesões Focais/genética , Conformação Proteica , Citoesqueleto de Actina/química , Citoesqueleto de Actina/genética , Motivos de Aminoácidos/genética , Sequência de Aminoácidos/genética , Proteínas Reguladoras de Apoptose/química , Proteínas Reguladoras de Apoptose/genética , Ácido Aspártico/genética , Sítios de Ligação/genética , Movimento Celular/genética , Quinase 1 de Adesão Focal/química , Adesões Focais/química , Regulação da Expressão Gênica/genética , Humanos , Lisina/química , Lisina/genética , Células MCF-7 , Proteínas de Membrana/química , Proteínas de Membrana/genética , Paxilina/química , Paxilina/genética , Ligação Proteica/genética , Engenharia de Proteínas , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/genética , Vinculina/química , Vinculina/genética
3.
Structure ; 27(11): 1686-1697.e5, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31590942

RESUMO

Activation of cell surface receptor integrin has been extensively studied as the first key step to trigger cell adhesion, but the subsequent events, widely regarded as integrin "outside-in" signaling to form supramolecular complexes (focal adhesions [FAs]) to promote dynamic cell adhesion, remain poorly elucidated. Integrin activator kindlin-2 was recently found to associate with paxillin in nascent FAs, implicating an early yet undefined integrin outside-in signaling event. Here we show structurally that kindlin-2 recognizes paxillin via a distinct interface involving the ubiquitin-like kindlin-2 F0 domain and the paxillin LIM4 domain. The interface is adjacent to the membrane binding site of kindlin-2 F0, suggesting a mechanism for kindlin-2 to recruit paxillin to the membrane-proximal site where FA assembly is initiated. Disruption of the interface impaired the localization of paxillin, causing strong defects in FA assembly and cell migration. These data unveil a structural basis of the kindlin-2/paxillin interaction in controlling dynamic cell adhesion.


Assuntos
Adesão Celular , Proteínas de Membrana/química , Proteínas de Neoplasias/química , Paxilina/química , Animais , Sítios de Ligação , Movimento Celular , Células Cultivadas , Células HEK293 , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Proteínas de Neoplasias/metabolismo , Paxilina/metabolismo , Ligação Proteica
4.
Molecules ; 24(18)2019 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-31540099

RESUMO

The Focal Adhesion Targeting (FAT) domain of Focal Adhesion Kinase (FAK) is a promising drug target since FAK is overexpressed in many malignancies and promotes cancer cell metastasis. The FAT domain serves as a scaffolding protein, and its interaction with the protein paxillin localizes FAK to focal adhesions. Various studies have highlighted the importance of FAT-paxillin binding in tumor growth, cell invasion, and metastasis. Targeting this interaction through high-throughput screening (HTS) provides a challenge due to the large and complex binding interface. In this report, we describe a novel approach to targeting FAT through fragment-based drug discovery (FBDD). We developed two fragment-based screening assays-a primary SPR assay and a secondary heteronuclear single quantum coherence nuclear magnetic resonance (HSQC-NMR) assay. For SPR, we designed an AviTag construct, optimized SPR buffer conditions, and created mutant controls. For NMR, resonance backbone assignments of the human FAT domain were obtained for the HSQC assay. A 189-compound fragment library from Enamine was screened through our primary SPR assay to demonstrate the feasibility of a FAT-FBDD pipeline, with 19 initial hit compounds. A final total of 11 validated hits were identified after secondary screening on NMR. This screening pipeline is the first FBDD screen of the FAT domain reported and represents a valid method for further drug discovery efforts on this difficult target.


Assuntos
Quinase 1 de Adesão Focal/química , Adesões Focais/química , Ressonância Magnética Nuclear Biomolecular , Ressonância de Plasmônio de Superfície , Descoberta de Drogas , Quinase 1 de Adesão Focal/genética , Adesões Focais/genética , Humanos , Paxilina/química , Paxilina/genética , Domínios Proteicos
5.
Genes Genomics ; 41(2): 241-248, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30604146

RESUMO

BACKGROUND: Ba/F3, a mouse pro-B cell line, is dependent on IL-3 for its survival and proliferation. IL-3 withdrawal causes cells to round, stop in G1 phase, then undergo apoptosis. Additionally, IL-3 is known to induce tyrosine phosphorylation of paxillin, a scaffold and signaling protein. We previously determined that overexpression of paxillin prohibited Ba/F3 cell apoptosis induced by IL-3 withdrawal. OBJECTIVE: Address whether phosphorylation is essential for the anti-apoptotic effect of overexpressed paxillin. METHODS: Mutations were introduced into paxillin cDNA at five phosphorylation sites-Y31F, Y40F, Y118F, Y181F, S273A, or S273D. After overexpression of paxillin mutants in Ba/F3 cells, the apoptotic proportions of cell populations were measured by an annexin V conjugation assay while cells were undergoing IL-3 withdrawal. RESULTS: The anti-apoptotic effect of paxillin overexpression was abolished by site-directed mutagenesis replacing Y31, Y40, Y118, and Y181 with phenylalanine, and S273 with aspartic acid. In contrast, the mutation replacing S273 with alanine had no effect on the anti-apoptotic effect. CONCLUSION: The above results suggest that paxillin-mediated phosphorylation at Y31, Y40, Y118, and Y181 is essential for the anti-apoptotic effect of paxillin overexpression in Ba/F3 cells and contributes to the cell survival signaling pathway triggered by IL-3. Conversely, phosphorylation at S273 is involved in the negative regulation of the anti-apoptotic action of overexpressed paxillin.


Assuntos
Apoptose , Paxilina/metabolismo , Motivos de Aminoácidos , Animais , Linfócitos B/metabolismo , Linhagem Celular Tumoral , Interleucina-3/metabolismo , Camundongos , Paxilina/química , Paxilina/genética , Fosforilação
6.
Methods ; 140-141: 85-96, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29605734

RESUMO

Quantitative fluorescence fluctuation spectroscopy from optical microscopy datasets is a very powerful tool to resolve multiple spatiotemporal cellular and subcellular processes at the molecular level. In particular, raster image correlation spectroscopy (RICS) and number and brightness analyses (N&B) yield molecular mobility and clustering dynamic information extracted from real-time cellular processes. This quantitative information can be inferred in a highly flexible and detailed manner, i.e. 1) at the localisation level: from full-frame datasets and multiple regions of interest within; and 2) at the temporal level: not only from full-frame and multiple regions, but also intermediate temporal events. Here we build on previous research in deciphering the molecular dynamics of paxillin, a main component of focal adhesions. Cells use focal adhesions to attach to the extracellular matrix and interact with their local environment. Through focal adhesions and other adhesion structures, cells sense their local environment and respond accordingly; due to this continuous communication, these structures can be highly dynamic depending on the extracellular characteristics. By using a previously well-characterised model like paxillin, we examine the powerful sensitivity and some limitations of RICS and N&B analyses. We show that cells upon contact to different surfaces show differential self-assembly dynamics in terms of molecular diffusion and oligomerisation. In addition, single-cell studies show that these dynamics change gradually following an antero-posterior gradient.


Assuntos
Adesões Focais/metabolismo , Análise de Célula Única/métodos , Espectrometria de Fluorescência/métodos , Imagem com Lapso de Tempo/métodos , Linhagem Celular Tumoral , Movimento Celular , Difusão , Proteínas de Fluorescência Verde/química , Humanos , Microscopia de Fluorescência/instrumentação , Microscopia de Fluorescência/métodos , Simulação de Dinâmica Molecular , Paxilina/química , Paxilina/metabolismo , Análise de Célula Única/instrumentação , Espectrometria de Fluorescência/instrumentação , Imagem com Lapso de Tempo/instrumentação
7.
Steroids ; 133: 87-92, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29097144

RESUMO

Paxillin is a group III LIM domain protein that is best characterized as a cytoplasmic scaffold/adaptor protein that functions primarily as a mediator of focal adhesion. However, emerging studies indicate that paxillin's functions are far broader. Not only does paxillin appear to regulate cytoplasmic kinase signaling, but it also cycles between the cytoplasm and nucleus, and may serve as an important regulator of mRNA trafficking and subsequent translation. Herein, we provide some insights suggesting that paxillin, like its relative Hic-5, has nuclear binding partners and mediates critical processes within the nucleus, at least in part functioning as coregulator of nuclear receptors and nuclear kinases to mediate genomic signaling.


Assuntos
Núcleo Celular/metabolismo , Paxilina/metabolismo , Motivos de Aminoácidos , Animais , Humanos , Terapia de Alvo Molecular , Sinais de Exportação Nuclear , Paxilina/química
8.
J Biol Chem ; 292(44): 18281-18289, 2017 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-28860193

RESUMO

The Cas family scaffolding protein p130Cas is a Src substrate localized in focal adhesions (FAs) and functions in integrin signaling to promote cell motility, invasion, proliferation, and survival. p130Cas targeting to FAs is essential for its tyrosine phosphorylation and downstream signaling. Although the N-terminal SH3 domain is important for p130Cas localization, it has also been reported that the C-terminal region is involved in p130Cas FA targeting. The C-terminal region of p130Cas or Cas family homology domain (CCHD) has been reported to adopt a structure similar to that of the focal adhesion kinase C-terminal focal adhesion-targeting domain. The mechanism by which the CCHD promotes FA targeting of p130Cas, however, remains unclear. In this study, using a calorimetry approach, we identified the first LD motif (LD1) of the FA-associated protein paxillin as the binding partner of the p130Cas CCHD (in a 1:1 stoichiometry with a Kd ∼4.2 µm) and elucidated the structure of the p130Cas CCHD in complex with the paxillin LD1 motif by X-ray crystallography. Of note, a comparison of the CCHD/LD1 complex with a previously solved structure of CCHD in complex with the SH2-containing protein NSP3 revealed that LD1 had almost identical positioning of key hydrophobic and acidic residues relative to NSP3. Because paxillin is one of the key scaffold molecules in FAs, we propose that the interaction between the p130Cas CCHD and the LD1 motif of paxillin plays an important role in p130Cas FA targeting.


Assuntos
Proteínas Aviárias/metabolismo , Proteína Substrato Associada a Crk/metabolismo , Modelos Moleculares , Paxilina/metabolismo , Motivos de Aminoácidos , Substituição de Aminoácidos , Animais , Proteínas Aviárias/química , Sítios de Ligação , Galinhas , Proteína Substrato Associada a Crk/química , Proteína Substrato Associada a Crk/genética , Cristalografia por Raios X , Interações Hidrofóbicas e Hidrofílicas , Cinética , Leucina , Camundongos , Mutação , Paxilina/química , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Estabilidade Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Homologia Estrutural de Proteína
9.
ACS Nano ; 11(4): 4028-4040, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28355484

RESUMO

Determining how cells generate and transduce mechanical forces at the nanoscale is a major technical challenge for the understanding of numerous physiological and pathological processes. Podosomes are submicrometer cell structures with a columnar F-actin core surrounded by a ring of adhesion proteins, which possess the singular ability to protrude into and probe the extracellular matrix. Using protrusion force microscopy, we have previously shown that single podosomes produce local nanoscale protrusions on the extracellular environment. However, how cellular forces are distributed to allow this protruding mechanism is still unknown. To investigate the molecular machinery of protrusion force generation, we performed mechanical simulations and developed quantitative image analyses of nanoscale architectural and mechanical measurements. First, in silico modeling showed that the deformations of the substrate made by podosomes require protrusion forces to be balanced by local traction forces at the immediate core periphery where the adhesion ring is located. Second, we showed that three-ring proteins are required for actin polymerization and protrusion force generation. Third, using DONALD, a 3D nanoscopy technique that provides 20 nm isotropic localization precision, we related force generation to the molecular extension of talin within the podosome ring, which requires vinculin and paxillin, indicating that the ring sustains mechanical tension. Our work demonstrates that the ring is a site of tension, balancing protrusion at the core. This local coupling of opposing forces forms the basis of protrusion and reveals the podosome as a nanoscale autonomous force generator.


Assuntos
Podossomos/química , Actinas/química , Actinas/metabolismo , Fenômenos Biomecânicos , Adesão Celular , Células Cultivadas , Simulação por Computador , Humanos , Macrófagos/citologia , Macrófagos/metabolismo , Mecanotransdução Celular , Monócitos/citologia , Monócitos/metabolismo , Nanoestruturas/química , Tamanho da Partícula , Paxilina/química , Paxilina/metabolismo , Podossomos/ultraestrutura , Propriedades de Superfície , Talina/química , Talina/metabolismo , Vinculina/química , Vinculina/metabolismo
10.
Cell Rep ; 15(8): 1660-72, 2016 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-27184837

RESUMO

Autophagy is a conserved catabolic process that plays a housekeeping role in eliminating protein aggregates and organelles and is activated during nutrient deprivation to generate metabolites and energy. Autophagy plays a significant role in tumorigenesis, although opposing context-dependent functions of autophagy in cancer have complicated efforts to target autophagy for therapeutic purposes. We demonstrate that autophagy inhibition reduces tumor cell migration and invasion in vitro and attenuates metastasis in vivo. Numerous abnormally large focal adhesions (FAs) accumulate in autophagy-deficient tumor cells, reflecting a role for autophagy in FA disassembly through targeted degradation of paxillin. We demonstrate that paxillin interacts with processed LC3 through a conserved LIR motif in the amino-terminal end of paxillin and that this interaction is regulated by oncogenic SRC activity. Together, these data establish a function for autophagy in FA turnover, tumor cell motility, and metastasis.


Assuntos
Autofagia , Movimento Celular , Adesões Focais/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Neoplasias/patologia , Paxilina/metabolismo , Quinases da Família src/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Autofagossomos/metabolismo , Proteínas Relacionadas à Autofagia/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Silenciamento de Genes , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Paxilina/química , Ligação Proteica , Estabilidade Proteica , Transporte Proteico
11.
PLoS One ; 11(2): e0150153, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26928467

RESUMO

Intrinsically disordered proteins (IDPs) play a major role in various cellular functions ranging from transcription to cell migration. Mutations/modifications in such IDPs are shown to be associated with various diseases. Current strategies to study the mode of action and regulatory mechanisms of disordered proteins at the structural level are time consuming and challenging. Therefore, using simple and swift strategies for identifying functionally important regions in unstructured segments and understanding their underlying mechanisms is critical for many applications. Here we propose a simple strategy that employs dissection of human paxillin (residues 1-313) that comprises intrinsically disordered regions, followed by its interaction study using FAT (Focal adhesion targeting domain of focal adhesion kinase) as its binding partner to retrace structural behavior. Our findings show that the paxillin interaction with FAT exhibits a masking and unmasking effect by a putative intra-molecular regulatory region. This phenomenon suggests how cancer associated mutations in paxillin affect its interactions with Focal Adhesion Kinase (FAK). The strategy could be used to decipher the mode of regulations and identify functionally relevant constructs for other studies.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/química , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Adesões Focais/metabolismo , Proteínas Intrinsicamente Desordenadas/química , Proteínas Intrinsicamente Desordenadas/metabolismo , Paxilina/química , Paxilina/metabolismo , Humanos , Modelos Moleculares , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína
12.
Nat Cell Biol ; 17(7): 880-92, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26053221

RESUMO

Focal adhesions (FAs) link the extracellular matrix to the actin cytoskeleton to mediate cell adhesion, migration, mechanosensing and signalling. FAs have conserved nanoscale protein organization, suggesting that the position of proteins within FAs regulates their activity and function. Vinculin binds different FA proteins to mediate distinct cellular functions, but how vinculin's interactions are spatiotemporally organized within FAs is unknown. Using interferometric photoactivation localization super-resolution microscopy to assay vinculin nanoscale localization and a FRET biosensor to assay vinculin conformation, we found that upward repositioning within the FA during FA maturation facilitates vinculin activation and mechanical reinforcement of FAs. Inactive vinculin localizes to the lower integrin signalling layer in FAs by binding to phospho-paxillin. Talin binding activates vinculin and targets active vinculin higher in FAs where vinculin can engage retrograde actin flow. Thus, specific protein interactions are spatially segregated within FAs at the nanoscale to regulate vinculin activation and function.


Assuntos
Adesões Focais/metabolismo , Nanoestruturas , Nanotecnologia/métodos , Vinculina/metabolismo , Actinas/química , Actinas/metabolismo , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Transferência Ressonante de Energia de Fluorescência , Adesões Focais/genética , Humanos , Integrinas/química , Integrinas/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microscopia de Fluorescência/métodos , Modelos Moleculares , Mutação , Paxilina/química , Paxilina/genética , Paxilina/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Interferência de RNA , Talina/química , Talina/genética , Talina/metabolismo , Vinculina/química , Vinculina/genética
13.
J Biol Chem ; 290(24): 15197-209, 2015 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-25922072

RESUMO

The guanine nucleotide exchange factor Rgnef (also known as ArhGEF28 or p190RhoGEF) promotes colon carcinoma cell motility and tumor progression via interaction with focal adhesion kinase (FAK). Mechanisms of Rgnef activation downstream of integrin or G protein-coupled receptors remain undefined. In the absence of a recognized G protein signaling homology domain in Rgnef, no proximal linkage to G proteins was known. Utilizing multiple methods, we have identified Rgnef as a new effector for Gα13 downstream of gastrin and the type 2 cholecystokinin receptor. In DLD-1 colon carcinoma cells depleted of Gα13, gastrin-induced FAK Tyr(P)-397 and paxillin Tyr(P)-31 phosphorylation were reduced. RhoA GTP binding and promoter activity were increased by Rgnef in combination with active Gα13. Rgnef co-immunoprecipitated with activated Gα13Q226L but not Gα12Q229L. The Rgnef C-terminal (CT, 1279-1582) region was sufficient for co-immunoprecipitation, and Rgnef-CT exogenous expression prevented Gα13-stimulated SRE activity. A domain at the C terminus of the protein close to the FAK binding domain is necessary to bind to Gα13. Point mutations of Rgnef-CT residues disrupt association with active Gα13 but not Gαq. These results show that Rgnef functions as an effector of Gα13 signaling and that this linkage may mediate FAK activation in DLD-1 colon carcinoma cells.


Assuntos
Neoplasias do Colo/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/fisiologia , Gastrinas/fisiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Células HEK293 , Humanos , Paxilina/química , Paxilina/metabolismo , Fosforilação , Receptor de Colecistocinina B/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/química , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Tirosina/metabolismo
14.
Biochem J ; 460(3): 317-29, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24870021

RESUMO

LD motifs (leucine-aspartic acid motifs) are short helical protein-protein interaction motifs that have emerged as key players in connecting cell adhesion with cell motility and survival. LD motifs are required for embryogenesis, wound healing and the evolution of multicellularity. LD motifs also play roles in disease, such as in cancer metastasis or viral infection. First described in the paxillin family of scaffolding proteins, LD motifs and similar acidic LXXLL interaction motifs have been discovered in several other proteins, whereas 16 proteins have been reported to contain LDBDs (LD motif-binding domains). Collectively, structural and functional analyses have revealed a surprising multivalency in LD motif interactions and a wide diversity in LDBD architectures. In the present review, we summarize the molecular basis for function, regulation and selectivity of LD motif interactions that has emerged from more than a decade of research. This overview highlights the intricate multi-level regulation and the inherently noisy and heterogeneous nature of signalling through short protein-protein interaction motifs.


Assuntos
Motivos de Aminoácidos/fisiologia , Ácido Aspártico/metabolismo , Leucina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas Reguladoras de Apoptose/fisiologia , Proteínas de Ciclo Celular/fisiologia , Quinase 2 de Adesão Focal/química , Humanos , Ligantes , Proteínas de Membrana/fisiologia , Proteínas dos Microfilamentos/metabolismo , Paxilina/química , Proteína I de Ligação a Poli(A)/metabolismo , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Vinculina/fisiologia
15.
Science ; 339(6120): 694-8, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23393263

RESUMO

E6 viral oncoproteins are key players in epithelial tumors induced by papillomaviruses in vertebrates, including cervical cancer in humans. E6 proteins target many host proteins by specifically interacting with acidic LxxLL motifs. We solved the crystal structures of bovine (BPV1) and human (HPV16) papillomavirus E6 proteins bound to LxxLL peptides from the focal adhesion protein paxillin and the ubiquitin ligase E6AP, respectively. In both E6 proteins, two zinc domains and a linker helix form a basic-hydrophobic pocket, which captures helical LxxLL motifs in a way compatible with other interaction modes. Mutational inactivation of the LxxLL binding pocket disrupts the oncogenic activities of both E6 proteins. This work reveals the structural basis of both the multifunctionality and the oncogenicity of E6 proteins.


Assuntos
Proteínas Oncogênicas Virais/química , Proteínas Oncogênicas Virais/metabolismo , Paxilina/química , Domínios e Motivos de Interação entre Proteínas , Proteínas Repressoras/química , Proteínas Repressoras/metabolismo , Ubiquitina-Proteína Ligases/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Papillomavirus Bovino 1 , Cristalografia por Raios X , Papillomavirus Humano 16 , Humanos , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Proteínas Oncogênicas Virais/genética , Paxilina/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Mutação Puntual , Estrutura Secundária de Proteína , Proteínas Repressoras/genética , Ubiquitina-Proteína Ligases/metabolismo
16.
FEBS Lett ; 586(16): 2294-9, 2012 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-22728435

RESUMO

Barrier-protective agonists induce association of focal adhesions (FA) and adherens junctions (AJ) in endothelial cells. Here we identified specific domains of FA protein paxillin interacting with AJ protein and examined regulation of paxillin domain interactions with ß-catenin by Rac GTPase. Co-expression of paxillin LD-1,2; LD-3,4; LIM-1,2; and LIM-3,4 domains with ß-catenin showed exclusive interaction of LIM-1,2 and LIM-3,4 with ß-catenin, which was enhanced by agonist-induced Rac activation or expression of activated Rac mutant. These results demonstrate a novel function of paxillin LIM domains in targeting ß-catenin in a Rac-dependent manner, which may play a role in Rac-dependent control of FA-AJ interactions and monolayer integrity.


Assuntos
Regulação da Expressão Gênica , Paxilina/química , beta Catenina/química , Junções Aderentes/química , Sítio Alostérico , Células Endoteliais/citologia , Escherichia coli/metabolismo , Adesões Focais/química , Glutationa Transferase/metabolismo , Células HEK293 , Humanos , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-akt/química , Proteínas Recombinantes/química , Transdução de Sinais
17.
J Biol Chem ; 286(29): 26138-47, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21632544

RESUMO

Cerebral cavernous malformation (CCM) is a disease that affects between 0.1 and 0.5% of the human population, with mutations in CCM3 accounting for ~ 15% of the autosomal dominant form of the disease. We recently reported that CCM3 contains an N-terminal dimerization domain (CCM3D) and a C-terminal focal adhesion targeting (FAT) homology domain. Intermolecular protein-protein interactions of CCM3 are mediated by a highly conserved surface on the FAT homology domain and are affected by CCM3 truncations in the human disease. Here we report the crystal structures of CCM3 in complex with three different leucine-aspartate repeat (LD) motifs (LD1, LD2, and LD4) from the scaffolding protein paxillin, at 2.8, 2.7, and 2.5 Å resolution. We show that CCM3 binds LD motifs using the highly conserved hydrophobic patch 1 (HP1) and that this binding is similar to the binding of focal adhesion kinase and Pyk2 FAT domains to paxillin LD motifs. We further show by surface plasmon resonance that CCM3 binds paxillin LD motifs with affinities in the micromolar range, similar to FAK family FAT domains. Finally, we show that endogenous CCM3 and paxillin co-localize in mouse cerebral pericytes. These studies provide a molecular-level framework to investigate the protein-protein interactions of CCM3.


Assuntos
Proteínas Reguladoras de Apoptose/química , Proteínas Reguladoras de Apoptose/metabolismo , Adesões Focais/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Paxilina/química , Paxilina/metabolismo , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Sequências Repetitivas de Aminoácidos , Homologia de Sequência de Aminoácidos , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Ácido Aspártico , Encéfalo/citologia , Cristalografia por Raios X , Quinase 2 de Adesão Focal/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Leucina , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Pericitos/metabolismo , Estrutura Terciária de Proteína , Transporte Proteico
18.
J Biol Chem ; 285(31): 24099-107, 2010 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-20489202

RESUMO

CCM3 mutations are associated with cerebral cavernous malformation (CCM), a disease affecting 0.1-0.5% of the human population. CCM3 (PDCD10, TFAR15) is thought to form a CCM complex with CCM1 and CCM2; however, the molecular basis for these interactions is not known. We have determined the 2.5 A crystal structure of CCM3. This structure shows an all alpha-helical protein containing two domains, an N-terminal dimerization domain with a fold not previously observed, and a C-terminal focal adhesion targeting (FAT)-homology domain. We show that CCM3 binds CCM2 via this FAT-homology domain and that mutation of a highly conserved FAK-like hydrophobic pocket (HP1) abrogates CCM3-CCM2 interaction. This CCM3 FAT-homology domain also interacts with paxillin LD motifs using the same surface, and partial CCM3 co-localization with paxillin in cells is lost on HP1 mutation. Disease-related CCM3 truncations affect the FAT-homology domain suggesting a role for the FAT-homology domain in the etiology of CCM.


Assuntos
Proteínas Reguladoras de Apoptose/química , Hemangioma Cavernoso do Sistema Nervoso Central/metabolismo , Proteínas de Membrana/química , Proteínas Proto-Oncogênicas/química , Ligação Competitiva , Encéfalo/metabolismo , Cristalografia por Raios X/métodos , Dimerização , Humanos , Cinética , Conformação Molecular , Mutação , Paxilina/química , Conformação Proteica , Dobramento de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
19.
Biochem Biophys Res Commun ; 383(3): 347-52, 2009 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-19358827

RESUMO

Focal adhesion targeting (FAT) domains target the non-receptor tyrosine kinases FAK and Pyk2 to cellular focal adhesion areas, where the signaling molecule paxillin is also located. Here, we report the crystal structures of the Pyk2 FAT domain alone or in complex with paxillin LD4 peptides. The overall structure of Pyk2-FAT is an antiparallel four-helix bundle with an up-down, up-down, right-handed topology. In the LD4-bound FAT complex, two paxillin LD4 peptides interact with two opposite sides of Pyk2-FAT, at the surfaces of the alpha1alpha4 and alpha2alpha3 helices of each FAT molecule. We also demonstrate that, while paxillin is phosphorylated by Pyk2, complex formation between Pyk2 and paxillin does not depend on Pyk2 tyrosine kinase activity. These experiments reveal the structural basis underlying the selectivity of paxillin LD4 binding to the Pyk2 FAT domain and provide insights about the molecular details which influence the different behavior of these two closely-related kinases.


Assuntos
Quinase 2 de Adesão Focal/metabolismo , Adesões Focais/enzimologia , Paxilina/metabolismo , Cristalografia por Raios X , Quinase 2 de Adesão Focal/química , Quinase 2 de Adesão Focal/genética , Humanos , Paxilina/química , Paxilina/genética , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
20.
Mol Carcinog ; 48(6): 532-44, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18973190

RESUMO

Transforming growth factor beta (TGF-beta) exerts an important role in the late steps of carcinogenesis by cooperating with Ras to induce cell motility and tumor invasion. The transcription complex AP-1 has been implicated in the regulation of genes involved in motility and invasion, by mechanisms not yet delineated. We utilized a model of immortalized human hepatocytes (IHH) overexpressing c-Fos (IHH-Fos) or not (IHH-C) to investigate the role of c-Fos on cell motility in response to a prolonged treatment with TGF-beta, EGF or a combination of both. Cotreatment with EGF and TGF-beta, but neither cytokine alone, induced the conversion of hepatocytes to a fibroblastoid phenotype and increased their motility in Boyden chambers. EGF/TGF-beta cotreatment induced a higher effect on ERK phosphorylation compared to TGF-beta treatment alone. It also induced an increase in total and phosphorylated Ser(178) paxillin, a protein previously implicated in cell motility. This response was inhibited by two specific MEK inhibitors, indicating the involvement of the ERK pathway in paxillin activation. Overexpression of c-Fos correlated with increased cell scattering and motility, higher levels of ERK activation and phospho Ser(178) paxillin, increased levels of EGF receptor (EGF-R) mRNA and higher EGF-R phosphorylation levels following EGF/TGF-beta cotreatment. Conversely, siRNA-mediated invalidation of c-Fos delayed the appearance of fibroblastoid cells, decreased EGF-R mRNA and downregulated ERK and Ser(178) paxillin phosphorylations, indicating that c-Fos activates hepatocyte motility through an EGF-R/ERK/paxillin pathway. Since c-Fos is frequently overexpressed in hepatocarcinomas, this newly identified mechanism might be involved in the progression of hepatic tumors in vivo.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hepatócitos/citologia , Paxilina/metabolismo , Proteínas Proto-Oncogênicas c-fos/fisiologia , Serina/metabolismo , Regulação para Cima , Sequência de Bases , Linhagem Celular Transformada , Primers do DNA , Ativação Enzimática , Fator de Crescimento Epidérmico/farmacologia , Fibroblastos/citologia , Humanos , Microscopia Confocal , Microscopia de Fluorescência , Paxilina/química , Fosforilação , Reação em Cadeia da Polimerase , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA