Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
bioRxiv ; 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38405732

RESUMO

The PEAK family of pseudokinases, comprising PEAK1-3, are signalling scaffolds that play oncogenic roles in several poor prognosis human cancers, including triple negative breast cancer (TNBC). However, therapeutic targeting of pseudokinases is challenging due to their lack of catalytic activity. To address this, we screened for PEAK1 effectors by affinity purification and mass spectrometry, identifying calcium/calmodulin-dependent protein kinase 2 (CAMK2)D and CAMK2G. PEAK1 promoted CAMK2D/G activation in TNBC cells via a novel feed-forward mechanism involving PEAK1/PLCγ1/Ca 2+ signalling and direct binding via a consensus CAMK2 interaction motif in the PEAK1 N-terminus. In turn, CAMK2 phosphorylated PEAK1 to enhance association with PEAK2, which is critical for PEAK1 oncogenic signalling. To achieve pharmacologic targeting of PEAK1/CAMK2, we repurposed RA306, a second generation CAMK2 inhibitor under pre-clinical development for treatment of cardiovascular disease. RA306 demonstrated on-target activity against CAMK2 in TNBC cells and inhibited PEAK1-enhanced migration and invasion in vitro . Moreover, RA306 significantly attenuated TNBC xenograft growth and blocked metastasis in a manner mirrored by CRISPR-mediated PEAK1 ablation. Overall, these studies establish PEAK1 as a critical cell signalling nexus, identify a novel mechanism for regulation of Ca 2+ signalling and its integration with tyrosine kinase signals, and identify CAMK2 as a therapeutically 'actionable' target downstream of PEAK1.

2.
Nat Commun ; 14(1): 3542, 2023 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-37336884

RESUMO

PEAK pseudokinases regulate cell migration, invasion and proliferation by recruiting key signaling proteins to the cytoskeleton. Despite lacking catalytic activity, alteration in their expression level is associated with several aggressive cancers. Here, we elucidate the molecular details of key PEAK signaling interactions with the adapter proteins CrkII and Grb2 and the scaffold protein 14-3-3. Our findings rationalize why the dimerization of PEAK proteins has a crucial function in signal transduction and provide biophysical and structural data to unravel binding specificity within the PEAK interactome. We identify a conserved high affinity 14-3-3 motif on PEAK3 and demonstrate its role as a molecular switch to regulate CrkII binding and signaling via Grb2. Together, our studies provide a detailed structural snapshot of PEAK interaction networks and further elucidate how PEAK proteins, especially PEAK3, act as dynamic scaffolds that exploit adapter proteins to control signal transduction in cell growth/motility and cancer.


Assuntos
Proteínas 14-3-3 , Proteínas do Citoesqueleto , Transdução de Sinais , Movimento Celular , Proliferação de Células , Transdução de Sinais/fisiologia , Proteínas do Citoesqueleto/metabolismo , Proteínas 14-3-3/metabolismo
3.
J Biophotonics ; 16(5): e202200190, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36722724

RESUMO

High-energy laser pulses used in laser angioplasty are challenging the laser cost, delivery system damage, efficiency, and laser catheter operating time. 355 nm nanosecond-pulsed laser in burst mode has shown potentials in reducing the system complexity and selective ablation of tissues. In this paper, burst mode laser ablation of porcine subcutaneous fat and porcine aorta is investigated. A histopathological analysis demonstrates that porcine subcutaneous fat can be ablated at a rate of greater than 0.2 mm/s when the number of pulses per burst is 1500 (corresponding to a fluence of 0.12 mJ/mm2 per pulse and 180 mJ/mm2 per burst), and the temperature of tissue during lasing is lower than 45°C. The porcine aorta remains nearly unaffected at the same laser parameter, and the tissue temperature during lasing is lower than 35°C. It shows the feasibility of using a burst-mode laser for selective ablation of tissue.


Assuntos
Terapia a Laser , Lasers de Estado Sólido , Suínos , Animais , Luz , Fatores de Tempo , Aorta/patologia
4.
Oncogene ; 42(11): 833-847, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36693952

RESUMO

We have determined that expression of the pseudokinase NRBP1 positively associates with poor prognosis in triple negative breast cancer (TNBC) and is required for efficient migration, invasion and proliferation of TNBC cells in culture as well as growth of TNBC orthotopic xenografts and experimental metastasis. Application of BioID/MS profiling identified P-Rex1, a known guanine nucleotide exchange factor for Rac1, as a NRBP1 binding partner. Importantly, NRBP1 overexpression enhanced levels of GTP-bound Rac1 and Cdc42 in a P-Rex1-dependent manner, while NRBP1 knockdown reduced their activation. In addition, NRBP1 associated with P-Rex1, Rac1 and Cdc42, suggesting a scaffolding function for this pseudokinase. NRBP1-mediated promotion of cell migration and invasion was P-Rex1-dependent, while constitutively-active Rac1 rescued the effect of NRBP1 knockdown on cell proliferation and invasion. Generation of reactive oxygen species via a NRBP1/P-Rex1 pathway was implicated in these oncogenic roles of NRBP1. Overall, these findings define a new function for NRBP1 and a novel oncogenic signalling pathway in TNBC that may be amenable to therapeutic intervention.


Assuntos
Neoplasias de Mama Triplo Negativas , Humanos , Proteínas rac1 de Ligação ao GTP/metabolismo , Transdução de Sinais , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Movimento Celular , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas de Transporte Vesicular/metabolismo
5.
Cell Commun Signal ; 20(1): 197, 2022 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-36550548

RESUMO

Specific members of the Nima-Related Kinase (NEK) family have been linked to cancer development and progression, and a role for NEK5, one of the least studied members, in breast cancer has recently been proposed. However, while NEK5 is known to regulate centrosome separation and mitotic spindle assembly, NEK5 signalling mechanisms and function in this malignancy require further characterization. To this end, we established a model system featuring overexpression of NEK5 in the immortalized breast epithelial cell line MCF-10A. MCF-10A cells overexpressing NEK5 exhibited an increase in clonogenicity under monolayer conditions and enhanced acinar size and abnormal morphology in 3D Matrigel culture. Interestingly, they also exhibited a marked reduction in Src activation and downstream signalling. To interrogate NEK5 signalling and function in an unbiased manner, we applied a variety of MS-based proteomic approaches. Determination of the NEK5 interactome by Bio-ID identified a variety of protein classes including the kinesins KIF2C and KIF22, the mitochondrial proteins TFAM, TFB2M and MFN2, RhoH effectors and the negative regulator of Src, CSK. Characterization of proteins and phosphosites modulated upon NEK5 overexpression by global MS-based (phospho)proteomic profiling revealed impact on the cell cycle, DNA synthesis and repair, Rho GTPase signalling, the microtubule cytoskeleton and hemidesmosome assembly. Overall, the study indicates that NEK5 impacts diverse pathways and processes in breast epithelial cells, and likely plays a multifaceted role in breast cancer development and progression. Video Abstract.


Assuntos
Neoplasias da Mama , Proteômica , Humanos , Feminino , Quinases Relacionadas a NIMA/metabolismo , Linhagem Celular , Neoplasias da Mama/metabolismo , Células Epiteliais/metabolismo , Proteínas de Ligação a DNA , Cinesinas
6.
Sci Signal ; 15(722): eabj3554, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35192416

RESUMO

The pseudokinase scaffolds PEAK1 and PEAK2 are implicated in cancer cell migration and metastasis. We characterized the regulation and role of the third family member PEAK3 in cell signaling. Similar to PEAK1 and PEAK2, PEAK3 formed both homotypic and heterotypic complexes. In addition, like PEAK1, it bound to the adaptors Grb2 and CrkII. However, unlike PEAK1 and PEAK2, homodimerized PEAK3 also interacted with the ARF GTPase-activating protein ASAP1, the E3 ubiquitin ligase Cbl, and the kinase PYK2. Dimerization and subsequent phosphorylation on Tyr24, likely by a Src family kinase, were required for the binding of PEAK3 to Grb2 and ASAP1. Interactions with Grb2, CrkII, ASAP1, Cbl, and PYK2 exhibited contrasting dynamics upon cell stimulation with epidermal growth factor (EGF), in part due to PEAK3 dephosphorylation mediated by the phosphatase PTPN12. Overexpressing PEAK3 in mesenchymal-like MDA-MB-231 breast cancer cells enhanced cell elongation in a manner dependent on PEAK3 dimerization, and manipulation of PEAK3 expression demonstrated a positive role for this scaffold in regulating cell migration. Overexpressing PEAK3 in PEAK1/2 double-knockout MCF-10A breast epithelial cells enhanced acinar growth, impaired basement membrane integrity, and promoted invasion in three-dimensional cultures, with the latter two effects dependent on the binding of PEAK3 to Grb2 and ASAP1. PEAK1 and PEAK2 quantitatively and temporally influenced PEAK3 function. These findings characterize PEAK3 as an integral, signal-diversifying member of the PEAK family with scaffolding roles that promote cell proliferation, migration, and invasion.


Assuntos
Proteínas Tirosina Quinases , Transdução de Sinais , Movimento Celular , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Quinases da Família src/metabolismo
7.
Molecules ; 24(20)2019 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-31627265

RESUMO

Grb7 is an adapter protein, overexpressed in HER2+ve breast and other cancers, and identified as a therapeutic target. Grb7 promotes both proliferative and migratory cellular pathways through interaction of its SH2 domain with upstream binding partners including HER2, SHC, and FAK. Here we present the evaluation of a series of monocyclic and bicyclic peptide inhibitors that have been developed to specifically and potently target the Grb7 SH2-domain. All peptides tested were found to inhibit signaling in both ERK and AKT pathways in SKBR-3 and MDA-MB-231 cell lines. Proliferation, migration, and invasion assays revealed, however, that the second-generation bicyclic peptides were not more bioactive than the first generation G7-18NATE peptide, despite their higher in vitro affinity for the target. This was found not to be due to steric hindrance by the cell-permeability tag, as ascertained by ITC, but to differences in the ability of the bicyclic peptides to interact with and penetrate cellular membranes, as determined using SPR and mass spectrometry. These studies reveal that just small differences to amino acid composition can greatly impact the effectiveness of peptide inhibitors to their intracellular target and demonstrate that G7-18NATE remains the most effective peptide inhibitor of Grb7 developed to date.


Assuntos
Antineoplásicos/farmacologia , Células Epiteliais/efeitos dos fármacos , Proteína Adaptadora GRB7/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica , Peptídeos Cíclicos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sequência de Aminoácidos , Antineoplásicos/síntese química , Sítios de Ligação , Linhagem Celular , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Proteína Adaptadora GRB7/genética , Proteína Adaptadora GRB7/metabolismo , Humanos , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Peptídeos Cíclicos/síntese química , Fosfatidilcolinas/química , Fosfatidilcolinas/metabolismo , Fosfatidilserinas/química , Fosfatidilserinas/metabolismo , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética , Relação Estrutura-Atividade , Domínios de Homologia de src/efeitos dos fármacos
8.
Mol Cell Proteomics ; 18(7): 1410-1427, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31061140

RESUMO

In prostate cancer, cancer-associated fibroblasts (CAF) exhibit contrasting biological properties to non-malignant prostate fibroblasts (NPF) and promote tumorigenesis. Resolving intercellular signaling pathways between CAF and prostate tumor epithelium may offer novel opportunities for research translation. To this end, the proteome and phosphoproteome of four pairs of patient-matched CAF and NPF were characterized to identify discriminating proteomic signatures. Samples were analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS) with a hyper reaction monitoring data-independent acquisition (HRM-DIA) workflow. Proteins that exhibited a significant increase in CAF versus NPF were enriched for the functional categories "cell adhesion" and the "extracellular matrix." The CAF phosphoproteome exhibited enhanced phosphorylation of proteins associated with the "spliceosome" and "actin binding." STRING analysis of the CAF proteome revealed a prominent interaction hub associated with collagen synthesis, modification, and signaling. It contained multiple collagens, including the fibrillar types COL1A1/2 and COL5A1; the receptor tyrosine kinase discoidin domain-containing receptor 2 (DDR2), a receptor for fibrillar collagens; and lysyl oxidase-like 2 (LOXL2), an enzyme that promotes collagen crosslinking. Increased activity and/or expression of LOXL2 and DDR2 in CAF were confirmed by enzymatic assays and Western blotting analyses. Pharmacological inhibition of CAF-derived LOXL2 perturbed extracellular matrix (ECM) organization and decreased CAF migration in a wound healing assay. Further, it significantly impaired the motility of co-cultured RWPE-2 prostate tumor epithelial cells. These results indicate that CAF-derived LOXL2 is an important mediator of intercellular communication within the prostate tumor microenvironment and is a potential therapeutic target.


Assuntos
Aminoácido Oxirredutases/metabolismo , Fibroblastos Associados a Câncer/metabolismo , Neoplasias da Próstata/metabolismo , Proteômica , Microambiente Tumoral , Comunicação Autócrina , Linhagem Celular Tumoral , Movimento Celular , Células Epiteliais/patologia , Matriz Extracelular/metabolismo , Humanos , Masculino , Proteínas de Neoplasias/metabolismo , Comunicação Parácrina , Fosfoproteínas/metabolismo , Fosforilação , Próstata/metabolismo , Próstata/patologia , Proteoma/metabolismo , Reprodutibilidade dos Testes , Transdução de Sinais
9.
Nat Commun ; 10(1): 296, 2019 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-30655532

RESUMO

Despite significant progress, our understanding of how specific oncogenes transform cells is still limited and likely underestimates the complexity of downstream signalling events. To address this gap, we use mass spectrometry-based chemical proteomics to characterize the global impact of an oncogene on the expressed kinome, and then functionally annotate the regulated kinases. As an example, we identify 63 protein kinases exhibiting altered expression and/or phosphorylation in Src-transformed mammary epithelial cells. An integrated siRNA screen identifies nine kinases, including SGK1, as being essential for Src-induced transformation. Accordingly, we find that Src positively regulates SGK1 expression in triple negative breast cancer cells, which exhibit a prominent signalling network governed by Src family kinases. Furthermore, combined inhibition of Src and SGK1 reduces colony formation and xenograft growth more effectively than either treatment alone. Therefore, this approach not only provides mechanistic insights into oncogenic transformation but also aids the design of improved therapeutic strategies.


Assuntos
Transformação Celular Neoplásica/genética , Regulação Neoplásica da Expressão Gênica , Proteínas Imediatamente Precoces/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Quinases da Família src/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Benzodioxóis/farmacologia , Benzodioxóis/uso terapêutico , Linhagem Celular Tumoral , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/patologia , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Proteínas Imediatamente Precoces/antagonistas & inibidores , Espectrometria de Massas/métodos , Camundongos Endogâmicos BALB C , Camundongos Nus , Oncogenes/genética , Mapeamento de Interação de Proteínas/métodos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteômica/métodos , Quinazolinas/farmacologia , Quinazolinas/uso terapêutico , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Quinases da Família src/antagonistas & inibidores
10.
J Med Chem ; 60(22): 9349-9359, 2017 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-29083893

RESUMO

Grb7 is a signaling protein with critical roles in tumor cell proliferation and migration and an established cancer therapeutic target. Here we explore chemical space to develop a new bicyclic peptide inhibitor, incorporating thioether and lactam linkers that binds with affinity (KD = 1.1 µM) and specificity to the Grb7-SH2 domain. Structural analysis of the Grb7-SH2/peptide complex revealed an unexpected binding orientation underlying the binding selectivity by this new scaffold. We further incorporated carboxymethylphenylalanine and carboxyphenylalanine phosphotyrosine mimetics and arrived at an optimized inhibitor that potently binds Grb7-SH2 (KD = 0.13 µM) under physiological conditions. X-ray crystal structures of these Grb7-SH2/peptide complexes reveal the structural basis for the most potent and specific inhibitors of Grb7 developed to date. Finally, we demonstrate that cell permeable versions of these peptides successfully block Grb7 mediated interactions in a breast cancer cell line, establishing the potential of these peptides in the development of novel therapeutics targeted to Grb7.


Assuntos
Proteína Adaptadora GRB7/antagonistas & inibidores , Peptídeos Cíclicos/farmacologia , Linhagem Celular Tumoral , Cristalografia por Raios X , Descoberta de Drogas , Quinase 1 de Adesão Focal/metabolismo , Proteína Adaptadora GRB7/química , Proteína Adaptadora GRB7/metabolismo , Humanos , Lactamas/síntese química , Lactamas/química , Lactamas/farmacologia , Ligantes , Peptídeos Cíclicos/síntese química , Peptídeos Cíclicos/química , Fosfatos/química , Conformação Proteica , Receptor ErbB-2/metabolismo , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Domínios de Homologia de src
11.
Nat Commun ; 8(1): 1157, 2017 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-29079850

RESUMO

The mammalian pseudokinase SgK223, and its structurally related homologue SgK269, are oncogenic scaffolds that nucleate the assembly of specific signalling complexes and regulate tyrosine kinase signalling. Both SgK223 and SgK269 form homo- and hetero-oligomers, a mechanism that underpins a diversity of signalling outputs. However, mechanistic insights into SgK223 and SgK269 homo- and heterotypic association are lacking. Here we present the crystal structure of SgK223 pseudokinase domain and its adjacent N- and C-terminal helices. The structure reveals how the N- and C-regulatory helices engage in a novel fold to mediate the assembly of a high-affinity dimer. In addition, we identified regulatory interfaces on the pseudokinase domain required for the self-assembly of large open-ended oligomers. This study highlights the diversity in how the kinase fold mediates non-catalytic functions and provides mechanistic insights into how the assembly of these two oncogenic scaffolds is achieved in order to regulate signalling output.


Assuntos
Proteínas de Transporte/química , Proteínas Tirosina Quinases/química , Trifosfato de Adenosina/química , Área Sob a Curva , Sítios de Ligação , Catálise , Linhagem Celular Tumoral , Análise por Conglomerados , Cristalografia por Raios X , Células HEK293 , Humanos , Interações Hidrofóbicas e Hidrofílicas , Peptídeos e Proteínas de Sinalização Intracelular , Ligação Proteica , Domínios Proteicos , Multimerização Proteica , Proteínas Recombinantes/química
12.
Cancer Res ; 77(16): 4279-4292, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28634201

RESUMO

Despite intensive multimodal treatment of sarcomas, a heterogeneous group of malignant tumors arising from connective tissue, survival remains poor. Candidate-based targeted treatments have demonstrated limited clinical success, urging an unbiased and comprehensive analysis of oncogenic signaling networks to reveal therapeutic targets and personalized treatment strategies. Here we applied mass spectrometry-based phosphoproteomic profiling to the largest and most heterogeneous set of sarcoma cell lines characterized to date and identified novel tyrosine phosphorylation patterns, enhanced tyrosine kinases in specific subtypes, and potential driver kinases. ALK was identified as a novel driver in the Aska-SS synovial sarcoma (SS) cell line via expression of an ALK variant with a large extracellular domain deletion (ALKΔ2-17). Functional ALK dependency was confirmed in vitro and in vivo with selective inhibitors. Importantly, ALK immunopositivity was detected in 6 of 43 (14%) of SS patient specimens, one of which exhibited an ALK rearrangement. High PDGFRα phosphorylation also characterized SS cell lines, which was accompanied by enhanced MET activation in Yamato-SS cells. Although Yamato-SS cells were sensitive to crizotinib (ALK/MET-inhibitor) but not pazopanib (VEGFR/PDGFR-inhibitor) monotherapy in vitro, synergistic effects were observed upon drug combination. In vivo, both drugs were individually effective, with pazopanib efficacy likely attributable to reduced angiogenesis. MET or PDGFRα expression was detected in 58% and 84% of SS patients, respectively, with coexpression in 56%. Consequently, our integrated approach has led to the identification of ALK and MET as promising therapeutic targets in SS. Cancer Res; 77(16); 4279-92. ©2017 AACR.


Assuntos
Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Sarcoma Sinovial/tratamento farmacológico , Sarcoma Sinovial/enzimologia , Quinase do Linfoma Anaplásico , Animais , Linhagem Celular Tumoral , Estudos de Coortes , Crizotinibe , Feminino , Humanos , Indazóis , Camundongos , Camundongos Endogâmicos BALB C , Terapia de Alvo Molecular , Fosforilação , Proteômica , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/genética , Pirazóis/farmacologia , Piridinas/farmacologia , Pirimidinas/farmacologia , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/biossíntese , Sarcoma Sinovial/genética , Transdução de Sinais , Sulfonamidas/farmacologia , Sulfonas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Biol Chem ; 291(41): 21571-21583, 2016 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-27531744

RESUMO

SgK269/PEAK1 is a pseudokinase and scaffolding protein that plays a critical role in regulating growth factor receptor signal output and is implicated in the progression of several cancers, including those of the breast, colon, and pancreas. SgK269 is structurally related to SgK223, a human pseudokinase that also functions as a scaffold but recruits a distinct repertoire of signaling proteins compared with SgK269. Structural similarities between SgK269 and SgK223 include a predicted α-helical region (designated CH) immediately preceding the conserved C-terminal pseudokinase (PK) domain. Structure-function analyses of SgK269 in MCF-10A mammary epithelial cells demonstrated a critical role for the CH and PK regions in promoting cell migration and Stat3 activation. Characterization of the SgK269 "interactome" by mass spectrometry-based proteomics identified SgK223 as a novel binding partner, and association of SgK269 with SgK223 in cells was dependent on the presence of the CH and PK domains of both pseudokinases. Homotypic association of SgK269 and SgK223 was also demonstrated and exhibited the same structural requirements. Further analysis using pulldowns and size-exclusion chromatography underscored the critical role of the CH region in SgK269/SgK223 association. Importantly, although SgK269 bridged SgK223 to Grb2, it was unable to activate Stat3 or efficiently enhance migration in SgK223 knock-out cells generated by CRISPR/Cas9. These results reveal previously unrecognized interplay between two oncogenic scaffolds and demonstrate a novel signaling mechanism for pseudokinases whereby homotypic and heterotypic association is used to assemble scaffolding complexes with distinct binding properties and hence qualitatively regulate signal output.


Assuntos
Proteínas de Transporte/metabolismo , Movimento Celular/fisiologia , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais/fisiologia , Animais , Proteínas de Transporte/genética , Linhagem Celular , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Domínios Proteicos , Proteínas Tirosina Quinases/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo
14.
Sci Signal ; 8(380): rs6, 2015 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-26060331

RESUMO

A major goal in signaling biology is the establishment of high-throughput quantitative methods for measuring changes in protein phosphorylation of entire signal transduction pathways across many different samples comprising temporal or dose data or patient samples. Data-independent acquisition (DIA) mass spectrometry (MS) methods, which involve tandem MS scans that are collected independently of precursor ion information and then are followed by targeted searching for known peptides, may achieve this goal. We applied DIA-MS to systematically quantify phosphorylation of components in the insulin signaling network in response to insulin as well as in stimulated cells exposed to a panel of kinase inhibitors targeting key downstream effectors in the network. We accurately quantified the effect of insulin on phosphorylation of 86 protein targets in the insulin signaling network using either stable isotope standards (SIS) or label-free quantification (LFQ) and mapped signal transmission through this network. By matching kinases to specific phosphorylation events (based on linear consensus motifs and temporal phosphorylation) to the quantitative phosphoproteomic data from cells exposed to inhibitors, we investigated predicted kinase-substrate relationships of AKT and mTOR in a targeted fashion. Furthermore, we applied this approach to show that AKT2-dependent phosphorylation of GAB2 promoted insulin signaling but inhibited epidermal growth factor (EGF) signaling in a manner dependent on 14-3-3 binding. Because DIA-MS can increase throughput and improve the reproducibility of peptide detection across multiple samples, this approach should facilitate more accurate, comprehensive, and quantitative assessment of signaling networks under various experimental conditions than are possible using other MS proteomic methods.


Assuntos
Insulina/metabolismo , Fosfoproteínas/metabolismo , Proteômica/métodos , Transdução de Sinais , Espectrometria de Massas em Tandem/métodos , Proteínas 14-3-3/metabolismo , Células 3T3-L1 , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Western Blotting , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Células HEK293 , Humanos , Insulina/farmacologia , Camundongos , Fosfopeptídeos/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Treonina/metabolismo
15.
PLoS One ; 9(4): e95598, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24752197

RESUMO

The Niemann-Pick disease, type C1 (NPC1) gene encodes a transmembrane protein involved in cholesterol efflux from the lysosome. SNPs within NPC1 have been associated with obesity and type 2 diabetes, and mice heterozygous or null for NPC1 are insulin resistant. However, the molecular mechanism underpinning this association is currently undefined. This study aimed to investigate the effects of inhibiting NPC1 function on insulin action in adipocytes. Both pharmacological and genetic inhibition of NPC1 impaired insulin action. This impairment was evident at the level of insulin signalling and insulin-mediated glucose transport in the short term and decreased GLUT4 expression due to reduced liver X receptor (LXR) transcriptional activity in the long-term. These data show that cholesterol homeostasis through NPC1 plays a crucial role in maintaining insulin action at multiple levels in adipocytes.


Assuntos
Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Insulina/farmacologia , Proteínas/metabolismo , Células 3T3-L1 , Adipócitos/enzimologia , Androstenos/farmacologia , Animais , Células CHO , Cricetinae , Cricetulus , Ativação Enzimática/efeitos dos fármacos , Deleção de Genes , Técnicas de Inativação de Genes , Glucose/metabolismo , Transportador de Glucose Tipo 4/genética , Transportador de Glucose Tipo 4/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Receptores X do Fígado , Camundongos , Proteína C1 de Niemann-Pick , Receptores Nucleares Órfãos/metabolismo , Fenótipo , Transporte Proteico/efeitos dos fármacos , Proteínas/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
16.
J Biol Chem ; 288(20): 14417-14427, 2013 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-23580655

RESUMO

Signal transducers and activators of transcription 1 (STAT1) transduces signals from cytokines and growth factors, particularly IFN-γ, and regulates expression of genes involved in cell survival/death, proliferation, and migration. STAT1 is activated through phosphorylation on its tyrosine 701 by JAKs and is inactivated through dephosphorylation by tyrosine phosphatases. We discovered a natural compound, wedelolactone, that increased IFN-γ signaling by inhibiting STAT1 dephosphorylation and prolonging STAT1 activation through specific inhibition of T-cell protein tyrosine phosphatase (TCPTP), an important tyrosine phosphatase for STAT1 dephosphorylation. More interestingly, wedelolactone inhibited TCPTP through interaction with the C-terminal autoinhibition domain of TCPTP. We also found that wedelolactone synergized with IFN-γ to induce apoptosis of tumor cells. Our data suggest a new target for anticancer or antiproliferation drugs, a new mechanism to regulate PTPs specifically, and a new drug candidate for treating cancer or other proliferation disorders.


Assuntos
Antineoplásicos/farmacologia , Cumarínicos/farmacologia , Regulação Neoplásica da Expressão Gênica , Interferon gama/metabolismo , Fator de Transcrição STAT1/antagonistas & inibidores , Linhagem Celular Tumoral , Sobrevivência Celular , Ensaios de Seleção de Medicamentos Antitumorais , Genes Reporter , Células Hep G2 , Humanos , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação , Extratos Vegetais/farmacologia , Regiões Promotoras Genéticas , Interferência de RNA
17.
Biochem Pharmacol ; 84(4): 468-76, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22687625

RESUMO

Selectively eradicating cancer cells with minimum adverse effects on normal cells is a major challenge in the development of anticancer therapy. We hypothesize that nutrient-limiting conditions frequently encountered by cancer cells in poorly vascularized solid tumors might provide an opportunity for developing selective therapy. In this study, we investigated the function and molecular mechanisms of a natural compound, arctigenin, in regulating tumor cell growth. We demonstrated that arctigenin selectively promoted glucose-starved A549 tumor cells to undergo necrosis by inhibiting mitochondrial respiration. In doing so, arctigenin elevated cellular level of reactive oxygen species (ROS) and blocked cellular energy metabolism in the glucose-starved tumor cells. We also demonstrated that cellular ROS generation was caused by intracellular ATP depletion and played an essential role in the arctigenin-induced tumor cell death under the glucose-limiting condition. Furthermore, we combined arctigenin with the glucose analogue 2-deoxyglucose (2DG) and examined their effects on tumor cell growth. Interestingly, this combination displayed preferential cell-death inducing activity against tumor cells compared to normal cells. Hence, we propose that the combination of arctigenin and 2DG may represent a promising new cancer therapy with minimal normal tissue toxicity.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Furanos/farmacologia , Glucose/metabolismo , Lignanas/farmacologia , Trifosfato de Adenosina/metabolismo , Antineoplásicos Fitogênicos/isolamento & purificação , Arctium/química , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Metabolismo Energético/efeitos dos fármacos , Furanos/isolamento & purificação , Glucose/deficiência , Humanos , Lignanas/isolamento & purificação , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Necrose , Espécies Reativas de Oxigênio/metabolismo , Microambiente Tumoral
18.
Chem Biol ; 18(11): 1474-81, 2011 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-22118681

RESUMO

Autophagy is a cellular lysosome-dependent catabolic mechanism mediating the turnover of intracellular organelles and long-lived proteins. We show that antimycin A, a known inhibitor of mETC complex III, can inhibit autophagy. A structural and functional study shows that four close analogs of antimycin A that have no effect on mitochondria inhibition also do not inhibit autophagy, whereas myxothiazol, another mETC complex III inhibitor with unrelated structure to antimycin A, inhibits autophagy. Additionally, antimycin A and myxothiazol cannot inhibit autophagy in mtDNA-depleted H4 and mtDNA-depleted HeLa cells. These data suggest that antimycin A inhibits autophagy through its inhibitory activity on mETC complex III. Our data suggest that mETC complex III may have a role in mediating autophagy induction.


Assuntos
Antibacterianos/farmacologia , Antimicina A/farmacologia , Autofagia/efeitos dos fármacos , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Mitocôndrias/metabolismo , Trifosfato de Adenosina/metabolismo , Antimicina A/análogos & derivados , Linhagem Celular , DNA Mitocondrial/metabolismo , Humanos , Mitocôndrias/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
19.
Cell Signal ; 23(8): 1404-12, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21511030

RESUMO

Signal transducer and activator of transcription 1 (STAT1) is an important mediator for cytokine signal transduction, particularly IFN-γ. Following IFN-γ stimulation, STAT1 is activated through tyrosine phosphorylation. Little is known about the function and regulation of STAT1 dephosphorylation after activation. We studied the regulation and function of STAT1 dephosphorylation in different types of cells and found that the phosphorylated STAT1 was quickly dephosphorylated in most of epithelial cells. Further studies revealed that the dephosphorylation of STAT1 was regulated by cell shape/adhesion. Actin cytoskeleton and extracellular matrix (ECM) proteins mediated the STAT1 dephosphorylation through the T-cell protein tyrosine phosphatase TCPTP. Inactivation of the dephosphorylation system by cell detachment rendered the cells more sensitive to IFN-γ-induced cell death. Our results revealed a novel mechanism in regulating IFN-γ/STAT1 signaling. This cell adhesion and cell cytoskeleton-dependent STAT1 dephosphorylation system may have a role in IFN-γ-mediated immunosurveillance for cancer cells by inducing anoikis of detached metastatic cancer cells.


Assuntos
Interferon gama/farmacologia , Fator de Transcrição STAT1/metabolismo , Actinas/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Forma Celular , Citoesqueleto/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Humanos , Interferon gama/metabolismo , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 2/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 2/genética , Proteína Tirosina Fosfatase não Receptora Tipo 2/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais
20.
Cancer Res ; 69(18): 7302-10, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19706767

RESUMO

Constitutive activation of the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway occurs frequently in cancer cells and contributes to oncogenesis. Among the members of STAT family, STAT3 plays a pivotal role in the development and progression of human tumors. The STAT3-mediated signaling pathway has been recognized as a promising anticancer target. Here, we show that 17-Hydroxy-jolkinolide B (HJB), a diterpenoid from the Chinese medicinal herb Euphorbia fischeriana Steud, strongly inhibits interleukin (IL)-6-induced as well as constitutive STAT3 activation. Furthermore, we show that HJB directly targets the JAK family kinases, JAK1, JAK2, and TYK2, by inducing dimerization of the JAKs via cross-linking. Addition of DTT or glutathione prevents the JAK cross-linking and blocks the inhibitory effects of HJB on IL-6-induced STAT3 activation, suggesting that HJB may react with cystein residues of JAKs to form covalent bonds that inactivate JAKs. Liquid chromatography/mass spectrometry analysis confirmed that each HJB reacted with two thiols. The effect of HJB on the JAK/STAT3 pathway is specific as HJB has no effect on platelet-derived growth factor, epidermal growth factor, or insulin-like growth factor I signaling pathways. Finally, we show that HJB inhibits growth and induces apoptosis of tumor cells, particularly those tumor cells with constitutively activated STAT3. We propose that the natural compound HJB is a promising anticancer drug candidate as a potent STAT3 signaling inhibitor.


Assuntos
Diterpenos/farmacologia , Janus Quinases/metabolismo , Neoplasias/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Linhagem Celular Tumoral , Medicamentos de Ervas Chinesas/farmacologia , Euphorbia/química , Humanos , Interleucina-6/farmacologia , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/metabolismo , Janus Quinases/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tirfostinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA