Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Biol Chem ; 402(11): 1441-1452, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34280958

RESUMO

Angiogenesis is an important physiological process playing a crucial role in wound healing and cancer progression. Vascular endothelial growth factor (VEGF) and platelet derived growth factor (PDGF) are key players in angiogenesis. Based on previous findings regarding the modulation of VEGF activity by glycosaminoglycans (GAG), here we explore the interaction of hyaluronan (HA)-based GAG with PDGF and its receptor PDGFR-ß by applying molecular modeling and dynamics simulations in combination with surface plasmon resonance (SPR). Computational analysis on the interaction of oligo-hyaluronan derivatives with different sulfation pattern and functionalization shows that these GAG interact with PDGF in relevant regions for receptor recognition, and that high sulfation as well as modification with the TAMRA group convey stronger binding. On the other hand, the studied oligo-hyaluronan derivatives are predicted to scarcely recognize PDGFR-ß. SPR results are in line with the computational predictions regarding the binding pattern of HA tetrasaccharide (HA4) derivatives to PDGF and PDGFR-ß. Furthermore, our experimental results also show that the complexation of PDGF to PDGFR-ß can be modulated by HA4 derivatives. The results found open the path for considering HA4 derivatives as potential candidates to be exploited for modulation of the PDGF/PDGFR-ß signaling system in angiogenesis and related disease conditions.


Assuntos
Ácido Hialurônico/química , Fator de Crescimento Derivado de Plaquetas/química , Receptor beta de Fator de Crescimento Derivado de Plaquetas/química , Configuração de Carboidratos , Humanos , Modelos Moleculares , Proteínas Recombinantes/química , Ressonância de Plasmônio de Superfície
2.
J Mol Neurosci ; 70(12): 2102-2106, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32613555

RESUMO

The platelet-derived growth factor receptor beta (PDGFRB) gene is involved in proliferative and developmental processes in mammals. Variations in this gene lead to several different syndromic conditions, such as infantile myofibromatosis I, sporadic port-wine stain, primary familial brain calcification, and the Penttinen and overgrowth syndromes. Our objective was to investigate PDGFRB's genetic relationship to clinical conditions and evaluate the protein interactions using GeneNetwork, GeneMANIA, and STRING network databases. We have evidenced the gene's pleiotropy through its many connections and its link to syndromic conditions. Therefore, PDGFRB may be an important therapeutic target for treating such conditions.


Assuntos
Pleiotropia Genética , Predisposição Genética para Doença , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Acro-Osteólise/genética , Sítios de Ligação , Calcinose/genética , Transtornos do Crescimento/genética , Humanos , Deformidades Congênitas dos Membros/genética , Miofibromatose/congênito , Miofibromatose/genética , Mancha Vinho do Porto/genética , Progéria/genética , Mapas de Interação de Proteínas , Receptor beta de Fator de Crescimento Derivado de Plaquetas/química , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo
3.
J Mol Biol ; 431(19): 3753-3770, 2019 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-31301406

RESUMO

Specific interactions between the helical membrane-spanning domains of transmembrane proteins play central roles in the proper folding and oligomerization of these proteins. However, the relationship between the hydrophobic amino acid sequences of transmembrane domains and their functional interactions is in most cases unknown. Here, we use ultra-simple artificial proteins to systematically study the sequence basis for transmembrane domain interactions. We show that most short homopolymeric polyleucine transmembrane proteins containing single amino acid substitutions can activate the platelet-derived growth factor ß receptor or the erythropoietin receptor in cultured mouse cells, resulting in cell transformation or proliferation. These proteins displayed complex patterns of activity that were markedly affected by seemingly minor sequence differences in the ultra-simple protein itself or in the transmembrane domain of the target receptor, and the effects of these sequence differences are not additive. In addition, specific leucine residues along the length of these proteins are required for activity, and the positions of these required leucines differ based on the identity and position of the central substituted amino acid. Our results suggest that these ultra-simple proteins use a variety of molecular mechanisms to activate the same target and that diversification of transmembrane domain sequences over the course of evolution minimized off-target interactions.


Assuntos
Mapeamento de Interação de Proteínas , Proteínas/química , Proteínas/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Linhagem Celular , Humanos , Camundongos , Mutagênese/genética , Mutação/genética , Peptídeos/metabolismo , Domínios Proteicos , Receptor beta de Fator de Crescimento Derivado de Plaquetas/química , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores da Eritropoetina/química , Receptores da Eritropoetina/metabolismo
4.
Exp Cell Res ; 380(1): 69-79, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30970237

RESUMO

The role of plasma membrane composition and dynamics in the activation process of receptor tyrosine kinases (RTKs) is still poorly understood. In this study we have investigated how signaling via the RTK, platelet-derived growth factor ß-receptor (PDGFR-ß) is affected by Dynasore or Dyngo-4a, which are commonly used dynamin inhibitors. PDGFR-ß preferentially internalizes via clathrin-coated pits and in this pathway, Dynamin II has a major role in the formation and release of vesicles from the plasma membrane by performing the membrane scission. We have found that dynamin inhibitors impedes the activation of PDGFR-ß by impairing ligand-induced dimerization of the receptor monomers, which leads to a subsequent lack of phosphorylation and activation both of receptors and downstream effectors, such as ERK1/2 and AKT. In contrast, dynamin inhibitors did not affect epidermal growth factor receptor (EGFR) dimerization and phosphorylation. Our findings suggest that there is a link between plasma membrane dynamics and PDGFR-ß activation, and that this link is not shared with the epidermal growth factor receptor.


Assuntos
Dinaminas/genética , Multimerização Proteica/efeitos dos fármacos , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Transdução de Sinais/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Dinaminas/antagonistas & inibidores , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Fibroblastos/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Humanos , Hidrazonas/farmacologia , Ligantes , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Naftóis/farmacologia , Fosforilação/efeitos dos fármacos , Multimerização Proteica/genética , Proteínas Proto-Oncogênicas c-akt/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/química , Transdução de Sinais/genética
5.
J Control Release ; 302: 63-78, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-30930215

RESUMO

The delivery of anticancer drugs is hampered by tumor vessels with abnormal structure and function, which requires that vessel normalization be mediated by pharmaceutics. The current strategies for vessel normalization focus on direct modulation of endothelial cells (ECs), which frequently affect vessels in normal tissues. Modulating EC-supporting cells, such as pericytes (PCs), is a new direction. Here, we produced a fusion protein, Z-TNFα, by fusing the platelet-derived growth factor receptor ß (PDGFRß)- antagonistic affibody ZPDGFRß to tumor necrosis factor α (TNFα). Owing to the affinity of fused ZPDGFRß for PDGFRß, Z-TNFα binds PDGFRß+ PCs but not PDGFRß- ECs. Low-dose (1 µg/mouse) Z-TNFα treatment remodeled the tumor vessels, thus reducing vessel permeability and increasing vessel perfusion. As a result, the Z-TNFα treatment improved the delivery of doxorubicin (DOX) and enhanced its antitumor effect, indicating that Z-TNFα induced normalization of tumor vessels. Mechanically, the tumor vessel normalization mediated by Z-TNFα might be attributed to the reduction of vascular endothelial growth factor (VEGF) secretion by PCs and the elevated expression of intercellular cell adhesion molecule-1 (ICAM-1) in PCs, which might suppress the proliferation and migration of ECs and simultaneously trigger interaction between perivascular macrophages and PCs. These results demonstrated that tumor-associated PCs could be considered novel target cells for vessel normalization, and Z-TNFα might be developed as a potential tool for antitumor combination therapy.


Assuntos
Antineoplásicos/química , Doxorrubicina/química , Portadores de Fármacos/química , Pericitos/química , Receptor beta de Fator de Crescimento Derivado de Plaquetas/química , Proteínas Recombinantes de Fusão/química , Fator de Necrose Tumoral alfa/química , Animais , Antineoplásicos/farmacologia , Transporte Biológico , Linhagem Celular Tumoral , Técnicas de Química Combinatória/métodos , Doxorrubicina/farmacologia , Liberação Controlada de Fármacos , Células Endoteliais/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Neoplasias Experimentais , Inibidores de Proteínas Quinases/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Resultado do Tratamento , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular
6.
J Mol Graph Model ; 82: 145-156, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29738888

RESUMO

PDGFRß/PDGF-B interaction plays a role in angiogenesis, and is mandatory in wound healing and cancer treatment. It has been reported that the PDGF-B aptamer was able to bind to PDGF-B, thus regulating the angiogenesis. However, the binding interaction between the aptamer and the growth factor, including the binding sites, has not been well investigated. This study applied a molecular dynamics (MD) simulation to investigate the aptamer-growth factor interaction in the presence or absence of a receptor (PDGFRß). Characterization of the structure of an aptamer-growth factor complex revealed binding sites from each section in the complex. Upon the complex formation, PDGF-B and its aptamer exhibited less flexibility in their molecular movement, as indicated by the minimum values of RMSD, RMSF, loop-to-loop distance, and the summation of PCA eigenvalues. Our study of residue pairwise interaction demonstrated that the binding interaction was mainly contributed by electrostatic interaction between the positively-charged amino acid and the negatively-charged phosphate backbone. The role of the PDGF-B aptamer in PDGFRß/PDGF-B interaction was also investigated. We demonstrated that the stability of the Apt-PDGF-B complex could prevent the presence of a competitor, of PDGFRß, interrupting the binding process. Because the aptamer was capable of binding with PDGF-B, and blocking the growth factor from the PDGFRß, it could down regulate the consequent signaling pathway. We provide evidence that the PDGF-BB aptamer is a promising molecule for regulation of angiogenesis. The MD study provides a molecular understanding to modification of the aptamer binding interaction, which could be used in a number of medical applications.


Assuntos
Aptâmeros de Nucleotídeos/química , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Receptor beta de Fator de Crescimento Derivado de Plaquetas/química , Sequência de Aminoácidos , Aptâmeros de Nucleotídeos/metabolismo , Sequência de Bases , Conformação Molecular , Ligação Proteica , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo
7.
Cell Mol Biol Lett ; 23: 24, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29785186

RESUMO

BACKGROUND: Patients with anti-ß2GPI antibodies display significantly higher platelet activation/aggregation and vascular endothelial cell damage. The mechanism underlying the correlation between platelet activation, vascular endothelial cell dysfunctions and anti-ß2GPI antibodies remains unknown. METHODS: In this study, we derived miR-96 and -26a from platelets activated by the anti-ß2GPI/ß2GPI complex and explored their role in modulating human umbilical vein endothelial cell (HUVEC) migration and tube formation. RESULTS: Anti-ß2GPI/ß2GPI complex induces the release of platelet-derived microparticles (p-MPs). The amounts of miR-96 and -26a in these p-MPs were also higher than for the control group. Co-incubation of HUVECs with p-MPs resulted in the transfer of miR-96 and -26a into HUVECs, where they inhibited migration and tube formation. The targeting role of these miRNAs was further validated by directly downregulating targeted selectin-P (SELP) and platelet-derived growth factor receptor alpha (PDGFRA) via luciferase activity assay. CONCLUSION: Our study suggests that miR-96 and -26a in p-MPs can inhibit HUVEC behavior by targeting SELP and PDGFRA.


Assuntos
Complexo Antígeno-Anticorpo/farmacologia , Plaquetas/efeitos dos fármacos , MicroRNAs/metabolismo , beta 2-Glicoproteína I/imunologia , Regiões 3' não Traduzidas , Antagomirs/metabolismo , Plaquetas/citologia , Plaquetas/metabolismo , Movimento Celular , Micropartículas Derivadas de Células/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Neovascularização Fisiológica , Selectina-P/química , Selectina-P/genética , Selectina-P/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/química , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , beta 2-Glicoproteína I/metabolismo
9.
J Am Chem Soc ; 139(22): 7456-7475, 2017 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-28471683

RESUMO

The platelet-derived growth factor receptor ß (PDGFR-ß) signaling pathway is a validated and important target for the treatment of certain malignant and nonmalignant pathologies. We previously identified a G-quadruplex-forming nuclease hypersensitive element (NHE) in the human PDGFR-ß promoter that putatively forms four overlapping G-quadruplexes. Therefore, we further investigated the structures and biological roles of the G-quadruplexes and i-motifs in the PDGFR-ß NHE with the ultimate goal of demonstrating an alternate and effective strategy for molecularly targeting the PDGFR-ß pathway. Significantly, we show that the primary G-quadruplex receptor for repression of PDGFR-ß is the 3'-end G-quadruplex, which has a GGA sequence at the 3'-end. Mutation studies using luciferase reporter plasmids highlight a novel set of G-quadruplex point mutations, some of which seem to provide conflicting results on effects on gene expression, prompting further investigation into the effect of these mutations on the i-motif-forming strand. Herein we characterize the formation of an equilibrium between at least two different i-motifs from the cytosine-rich (C-rich) sequence of the PDGFR-ß NHE. The apparently conflicting mutation results can be rationalized if we take into account the single base point mutation made in a critical cytosine run in the PDGFR-ß NHE that dramatically affects the equilibrium of i-motifs formed from this sequence. We identified a group of ellipticines that targets the G-quadruplexes in the PDGFR-ß promoter, and from this series of compounds, we selected the ellipticine analog GSA1129, which selectively targets the 3'-end G-quadruplex, to shift the dynamic equilibrium in the full-length sequence to favor this structure. We also identified a benzothiophene-2-carboxamide (NSC309874) as a PDGFR-ß i-motif-interactive compound. In vitro, GSA1129 and NSC309874 downregulate PDGFR-ß promoter activity and transcript in the neuroblastoma cell line SK-N-SH at subcytotoxic cell concentrations. GSA1129 also inhibits PDGFR-ß-driven cell proliferation and migration. With an established preclinical murine model of acute lung injury, we demonstrate that GSA1129 attenuates endotoxin-mediated acute lung inflammation. Our studies underscore the importance of considering the effects of point mutations on structure formation from the G- and C-rich sequences and provide further evidence for the involvement of both strands and associated structures in the control of gene expression.


Assuntos
Motivos de Aminoácidos , Desoxirribonucleases/química , Sistemas de Liberação de Medicamentos , Quadruplex G , Receptor beta de Fator de Crescimento Derivado de Plaquetas/química , Sequência de Bases , Regulação para Baixo , Quadruplex G/efeitos dos fármacos , Redes Reguladoras de Genes , Humanos , Mutação , Regiões Promotoras Genéticas
11.
Oncotarget ; 7(38): 62607-62618, 2016 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-27566585

RESUMO

Effective delivery of nanomedicines to tumor tissues depends on both the tumor microenvironment and nanomedicine properties. Accordingly, tumor microenvironment modification or advanced design of nanomedicine was emerging to improve nanomedicine delivery to tumors. However, few studies have emphasized the necessity to optimize the tumor microenvironment and nanomedicine properties simultaneously to improve tumor treatment. In the present study, imatinib mesylate (IMA) was used to normalize the tumor microenvironment including platelet-derived growth factor receptor-ß expression inhibition, tumor vessel normalization, and tumor perfusion improvement as demonstrated by immunofluorescence staining. In addition, the effect of tumor microenvironment normalization on tumor delivery of nanomedicines with different sizes was carefully investigated. It was shown that IMA treatment significantly reduced the accumulation of nanoparticles (NPs) around 110 nm but enhanced the accumulation of micelles around 23 nm by in vivo fluorescence imaging experiment. Furthermore, IMA treatment limited the distribution of NPs inside tumors but increased that of micelles with a more homogeneous pattern. Finally, the anti-tumor efficacy study displayed that IMA pretreatment could significantly increase the therapeutic effects of paclitaxel-loaded micelles. All-together, a new strategy to improve nanomedicine delivery to tumor was provided by optimizing both nanomedicine size and the tumor microenvironment simultaneously, and it will have great potential in clinics for tumor treatment.


Assuntos
Sistemas de Liberação de Medicamentos , Nanomedicina/métodos , Microambiente Tumoral/efeitos dos fármacos , Células A549 , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Mesilato de Imatinib/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Micelas , Microscopia de Fluorescência , Nanopartículas , Transplante de Neoplasias , Neovascularização Patológica , Paclitaxel/administração & dosagem , Perfusão , Receptor beta de Fator de Crescimento Derivado de Plaquetas/química
12.
J Med Chem ; 59(17): 7901-14, 2016 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-27502700

RESUMO

A series of potent PDGFR inhibitors has been identified. The series was optimized for duration of action in the lung. A novel kinase occupancy assay was used to directly measure target occupancy after i.t. dosing. Compound 25 shows 24 h occupancy of the PDGFR kinase domain, after a single i.t. dose and has efficacy at 0.03 mg/kg, in the rat moncrotaline model of pulmonary arterial hypertension. Examination of PK/PD data from the optimization effort has revealed in vitro:in vivo correlations which link duration of action in vivo with low permeability and high basicity and demonstrate that nonspecific binding to lung tissue increases with lipophilicity.


Assuntos
Remodelação das Vias Aéreas/efeitos dos fármacos , Hipertensão Pulmonar/tratamento farmacológico , Niacinamida/análogos & derivados , Pirazóis/química , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Remodelação Vascular/efeitos dos fármacos , Administração por Inalação , Animais , Linhagem Celular , Proliferação de Células , Hipertensão Pulmonar/patologia , Pulmão/irrigação sanguínea , Membranas Artificiais , Simulação de Acoplamento Molecular , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Niacinamida/síntese química , Niacinamida/química , Niacinamida/farmacologia , Permeabilidade , Proteínas Proto-Oncogênicas c-kit/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-kit/química , Pirazóis/síntese química , Pirazóis/farmacologia , Ratos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/química , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptor beta de Fator de Crescimento Derivado de Plaquetas/química , Receptores do Fator de Crescimento Derivado de Plaquetas/química , Relação Estrutura-Atividade
13.
J Mol Biol ; 427(24): 3921-34, 2015 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-26463591

RESUMO

Members of the receptor tyrosine kinases (RTKs) regulate important cellular functions such as cell growth and migration, which are key steps in angiogenesis, in organ morphogenesis and in the unregulated states, cancer formation. One long-standing puzzle regarding RTKs centers on how the extracellular domain (ECD), which detects and binds to growth factors, is coupled with the intracellular domain kinase activation. While extensive structural works on the soluble portions of RTKs have provided critical insights into RTK structures and functions, lack of a full-length receptor structure has hindered a comprehensive overview of RTK activation. In this study, we successfully purified and determined a 27-Å-resolution structure of PDGFRß [a full-length human platelet-derived growth factor receptor], in complex with its ligand PDGF-B. In the ligand-stimulated complex, two PDGFRßs assemble into a dimer via an extensive interface essentially running along the full-length of the receptor, suggesting that the membrane-proximal region, the transmembrane helix and the kinase domain of PDGFRß are involved in dimerization. Major structural differences are seen between the full-length and soluble ECD structures, rationalizing previous experimental data on how membrane-proximal domains modulate receptor ligand-binding affinity and dimerization efficiency. Also, in contrast to the 2-fold symmetry of the ECD, the intracellular kinase domains adopt an asymmetric dimer arrangement, in agreement with prior observations for the closely related KIT receptor. In essence, the structure provides a first glimpse into how platelet-derived growth factor receptor ECD, upon ligand stimulation, is coupled to its intracellular domain kinase activation.


Assuntos
Proteínas Proto-Oncogênicas c-sis/química , Receptor beta de Fator de Crescimento Derivado de Plaquetas/química , Domínio Catalítico , Células HEK293 , Humanos , Microscopia Eletrônica , Modelos Moleculares , Ligação Proteica , Estrutura Quaternária de Proteína , Proteínas Proto-Oncogênicas c-sis/ultraestrutura , Receptor beta de Fator de Crescimento Derivado de Plaquetas/ultraestrutura , Coloração e Rotulagem
14.
J Natl Compr Canc Netw ; 13(10): 1181-5, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26483058

RESUMO

With the advent of widespread tumor genetic profiling, an increased number of mutations with unknown significance are being identified. Often, a glut of uninterpretable findings may confuse the clinician and provide little or inappropriate guidance in therapeutic decision-making. This report describes a method of protein modeling by in silico analysis (ie, using computer simulation) that is easily accessible to the practicing clinician without need for further laboratory analysis, which can potentially serve as a guide in therapeutic decisions based on poorly characterized tumor mutations. An example of this model is given wherein poorly characterized KIT, PDGFRB, and ERBB2 mutations were discovered in a patient with treatment-refractory metastatic transitional cell carcinoma of the renal pelvis. The KIT and PDGFRB mutations were predicted to be pathogenic using in silico analysis, whereas the ERBB2 mutation was predicted to be benign. Based on these findings, the patient was treated with pazopanib and achieved a partial response that lasted for 7.5 months. We propose that in silico analysis be explored as a potential means to further characterize genetic abnormalities found by tumor profiling assays, such as next-generation sequencing.


Assuntos
Carcinoma de Células de Transição/genética , Proteínas Proto-Oncogênicas c-kit/genética , Receptor ErbB-2/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Carcinoma de Células de Transição/tratamento farmacológico , Carcinoma de Células de Transição/patologia , Simulação por Computador , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Indazóis , Pelve Renal/efeitos dos fármacos , Pelve Renal/patologia , Pessoa de Meia-Idade , Modelos Moleculares , Mutação , Metástase Neoplásica , Proteínas Proto-Oncogênicas c-kit/química , Pirimidinas/uso terapêutico , Receptor ErbB-2/química , Receptor beta de Fator de Crescimento Derivado de Plaquetas/química , Sulfonamidas/uso terapêutico , Urotélio/efeitos dos fármacos , Urotélio/patologia
15.
ChemMedChem ; 10(8): 1335-8, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26076342

RESUMO

Imatinib is the first protein kinase inhibitor approved for clinical use and is a seminal drug for the concept of targeted therapy. Herein we report on the design, synthesis, photokinetic properties, and in vitro enzymatic evaluation of a photoactivatable caged prodrug of imatinib. This approach allows spatial and temporal control over the activation of imatinib triggered by ultraviolet light. The successful application of the photoactivation concept to this significant kinase inhibitor provides further evidence for the caging technique as a feasible approach in the kinase field. The presented photoactivatable imatinib prodrug will be highly useful as a pharmacological tool to study the impact of imatinib toward biological systems in greater detail.


Assuntos
Desenho de Fármacos , Mesilato de Imatinib/química , Pró-Fármacos/química , Inibidores de Proteínas Quinases/síntese química , Sítios de Ligação , Domínio Catalítico , Mesilato de Imatinib/síntese química , Simulação de Acoplamento Molecular , Pró-Fármacos/síntese química , Inibidores de Proteínas Quinases/química , Receptor beta de Fator de Crescimento Derivado de Plaquetas/química , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Raios Ultravioleta
16.
Front Biosci (Elite Ed) ; 7(3): 434-46, 2015 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-25961423

RESUMO

Platelet-derived growth factor receptor alpha (PDGFRalpha) interacts with PDGFs A, B, C and AB, while PDGFRbeta binds to PDGFs B and D, thus suggesting that PDGFRalpha is more promiscuous than PDGFRbeta. The structural analysis of PDGFRalpha-PDGFA and PDGFRalpha-PDGFB complexes and a molecular explanation for the promiscuity of PDGFRalpha and the specificity of PDGFRbeta remain unclear. In the present study, we modeled the three extracellular domains of PDGFRalpha using a previous crystallographic structure of PDGFRbeta as a template. Additionally, we analyzed the interacting residues of PDGFRalpha-PDGFA and PDGFRalpha-PDGFB complexes using docking simulations. The validation of the resulting complexes was evaluated by molecular dynamics simulations. Structural analysis revealed that changes of non-aromatic amino acids in PDGFRalpha to aromatic amino acids in PDGFRbeta (I139F, P267F and N204Y) may be involved in the promiscuity of PDGFRalpha. Indeed, substitution of amino acids with few probabilities of rotamer changes in PDGFRbeta (M133A, N163E and N179S) and energy stability due to the formation of hydrogen bond in PDGFRbeta could explain the specificity of PDGFRbeta. These results may be used as an input for a better and more specific drug and peptide design targeting diseases related with the malfunction of PDGFs and PDGFRalpha such as cancer and atherosclerosis.


Assuntos
Receptor beta de Fator de Crescimento Derivado de Plaquetas/química , Sítios de Ligação , Humanos , Modelos Moleculares , Simulação de Acoplamento Molecular , Estrutura Terciária de Proteína , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Alinhamento de Sequência , Análise de Sequência de Proteína , Transdução de Sinais
17.
Bioorg Med Chem ; 23(10): 2408-23, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25882519

RESUMO

A series of eleven conformationally restricted, 4-substituted 2,6-dimethylfuro[2,3-d]pyrimidines was designed to explore the bioactive conformation required for dual inhibition of microtubule assembly and receptor tyrosine kinases (RTKs), and their biological activities are reported. All three rotatable single bonds in the lead compound 1 were sequentially restricted to address the role of each in SAR for microtubule and RTK inhibitory effects. Compounds 2, 3, 7 and 10 showed microtubule depolymerizing activity comparable to or better than the lead 1, some with nanomolar EC50 values. While compound 8 had no effect on microtubules, 8 and 10 both showed potent RTK inhibition with nanomolar IC50s. These compounds confirm that the bioactive conformation for RTK inhibition is different from that for tubulin inhibition. The tetrahydroquinoline analog 10 showed the most potent dual tubulin and RTK inhibitory activities (low nanomolar inhibition of EGFR, VEGFR2 and PDGFR-ß). Compound 10 has highly potent activity against many NCI cancer cell lines, including several chemo-resistant cell lines, and could serve as a lead for further preclinical studies.


Assuntos
Inibidores da Angiogênese/síntese química , Furanos/síntese química , Microtúbulos/efeitos dos fármacos , Inibidores de Proteínas Quinases/síntese química , Pirimidinas/síntese química , Moduladores de Tubulina/síntese química , Inibidores da Angiogênese/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Embrião de Galinha , Desenho de Fármacos , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/química , Receptores ErbB/metabolismo , Furanos/farmacologia , Humanos , Concentração Inibidora 50 , Microtúbulos/química , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Neovascularização Fisiológica/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptor beta de Fator de Crescimento Derivado de Plaquetas/química , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Relação Estrutura-Atividade , Moduladores de Tubulina/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/química , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
18.
Biochem Biophys Res Commun ; 459(3): 443-9, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25744030

RESUMO

Neuropilin-1 (NRP-1) is involved in angiogenesis, but the role of NRP-1 in megakaryocytopoiesis is not yet fully understood. In this study, we investigated whether thrombopoietin (TPO) regulates the expression of platelet-derived growth factor (PDGF) and its receptors (PDGFRs) on TPO-dependent UT-7/TPO cells and whether PDGFRs and NRP-1 on UT-7/TPO cells form complexes during megakaryocytic differentiation. When UT-7/TPO cells were starved of TPO for 24 h and then stimulated with 5 ng/ml TPO, the expression of PDGF-B, PDGFRα, and PDGFRß were significantly up-regulated after the addition of TPO. TPO also induced tyrosine phosphorylation of PDGFRα but not PDGFRß, and promoted the formation of PDGFRαß heterodimer complexes. Furthermore, megakaryocytic differentiation of UT-7/TPO cells on treatment with phorbol myristate acetate (PMA) was accompanied by a marked up-regulation of PDGFRß and NRP-1 protein expression, complex formation between PDGFRs and NRP-1, PDGFRαß heterodimer complexes, and an increase in PDGF-BB-binding activity. Immunocytochemistry confirmed complex formation between PDGFRs and NRP-1 and PDGFRαß heterodimer complexes in PMA-differentiated UT-7/TPO cells. Our observations suggest that NRP-1 is involved in megakaryocytopoiesis through complex formation with PDGFRs, and that NRP-1-PDGFR-complexes may contribute to effective cellular functions mediated by TPO and PDGF in megakaryocytic cells.


Assuntos
Megacariócitos/citologia , Megacariócitos/metabolismo , Neuropilina-1/química , Neuropilina-1/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/química , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/química , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Trombopoese/fisiologia , Trombopoetina/metabolismo , Becaplermina , Linhagem Celular , Humanos , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas c-sis/química , Proteínas Proto-Oncogênicas c-sis/genética , Proteínas Proto-Oncogênicas c-sis/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Proteínas Recombinantes/metabolismo , Trombopoese/genética
19.
Cell Signal ; 27(2): 267-74, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25446260

RESUMO

SLAP (Src like adaptor protein) contains adjacent Src homology 3 (SH3) and Src homology 2 (SH2) domains closely related in sequence to that of cytoplasmic Src family tyrosine kinases. Expressed most abundantly in the immune system, SLAP function has been predominantly studied in the context of lymphocyte signaling, where it functions in the Cbl dependent downregulation of antigen receptor signaling. However, accumulating evidence suggests that SLAP plays a role in the regulation of a broad range of membrane receptors including members of the receptor tyrosine kinase (RTK) family. In this review we highlight the role of SLAP in the ubiquitin dependent regulation of type III RTKs PDGFR, CSF-1R, KIT and Flt3, as well as Eph family RTKs. SLAP appears to bind activated type III and Eph RTKs via a conserved autophosphorylated juxtamembrane tyrosine motif in an SH2-dependent manner, suggesting that SLAP is important in regulating RTK signaling.


Assuntos
Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Humanos , Proteínas Proto-Oncogênicas c-kit/química , Proteínas Proto-Oncogênicas c-kit/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/química , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/química , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/química , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Tirosina Quinase 3 Semelhante a fms/química , Tirosina Quinase 3 Semelhante a fms/metabolismo , Domínios de Homologia de src
20.
J Med Chem ; 58(1): 170-82, 2015 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-25007344

RESUMO

In this study we report on the hit optimization of substituted 3,5-diaryl-pyrazin-2(1H)-ones toward potent and effective platelet-derived growth factor receptor (PDGF-R) ß-inhibitors. Originally, the 3,5-diaryl-pyrazin-2-one core was derived from the marine sponge alkaloid family of hamacanthins. In our first series compound 2 was discovered as a promising hit showing strong activity against PDGF-Rß in the kinase assay (IC50 = 0.5 µM). Furthermore, 2 was shown to be selective for PDGF-Rß in a panel of 24 therapeutically relevant protein kinases. Molecular modeling studies on a PDGF-Rß homology model using prediction of water thermodynamics suggested an optimization strategy for the 3,5-diaryl-pyrazin-2-ones as DFG-in binders by using a phenolic OH function to replace a structural water molecule in the ATP binding site. Indeed, we identified compound 38 as a highly potent inhibitor with an IC50 value of 0.02 µM in a PDGF-Rß enzymatic assay also showing activity against PDGF-R dependent cancer cells.


Assuntos
Inibidores de Proteínas Quinases/química , Receptor beta de Fator de Crescimento Derivado de Plaquetas/química , Bibliotecas de Moléculas Pequenas/química , Termodinâmica , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Sítios de Ligação , Ligação Competitiva , Humanos , Modelos Químicos , Modelos Moleculares , Estrutura Molecular , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Estrutura Terciária de Proteína , Pirazinas/química , Pirazinas/metabolismo , Pirazinas/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Água/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA