Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
1.
Methods Enzymol ; 667: 633-662, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35525557

RESUMO

Obtaining high-resolution structures of Receptor Tyrosine Kinases that visualize extracellular, transmembrane and intracellular kinase regions simultaneously is an eagerly pursued but still unmet challenge of structural biology. The Human Epidermal Growth Factor Receptor 3 (HER3) that has a catalytically inactive kinase domain (pseudokinase) forms a potent signaling complex upon binding of growth factor neuregulin 1ß (NRG1ß) and upon dimerization with a close homolog, the HER2 receptor. The HER2/HER3/NRG1ß complex is often referred to as an oncogenic driver in breast cancer and is an attractive target for anti-cancer therapies. After overcoming significant hurdles in isolating sufficient amounts of the HER2/HER3/NRG1ß complex for structural studies by cryo-electron microscopy (cryo-EM), we recently obtained the first high-resolution structures of the extracellular portion of this complex. Here we describe a step-by-step protocol for obtaining a stable and homogenous HER2/HER3/NRG1ß complex for structural studies and our recommendation for collecting and processing cryo-EM data for this sample. We also show improved EM density for the transmembrane and kinase domains of the receptors, which continue to evade structural determination at high resolution. The discussed strategies are tunable and applicable to other membrane receptor complexes.


Assuntos
Neoplasias da Mama , Receptor ErbB-3 , Neoplasias da Mama/metabolismo , Microscopia Crioeletrônica , Feminino , Humanos , Ligantes , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Receptor ErbB-3/química , Receptor ErbB-3/metabolismo
2.
Nature ; 600(7888): 339-343, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34759323

RESUMO

Human epidermal growth factor receptor 2 (HER2) and HER3 form a potent pro-oncogenic heterocomplex1-3 upon binding of growth factor neuregulin-1ß (NRG1ß). The mechanism by which HER2 and HER3 interact remains unknown in the absence of any structures of the complex. Here we isolated the NRG1ß-bound near full-length HER2-HER3 dimer and, using cryo-electron microscopy, reconstructed the extracellulardomain module, revealing unexpected dynamics at the HER2-HER3 dimerization interface. We show that the dimerization arm of NRG1ß-bound HER3 is unresolved because the apo HER2 monomer does not undergo a ligand-induced conformational change needed to establish a HER3 dimerization arm-binding pocket. In a structure of the oncogenic extracellular domain mutant HER2(S310F), we observe a compensatory interaction with the HER3 dimerization arm that stabilizes the dimerization interface. Both HER2-HER3 and HER2(S310F)-HER3 retain the capacity to bind to the HER2-directed therapeutic antibody trastuzumab, but the mutant complex does not bind to pertuzumab. Our structure of the HER2(S310F)-HER3-NRG1ß-trastuzumab Fab complex reveals that the receptor dimer undergoes a conformational change to accommodate trastuzumab. Thus, similar to oncogenic mutations, therapeutic agents exploit the intrinsic dynamics of the HER2-HER3 heterodimer. The unique features of a singly liganded HER2-HER3 heterodimer underscore the allosteric sensing of ligand occupancy by the dimerization interface and explain why extracellular domains of HER2 do not homo-associate via a canonical active dimer interface.


Assuntos
Microscopia Crioeletrônica , Neuregulina-1/química , Multimerização Proteica , Receptor ErbB-2/química , Receptor ErbB-3/química , Regulação Alostérica , Anticorpos Monoclonais Humanizados/química , Anticorpos Monoclonais Humanizados/ultraestrutura , Sítios de Ligação , Humanos , Fragmentos Fab das Imunoglobulinas/química , Modelos Moleculares , Mutação , Neuregulina-1/ultraestrutura , Oncogenes/genética , Estabilidade Proteica , Receptor ErbB-2/ultraestrutura , Receptor ErbB-3/ultraestrutura , Trastuzumab/química , Trastuzumab/ultraestrutura
3.
PLoS One ; 16(10): e0259426, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34714866

RESUMO

ERBB family members and their ligands play an essential role in embryonic heart development and adult heart physiology. Among them, ERBB3 is a binding partner of ERBB2; the ERBB2/3 complex mediates downstream signaling for cell proliferation. ERBB3 has seven consensus binding sites to the p85 regulatory subunit of PI3K, which activates the downstream AKT pathway, leading to the proliferation of various cells. This study generated a human ERBB3 knock-in mouse expressing a mutant ERBB3 whose seven YXXM p85 binding sites were replaced with YXXA. Erbb3 knock-in embryos exhibited lethality between E12.5 to E13.5, and showed a decrease in mesenchymal cell numbers and density in AV cushions. We determined that the proliferation of mesenchymal cells in the atrioventricular (AV) cushion in Erbb3 knock-in mutant embryos was temporarily reduced due to the decrease of AKT and ERK1/2 phosphorylation. Overall, our results suggest that AKT/ERK activation by the ERBB3-dependent PI3K signaling is crucial for AV cushion morphogenesis during embryonic heart development.


Assuntos
Comunicação Atrioventricular/genética , Coxins Endocárdicos/metabolismo , Receptor ErbB-3/metabolismo , Animais , Sítios de Ligação , Células CHO , Cricetinae , Cricetulus , Comunicação Atrioventricular/metabolismo , Coxins Endocárdicos/embriologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor ErbB-3/química , Receptor ErbB-3/genética , Transdução de Sinais
4.
Acta Crystallogr F Struct Biol Commun ; 77(Pt 7): 192-201, 2021 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-34196609

RESUMO

The members of the human epidermal growth factor receptor (HER) family are among the most intensely studied oncological targets. HER3 (ErbB3), which had long been neglected, has emerged as a key oncogene, regulating the activity of other receptors and being involved in progression and tumor escape in multiple types of cancer. Designed ankyrin-repeat proteins (DARPins) serve as antibody mimetics that have proven to be useful in the clinic, in diagnostics and in research. DARPins have previously been selected against EGFR (HER1), HER2 and HER4. In particular, their combination into bivalent binders that separate or lock receptors in their inactive conformation has proved to be a promising strategy for the design of potent anticancer therapeutics. Here, the selection of DARPins targeting extracellular domain 4 of HER3 (HER3d4) is described. One of the selected DARPins, D5, in complex with HER3d4 crystallized in two closely related crystal forms that diffracted to 2.3 and 2.0 Šresolution, respectively. The DARPin D5 epitope comprises HER3d4 residues 568-577. These residues also contribute to interactions within the tethered (inactive) and extended (active) conformations of the extracellular domain of HER3.


Assuntos
Repetição de Anquirina/genética , Cristalografia por Raios X/métodos , Espaço Extracelular/genética , Receptor ErbB-3/química , Receptor ErbB-3/genética , Sequência de Aminoácidos , Espaço Extracelular/metabolismo , Humanos , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptor ErbB-3/metabolismo
5.
Biochimie ; 190: 132-142, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34293452

RESUMO

The human EGF receptor family plays pivotal roles in physiology and cancer, which contains four closely-related members: HER1/EGFR, HER2, HER3 and HER4. Previously, it was found that the mitogen-inducible gene 6 (Mig6) protein is a negative regulator of EGFR and HER2 by using its S1 segment to bind at the kinase dimerization interface. However, it is still unclear whether the S1 segment can also effectively target HER3 and HER4? Here, we performed a systematic investigation to address this issue. The segment can bind to all the four HER kinases with a varying affinity and moderate selectivity; breaking of the segment into shorter hotspot peptides would largely impair the affinity and selectivity, indicating that the full-length sequence is required for the effective binding of S1 to these kinases. The hs2 peptide, which corresponds to the middle hotspot region of S1 segment, can partially retain the affinity to HER kinases, can moderately compete with S1 segment at the dimerization interfaces, and can mimic the biological function of Mig6 protein to suppress HER4+ esophageal cancer at cellular level. In addition, we also analyzed the binding potency of S1 segment and hs2 peptide to the kinase domains of other five widely documented growth factor receptors (GFRs). It was showed that both the S1 and hs2 cannot effectively interact with these receptors. Overall, the Mig6 is suggested as a specific pan-HER inhibitor, which can target and suppress HER family members with a broad selectivity, but exhibits weak or no activity towards other GFRs.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias Esofágicas/metabolismo , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-3/metabolismo , Receptor ErbB-4/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Simulação por Computador , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/química , Receptores ErbB/metabolismo , Neoplasias Esofágicas/tratamento farmacológico , Humanos , Peptídeos/farmacologia , Ligação Proteica , Domínios Proteicos , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Receptor ErbB-3/química , Receptor ErbB-4/química , Especificidade por Substrato , Proteínas Supressoras de Tumor/química
6.
Cancer Cell ; 39(8): 1099-1114.e8, 2021 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-34171264

RESUMO

Activating mutations in HER2 (ERBB2) drive the growth of a subset of breast and other cancers and tend to co-occur with HER3 (ERBB3) missense mutations. The HER2 tyrosine kinase inhibitor neratinib has shown clinical activity against HER2-mutant tumors. To characterize the role of HER3 mutations in HER2-mutant tumors, we integrate computational structural modeling with biochemical and cell biological analyses. Computational modeling predicts that the frequent HER3E928G kinase domain mutation enhances the affinity of HER2/HER3 and reduces binding of HER2 to its inhibitor neratinib. Co-expression of mutant HER2/HER3 enhances HER2/HER3 co-immunoprecipitation and ligand-independent activation of HER2/HER3 and PI3K/AKT, resulting in enhanced growth, invasiveness, and resistance to HER2-targeted therapies, which can be reversed by combined treatment with PI3Kα inhibitors. Our results provide a mechanistic rationale for the evolutionary selection of co-occurring HER2/HER3 mutations and the recent clinical observations that HER3 mutations are associated with a poor response to neratinib in HER2-mutant cancers.


Assuntos
Neoplasias da Mama/genética , Mutação com Ganho de Função , Quinolinas/farmacologia , Receptor ErbB-2/genética , Receptor ErbB-3/genética , Aminopiridinas/administração & dosagem , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Humanos , Camundongos Nus , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Morfolinas/administração & dosagem , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase/administração & dosagem , Multimerização Proteica , Quinolinas/administração & dosagem , Quinolinas/química , Quinolinas/metabolismo , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Receptor ErbB-3/química , Receptor ErbB-3/metabolismo , Trastuzumab/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Biochem Biophys Res Commun ; 553: 148-153, 2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33770580

RESUMO

The interaction of human epidermal growth factor receptor 3 (HER3) and heregulin (HRG) is involved in resistance to human epidermal growth factor receptor 2 (HER2)-targeted cancer treatment, such as therapies using anti-HER2 monoclonal antibody. Therefore, inhibition of the HER3/HRG interaction is potentially valuable therapeutic target for cancer treatment. In this study, we used in vitro selection, also known as systematic evolution of ligands by exponential enrichment (SELEX) against the extracellular domain of human HER3, and discovered a novel RNA aptamer. Pull-down and bio-layer interferometry assays showed that RNA aptamer discovered specifically bound to HER3 with a dissociation constant (KD) of 700 nM. Pull-down assays using chemiluminescence detection also revealed that the HER3-binding RNA aptamer inhibited interactions between HER3 and human HRG. These results indicated that the novel HER3-binding RNA aptamer has potential to be used as basic tool in a range of applications involving HER3/HRG interactions, including research, therapeutic, and diagnostic applications.


Assuntos
Aptâmeros de Nucleotídeos/análise , Receptores ErbB/antagonistas & inibidores , Neuregulina-1/antagonistas & inibidores , Neuregulina-1/metabolismo , Receptor ErbB-3/antagonistas & inibidores , Receptor ErbB-3/metabolismo , Técnica de Seleção de Aptâmeros , Aptâmeros de Nucleotídeos/isolamento & purificação , Sequência de Bases , Receptores ErbB/metabolismo , Humanos , Cinética , Luminescência , Ligação Proteica/efeitos dos fármacos , Receptor ErbB-3/química
8.
J Clin Invest ; 131(6)2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33497358

RESUMO

Hirschsprung disease (HSCR) is the most frequent developmental anomaly of the enteric nervous system, with an incidence of 1 in 5000 live births. Chronic intestinal pseudo-obstruction (CIPO) is less frequent and classified as neurogenic or myogenic. Isolated HSCR has an oligogenic inheritance with RET as the major disease-causing gene, while CIPO is genetically heterogeneous, caused by mutations in smooth muscle-specific genes. Here, we describe a series of patients with developmental disorders including gastrointestinal dysmotility, and investigate the underlying molecular bases. Trio-exome sequencing led to the identification of biallelic variants in ERBB3 and ERBB2 in 8 individuals variably associating HSCR, CIPO, peripheral neuropathy, and arthrogryposis. Thorough gut histology revealed aganglionosis, hypoganglionosis, and intestinal smooth muscle abnormalities. The cell type-specific ErbB3 and ErbB2 function was further analyzed in mouse single-cell RNA sequencing data and in a conditional ErbB3-deficient mouse model, revealing a primary role for ERBB3 in enteric progenitors. The consequences of the identified variants were evaluated using quantitative real-time PCR (RT-qPCR) on patient-derived fibroblasts or immunoblot assays on Neuro-2a cells overexpressing WT or mutant proteins, revealing either decreased expression or altered phosphorylation of the mutant receptors. Our results demonstrate that dysregulation of ERBB3 or ERBB2 leads to a broad spectrum of developmental anomalies, including intestinal dysmotility.


Assuntos
Deficiências do Desenvolvimento/genética , Pseudo-Obstrução Intestinal/genética , Mutação , Neuregulina-1/genética , Receptor ErbB-2/genética , Receptor ErbB-3/genética , Adolescente , Animais , Pré-Escolar , Deficiências do Desenvolvimento/patologia , Modelos Animais de Doenças , Feminino , Motilidade Gastrointestinal/genética , Doença de Hirschsprung/genética , Doença de Hirschsprung/patologia , Humanos , Recém-Nascido , Pseudo-Obstrução Intestinal/patologia , Masculino , Camundongos , Modelos Moleculares , Linhagem , Fenótipo , Gravidez , Receptor ErbB-2/química , Receptor ErbB-3/química , Receptor ErbB-3/deficiência
9.
Biochem J ; 477(17): 3329-3347, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32815546

RESUMO

Despite being catalytically defective, pseudokinases are typically essential players of cellular signalling, acting as allosteric regulators of their active counterparts. Deregulation of a growing number of pseudokinases has been linked to human diseases, making pseudokinases therapeutic targets of interest. Pseudokinases can be dynamic, adopting specific conformations critical for their allosteric function. Interfering with their allosteric role, with small molecules that would lock pseudokinases in a conformation preventing their productive partner interactions, is an attractive therapeutic strategy to explore. As a well-known allosteric activator of epidermal growth factor receptor family members, and playing a major part in cancer progression, the pseudokinase HER3 is a relevant context in which to address the potential of pseudokinases as drug targets for the development of allosteric inhibitors. In this proof-of-concept study, we developed a multiplex, medium-throughput thermal shift assay screening strategy to assess over 100 000 compounds and identify selective small molecule inhibitors that would trap HER3 in a conformation which is unfavourable for the formation of an active HER2-HER3 heterodimer. As a proof-of-concept compound, AC3573 bound with some specificity to HER3 and abrogated HER2-HER3 complex formation and downstream signalling in cells. Our study highlights the opportunity to identify new molecular mechanisms of action interfering with the biological function of pseudokinases.


Assuntos
Inibidores de Proteínas Quinases , Receptor ErbB-3 , Regulação Alostérica , Animais , Células CHO , Cricetulus , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Estudo de Prova de Conceito , Ligação Proteica , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Receptor ErbB-2/química , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptor ErbB-3/antagonistas & inibidores , Receptor ErbB-3/química , Receptor ErbB-3/genética , Receptor ErbB-3/metabolismo
10.
Elife ; 92020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32267234

RESUMO

Human epidermal growth factor receptors (HERs) are the primary targets of many directed cancer therapies. However, the reason a specific dimer of HERs generates a stronger proliferative signal than other permutations remains unclear. Here, we used single-molecule immunoprecipitation to develop a biochemical assay for endogenously-formed, entire HER2-HER3 heterodimers. We observed unexpected, large conformational fluctuations in juxta-membrane and kinase domains of the HER2-HER3 heterodimer. Nevertheless, the individual HER2-HER3 heterodimers catalyze tyrosine phosphorylation at an unusually high rate, while simultaneously interacting with multiple copies of downstream signaling effectors. Our results suggest that the high catalytic rate and multi-tasking capability make a concerted contribution to the strong signaling potency of the HER2-HER3 heterodimers.


Assuntos
Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Receptor ErbB-3/química , Receptor ErbB-3/metabolismo , Transdução de Sinais , Dimerização , Células HEK293 , Humanos , Modelos Moleculares , Fosforilação , Conformação Proteica , Domínios Proteicos , Receptor ErbB-2/genética , Receptor ErbB-3/genética , Imagem Individual de Molécula , Tirosina/metabolismo
11.
Macromol Biosci ; 19(11): e1900159, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31531954

RESUMO

Despite the initial successes of gene delivery applications, they faced on several intrinsic drawbacks including toxicity and immunogenicity. Therefore, alternative gene-delivery systems derived from recombinant peptides have emerged and is rapidly developing. Human epidermal growth factor receptor-3 (HER3) shows high activity in tumor resistance to anti-human epidermal growth factor receptor 2 (HER2) therapies. In this study, an affibody molecule against HER3 is conjugated to a biomimetic peptide RALA (an amphipathic and cationic peptide enriched with arginine) and the ability of the fusion vector for targeting HER3 and afterward delivering specific genes in breast cancer cells is evaluated. The results demonstrate that the biopolymeric platform, which contains an affibody-conjugated RALA peptide, can effectively condense DNA into nanoparticles and target the overexpressed HER3 receptors in breast cancer cells and transfer specific genes. The use of such a recombinant biopolymer may pave the way for the development of sensitive and effective diagnostic and treatment tool for breast cancer.


Assuntos
Neoplasias da Mama/terapia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Peptídeos/genética , Receptor ErbB-3/genética , Proteínas Recombinantes de Fusão/genética , Sequência de Aminoácidos , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Clonagem Molecular , Feminino , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Células MCF-7 , Nanoconjugados/química , Nanotecnologia/métodos , Peptídeos/química , Receptor ErbB-3/química , Receptor ErbB-3/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo
12.
Neoplasia ; 21(4): 343-352, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30831376

RESUMO

There are seven ligands for the epidermal growth factor receptor (EGFR) ErbB1 and two ligands for ErbB3. EGFR can form a homodimer or a heterodimer with ErbB3. In this study, we investigated whether homodimers or heterodimers, and which ligand, play a major role in cancer development, with the goal of ultimately identifying therapeutic targets. We demonstrated that the ErbB3 ligand heregulin1 is the strongest mitogenic factor for non-small cell lung cancer cells and is more potent in activating EGFRmut-ErbB3 heterodimers than EGFRwt-ErbB3 heterodimers. We discovered that four of the seven EGFR ligands inhibited heregulin1-induced EGFRwt-ErbB3 activation and cell proliferation by promoting dephosphorylation of heregulin1-induced ErbB3 phosphorylation, whereas the other three did not exhibit such inhibition. Importantly, those four EGFR ligands did not inhibit heregulin1-induced EGFRmut-ErbB3 activation and proliferation of cells with EGFR mutants. We demonstrated that ErbB3 was overexpressed in the lung cancer cells but not in the adjacent normal alveoli or stromal tissue. EGFR and heregulin1 were also highly expressed in lung cancer cells. We conclude that the overexpression of heregulin1, ErbB3, and EGFR mutant renders uncontrolled cell proliferation.


Assuntos
Hemeproteínas/metabolismo , Neoplasias Pulmonares/metabolismo , Receptor ErbB-3/metabolismo , Animais , Células CHO , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Cricetulus , Expressão Gênica , Hemeproteínas/química , Humanos , Imuno-Histoquímica , Ligantes , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Mutação , Ligação Proteica , Multimerização Proteica , Receptor ErbB-3/química
13.
J Theor Biol ; 464: 63-71, 2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30593826

RESUMO

Lung cancer is the predominant cause of cancer deaths on a worldwide scale. A mutation in the epidermal growth factor receptor (EGFR) can cause non-small cell lung cancer (NSCLC). The L858R one-point mutation in exon 21 in EGFR is the most prevalent in NSCLC. For over 60% of EGFR-muted NSCLC, another mutation T790M can cause drug resistance. In this paper, we consider EGFR and ErbB3 heterodimers involving three structures of EGFR, wild-type, with L858R mutation, and with L858R and T790M mutations. We perform molecular dynamics (MD) simulations to analyze hydrogen bonds in all three instances. The hydrogen bonds contribute to the conformational stability of the protein and molecular recognition. Several other parameters are also investigated in the present study, which reveals significant changes in the dimer at different levels of mutation. The knowledge and results obtained from this study lead to useful insight into the mechanism of NSCLC drug resistance.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/enzimologia , Resistencia a Medicamentos Antineoplásicos , Neoplasias Pulmonares/enzimologia , Multimerização Proteica , Receptor ErbB-3/química , Substituição de Aminoácidos , Receptores ErbB/química , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Ligação de Hidrogênio , Mutação de Sentido Incorreto , Estrutura Quaternária de Proteína , Receptor ErbB-3/genética , Receptor ErbB-3/metabolismo
14.
J Photochem Photobiol B ; 186: 160-168, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30064062

RESUMO

To overcome the problem of breast cancer, silver nanoparticles (AgNPs) synthesized using Indian medicinal plant Madhuca longifolia could be explored as an alternative anticancer medicine. Synthesized AgNPs were studied their characteristics and their anti-proliferative property was investigated in breast cancer cell line (4T1). Based on zeta sizer analysis, the size of the AgNPs was 103.5 nm and potential -9.57 eV. Fe-SEM results showed particle size of 69.4-99.4 nm while TEM images indicated the particle size of 18-24 nm. In dose-dependent study, AgNPs showed 93% of anti-proliferative activity at 50 µg/ml whereas the methanolic extract of M. longifolia showed 80% activity only at 10-fold increased concentration (500 µg/ml). AgNPs exhibited higher level of cytotoxicity in breast cancer cell line than extract through cell wall degradation and ROS generation. Such effective AgNPs could be investigated further through in vivo models with a view to develop anticancer drug.


Assuntos
Membrana Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Madhuca/química , Nanopartículas Metálicas/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Prata/química , Sítios de Ligação , Linhagem Celular , Membrana Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Flavonoides/química , Flavonoides/metabolismo , Flavonoides/farmacologia , Glucosídeos/química , Glucosídeos/metabolismo , Glucosídeos/farmacologia , Química Verde , Humanos , Madhuca/metabolismo , Nanopartículas Metálicas/química , Microscopia de Fluorescência , Simulação de Acoplamento Molecular , Tamanho da Partícula , Extratos Vegetais/química , Folhas de Planta/química , Folhas de Planta/metabolismo , Receptor ErbB-3/química , Receptor ErbB-3/metabolismo , Transdução de Sinais/efeitos dos fármacos
15.
Elife ; 72018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29712619

RESUMO

While targeted therapy against HER2 is an effective first-line treatment in HER2+ breast cancer, acquired resistance remains a clinical challenge. The pseudokinase HER3, heterodimerisation partner of HER2, is widely implicated in the resistance to HER2-mediated therapy. Here, we show that lapatinib, an ATP-competitive inhibitor of HER2, is able to induce proliferation cooperatively with the HER3 ligand neuregulin. This counterintuitive synergy between inhibitor and growth factor depends on their ability to promote atypical HER2-HER3 heterodimerisation. By stabilising a particular HER2 conformer, lapatinib drives HER2-HER3 kinase domain heterocomplex formation. This dimer exists in a head-to-head orientation distinct from the canonical asymmetric active dimer. The associated clustering observed for these dimers predisposes to neuregulin responses, affording a proliferative outcome. Our findings provide mechanistic insights into the liabilities involved in targeting kinases with ATP-competitive inhibitors and highlight the complex role of protein conformation in acquired resistance.


Assuntos
Neoplasias da Mama/metabolismo , Proliferação de Células , Lapatinib/farmacologia , Neuregulina-1/metabolismo , Multimerização Proteica , Receptor ErbB-2/química , Receptor ErbB-3/química , Trifosfato de Adenosina/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Feminino , Humanos , Fosforilação , Conformação Proteica , Inibidores de Proteínas Quinases/farmacologia , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas
16.
Cancer Cell ; 33(5): 922-936.e10, 2018 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-29763625

RESUMO

HER2-driven cancers require phosphatidylinositide-3 kinase (PI3K)/Akt signaling through HER3 to promote tumor growth and survival. The therapeutic benefit of HER2-targeting agents, which depend on PI3K/Akt inhibition, can be overcome by hyperactivation of the heregulin (HRG)/HER3 pathway. Here we describe an unbiased phenotypic combinatorial screening approach to identify a bispecific immunoglobulin G1 (IgG1) antibody against HER2 and HER3. In tumor models resistant to HER2-targeting agents, the bispecific IgG1 potently inhibits the HRG/HER3 pathway and downstream PI3K/Akt signaling via a "dock & block" mechanism. This bispecific IgG1 is a potentially effective therapy for breast cancer and other tumors with hyperactivated HRG/HER3 signaling.


Assuntos
Anticorpos Biespecíficos/administração & dosagem , Imunoglobulina G/administração & dosagem , Neoplasias/tratamento farmacológico , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Anticorpos Biespecíficos/farmacologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Imunoglobulina G/farmacologia , Células MCF-7 , Camundongos , Modelos Moleculares , Neoplasias/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor ErbB-2/química , Receptor ErbB-3/química , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Nature ; 554(7691): 189-194, 2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29420467

RESUMO

Somatic mutations of ERBB2 and ERBB3 (which encode HER2 and HER3, respectively) are found in a wide range of cancers. Preclinical modelling suggests that a subset of these mutations lead to constitutive HER2 activation, but most remain biologically uncharacterized. Here we define the biological and therapeutic importance of known oncogenic HER2 and HER3 mutations and variants of unknown biological importance by conducting a multi-histology, genomically selected, 'basket' trial using the pan-HER kinase inhibitor neratinib (SUMMIT; clinicaltrials.gov identifier NCT01953926). Efficacy in HER2-mutant cancers varied as a function of both tumour type and mutant allele to a degree not predicted by preclinical models, with the greatest activity seen in breast, cervical and biliary cancers and with tumours that contain kinase domain missense mutations. This study demonstrates how a molecularly driven clinical trial can be used to refine our biological understanding of both characterized and new genomic alterations with potential broad applicability for advancing the paradigm of genome-driven oncology.


Assuntos
Mutação , Neoplasias/tratamento farmacológico , Neoplasias/genética , Quinolinas/farmacologia , Quinolinas/uso terapêutico , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-3/antagonistas & inibidores , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Estudos de Coortes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Mutação de Sentido Incorreto , Neoplasias/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Quinolinas/efeitos adversos , Receptor ErbB-2/química , Receptor ErbB-2/genética , Receptor ErbB-3/química , Receptor ErbB-3/genética , Resultado do Tratamento
18.
Mol Imaging Biol ; 20(2): 300-308, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28733706

RESUMO

PURPOSE: HER3 (ERBB3) is a receptor tyrosine kinase that is implicated in treatment resistance across multiple cancers, including those of the breast, lung, and prostate. Overexpression of HER3 following targeted therapy can occur rapidly and heterogeneously both within a single lesion and across sites of metastasis, making protein quantification by biopsy highly challenging. A global, non-invasive methodology such as positron emission tomography (PET) imaging can permit serial quantification of HER3, providing a useful approach to monitor HER3 expression across the entire tumor burden both prior to and following treatment. PET imaging of HER3 expression may permit a more personalized approach to targeted therapy by allowing for detection of HER3-mediated resistance, in addition to informing clinical trial patient selection for novel therapies targeting HER3. PROCEDURES: Phage display selection targeting the HER3 extracellular domain was performed in order to develop a peptide with optimal blood clearance and highly accurate HER3 quantification. RESULTS: The selection converged to a consensus peptide sequence that was subsequently found to bind HER3 with an affinity of 270 ± 151 nM. The peptide, termed HER3P1, was bound with high selectivity to HER3 over other similar receptor tyrosine kinases such as EGFR and HER2. Furthermore, HER3P1 was able to distinguish between high and low HER3-expressing cells in vitro. The peptide was radiolabeled with Ga-68 and demonstrated to specifically bind HER3 by in vivo PET imaging. Uptake of [68Ga]HER3P1 was highly specific for HER3-positive tumors, with tumor-to-background ratios ranging from 1.59-3.32, compared to those of HER3-negative tumors, ranging from 0.84-0.93. The uptake of [68Ga]HER3P1 also demonstrated high (P < 0.001) correlation with protein expression as quantified by Western blot and confirmed by biodistribution. CONCLUSIONS: HER3P1 accurately quantifies expression of HER3 by PET imaging and has potential utility as a clinical imaging agent.


Assuntos
Técnicas de Visualização da Superfície Celular , Peptídeos/química , Tomografia por Emissão de Pósitrons , Receptor ErbB-3/química , Animais , Biotina/metabolismo , Linhagem Celular Tumoral , Radioisótopos de Gálio/química , Camundongos , Ligação Proteica , Distribuição Tecidual
19.
Sci Rep ; 7(1): 11375, 2017 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-28900130

RESUMO

The ligand-induced dimerization of cell surface single-transmembrane receptors is essential for their activation. However, physiological molecules that inhibit their dimerization and activation have not been identified. ErbB3 dimerizes with ErbB2 upon binding of heregulin (HRG) to ErbB3, causing the ErbB2-catalyzed tyrosine phosphorylation of ErbB3, which leads to the activation of the signalling pathways for cell movement and survival. Genetic disorders of this receptor cause tumorigenesis and metastasis of cancers. We show here that nectin-like molecule-4/cell adhesion molecule 4, known to serve as a tumour suppressor, interacts with ErbB3 in the absence of HRG and inhibits the HRG-induced dimerization of ErbB3 with ErbB2 and its activation. The third immunoglobulin-like domain of nectin-like molecule-4 cis-interacts with the extracellular domain 3 of ErbB3. We describe here a novel regulatory mechanism for the activation and signalling of cell surface single-transmembrane receptors.


Assuntos
Moléculas de Adesão Celular/metabolismo , Imunoglobulinas/metabolismo , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Moléculas de Adesão Celular/química , Linhagem Celular , Espaço Extracelular/metabolismo , Humanos , Imunoglobulinas/química , Ligantes , Neuregulina-1/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Receptor ErbB-2/química , Receptor ErbB-3/química
20.
J Biol Chem ; 292(31): 12772-12782, 2017 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-28615452

RESUMO

The iron-regulated metastasis suppressor N-myc downstream-regulated gene 1 (NDRG1) has been shown to inhibit numerous oncogenic signaling pathways in cancer cells. Recent findings have demonstrated that NDRG1 inhibits the ErbB family of receptors, which function as key inducers of carcinogenesis. NDRG1 attenuates ErbB signaling by inhibiting formation of epidermal growth factor receptor (EGFR)/human epidermal growth factor receptor 2 (HER2) and HER2/HER3 heterodimers and by down-regulating EGFR via a mechanism involving its degradation. Understanding the complex interplay between NDRG1, iron, and ErbB signaling is vital for identifying novel, more effective targets for cancer therapy.


Assuntos
Carcinogênese , Proteínas de Ciclo Celular/metabolismo , Receptores ErbB/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Modelos Biológicos , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-3/antagonistas & inibidores , Transdução de Sinais , Animais , Antígenos CD/metabolismo , Proteínas de Ciclo Celular/química , Vesículas Revestidas por Clatrina , Endocitose , Endossomos/enzimologia , Receptores ErbB/química , Receptores ErbB/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Ferro/metabolismo , Ligantes , Multimerização Proteica , Proteólise , Receptor ErbB-2/agonistas , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Receptor ErbB-3/agonistas , Receptor ErbB-3/química , Receptor ErbB-3/metabolismo , Receptores da Transferrina/agonistas , Receptores da Transferrina/metabolismo , Transferrina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...