Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 614
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
EMBO J ; 39(2): e103637, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31803974

RESUMO

Although adoptive T-cell therapy has shown remarkable clinical efficacy in haematological malignancies, its success in combating solid tumours has been limited. Here, we report that PTPN2 deletion in T cells enhances cancer immunosurveillance and the efficacy of adoptively transferred tumour-specific T cells. T-cell-specific PTPN2 deficiency prevented tumours forming in aged mice heterozygous for the tumour suppressor p53. Adoptive transfer of PTPN2-deficient CD8+ T cells markedly repressed tumour formation in mice bearing mammary tumours. Moreover, PTPN2 deletion in T cells expressing a chimeric antigen receptor (CAR) specific for the oncoprotein HER-2 increased the activation of the Src family kinase LCK and cytokine-induced STAT-5 signalling, thereby enhancing both CAR T-cell activation and homing to CXCL9/10-expressing tumours to eradicate HER-2+ mammary tumours in vivo. Our findings define PTPN2 as a target for bolstering T-cell-mediated anti-tumour immunity and CAR T-cell therapy against solid tumours.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunoterapia Adotiva/métodos , Ativação Linfocitária/imunologia , Neoplasias/terapia , Proteína Tirosina Fosfatase não Receptora Tipo 2/fisiologia , Receptor ErbB-2/fisiologia , Receptores de Antígenos de Linfócitos T/imunologia , Transferência Adotiva , Animais , Apresentação de Antígeno/imunologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neoplasias/genética , Neoplasias/imunologia , Transdução de Sinais
2.
Proc Natl Acad Sci U S A ; 115(16): 4176-4181, 2018 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-29610308

RESUMO

Allelic loss of the autophagy gene, beclin 1/BECN1, increases the risk of patients developing aggressive, including human epidermal growth factor receptor 2 (HER2)-positive, breast cancers; however, it is not known whether autophagy induction may be beneficial in preventing HER2-positive breast tumor growth. We explored the regulation of autophagy in breast cancer cells by HER2 in vitro and the effects of genetic and pharmacological strategies to increase autophagy on HER2-driven breast cancer growth in vivo. Our findings demonstrate that HER2 interacts with Beclin 1 in breast cancer cells and inhibits autophagy. Mice with increased basal autophagy due to a genetically engineered mutation in Becn1 are protected from HER2-driven mammary tumorigenesis, and HER2 fails to inhibit autophagy in primary cells derived from these mice. Moreover, treatment of mice with HER2-positive human breast cancer xenografts with the Tat-Beclin 1 autophagy-inducing peptide inhibits tumor growth as effectively as a clinically used HER2 tyrosine kinase inhibitor (TKI). This inhibition of tumor growth is associated with a robust induction of autophagy, a disruption of HER2/Beclin 1 binding, and a transcriptional signature in the tumors distinct from that observed with HER2 TKI treatment. Taken together, these findings indicate that the HER2-mediated inhibition of Beclin 1 and autophagy likely contributes to HER2-mediated tumorigenesis and that strategies to block HER2/Beclin 1 binding and/or increase autophagy may represent a new therapeutic approach for HER2-positive breast cancers.


Assuntos
Autofagia , Proteína Beclina-1/fisiologia , Proteínas de Neoplasias/fisiologia , Receptor ErbB-2/fisiologia , Substituição de Aminoácidos , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Autofagia/efeitos dos fármacos , Proteína Beclina-1/deficiência , Proteína Beclina-1/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Técnicas de Introdução de Genes , Humanos , Lapatinib , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Terapia de Alvo Molecular , Mutação , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/genética , Fragmentos de Peptídeos/uso terapêutico , Ligação Proteica/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Quinazolinas/farmacologia , Distribuição Aleatória , Receptor ErbB-2/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Biochemistry (Mosc) ; 85(10): 1277-1287, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33202212

RESUMO

HER2 (Human Epidermal Growth Factor Receptor 2), also known as ERBB2, CD340, and Neu protooncogene, is a member of the epidermal growth factor receptor (EGRF) family. Members of the ERBB family, including HER2, activate molecular cascades that stimulate proliferation and migration of cancer cells, as well as their resistance to the anticancer therapy. These proteins are often overexpressed and/or mutated in various cancer types and represent promising targets for the anti-cancer therapy. Currently, anti-HER2 drugs have been approved for the treatment of several types of solid tumors. HER2-specific therapy includes monoclonal antibodies and low-molecular weight inhibitors of tyrosine kinase receptors, such as lapatinib, neratinib, and pyrotinib. In addition to the activation of molecular pathways responsible for cell proliferation and survival under stress conditions, HER2 directly regulates programmed cell death. Here, we review the studies focused on the involvement of HER2 in various signaling pathways and its role in the regulation of apoptosis.


Assuntos
Antineoplásicos , Apoptose , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias , Receptor ErbB-2/fisiologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Morte Celular/efeitos dos fármacos , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
4.
Hepatology ; 67(2): 762-773, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28671339

RESUMO

The ErbB/HER family comprises four distinct tyrosine kinase receptors, EGFR/ErbB1/HER1, ErbB2/HER2, ErbB3/HER3, and ErbB4/HER4, which trigger intracellular signals at the origin of essential cellular functions, including differentiation, proliferation, survival, and migration. Epithelial cells, named cholangiocytes, that line intrahepatic and extrahepatic bile ducts, contribute substantially to biliary secretory functions and bile transport. Although ErbB receptors have been widely studied in cholangiocarcinoma (CCA), a malignancy of the biliary tract, knowledge of these receptors in biliary epithelium physiology and in non-malignant cholangiopathies is far from complete. Current knowledge suggests a role for epidermal growth factor receptor (EGFR) in cholangiocyte specification and proliferation, and in hepatocyte transdifferentiation into cholangiocytes during liver regeneration to restore biliary epithelium integrity. High expression and activation of EGFR and/or ErbB2 were recently demonstrated in biliary lithiasis and primary sclerosing cholangitis, two cholangiopathies regarded as risk factors for CCA. In CCA, ErbB receptors are frequently overexpressed, leading to tumor progression and low prognosis. Anti-ErbB therapies were efficient only in preclinical trials and have suggested the existence of resistance mechanisms with the need to identify predictive factors of therapy response. This review aims to compile the current knowledge on the functions of ErbB receptors in physiology and physiopathology of the biliary epithelium. (Hepatology 2018;67:762-773).


Assuntos
Ductos Biliares/fisiologia , Células Epiteliais/fisiologia , Receptores ErbB/fisiologia , Animais , Doenças dos Ductos Biliares/etiologia , Ductos Biliares/citologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Humanos , Regeneração Hepática , Receptor ErbB-2/fisiologia , Receptor ErbB-3/fisiologia , Receptor ErbB-4/fisiologia , Transdução de Sinais/fisiologia , Microambiente Tumoral
5.
Oncology ; 97(5): 311-318, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31550723

RESUMO

INTRODUCTION: Human epidermal growth factor 2 (HER2) gene overexpression in breast carcinoma cell lines has been shown to drive mammary carcinogenesis and tumor growth and invasion through its effects on mammary stem cells. OBJECTIVE: Therefore, we investigated the mechanism by which HER2 regulates cancer stem cell (CSC) activity in gastric cancer cells. METHODS: HER2 was transfected into MKN28 gastric cancer cells, and its role in regulating CSC activity was determined by characterizing the HER2-overexpressing cells. RESULTS: The sphere formation assay revealed that the sphere sizes and frequency of sphere formation were significantly greater for the HER2-overexpressing cells than for the MKN28 control cells. The CSC markers Oct-4 and BMI1 were more highly expressed in the HER2-overexpressing cells, as were the EMT markers. This was accompanied by a significant enhancement in cellular invasion of the Matrigel and migration. The E-cadherin level was significantly downregulated, and the mesenchymal marker Snail upregulated, in the HER2-transfected cells. HER2 overexpression activated the well-characterized CSC-associated Wnt/ß-catenin signaling pathway, as shown by the luciferase assay. After treatment of these cells with the Wnt signal inhibitor PRI-724, the BMI1 and Oct-4 levels were decreased for 24 h and Snail was also downregulated. Immunofluorescence staining revealed the significant restoration of E-cadherin levels in the HER2-transfected cells after PRI-724 treatment. CONCLUSIONS: These results established a role for HER2 in regulating gastric CSC activity, with Wnt/ß-catenin signaling being mediated via a HER2-dependent pathway. In summary, HER2-overexpressing gastric cancer cells exhibited increased stemness and invasiveness and were regulated by Wnt/ß-catenin signaling.


Assuntos
Células-Tronco Neoplásicas/fisiologia , Receptor ErbB-2/fisiologia , Neoplasias Gástricas/patologia , Via de Sinalização Wnt/fisiologia , Antígenos CD/análise , Caderinas/análise , Linhagem Celular Tumoral , Humanos , Invasividade Neoplásica , Fator 3 de Transcrição de Octâmero/análise , Complexo Repressor Polycomb 1/análise , Receptor ErbB-2/análise , Neoplasias Gástricas/química , beta Catenina/análise
6.
Proc Natl Acad Sci U S A ; 113(11): 3030-5, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26929372

RESUMO

Loss of the tumor suppressor gene PTEN is implicated in breast cancer progression and resistance to targeted therapies, and is thought to promote tumorigenesis by activating PI3K signaling. In a transgenic model of breast cancer, Pten suppression using a tetracycline-regulatable short hairpin (sh)RNA cooperates with human epidermal growth factor receptor 2 (HER2/neu), leading to aggressive and metastatic disease with elevated signaling through PI3K and, surprisingly, the mitogen-activated protein kinase (MAPK) pathway. Restoring Pten function is sufficient to down-regulate both PI3K and MAPK signaling and triggers dramatic tumor regression. Pharmacologic inhibition of MAPK signaling produces similar effects to Pten restoration, suggesting that the MAPK pathway contributes to the maintenance of advanced breast cancers harboring Pten loss.


Assuntos
Sistema de Sinalização das MAP Quinases/fisiologia , Neoplasias Mamárias Experimentais/fisiopatologia , Proteínas de Neoplasias/fisiologia , PTEN Fosfo-Hidrolase/deficiência , Receptor ErbB-2/fisiologia , Animais , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Genes erbB-2 , Humanos , MAP Quinase Quinase 1/fisiologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Vírus do Tumor Mamário do Camundongo/fisiologia , Camundongos , Camundongos Nus , Camundongos Transgênicos , Metástase Neoplásica , Proteínas de Neoplasias/deficiência , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-akt/fisiologia , Interferência de RNA , RNA Interferente Pequeno/genética , Transdução de Sinais/fisiologia
7.
J Am Soc Nephrol ; 29(4): 1326-1335, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29511113

RESUMO

Many biomarkers have been epidemiologically linked with CKD; however, the possibility that such associations are due to reverse causation or confounding limits the utility of these biomarkers. To overcome this limitation, we used a Mendelian randomization (MR) approach to identify causal mediators of CKD. We performed MR by first identifying genetic determinants of 227 serum protein biomarkers assayed in 4147 participants of the Outcome Reduction with Initial Glargine Intervention (ORIGIN) trial who had early or prediabetes, and assessing the effects of these biomarkers on CKD in the CKD genetics consortium (n=117,165; 12,385 cases) using the inverse-variance weighted (fixed-effects) method. We then estimated the relationship between the serum concentration of each biomarker identified and incident CKD in ORIGIN participants. MR identified uromodulin (UMOD) and human EGF receptor 2 (HER2) as novel, causal mediators of CKD (UMOD: odds ratio [OR], 1.30 per SD; 95% confidence interval [95% CI], 1.25 to 1.35; P<5×10-20; HER2: OR, 1.30 per SD; 95% CI, 1.14 to 1.48; P=8.0×10-5). Consistent with these findings, blood HER2 concentration associated with CKD events in ORIGIN participants (OR, 1.07 per SD; 95% CI, 1.01 to 1.13; P=0.01). Additional exploratory MR analyses identified angiotensin-converting enzyme (ACE) as a regulator of HER2 levels (ß=0.13 per SD; 95% CI, 0.08 to 0.16; P=2.5×10-7). This finding was corroborated by an inverse relationship between ACE inhibitor use and HER2 levels. Thus, UMOD and HER2 are independent causal mediators of CKD in humans, and serum HER2 levels are regulated in part by ACE. These biomarkers are potential therapeutic targets for CKD prevention.


Assuntos
Estado Pré-Diabético/sangue , Receptor ErbB-2/sangue , Insuficiência Renal Crônica/etiologia , Uromodulina/sangue , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Anti-Hipertensivos/farmacologia , Biomarcadores , Causalidade , Feminino , Seguimentos , Genes erbB-2 , Humanos , Rim/anatomia & histologia , Doadores Vivos , Masculino , Análise da Randomização Mendeliana , Pessoa de Meia-Idade , Nefrectomia , Tamanho do Órgão , Peptidil Dipeptidase A/fisiologia , Polimorfismo de Nucleotídeo Único , Estado Pré-Diabético/genética , Receptor ErbB-2/genética , Receptor ErbB-2/fisiologia , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/epidemiologia , Insuficiência Renal Crônica/genética , Uromodulina/genética , Uromodulina/fisiologia
8.
Eur J Neurosci ; 48(10): 3299-3316, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30270571

RESUMO

In mammals, cochlear hair cells are not regenerated once they are lost, leading to permanent hearing deficits. In other vertebrates, the adjacent supporting cells act as a stem cell compartment, in that they both proliferate and differentiate into de novo auditory hair cells. Although there is evidence that mammalian cochlear supporting cells can differentiate into new hair cells, the signals that regulate this process are poorly characterized. We hypothesize that signaling from the epidermal growth factor receptor (EGFR) family may play a role in cochlear regeneration. We focus on one such member, ERBB2, and report the effects of expressing a constitutively active ERBB2 receptor in neonatal mouse cochlear supporting cells, using viruses and transgenic expression. Lineage tracing with fluorescent reporter proteins was used to determine the relationships between cells with active ERBB2 signaling and cells that divided or differentiated into hair cells. In vitro, individual supporting cells harbouring a constitutively active ERBB2 receptor appeared to signal to their neighbouring supporting cells, inducing them to down-regulate a supporting cell marker and to proliferate. In vivo, we found supernumerary hair cell-like cells near supporting cells that expressed ERBB2 receptors. Both supporting cell proliferation and hair cell differentiation were largely reproduced in vitro using small molecules that we show also activate ERBB2. Our data suggest that signaling from the receptor tyrosine kinase ERBB2 can drive the activation of secondary signaling pathways to regulate regeneration, suggesting a new model where an interplay of cell signaling regulates regeneration by endogenous stem-like cells.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células Ciliadas Auditivas/fisiologia , Receptor ErbB-2/fisiologia , Regeneração/fisiologia , Transdução de Sinais/fisiologia , Animais , Animais Recém-Nascidos , Camundongos , Camundongos Transgênicos
9.
Mol Biol Rep ; 45(6): 2247-2256, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30225583

RESUMO

HER2 is a member of epidermal factor receptor (EGFR) family which is overexpressed in breast cancer, ovarian cancer and gastric cancer. Development of new binders for cancer cell surface receptors and expressing them at the surface of exosomes would be a great approach in targeted cancer therapy. We found a high affinity scFv against HER2 using ribosome display with the approach of applying it as a targeting moiety at the surface of exosomes by fusion to lysosomal associated membrane protein 2B (LAMP2B). We also provide some structural information about the ribosome display selected scFv (scFv HFS2) through modeling the 3D structure of scFv HFS2 using RosettaAntibody and docked it at the extracellular domain of HER2. We also evaluated the structure of scFv HFS2 and its binding to HER2 after fusion to LAMP2B. Our results showed no significant change in 3D structure of scFv HFS2 when fused to LAMP2B (RMSD 1.3) and interaction analysis represented that scFv HFS2 binds HER2 domain III before and after fusion to LAMP2B. Although binding domain of scFv HFS2 on HER2 was the same at both state, residues involved in their interactions showed significant differences as it was probably due to the spatial hindrance of scFv HFS2 when fused to LAMP2B through a short linker and it should be considered before proceeding to experiment.


Assuntos
Engenharia de Proteínas/métodos , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia , Animais , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo , Humanos , Proteína 2 de Membrana Associada ao Lisossomo/genética , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Ovarianas/metabolismo , Receptor ErbB-2/fisiologia , Ribossomos/metabolismo , Anticorpos de Cadeia Única/genética
10.
Dev Biol ; 397(1): 116-28, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25446033

RESUMO

The study of molecular regulation in neural development provides information to understand how diverse neural cells are generated. It also helps to establish therapeutic strategies for the treatment of neural degenerative disorders and brain tumors. The Hairy/E(spl) family members are potential targets of Notch signaling, which is fundamental to neural cell maintenance, cell fate decisions, and compartment boundary formation. In this study, we isolated a zebrafish homolog of Hairy/E(spl), her2, and showed that this gene is expressed in neural progenitor cells and in the developing nervous system. The expression of her2 required Notch activation, as revealed by a Notch-defective mutant and a chemical inhibitor, N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT). The endogenous expression of Her2 was altered by both overexpression and morpholino-knockdown approaches, and the results demonstrated that Her2 was both necessary and sufficient to promote the proliferation of neural progenitors by inhibiting the transcription of the cell cycle inhibitors cdkn1a, cdkn1ba, and cdkn1bb. Her2 knockdown caused premature neuronal differentiation, which indicates that Her2 is essential for inhibiting neuronal differentiation. At a later stage of neural development, Her2 could induce glial differentiation. The overexpression of Her2 constructs lacking the bHLH or WRPW domain phenocopied the effect of the morpholino knockdown, demonstrating the essential function of these two domains and further confirming the knockdown specificity. In conclusion, our data reveal that Her2 promotes progenitor proliferation and maintains progenitor characteristics by inhibiting neuronal differentiation. Together, these two mechanisms ensure the proper development of the neural progenitor cell pool.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Neurogênese/fisiologia , Neuroglia/metabolismo , Neurônios/metabolismo , Receptor ErbB-2/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Ciclo Celular , Diferenciação Celular , Proliferação de Células , Dipeptídeos/química , Perfilação da Expressão Gênica , Genes Dominantes , Receptor ErbB-2/genética , Transdução de Sinais , Fatores de Tempo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
11.
Exp Cell Res ; 333(1): 116-26, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25743092

RESUMO

Fibronectin (FN), an extracellular matrix ligand, plays a pivotal role in cell adhesion, migration, and oncogenic transformation. Aberrant FN expression is associated with poor prognoses in various types of cancer, including breast cancer. In the current study, we investigated the relationship between FN induction and HER2 expression in breast cancer cells. Our results showed that the level of FN expression increased in response to HER family ligands, EGF and TGF-α in a time- and dose-dependent manner. On the other hand, EGF-induced FN expression decreased in response to trastuzumab, which is a HER2-targeted monoclonal antibody. However, EGF-induced FN expression was not affected by trastuzumab in JIMT-1 breast cancer cells, which are trastuzumab insensitive cells. Next, we introduced the HER2 gene into MDA-MB231 cells to verify the relationship between FN and HER2. The level of FN expression significantly increased in HER2-overexpressed MDA-MB231 cells. In contrast, the induction of FN by HER2 was significantly decreased in response to trastuzumab treatment. In addition, the induction of FN by HER2 was down-regulated by the MEK 1/2 specific inhibitor, U0126. Using conditioned culture media of vec- and HER2-overexpressed MDA-MB231 cells, we observed the cell morphology, adhesion, and invasion of MDA-MB231 cells. Interestingly, in conditioned culture media of HER2-overexpressed MDA-MB231 cells, the cell morphology was altered, and adhesion and invasion of MDA-MB231 cells significantly increased. In addition, our results showed that recombinant human FN augmented cell adhesion and invasion of MDA-MB231 cells while these inductions decreased in response to an FN inhibitor. Therefore, we demonstrated that the induction of FN by HER2 triggers cell adhesion and invasion capacities.


Assuntos
Adesão Celular , Fibronectinas/metabolismo , Receptor ErbB-2/fisiologia , Ativação Transcricional , Neoplasias da Mama , Linhagem Celular Tumoral , Forma Celular , Fator de Crescimento Epidérmico/fisiologia , Feminino , Fibronectinas/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Sistema de Sinalização das MAP Quinases , Invasividade Neoplásica , Fator de Crescimento Transformador alfa/fisiologia
12.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 41(11): 1143-1147, 2016 Nov 28.
Artigo em Zh | MEDLINE | ID: mdl-27932758

RESUMO

OBJECTIVE: To detect the expressions of human epidermal growth factor receptor 2 (HER2) and Topo IIα in breast cancer, and to analyze the clinical significance of neoadjuvant chemotherapy for the anthracycline-based drugs.
 Methods: The HER2 and Topo IIα gene and protein expressions in cancer tissues from 189 patients with breast cancer were detected by fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC). And the objective response rate (ORR) and pathological complete rate (pCR) were analyzed.
 Results: The HER2 protein expression in 46 patients (24.3%) and Topo IIα protein expression in 55 patients (29.1%) was 3+ by IHC or they were 49 (25.9%) and 94 (49.0%) by FISH, respectively. The ORR and pCR in HER2 negative or positive patients were 47.4% and 20.3% or 32.7% and 16.3%, respectively, with significant differences (All P<0.05). The ORR and pCR in Topo IIα positive or negative patients were 69.1% and 36.0% or 28.4% and 2.2%, respectively, with significant differences (All P<0.05).
 Conclusion: FISH and IHC were consistent in the determination of HER2 expression whereas they were inconsistent in the determination of Topo IIα expression. The amplification of Topo IIα can effectively improve the effect of the adjuvant treatment effect of the anthracyclines.


Assuntos
Antraciclinas/farmacologia , Neoplasias da Mama/química , Neoplasias da Mama/genética , Neoplasias da Mama/terapia , DNA Topoisomerases Tipo II/fisiologia , Receptor ErbB-2/fisiologia , Antraciclinas/uso terapêutico , Antibióticos Antineoplásicos , Feminino , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Terapia Neoadjuvante , Resultado do Tratamento
13.
J Biol Chem ; 289(44): 30318-30332, 2014 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-25202019

RESUMO

Epithelial cells are highly dependent during wound healing and tumorigenesis on the α6ß4 integrin and its association with receptor tyrosine kinases. Previous work showed that phosphorylation of the ß4 subunit upon matrix engagement depends on the matrix receptor syndecan (Sdc)-1 engaging the cytoplasmic domain of the ß4 integrin and coupling of the integrin to human epidermal growth factor receptor-2 (HER2). In this study, HER2-dependent migration activated by matrix engagement is compared with migration stimulated by EGF. We find that whereas HER2-dependent migration depends on Sdc1, EGF-dependent migration depends on a complex consisting of human epidermal growth factor receptor-1 (HER1, commonly known as EGFR), α6ß4, and Sdc4. The two syndecans recognize distinct sites at the extreme C terminus of the ß4 integrin cytoplasmic domain. The binding motif in Sdc1 is QEEXYX, composed in part by its syndecan-specific variable (V) region and in part by the second conserved (C2) region that it shares with other syndecans. A cell-penetrating peptide containing this sequence competes for HER2-dependent epithelial migration and carcinoma survival, although it is without effect on the EGFR-stimulated mechanism. ß4 mutants bearing mutations specific for Sdc1 and Sdc4 recognition act as dominant negative mutants to block cell spreading or cell migration that depends on HER2 or EGFR, respectively. The interaction of the α6ß4 integrin with the syndecans appears critical for it to be utilized as a signaling platform; migration depends on α3ß1 integrin binding to laminin 332 (LN332; also known as laminin 5), whereas antibodies that block α6ß4 binding are without effect. These findings indicate that specific syndecan family members are likely to have key roles in α6ß4 integrin activation by receptor tyrosine kinases.


Assuntos
Movimento Celular , Sobrevivência Celular , Integrina alfa6beta4/metabolismo , Sindecana-1/metabolismo , Sindecana-4/metabolismo , Sequência de Aminoácidos , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Citoplasma/metabolismo , Receptores ErbB/metabolismo , Receptores ErbB/fisiologia , Humanos , Integrina alfa6beta4/química , Integrina alfa6beta4/genética , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Receptor ErbB-2/fisiologia , Transdução de Sinais , Sindecana-1/química , Sindecana-4/química , Calinina
14.
J Biol Chem ; 289(23): 16057-71, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24737320

RESUMO

Previous studies have demonstrated that Artemin (ARTN) functions as a cancer stem cell (CSC) and metastatic factor in mammary carcinoma. Herein, we report that ARTN mediates acquired resistance to trastuzumab in HER2-positive mammary carcinoma cells. Ligands that increase HER2 activity increased ARTN expression in HER2-positive mammary carcinoma cells, whereas trastuzumab inhibited ARTN expression. Forced expression of ARTN decreased the sensitivity of HER2-positive mammary carcinoma cells to trastuzumab both in vitro and in vivo. Conversely, siRNA-mediated depletion of ARTN enhanced trastuzumab efficacy. Cells with acquired resistance to trastuzumab exhibited increased ARTN expression, the depletion of which restored trastuzumab sensitivity. Trastuzumab resistance produced an increased CSC population concomitant with enhanced mammospheric growth. ARTN mediated the enhancement of the CSC population by increased BCL-2 expression, and the CSC population in trastuzumab-resistant cells was abrogated upon inhibition of BCL-2. Hence, we conclude that ARTN is one mediator of acquired resistance to trastuzumab in HER2-positive mammary carcinoma cells.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Proteínas do Tecido Nervoso/fisiologia , Receptor ErbB-2/fisiologia , Anticorpos Monoclonais Humanizados/farmacologia , Sequência de Bases , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Primers do DNA , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Células-Tronco Neoplásicas/patologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Reação em Cadeia da Polimerase , Trastuzumab
16.
Arterioscler Thromb Vasc Biol ; 34(9): 1831-7, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24925972

RESUMO

Receptor tyrosine kinases are involved in regulation of key processes in endothelial biology, including proliferation, migration, and angiogenesis. It is now generally accepted that receptor tyrosine kinase signaling occurs intracellularly and on the plasma membrane, although many important details remain to be worked out. Endocytosis and subsequent intracellular trafficking spatiotemporally regulate receptor tyrosine kinase signaling, whereas signaling endosomes provide a platform for the compartmentalization of signaling events. This review summarizes recent advances in our understanding of endothelial receptor tyrosine kinase endocytosis and signaling using vascular endothelial growth factor receptor-2 as a paradigm.


Assuntos
Endocitose/fisiologia , Endotélio Vascular/enzimologia , Receptores Proteína Tirosina Quinases/fisiologia , Transdução de Sinais/fisiologia , Animais , Membrana Celular/enzimologia , Endossomos/fisiologia , Células Endoteliais/metabolismo , Endotélio Vascular/fisiopatologia , Receptores ErbB/fisiologia , Humanos , Comunicação Parácrina , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptor ErbB-2/fisiologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Quinases da Família src/fisiologia
17.
Cancer Cell ; 11(3): 214-6, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17349579

RESUMO

The protein tyrosine phosphatase PTP1B, previously recognized for its role in downregulating insulin and leptin signaling, has now been shown to function as a positive regulator of signaling events associated with breast tumorigenesis. Inhibitors of PTP1B that have been developed as drug candidates for treatment of diabetes and obesity may offer new avenues for the treatment of breast cancer.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Proteínas Tirosina Fosfatases/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Antineoplásicos/uso terapêutico , Apoptose , Neoplasias da Mama/tratamento farmacológico , Proliferação de Células , Transformação Celular Neoplásica , Feminino , Humanos , Camundongos , Camundongos Knockout , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/fisiologia , Transdução de Sinais , Trastuzumab
18.
Biochem J ; 458(2): 335-41, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24364879

RESUMO

NRG-1ß (neuregulin-1ß) serves multiple functions during embryonic heart development by signalling through ErbB family receptor tyrosine kinases (ErbB2, ErbB3 and ErbB4). Previous studies reported that NRG-1ß induces cardiomyogenesis of mESCs (mouse embryonic stem cells) at the later stages of differen-tiation through ErbB4 receptor activation. In the present study we systematically examined NRG-1ß induction of cardiac myocytes in mESCs and identified a novel time window, the first 48 h, for NRG-1ß-based cardiomyogenesis. At this time point ErbB3, but not ErbB4, is expressed. In contrast with the later differentiation of mESCs in which NRG-1ß induces cardiomyogenesis via the ErbB4 receptor, we found that knocking down ErbB3 or ErbB2 with siRNA during the early differentiation inhibited NRG-1ß-induced cardiomyogenesis in mESCs. Microarray analysis of RNA expression at this early time point indicated that NRG-1ß treatment in mESCs resulted in gene expression changes important to differentiation including up-regulation of components of PI3K (phosphoinositide 3-kinase), a known mediator of the NRG-1ß/ErbB signalling pathway, as well as activation of CREB (cAMP-response-element-binding protein). Further study demonstrated that the NRG-1ß-induced phosphorylation of CREB was required for cardiomyogenesis of mESCs. In summary, we report a previously unrecognized role for NRG-1ß/ErbB3/CREB signalling at the pre-mesoderm stage for stem cell cardiac differentiation.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/fisiologia , Miócitos Cardíacos/fisiologia , Neuregulina-1/fisiologia , Receptor ErbB-2/deficiência , Receptor ErbB-2/fisiologia , Receptor ErbB-3/fisiologia , Animais , Diferenciação Celular/genética , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Técnicas de Silenciamento de Genes , Camundongos , Proteínas do Tecido Nervoso/fisiologia , RNA Interferente Pequeno/genética , Receptor ErbB-3/deficiência , Receptor ErbB-3/genética , Transdução de Sinais/fisiologia
19.
Biochim Biophys Acta ; 1835(2): 230-42, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23403165

RESUMO

The analysis of clinical breast samples using biomarkers is integral to current breast cancer management. Currently, a limited number of targeted therapies are standard of care in breast cancer treatment. However, these targeted therapies are only suitable for a subset of patients and resistance may occur. Strategies to prevent the occurrence of invasive lesions are required to reduce the morbidity and mortality associated with the development of cancer. In theory, application of targeted therapies to pre-invasive lesions will prevent their progression to invasive lesions with full malignant potential. The diagnostic challenge for pathologists is to make interpretative decisions on early detected pre-invasive lesions. Overall, only a small proportion of these pre-invasive lesions will progress to invasive carcinoma and morphological assessment is an imprecise and subjective means to differentiate histologically identical lesions with varying malignant potential. Therefore differential biomarker analysis in pre-invasive lesions may prevent overtreatment with surgery and provide a predictive indicator of response to therapy. There follows a review of established and emerging potential druggable targets in pre-invasive lesions and correlation with lesion morphology.


Assuntos
Neoplasias da Mama/química , Lesões Pré-Cancerosas/química , Antígenos de Neoplasias/análise , Antígenos de Neoplasias/fisiologia , Biomarcadores , Neoplasias da Mama/tratamento farmacológico , DNA Topoisomerases Tipo II/análise , DNA Topoisomerases Tipo II/fisiologia , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/fisiologia , Feminino , Humanos , PTEN Fosfo-Hidrolase/análise , PTEN Fosfo-Hidrolase/química , PTEN Fosfo-Hidrolase/fisiologia , Fosfatidilinositol 3-Quinases/análise , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/fisiologia , Receptor ErbB-2/análise , Receptor ErbB-2/química , Receptor ErbB-2/fisiologia , Receptores de Estrogênio/análise , Receptores de Progesterona/análise , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/fisiologia
20.
Prostate ; 74(11): 1107-17, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24913716

RESUMO

BACKGROUND: The endocannabinoid system regulates cancer cell proliferation, and in prostate cancer a high cannabinoid CB1 receptor expression is associated with a poor prognosis. Down-stream mediators of CB1 receptor signaling in prostate cancer are known, but information on potential upstream regulators is lacking. RESULTS: Data from a well-characterized tumor tissue microarray were used for a Bayesian network analysis using the max-min hill-climbing method. In non-malignant tissue samples, a directionality of pEGFR (the phosphorylated form of the epidermal growth factor receptor) → CB1 receptors were found regardless as to whether the endocannabinoid metabolizing enzyme fatty acid amide hydrolase (FAAH) was included as a parameter. A similar result was found in the tumor tissue, but only when FAAH was included in the analysis. A second regulatory pathway, from the growth factor receptor ErbB2 → FAAH was also identified in the tumor samples. Transfection of AT1 prostate cancer cells with CB1 receptors induced a sensitivity to the growth-inhibiting effects of the CB receptor agonist CP55,940. The sensitivity was not dependent upon the level of receptor expression. Thus a high CB1 receptor expression alone does not drive the cells towards a survival phenotype in the presence of a CB receptor agonist. CONCLUSIONS: The data identify two potential regulators of the endocannabinoid system in prostate cancer and allow the construction of a model of a dysregulated endocannabinoid signaling network in this tumor. Further studies should be designed to test the veracity of the predictions of the network analysis in prostate cancer and other solid tumors.


Assuntos
Neoplasias da Próstata/patologia , Neoplasias da Próstata/fisiopatologia , Receptor CB1 de Canabinoide/fisiologia , Transdução de Sinais/fisiologia , Análise Serial de Tecidos/métodos , Amidoidrolases/fisiologia , Teorema de Bayes , Proliferação de Células , Receptores ErbB/fisiologia , Humanos , Masculino , Glicoproteínas de Membrana/fisiologia , Prognóstico , Neoplasias da Próstata/diagnóstico , Receptor ErbB-2/fisiologia , Estudos Retrospectivos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA