Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
PLoS One ; 16(12): e0261660, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34941939

RESUMO

Earlier, we proposed the "mechanosome" concept as a testable model for understanding how mechanical stimuli detected by cell surface adhesion molecules are transmitted to modulate gene expression inside cells. Here, for the first time we document a putative mechanosome involving Src, Pyk2 and MBD2 in MLO-Y4 osteocytes with high spatial resolution using FRET-FLIM. Src-Pyk2 complexes were concentrated at the periphery of focal adhesions and the peri-nuclear region. Pyk2-MBD2 complexes were located primarily in the nucleus and peri-nuclear region. Lifetime measurements indicated that Src and MBD2 did not interact directly. Finally, mechanical stimulation by fluid flow induced apparent accumulation of Src-Pyk2 protein complexes in the peri-nuclear/nuclear region, consistent with the proposed behavior of a mechanosome in response to a mechanical stimulus.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Osteócitos/metabolismo , Quinases da Família src/metabolismo , Animais , Linhagem Celular , Proteínas de Ligação a DNA/análise , Transferência Ressonante de Energia de Fluorescência , Quinase 2 de Adesão Focal/análise , Adesões Focais/metabolismo , Mecanotransdução Celular , Camundongos , Osteócitos/citologia , Quinases da Família src/análise
2.
Breast Cancer Res ; 20(1): 130, 2018 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-30359299

RESUMO

BACKGROUND: De novo or acquired resistance in breast cancer leads to treatment failures and disease progression. In human epidermal growth factor receptor 2 (HER2)-positive (HER2+) breast cancer, Src, a non-receptor tyrosine kinase, is identified as a major mechanism of trastuzumab resistance, with its activation stabilizing aberrant HER2 signaling, thus making it an attractive target for inhibition. Here, we explored the causal relationship between Src and HER2 by examining the potential of 89Zr-trastuzumab as a surrogate imaging marker of Src activity upon inhibition with dasatinib in HER2+ breast cancer. METHODS: HER2+ primary breast cancer cell lines BT-474 and trastuzumab-resistant JIMT-1 were treated with dasatinib and assessed for expression and localization of HER2, Src, and phosphorylated Src (pSrc) (Y416) through western blots and binding assays. Mice bearing BT-474 or JIMT-1 tumors were treated for 7 or 14 days with dasatinib. At the end of each treatment, tumors were imaged with 89Zr-trastuzumab. The results of 89Zr-trastuzumab positron emission tomography (PET) was compared against tumor uptake of fluorodeoxyglucose (18F-FDG) obtained the day before in the same group of mice. Ex vivo western blots and immunohistochemical staining (IHC) were performed for validation. RESULTS: In BT-474 and JIMT-1 cells, treatment with dasatinib resulted in a decrease in internalized 89Zr-trastuzumab. Confirmation with immunoblots displayed abrogation of pSrc (Y416) signaling; binding assays in both cell lines demonstrated a decrease in cell surface and internalized HER2-bound tracer. In xenograft models, dasatinib treatment for 7 days (BT-474, 11.05 ± 2.10 % injected dose per gram of tissue %(ID)/g; JIMT-1, 3.88 ± 1.47 %ID/g)) or 14 days (BT-474, 9.20 ± 1.85 %ID/g; JIMT-1, 4.45 ± 1.23 %ID/g) resulted in a significant decrease in 89Zr-trastuzumab uptake on PET compared to untreated control (BT-474, 17.88 ± 2.18 %ID/g; JIMT-1, 8.04 ± 1.47 %ID/g). No difference in 18F-FDG uptake was observed between control and treated cohorts. A parallel decrease in membranous HER2 and pSrc (Y416) staining was observed in tumors post treatment on IHC. Immunoblots further validated the 89Zr-trastuzumab-PET readout. Positive correlation was established between 89Zr-trastuzumab tumor uptake versus tumor regression, pSrc and pHER2 expression. CONCLUSIONS: 89Zr-trastuzumab can potentially assess tumor response to dasatinib in HER2+ breast cancer and could be used as a surrogate tool to monitor early changes in Src signaling downstream of HER2.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Imagem Molecular/métodos , Tomografia por Emissão de Pósitrons/métodos , Inibidores de Proteínas Quinases/farmacologia , Quinases da Família src/análise , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/farmacocinética , Mama/diagnóstico por imagem , Mama/patologia , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Dasatinibe/farmacologia , Dasatinibe/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Estudos de Viabilidade , Feminino , Fluordesoxiglucose F18/administração & dosagem , Fluordesoxiglucose F18/farmacocinética , Humanos , Camundongos , Camundongos Nus , Inibidores de Proteínas Quinases/uso terapêutico , Receptor ErbB-2/metabolismo , Distribuição Tecidual , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto , Quinases da Família src/antagonistas & inibidores
3.
JAMA Oncol ; 4(6): 814-820, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29710216

RESUMO

Importance: Gastrointestinal stromal tumors (GISTs) are life-threatening when metastatic or not amenable to surgical removal. In a few patients with advanced GISTs refractory to imatinib mesylate, treatment with sunitinib malate followed by regorafenib provides tumor control; however, additional active treatments are needed for most patients. Objective: To evaluate the 6-month progression-free survival (PFS), tumor objective response, and overall survival rates in patients with GISTs treated with dasatinib. Design, Setting, and Participants: This single-arm clinical trial used a Bayesian design to enroll patients 13 years or older with measurable imatinib-refractory metastatic GISTs treated at 14 sarcoma referral centers from June 1, 2008, through December 31, 2009. A control group was not included. Patients were followed up for survival for a minimum of 5 years from date of enrollment. Tumor imaging using computed tomography or magnetic resonance imaging was performed every 8 weeks for the first 24 weeks and every 12 weeks thereafter. Tumor response was assessed by local site using the Choi criteria. Treatment was continued until tumor progression, unacceptable toxic effects after reduction in drug dose, or patient or physician decision. Archival tumor tissue was evaluated for expression of the proto-oncogene tyrosine-protein kinase Src (SRC), phosphorylated SRC (pSRC), and succinate dehydrogenase complex iron sulfur subunit B (SDHB) proteins and for mutation in the V-Kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog (KIT) and platelet-derived growth factor receptor α (PDGFRA) genes. Data analysis was performed from May 19, 2017, through December 20, 2017. Interventions: Dasatinib, 70 mg orally twice daily. Main Outcomes and Measures: The primary end point was the 6-month PFS estimate using greater than 30% as evidence of an active drug and less than 10% as evidence of inactive treatment. Results: In this study, 50 patients were enrolled (median age, 60 years; age range, 19-78 years; 31 [62%] male and 19 [38%] female; 41 [82%] white), and 48 were evaluable for response. The estimated 6-month PFS rate was 29% in the overall population and 50% in a subset of 14 patients with pSRC in GISTs. Objective tumor response was observed in 25%, including 1 patient with an imatinib-resistant mutation in PDGFRA exon 18. Conclusions and Relevance: Dasatinib may have activity in a subset of patients with imatinib-resistant GISTs. Further study is needed to determine whether pSRC is a prognostic biomarker.


Assuntos
Antineoplásicos/uso terapêutico , Dasatinibe/uso terapêutico , Neoplasias Gastrointestinais/tratamento farmacológico , Tumores do Estroma Gastrointestinal/secundário , Mesilato de Imatinib/uso terapêutico , Terapia de Alvo Molecular , Inibidores de Proteínas Quinases/uso terapêutico , Adulto , Idoso , Resistencia a Medicamentos Antineoplásicos , Substituição de Medicamentos , Feminino , Seguimentos , Neoplasias Gastrointestinais/química , Neoplasias Gastrointestinais/mortalidade , Tumores do Estroma Gastrointestinal/química , Tumores do Estroma Gastrointestinal/tratamento farmacológico , Tumores do Estroma Gastrointestinal/mortalidade , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/análise , Intervalo Livre de Progressão , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-kit/análise , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/análise , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Succinato Desidrogenase/análise , Adulto Jovem , Quinases da Família src/análise
4.
J Proteome Res ; 17(4): 1415-1425, 2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29457907

RESUMO

Adipose triglyceride lipase (ATGL) catalyzes the rate limiting step in triacylglycerol breakdown in adipocytes but is expressed in most tissues. The enzyme was shown to be lost in many human tumors, and its loss may play a role in early stages of cancer development. Here, we report that loss of ATGL supports a more-aggressive cancer phenotype in a model system in which ATGL was deleted in A549 lung cancer cells by CRISPR/Cas9. We observed that loss of ATGL led to triacylglycerol accumulation in lipid droplets and higher levels of cellular phospholipid and bioactive lipid species (lyso- and ether-phospholipids). Label-free quantitative proteomics revealed elevated expression of the pro-oncogene SRC kinase in ATGL depleted cells, which was also found on mRNA level and confirmed on protein level by Western blot. Consistently, higher expression of phosphorylated (active) SRC (Y416 phospho-SRC) was observed in ATGL-KO cells. Cells depleted of ATGL migrated faster, which was dependent on SRC kinase activity. We propose that loss of ATGL may thus increase cancer aggressiveness by activation of pro-oncogenic signaling via SRC kinase and increased levels of bioactive lipids.


Assuntos
Lipase/deficiência , Neoplasias Pulmonares/patologia , Triglicerídeos/metabolismo , Células A549 , Movimento Celular/efeitos dos fármacos , Deleção de Genes , Humanos , Lipase/genética , Metabolismo dos Lipídeos , Fenótipo , Proteômica , Transdução de Sinais/efeitos dos fármacos , Quinases da Família src/análise , Quinases da Família src/metabolismo , Quinases da Família src/farmacologia
5.
Hum Pathol ; 67: 119-125, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28601656

RESUMO

Src belongs to a family of cytoplasmic tyrosine kinases that play a key role in tumor initiation and progression. Src activation has been associated with a more aggressive neoplastic phenotype and induces resistance to platinum agents in preclinical models. The aim of our study was to assess the prognostic and/or predictive value of Src activation in patients with stage II-III colon cancer. pSrc expression was assessed in paraffin-embedded tumor samples by immunohistochemistry (phospho-Y418, ab4816; Abcam). Cases were classified by staining intensity in 4 categories: no staining (0), weak (1+), moderate (2+), and intense (3+) staining. A total of 487 patients were evaluated (240 stage II, 247 stage III), of whom 298 (61%) had received adjuvant chemotherapy. Staining was absent in 78 (16%), weak in 262 (54%), moderate in 103 (21%), and intense in 44 (9%). High pSrc expression was significantly associated with decreased 5-year disease-free survival (39% versus 63% for patients with high versus low pSrc expression; hazard ratio, 0.56; P=.005) and overall survival (58% versus 74%; hazard ratio, 0.55; P=.02). Multivariate analysis confirmed pSrc expression as a significant prognostic factor both for disease-free survival and overall survival, independent of age, sex, tumor stage, bowel obstruction/perforation, or adjuvant chemotherapy. These findings illustrate the relevance of Src activation in colon cancer biology, conferring a poor prognosis to patients with early stage colon cancer regardless of adjuvant chemotherapy. Our findings may help improve prognostic stratification of patients for clinical decisions and open new avenues for potential pharmacologic manipulation that may eventually improve patients' outcomes.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias do Colo/enzimologia , Quinases da Família src/análise , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Quimioterapia Adjuvante , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/mortalidade , Neoplasias do Colo/patologia , Progressão da Doença , Intervalo Livre de Doença , Resistencia a Medicamentos Antineoplásicos , Ativação Enzimática , Feminino , Fluoruracila/uso terapêutico , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Compostos Organoplatínicos/uso terapêutico , Oxaliplatina , Fosforilação , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento
6.
Oncotarget ; 7(19): 28340-55, 2016 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-27078847

RESUMO

Aberrant activation of cellular Src (c-Src), a non-receptor tyrosine kinase, could promote cancer progression through activating its downstream signaling pathways. However, the roles of c-Src and phosphorylated-Src (p-Src) in nasopharyngeal carcinoma (NPC) progression are rarely investigated. Herein, we have identified high c-Src concentrations in the serum of NPC patients with distant metastasis using high-throughput protein microarrays. Levels of c-Src in serum and p-Src in human primary NPC samples were unfavorable independent prognostic factors for cancer-specific survival, disease-free survival, and distant metastasis-free survival. Depletion or inactivation of c-Src in NPC cells using sgRNA with CRISPR/Cas9 system or PP2 decreased cell viability, colony formation, migration and invasion in vitro and metastasis in vivo. In contrast, these malignancies could be up-regulated by overexpressed c-Src in a NPC cell line with low-metastasis potential. Furthermore, p-Src was involved in promoting NPC cell metastasis by inducing the epithelial-mesenchymal transition (EMT) process via activating the PI3K/Akt pathway and cytoskeleton remodeling. The p-Src-induced EMT process could be retarded by PP2, which mediated by down-regulating the PI3K/Akt pathway. In conclusion, elevated levels of c-Src in serum and p-Src in primary NPC tissue correlated with poor outcomes of NPC patients. And aberrant activation of c-Src facilitated NPC cells with malignant potential, especially metastasis ability, which mediated by the PI3K/Akt pathway activation and sequentially induced the EMT process. These findings unveiled a promising approach for targeted therapy of advanced NPC.


Assuntos
Carcinoma/patologia , Transição Epitelial-Mesenquimal/fisiologia , Neoplasias Nasofaríngeas/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinases da Família src/metabolismo , Animais , Biomarcadores Tumorais/análise , Proteína Tirosina Quinase CSK , Carcinoma/metabolismo , Carcinoma/mortalidade , Intervalo Livre de Doença , Xenoenxertos , Humanos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Nus , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/mortalidade , Invasividade Neoplásica/patologia , Transdução de Sinais/fisiologia , Quinases da Família src/análise
7.
Mol Med Rep ; 13(6): 4774-8, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27082851

RESUMO

A previous study by our group demonstrated that overexpression of KAI1 was associated with lymphatic metastasis in pancreatic cancer. The present study further investigated the signaling pathways involved in KAI1­induced downregulation of vascular endothelial growth factor C (VEGF­C) and lymphatic metastasis in pancreatic cancer. Immunohistochemistry was performed to examine KAI1 and VEGF­C expression in 28 surgically resected pancreatic cancer tissues. MIA PaCa­2 and PCAN1 pancreatic cancer cell lines were transfected with KAI1 overexpression vector. VEGF­C expression as well as phosphorylation of Src and signal transducer and activator of transcription (STAT)3 were assessed by western blot analysis. Furthermore, the signal transduction inhibitors PP2 and AG490 were used to block the Src and STAT3 signaling pathways, respectively. KAI1 was negatively correlated with VEGF­C expression in pancreatic tumor samples. In MIA PaCa­2 cells, VEGF­C expression was more significantly inhibited by restoration of KAI1 than that in PCAN1 cells. In addition, Src and STAT3 phosphorylation was decreased by KAI1 in MIA PaCa­2 cells. Of note, pre­treatment with PP2 efficiently reversed the KAI1-induced enhancement of Src and STAT3 phosphorylation as well as VEGF­C expression. Pre­treatment with AG490 efficiently reversed the KAI1-induced enhancement of STAT3 phosphorylation and VEGF­C expression, but had no effect on the upregulation of Src phosphorylation. The present study identified the involvement of Src/STAT3 signaling pathways in KAI1­induced downregulation of VEGF­C expression and suggested the implication of these pathways in lymphatic metastasis of pancreatic cancer.


Assuntos
Proteína Kangai-1/metabolismo , Neoplasias Pancreáticas/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Fator C de Crescimento do Endotélio Vascular/metabolismo , Quinases da Família src/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , Humanos , Proteína Kangai-1/análise , Pâncreas/metabolismo , Pâncreas/patologia , Neoplasias Pancreáticas/patologia , Fosforilação , Fator de Transcrição STAT3/análise , Fator C de Crescimento do Endotélio Vascular/análise , Quinases da Família src/análise
8.
Theranostics ; 6(4): 594-609, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26941850

RESUMO

Aberrant activation of the Src kinase is implicated in the development of a variety of human malignancies. However, it is almost impossible to monitor Src activity in an in vivo setting with current biochemical techniques. To facilitate the noninvasive investigation of the activity of Src kinase both in vitro and in vivo, we developed a genetically engineered, activatable bioluminescent reporter using split-luciferase complementation. The bioluminescence of this reporter can be used as a surrogate for Src activity in real time. This hybrid luciferase reporter was constructed by sandwiching a Src-dependent conformationally responsive unit (SH2 domain-Srcpep) between the split luciferase fragments. The complementation bioluminescence of this reporter was dependent on the Src activity status. In our study, Src kinase activity in cultured cells and tumor xenografts was monitored quantitatively and dynamically in response to clinical small-molecular kinase inhibitors, dasatinib and saracatinib. This system was also applied for high-throughput screening of Src inhibitors against a kinase inhibitor library in living cells. These results provide unique insights into drug development and pharmacokinetics/phoarmocodynamics of therapeutic drugs targeting Src signaling pathway enabling the optimization of drug administration schedules for maximum benefit. Using both Firefly and Renilla luciferase imaging, we have successfully monitored Src tyrosine kinase activity and Akt serine/threonine kinase activity concurrently in one tumor xenograft. This dual luciferase reporter imaging system will be helpful in exploring the complex signaling networks in vivo. The strategies reported here can also be extended to study and image other important kinases and the cross-talks among them.


Assuntos
Genes Reporter , Luciferases de Renilla/análise , Medições Luminescentes , Neoplasias/diagnóstico por imagem , Quinases da Família src/análise , Animais , Linhagem Celular Tumoral , Xenoenxertos , Luciferases de Renilla/genética , Camundongos , Análise de Sequência de DNA
9.
J Oral Pathol Med ; 45(8): 591-8, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26752341

RESUMO

BACKGROUND: Cell migration and invasion through interstitial tissues are dependent upon several specialized characteristics of the migratory cell notably generation of proteolytic membranous protrusions or invadopodia. Ameloblastoma is a benign odontogenic epithelial neoplasm with a locally infiltrative behaviour. Cortactin and MMT1-MMP are two invadopodia proteins implicated in its local invasiveness. Other invadopodia regulators, namely N-WASP, WIP and Src kinase remain unclarified. This study addresses their roles in ameloblastoma. MATERIALS AND METHOD: Eighty-seven paraffin-embedded ameloblastoma cases (20 unicystic, 47 solid/multicystic, 3 desmoplastic and 17 recurrent) were subjected to immunohistochemistry for expression of cortactin, N-WASP, WIP, Src kinase and F-actin, and findings correlated with clinicopathological parameters. RESULTS: Invadopodia proteins (except Src kinase) and F-actin were widely detected in ameloblastoma (cortactin: n = 73/87, 83.9%; N-WASP: n = 59/87; 67.8%; WIP: n = 77/87; 88.5%; and F-actin: n = 87/87, 100%). Protein localization was mainly cytoplasmic and/or membranous, and occasionally nuclear for F-actin. Cortactin, which functions as an actin-scaffolding protein, demonstrated significantly higher expression levels within ameloblastoma tumoral epithelium than in stroma (P < 0.05). N-WASP, which coordinates actin polymerization and invadopodia-mediated extracellular matrix degradation, was overexpressed in the solid/multicystic subtype (P < 0.05). WIP, an upstream regulator of N-WASP, and F-actin were significantly upregulated along the tumour invasive front compared to tumour centres (P < 0.05). Except for males with cortactin overexpression, other clinical parameters (age, ethnicity and anatomical site) showed no significant correlations. CONCLUSIONS: Present results suggest that local invasiveness of ameloblastoma is dependent upon the migratory potential of its tumour cells as defined by their distribution of cortactin, N-WASP and WIP in correlation with F-actin cytoskeletal dynamics.


Assuntos
Ameloblastoma/metabolismo , Cortactina/fisiologia , Proteínas do Citoesqueleto/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neoplasias Maxilomandibulares/metabolismo , Podossomos/fisiologia , Proteína Neuronal da Síndrome de Wiskott-Aldrich/fisiologia , Actinas/análise , Actinas/biossíntese , Actinas/fisiologia , Adolescente , Adulto , Idoso , Ameloblastoma/patologia , Movimento Celular/fisiologia , Criança , Cortactina/biossíntese , Proteínas do Citoesqueleto/biossíntese , Feminino , Humanos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Neoplasias Maxilomandibulares/patologia , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Neoplasias Epiteliais e Glandulares/metabolismo , Células Estromais/metabolismo , Células Estromais/patologia , Proteína Neuronal da Síndrome de Wiskott-Aldrich/biossíntese , Adulto Jovem , Quinases da Família src/análise , Quinases da Família src/fisiologia
10.
Artigo em Inglês | MEDLINE | ID: mdl-26297394

RESUMO

OBJECTIVE: To describe the clinicopathologic and immunohistochemical features of five cases of adenoid ameloblastoma. STUDY DESIGN: Clinicopathologic data were gathered from medical records and compared with those compiled from a systematic review. Slides were also immunohistochemically stained for Ki-67, p16, p53, and cytokeratins (7, 8, 14, 18, and 19). RESULTS: There were 3 males (60%) and 2 (40%) females. The mean age was 44 ± 10 years. Of the five adenoid ameloblastomas, 4 (80%) occurred in the posterior maxilla. Patients typically complained of asymptomatic swelling. All patients received surgical resection as primary therapy; 1 (20%) patient also received adjuvant radiotherapy. Recurrence was diagnosed in all patients. Immunohistochemically, the tumors stained focally positive for CK7, 8, 14, and 18 and diffusely positive for CK-19, p16, and p53. The mean Ki-67-positive cells were 72.4 ± 24.9 positive cells per high-power field (range 53-111). CONCLUSIONS: To our knowledge, this is the largest series of adenoid ameloblastoma reported in the literature. Our data suggest that this entity demonstrates aggressive behavior characterized by a high likelihood of recurrence.


Assuntos
Tonsila Faríngea/patologia , Ameloblastoma/patologia , Neoplasias Nasofaríngeas/patologia , Adulto , Ameloblastoma/terapia , Biomarcadores Tumorais/análise , Inibidor p16 de Quinase Dependente de Ciclina , Feminino , Humanos , Técnicas Imunoenzimáticas , Queratina-19/análise , Masculino , Neoplasias Nasofaríngeas/terapia , Proteínas de Neoplasias/análise , Recidiva Local de Neoplasia/patologia , Proteínas Proto-Oncogênicas/análise , Quinases da Família src/análise
11.
PLoS One ; 9(10): e109304, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25275584

RESUMO

In vitro expanded bone marrow stromal cells contain at least two populations of fibroblasts, a CD146/MCAM positive population, previously reported to be critical for establishing the stem cell niche and a CD146-negative population that expresses CUB domain-containing protein 1 (CDCP1)/CD318. Immunohistochemistry of marrow biopsies shows that clusters of CDCP1+ cells are present in discrete areas distinct from areas of fibroblasts expressing CD146. Using a stromal cell line, HS5, which approximates primary CDCP1+ stromal cells, we show that binding of an activating antibody against CDCP1 results in tyrosine-phosphorylation of CDCP1, paralleled by phosphorylation of Src Family Kinases (SFKs) Protein Kinase C delta (PKC-δ). When CDCP1 expression is knocked-down by siRNA, the expression and secretion of myelopoietic cytokines is increased. These data suggest CDCP1 expression can be used to identify a subset of marrow fibroblasts functionally distinct from CD146+ fibroblasts. Furthermore the CDCP1 protein may contribute to the defining function of these cells by regulating cytokine expression.


Assuntos
Antígenos CD/análise , Células da Medula Óssea/imunologia , Antígeno CD146/análise , Moléculas de Adesão Celular/análise , Fibroblastos/imunologia , Proteínas de Neoplasias/análise , Adulto , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos de Neoplasias , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Antígeno CD146/imunologia , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Células Cultivadas , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Imunofenotipagem , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Proteína Quinase C-delta/análise , Proteína Quinase C-delta/imunologia , Interferência de RNA , RNA Interferente Pequeno/genética , Células Estromais/citologia , Células Estromais/imunologia , Células Estromais/metabolismo , Quinases da Família src/análise , Quinases da Família src/imunologia
12.
Adv Exp Med Biol ; 759: 33-56, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25030759

RESUMO

The oocyte is a highly specialized cell poised to respond to fertilization with a unique set of actions needed to recognize and incorporate a single sperm, complete meiosis, reprogram maternal and paternal genomes and assemble them into a unique zygotic genome, and finally initiate the mitotic cell cycle. Oocytes accomplish this diverse series of events through an array of signal transduction pathway components that include a characteristic collection of protein tyrosine kinases. The src-family protein kinases (SFKs) figure importantly in this signaling array and oocytes characteristically express certain SFKs at high levels to provide for the unique actions that the oocyte must perform. The SFKs typically exhibit a distinct pattern of subcellular localization in oocytes and perform critical functions in different subcellular compartments at different steps during oocyte maturation and fertilization. While many aspects of SFK signaling are conserved among oocytes from different species, significant differences exist in the extent to which src-family-mediated pathways are used by oocytes from species that fertilize externally vs those which are fertilized internally. The observation that several oocyte functions which require SFK signaling appear to represent common points of failure during assisted reproductive techniques in humans, highlights the importance of these signaling pathways for human reproductive health.


Assuntos
Fertilização/fisiologia , Oócitos/fisiologia , Oogênese/fisiologia , Quinases da Família src/fisiologia , Animais , Evolução Biológica , Humanos , Transdução de Sinais/fisiologia , Quinases da Família src/análise , Quinases da Família src/química
13.
Oncotarget ; 5(5): 1382-9, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24675526

RESUMO

Blood borne metastatic tumor cell adhesion to endothelial cells constitutes a critical rate-limiting step in hematogenous cancer metastasis. Interactions between cancer associated carbohydrate Thomsen-Friedenreich antigen (TF-Ag) and endothelium-expressed galectin-3 (Gal-3) have been identified as the leading molecular mechanism initiating tumor/endothelial cell adhesion in several types of cancer. However, it is unknown how these rather weak and transient carbohydrate/lectin mediated interactions are stabilized. Here, using Western blot and LC tandem mass spectrometry analyses of pull-downs utilizing TF-Ag loaded gold nanoparticles, we identified Gal-3, endothelial integrin α3ß1, Src kinase, as well as 5 additional molecules mapping onto focal adhesion pathway as parts of the macromolecular complexes formed at the endothelial cell membranes downstream of TF-Ag/Gal-3 interactions. In a modified parallel flow chamber assay, inhibiting α3ß1 integrin greatly reduced the strength of tumor/endothelial cell interactions without affecting the initial cancer cell adhesion. Further, the macromolecular complex induced by TF-Ag/Gal-3/α3ß1 interactions activates Src kinase, p38, and ERK1/2, pathways in endothelial cells in a time- and α3ß1-dependent manner. We conclude that, following the initial metastatic cell attachment to endothelial cells mediated by TF-Ag/Gal-3 interactions, endothelial integrin α3ß1 stabilizes tumor/endothelial cell adhesion and induces the formation of macromolecular signaling complex activating several major signaling pathways in endothelial cells.


Assuntos
Antígenos Glicosídicos Associados a Tumores/metabolismo , Adesão Celular/fisiologia , Células Endoteliais/fisiologia , Galectina 3/metabolismo , Integrina alfa3beta1/metabolismo , Sistema de Sinalização das MAP Quinases , Metástase Neoplásica/fisiopatologia , Neoplasias da Próstata/metabolismo , Linhagem Celular Tumoral , Membrana Celular/química , Membrana Celular/metabolismo , Células Endoteliais/química , Galectina 3/análise , Humanos , Integrina alfa3beta1/análise , Substâncias Macromoleculares/metabolismo , Masculino , Quinases da Família src/análise , Quinases da Família src/metabolismo
14.
Anal Sci ; 29(5): 491-7, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23665620

RESUMO

Src homology 2 (SH2) domains are modules of approximately 100 amino acids and are known to bind phosphotyrosine-containing sequences with high affinity and specificity. In the present work, we developed an SH2 domain-based assay for Src tyrosine kinase using a unique biotinylation reaction from archaeon Sulfolobus tokodaii. S. tokodaii biotinylation has a unique property that biotin protein ligase (BPL) forms a stable complex with its biotinylated substrate protein (BCCP). Here, an SH2 domain from lymphocyte-specific tyrosine kinase was genetically fused to a truncated BCCP, and the resulting fusion protein was labeled through biotinylation with BPL carrying multiple copies of a luminescent Tb(3+) complex. The labeled SH2 fusion proteins were employed to detect a phosphorylated peptide immobilized on the surface of the microtiter plate, where the phosphorylated peptide was produced by phosphorylation to the substrate peptide by Src tyrosine kinase. Our assay allows for a reliable determination of the activity of Src kinase lower than 10 pg/µL by a simple procedure.


Assuntos
Biotina/química , Ligases/química , Térbio/química , Domínios de Homologia de src , Quinases da Família src/análise , Modelos Moleculares , Reação em Cadeia da Polimerase , Sulfolobus/enzimologia
15.
Int J Clin Exp Pathol ; 6(6): 1121-7, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23696930

RESUMO

AIMS: The molecular mechanisms of the tumorigenesis and recurrence of cervical cancer are poorly understood. The objective of this study was to analyze the expression of phosphorylated c-Src (phospho-c-Src) and its clinical significance in human cervical cancer. METHODS: The expression of phospho-c-Src was determined by immunohistochemistry in a total of 127 cervical specimens including 20 normal cervix tissues, 20 cases of carcinoma in situ of cervix (CIS), and 87 cases of cervical squamous cell carcinoma (CSCC). RESULTS: The expression of phospho-Src in normal cervix, CIS, and CSCC increased gradually in ascending order (p=0.026). In addition, the expression of phospho-Src was correlated with overall (p=0.037) and recurrence (p=0.001) survival of cervical cancer. In multivariate Cox regression analysis, phospho-Src expression was an independent prognosis factor for recurrence-free survival (p=0.004). CONCLUSION: Our present study suggests that Src signaling may play essential role in cervical cancer progression. Phospho-Src expression may be considered as a prognostic marker to predict recurrence in CSCC.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma in Situ/enzimologia , Carcinoma de Células Escamosas/enzimologia , Recidiva Local de Neoplasia/enzimologia , Neoplasias do Colo do Útero/enzimologia , Quinases da Família src/análise , Adulto , Biópsia , Carcinoma in Situ/mortalidade , Carcinoma in Situ/patologia , Carcinoma de Células Escamosas/mortalidade , Carcinoma de Células Escamosas/patologia , Estudos de Casos e Controles , Intervalo Livre de Doença , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Análise Multivariada , Invasividade Neoplásica , Recidiva Local de Neoplasia/mortalidade , Recidiva Local de Neoplasia/patologia , Fosforilação , Valor Preditivo dos Testes , Prognóstico , Modelos de Riscos Proporcionais , Fatores de Risco , Fatores de Tempo , Neoplasias do Colo do Útero/mortalidade , Neoplasias do Colo do Útero/patologia
16.
Cell Signal ; 25(9): 1852-60, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23707526

RESUMO

Type III receptor tyrosine kinases (RTKs), FLT3 and c-Kit play important roles in a variety of cellular processes. A number of SH2-domain containing proteins interact with FLT3 and c-Kit and regulate downstream signaling. The SH2-domain containing non-receptor protein tyrosine kinase CSK is mainly studied in the context of regulating Src family kinases. Here we present an additional role of this kinase in RTK signaling. We show that CSK interacts with FLT3 and c-Kit in a phosphorylation dependent manner. This interaction is facilitated through the SH2-domain of CSK. Under basal conditions CSK is mainly localized throughout the cytosolic compartment but upon ligand stimulation it is recruited to the inner side of cell membrane. CSK association did not alter receptor ubiquitination or phosphorylation but disrupted downstream signaling. Selective depletion of CSK using siRNA, or inhibition with CSK inhibitor, led to increased phosphorylation of Akt and Erk, but not p38, upon FLT3 ligand (FL) stimulation. Stem cell factor (SCF)-mediated Akt and Erk activation was also elevated by CSK inhibition. However, siRNA mediated CSK knockdown increased SCF stimulated Akt phosphorylation but decreased Erk phosphorylation. CSK depletion also significantly increased both FL- and SCF-induced SHC, Gab2 and SHP2 phosphorylation. Furthermore, CSK depletion contributed to oncogenic FLT3- and c-Kit-mediated cell proliferation, but not to cell survival. Thus, the results indicate that CSK association with type III RTKs, FLT3 and c-Kit can have differential impact on receptor downstream signaling.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Transdução de Sinais , Quinases da Família src/metabolismo , Animais , Células COS , Proteína Tirosina Quinase CSK , Linhagem Celular , Chlorocebus aethiops , Humanos , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Mapas de Interação de Proteínas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Ubiquitinação , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Domínios de Homologia de src , Quinases da Família src/análise , Quinases da Família src/química , Quinases da Família src/genética
17.
Biosens Bioelectron ; 46: 97-101, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23517824

RESUMO

Fluorescent protein (FP)-based Förster resonance energy transfer (FRET) biosensors are powerful tools for dynamically measuring cellular molecular events because they offer high spatial and temporal resolution in living cells. Despite the broad use of FP-based FRET biosensors in cell biology, imaging of multiple molecular events (multi-parameter molecular imaging) in single cells using current FRET pairs remains difficult because it usually requires a control group for additional data calibration. Hence, spectrally compatible FRET pairs that do not require complex image calibration are the key to widespread applications of FP-based FRET biosensors in multi-parameter molecular imaging. Here, we report a new combination of spectrally distinguishable FRET pairs for dual-parameter molecular imaging: mTagBFP/sfGFP (blue and green FP, B/G) and mVenus/mKOκ (yellow and orange FP, Y/O). We demonstrate that additional image correction is not necessary for these dual FRET pairs. Using these dual FRET pairs, we achieve simultaneous imaging of Src and Ca(2+) signaling in single living cells stimulated with epithelial growth factor (EGF). By converting traditional FRET biosensors into B/G and Y/O-based biosensors, additional applications are available to elucidate the dynamic relationships of multiple molecular events within a single living cell.


Assuntos
Sinalização do Cálcio , Transferência Ressonante de Energia de Fluorescência/métodos , Substâncias Luminescentes/análise , Proteínas Luminescentes/análise , Imagem Molecular/métodos , Quinases da Família src/análise , Cálcio/análise , Cálcio/metabolismo , Células HeLa , Humanos , Microscopia Confocal/métodos , Análise de Célula Única/métodos , Quinases da Família src/metabolismo
18.
Radiother Oncol ; 105(2): 241-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23010482

RESUMO

BACKGROUND AND PURPOSE: Although inhibition of epidermal growth factor receptor (EGFR) signaling during radiation led to improvement of tumor control and survival, novel strategies are needed to further improve the outcome of patients with locally advanced head and neck carcinoma. Because EGFR is known to interact with c-Src kinases, the present study investigated dasatinib (BMS-354825), an inhibitor of c-Src kinases, for its efficacy in enhancing radiosensitivity of human head and neck squamous cell carcinomas (HNSCC) in vitro and examined the underlying mechanisms for this effect. MATERIALS AND METHODS: Six HNSCC lines were exposed to dasatinib, radiation, or both, and assessed for c-Src and EGFR expression, cell survival and colony forming ability. Among these cell lines, HN-5 and FaDu lines were analyzed for induction of apoptosis, cell cycle re-distribution and for nuclear localization of EGFR, γ-H2AX and 53BP1 proteins. Immuno-precipitation and Western blots were performed to analyze the levels and binding of proteins involved in cell survival, apoptosis and DNA repair pathways. Suppression of c-Src by siRNA and subsequent clonogenic assay was performed in HN-5 cells. RESULTS: All six HNSCC lines that were examined expressed high levels of c-Src. Two (HN-5 and MDA-183) expressed higher levels of EGFR than other lines. Dasatinib suppressed cell survival of all cell lines tested independent of c-Src or EGFR levels but enhanced the radiosensitivity of HN-5 and MDA-183. HN-5 and FaDu were analyzed further. Dasatinib suppressed phosphorylation of c-Src in both cell lines, but decreased repair of radiation-induced DNA damage in HN-5 cells only as evidenced by suppression of c-Abl and Nbs-1 activity, inhibition of the association between c-Src and EGFR or Her-2, prolongation of nuclear γ-H2AX and 53BP1 foci and inhibition of EGFR nuclear localization and its association with DNA-PKcs. Finally, partial suppression of c-Src resulted in a small increase in HN-5 cell radiosensitivity. CONCLUSIONS: Our data demonstrate that dasatinib induces apoptosis and blocks DNA repair in EGFR-expressing HNSCC cells and improves radiotherapy outcome. These findings warrant further investigation using in vivo tumor models for potential translation into clinical testing.


Assuntos
Carcinoma de Células Escamosas/radioterapia , Núcleo Celular/metabolismo , Reparo do DNA/efeitos dos fármacos , Receptores ErbB/metabolismo , Neoplasias de Cabeça e Pescoço/radioterapia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Tiazóis/farmacologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Apoptose/efeitos da radiação , Proteína Tirosina Quinase CSK , Ciclo Celular , Dasatinibe , Receptores ErbB/análise , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço , Quinases da Família src/análise , Quinases da Família src/metabolismo
19.
Placenta ; 33(9): 704-11, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22766276

RESUMO

OBJECTIVES: Our previous studies have shown that Cyclosporin A (CsA) promotes human trophoblast invasion via mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathway. E-cadherin and matrix metalloproteinases (MMPs) are important mediators in trophoblast migration and invasion. Here, we further investigate the role of focal adhesion kinase (FAK) signaling in the CsA-induced trophoblast migration and invasion and ERK activation. STUDY DESIGN: The migration and invasion of human primary trophoblasts and JEG-3 cells were measured by transwell migration and matrigel invasion assays. The activation of FAK, Src and ERK induced by CsA were examined by western blot. The colocalization of FAK and Src was detected by dual immunofluorescence assay. The regulation of E-cadherin expression and matrix metalloproteinases (MMPs) activity was evaluated by western blot and gelatin zymography, respectively. RESULTS: CsA increased the phosphorylation of FAK and Src in human primary trophoblasts and JEG-3 cells. Meanwhile, the activated FAK and Src colocalized in the cytoplasm of JEG-3 cells. The FAK inhibitor Y15 or Src inhibitor PP2 could abrogate the phosphorylation of ERK, the enhanced migration and invasion and the activity of MMP2, 9 induced by CsA. In addition, these inhibitors also restored the expression of E-cadherin which is down-regulated by CsA. However, U0126, an inhibitor of ERK, had no significant effect on the CsA-induced activation of FAK and Src. CONCLUSIONS: FAK-Src signaling, the upstream signaling cascade of ERK activation, plays an important role in the CsA-induced migration and invasion via down-regulating expression of E-cadherin and up-regulating activity of MMP2, 9 in trophoblast cells. These results may help provide a rationale to develop a novel therapeutic strategy for pregnancy disorders from insufficient trophoblast invasion.


Assuntos
Movimento Celular/fisiologia , Ciclosporina/farmacologia , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Transdução de Sinais/fisiologia , Trofoblastos/fisiologia , Caderinas/análise , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Citoplasma/química , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Proteína-Tirosina Quinases de Adesão Focal/análise , Humanos , Imunossupressores/farmacologia , Metaloproteinase 2 da Matriz/análise , Metaloproteinase 9 da Matriz/análise , Gravidez , Transdução de Sinais/efeitos dos fármacos , Trofoblastos/efeitos dos fármacos , Trofoblastos/ultraestrutura , Quinases da Família src/análise , Quinases da Família src/metabolismo
20.
Reproduction ; 144(1): 67-75, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22573827

RESUMO

During the capacitation process, spermatozoa acquire the ability to fertilize an oocyte, and upregulation of cAMP-dependent protein tyrosine phosphorylation occurs. Recently, Src family tyrosine kinase (SFK) has been involved in spermatozoa capacitation as a key PKA-dependent tyrosine kinase in several species. This work investigates the expression and role of SFK in porcine spermatozoa. SFK members Lyn and Yes are identified in porcine spermatozoa by western blotting as well as two proteins named SFK1 and SFK2 were also detected by their tyrosine 416 phosphorylation, a key residue for SFK activation. Spermatozoa with SFK1 and SFK2 increase their Y416 phosphorylation time-dependently under capacitating conditions compared with noncapacitating conditions. The specific SFK inhibitor SU6656 unaffected porcine spermatozoa motility or viability. Moreover, SFK inhibition in spermatozoa under capacitating conditions leads to a twofold increase in both nonstimulated and calcium-induced acrosome reaction. Our data show that capacitating conditions lead to a time-dependent increase in actin polymerization in boar spermatozoa and that long-term incubation with SFK inhibitor causes a reduction in the F-actin content. In summary, this work shows that the SFK members Lyn and Yes are expressed in porcine spermatozoa and that SFK1 and SFK2 are phosphorylated (activated) during capacitation. Our results point out the important role exerted by SFK in the acrosome reaction, likely mediated in part by its involvement in the actin polymerization process that accompanies capacitation, and rule out its involvement in porcine spermatozoa motility.


Assuntos
Reação Acrossômica/fisiologia , Motilidade dos Espermatozoides/fisiologia , Suínos , Quinases da Família src/fisiologia , Reação Acrossômica/efeitos dos fármacos , Actinas/análise , Animais , Cálcio/farmacologia , Ativação Enzimática , Indóis/farmacologia , Masculino , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-yes/análise , Capacitação Espermática/fisiologia , Espermatozoides/enzimologia , Sulfonamidas/farmacologia , Quinases da Família src/análise , Quinases da Família src/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...