Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 132(22)2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36377656

RESUMO

Cancer immunotherapy targeting the TIGIT/PVR pathway is currently facing challenges. KIR2DL5, a member of the human killer cell, immunoglobulin-like receptor (KIR) family, has recently been identified as another binding partner for PVR. The biology and therapeutic potential of the KIR2DL5/PVR pathway are largely unknown. Here we report that KIR2DL5 was predominantly expressed on human NK cells with mature phenotype and cytolytic function and that it bound to PVR without competition with the other 3 known PVR receptors. The interaction between KIR2DL5 on NK cells and PVR on target cells induced inhibitory synapse formation, whereas new monoclonal antibodies blocking the KIR2DL5-PVR interaction robustly augmented the NK cytotoxicity against PVR+ human tumors. Mechanistically, both intracellular ITIM and ITSM of KIR2DL5 underwent tyrosine phosphorylation after engagement, which was essential for KIR2DL5-mediated NK suppression by recruiting SHP-1 and/or SHP-2. Subsequently, ITIM/SHP-1/SHP-2 and ITSM/SHP-1 downregulated the downstream Vav1/ERK1/2/p90RSK/NF-κB signaling. KIR2DL5+ immune cells infiltrated in various types of PVR+ human cancers. Markedly, the KIR2DL5 blockade reduced tumor growth and improved overall survival across multiple NK cell-based humanized tumor models. Thus, our results revealed functional mechanisms of KIR2DL5-mediated NK cell immune evasion, demonstrated blockade of the KIR2DL5/PVR axis as a therapy for human cancers, and provided an underlying mechanism for the clinical failure of anti-TIGIT therapies.


Assuntos
Células Matadoras Naturais , Neoplasias , Humanos , Transdução de Sinais , Fosforilação , Neoplasias/terapia , Neoplasias/metabolismo
2.
Adv Ther ; 39(6): 2946-2960, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35482249

RESUMO

INTRODUCTION: C8orf76 is a nuclear protein-encoding gene highly expressed in gastric tumor tissues that contributes to poor overall survival (OS). However, its function in breast cancer is unknown. We used The Cancer Genome Atlas (TCGA) database to investigate the role of C8orf76 in breast cancer. METHODS: We used logistic regression to estimate the relationship between C8orf76 and clinical-pathological features, followed by Kaplan-Meier and Cox regression analyses to assess the prognostic value of the gene regarding survival of patients with breast cancer from TCGA database. To explore the biological functions of C8orf76, gene ontology (GO), Kyoto Genome Encyclopedia (KEGG) enrichment analysis, and gene set enrichment analysis (GSEA) were used. We established a nomogram for high or low C8orf76 expression in breast cancer patients based on Cox multivariate analysis. Finally, we verified the proliferation-promoting effect of C8orf76 on breast cancer cells with CCK8 kit and further explored its role of apoptosis by flow cytometry. RESULTS: High C8orf76 expression was observed in breast cancer tissues and significantly correlated with clinical stage. Kaplan-Meier survival analysis showed that elevated C8orf76 expression was associated with poor OS. Among the highly expressed C8orf76 phenotypes, GO and KEGG analysis showed significant enrichment in aromatase activity and PPAR signaling pathway, whereas GSEA showed differential enrichment in receptor tyrosine signaling, PI3K/AKT signaling pathway, and some data sets in extracellular matrix and adhesion. Lower expression of C8orf76 leads to reduced proliferation, increased apoptosis, and downregulation of pAKT and Bcl-2 in breast cancer cells. CONCLUSIONS: High C8orf76 expression may be a valuable biomarker related to the diagnosis and prognosis of patients with breast cancer.


Assuntos
Neoplasias da Mama , Neoplasias da Mama/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Fosfatidilinositol 3-Quinases/genética , Prognóstico
3.
J Cancer ; 11(12): 3645-3654, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32284761

RESUMO

Breast cancer is the most prevalent malignant neoplasm among women worldwide. Despite continuous improvement of breast cancer individualized comprehensive therapy, local recurrence and distant metastasis still remain the challenges due to the development of acquired drug-resistance. Long non-coding RNAs (LncRNAs) is known to participated in the development of breast cancer. However, the mechanisms of LncRNAs involving in drug-resistance of breast cancer during chemotherapy remain to be further elucidated. Aiming to screen for candidate LncRNAs responsible for breast cancer mechanism, we first investigated the expression patterns of LncRNAs and mRNAs in paired breast cancer tissues and normal tissues using Agilent Human lncRNA array. The microarray results clearly demonstrated multiple differentially expressed mRNAs and LncRNAs including LncRNA NONHSAT141924. The remarkable up-regulation of LncRNA NONHSAT141924 in breast cancer MCF-7 was further confirmed by quantitative real-time PCR. GO and KEGG pathway analysis demonstrated that LncRNA NONHSAT141924 was most closely associated with paclitaxel (PTX)-resistant phenotype. To further explore the mechanism by which LncRNA NONHSAT141924 contributes to PTX-resistant characteristics, LncRNA NONHSAT141924 was transfected into MCF-7 breast cancer cell line. Overexpression of LncRNA NONHSAT141924 significantly reduced MCF-7 cell survivability through modulation of p-CREB/Bcl-2 apoptosis signaling pathway, one of the major pathways participated in LncRNAs-mediated chemotherapy resistance. Taken together, our study provides a new LncRNA-mediated regulatory mechanism for PTX-resistance of breast cancer and suggests that therapeutic inhibition of LncRNA NONHSAT141924 might be an efficient strategy for PTX-resistant breast cancer treatment.

4.
J Cancer ; 10(25): 6341-6348, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31772666

RESUMO

Retinoic acid-induced 14 (RAI14) is involved in the development of different tumor types, however, its expression and biological function in breast cancer are yet unknown. In the current study, we demonstrated that RAI14 was highly expressed in breast cancer. The high expression of RAI14 is positively correlated with the malignant progression of breast cancer and suggests a worse prognosis. Further, we found that knockdown RAI14 inhibits the proliferation, migration and invasion of breast cancer cells by regulating cell cycle and EMT through Akt/Cyclin D1, MMP2, MMP9 and ZEB1/E-cadhrin/Vimentin pathway. These findings revealed a novel function for RAI14 in breast cancer progression and suggest that RAI14 may become a promising diagnostic and therapeutic target for breast cancer.

5.
Front Oncol ; 9: 1403, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31921660

RESUMO

Methyltransferase-like 3 (METTL3), a predominantly catalytic enzyme in the N6-methyladenosine (m6A) methyltransferase system, is dysregulated and plays a dual role (oncogene or tumor suppressor) in different human cancers. The expression and pro- or anticancer role of METTL3 in different cancers remain controversial. METTL3 is implicated in many aspects of tumor progression, including tumorigenesis, proliferation, invasion, migration, cell cycle, differentiation, and viability. Most underlying mechanisms involve multiple signaling pathways that rely on m6A-dependent modification. However, METTL3 can also modulate the cancer process by directly promoting the translation of oncogenes via interaction with the translation initiation machinery through recruitment of eukaryotic translation initiation factor 3 subunit h (eIF3h). In this review, we summarized the current evidence on METTL3 in diverse human malignancies and its potential as a prognostic/ therapeutic target.

6.
Oncol Rep ; 34(3): 1487-93, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26134602

RESUMO

Heterologous expression of the Drosophila melanogaster multi-substrate deoxyribonucleoside kinase (Dm-dNK) increases the sensitivity of cancer cells to several cytotoxic nucleoside analogs. Thus, it may be used as a suicide gene in combined gene/chemotherapy treatment of cancer. To further characterize this potential suicide gene, we constructed two retroviral vectors that enabled the expression of Dm-dNK in cancer cells. One vector harbored the wild­type enzyme that localized to the nucleus. The other vector harbored a mitochondrial localized mutant enzyme that was constructed by deleting the nuclear localization signal and fusing it to a mitochondrial import signal of cytochrome c oxidase. A thymidine kinase-deficient osteosarcoma cell line was transduced with the recombinant viruses. The sensitivity and bystander cell killing in the presence of pyrimidine nucleoside analogs (E)-5-(2-bromovinyl)­2'­deoxyuridine and 1-ß-D-arabinofuranosylthymine were investigated. Tanshinone IIA is a constituent of Danshen; a traditional Chinese medicine used in the treatment of cardiovascular diseases. This study also looked at the influence of Tanshinone IIA on the bystander effect and the underlying mechanisms. We showed that sensitivity of the osteosarcoma cell line to the nucleoside analogs and the efficiency of bystander cell killing were independent of the subcellular localization of Dm-dNK. The enhanced effect of tanshinone IIA on the bystander effect was related to the increased expression of Cx43 and Cx26.


Assuntos
Abietanos/administração & dosagem , Terapia Genética , Osteossarcoma/tratamento farmacológico , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Animais , Efeito Espectador , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/genética , Conexina 26 , Conexinas , Desoxiuridina/administração & dosagem , Desoxiuridina/análogos & derivados , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Galactose/administração & dosagem , Galactose/análogos & derivados , Vetores Genéticos , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Sinais de Localização Nuclear/genética , Osteossarcoma/genética , Osteossarcoma/patologia , Fosfotransferases (Aceptor do Grupo Álcool)/biossíntese , Retroviridae/genética , Timina/administração & dosagem , Timina/análogos & derivados
7.
Surgery ; 158(1): 211-24, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25979438

RESUMO

BACKGROUND: The Hedgehog signaling pathway and its key target effector Gli1 are linked closely to the development of the epithelial to mesenchymal transition (EMT) in many cancers. The definite function of Gli1 in regulating the EMT of pancreatic cancer (PC), however, is still unclear. METHODS: At the cell and tissue levels, we investigated the role of Gli1 in the initiation of EMT in PC with and without external stimulus treatments. RESULTS: The immunohistochemistry results showed that Gli1 was associated positively with MMP9 but not with E-cad or Vimentin. Gli1 expression was associated positively with tumor T (P = .025) and Union for International Cancer Control stage (P = .032), whereas MMP9 expression was associated positively with lymph node metastasis (P = .017) and Union for International Cancer Control stage (P = .006). Furthermore, patients with Gli1 and MMP9 coexpression had poor overall survival (P = .015). Silencing of Gli1 alone without external stimulus had no effect on EMT but inhibited transforming growth factor-beta1 (TGFß1)- and epidermal growth factor (EGF)-induced EMT in PANC-1, AsPC-1, and BxPC-3 PC cell lines, along with the inhibition of TGFß1- and EGF-induced EMT-like cell morphology and invasion, down-regulation of E-cad, and up-regulation of MMP9 and Vimentin in those 3 cell lines, respectively. CONCLUSION: Gli1 silencing alone has no effect on EMT initiation; however, it exerts a protumor role in the aggressive invasion of PC cells by promoting TGFß1- and EGF-induced EMT.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Neoplasias Pancreáticas/metabolismo , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Caderinas/biossíntese , Linhagem Celular Tumoral , Inativação Gênica , Humanos , Metaloproteinase 9 da Matriz/biossíntese , Invasividade Neoplásica , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/patologia , Análise de Sobrevida , Fatores de Transcrição/genética , Células Tumorais Cultivadas , Vimentina/biossíntese , Proteína GLI1 em Dedos de Zinco
8.
J Exp Clin Cancer Res ; 33: 47, 2014 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-24886365

RESUMO

INTRODUCTION: Epidermal growth factor receptor (EGFR) overexpression has been associated with prognostic and predictive value in inflammatory breast cancer (IBC). Epidermal growth factor receptor 2 (HER2) overexpression is observed at a higher rate in IBC compared with noninflammatory breast cancer. Current clinically available anti-HER2 therapies are effective only in patients with HER2 amplified breast cancer, including IBC. AZD8931 is a novel small-molecule equipotent inhibitor of EGFR, HER2, and HER3 signaling. In this study, we investigated the antitumor activity of AZD8931 alone or in combination with paclitaxel using preclinical models of EGFR-overexpressed and HER2 non-amplified IBC cells. METHODS: Two IBC cell lines SUM149 and FC-IBC-02 derived from pleural effusion of an IBC patient were used in this study. Cell growth and apoptotic cell death were examined in vitro. For the in vivo tumor growth studies, IBC cells were orthotopically transplanted into the mammary fat pads of immunodeficient mice. AZD8931 was given by daily oral gavage at doses of 25 mg/kg, 5 days/week for 4 weeks. Paclitaxel was subcutaneously injected twice weekly. RESULTS: AZD8931 significantly suppressed cell growth of IBC cells and induced apoptosis of human IBC cells in vitro. Significantly, we showed that AZD8931 monotherapy inhibited xenograft growth and the combination of paclitaxel + AZD8931 was demonstrably more effective than paclitaxel or AZD8931 alone treatment at delaying tumor growth in vivo in orthotopic IBC models. CONCLUSION: AZD8931 single agent and in combination with paclitaxel demonstrated signal inhibition and antitumor activity in EGFR-overexpressed and HER2 non-amplified IBC models. These results suggest that AZD8931 may provide a novel therapeutic strategy for the treatment of IBC patients with HER2 non-amplified tumors.


Assuntos
Antineoplásicos/farmacologia , Receptores ErbB/metabolismo , Neoplasias Inflamatórias Mamárias/tratamento farmacológico , Quinazolinas/farmacologia , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Receptores ErbB/antagonistas & inibidores , Feminino , Amplificação de Genes , Humanos , Neoplasias Inflamatórias Mamárias/metabolismo , Camundongos , Camundongos SCID , Quinazolinas/uso terapêutico , Receptor ErbB-2/genética , Transdução de Sinais , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Cancer Lett ; 328(1): 95-103, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23000515

RESUMO

Currently, no curative treatments are available for late-stage metastatic or recurrent breast cancer, because the cancer tolerates both chemotherapy and endocrine therapy. In this study, we investigated the feasibility of a dual-regulated oncolytic adenoviral vector with a novel suicide gene to treat breast cancer. Following targeted gene virotherapy of conditionally replicating adenoviruses (CRAds), the novel suicide gene of multisubstrate deoxyribonucleoside kinase of Drosophila melanogaster (Dm-DNK) was inserted into the double-regulated oncolytic adenovirus SG500 to ensure more safety and enhanced antitumor activity against breast cancer both in vitro and in vivo. Selective replication, cell-killing efficacy, and cytotoxicity, combined with chemotherapeutics were investigated in several breast cell lines (MDA-MB-231 and MCF-7), normal cells (WI-38 and MRC-5), and human (MDA-MB-231) tumor models in vivo. The double-regulated SG500-dNK had high cell-killing activity in breast cancer. Replication was similar to wild-type in breast cells and was attenuated in normal cells. SG500-dNK combined with the chemotherapeutics (E)-5-(2-bromovinyl)-2'-deoxyuridine (Bvdu) and 2',2'-difluoro-deoxycytidine (dFdC) resulted in synergistically enhanced cell killing and greatly improved antitumor efficacy in vitro or in breast xenografts in vivo. These data suggest that the novel oncolytic variant SG500-dNK is a promising candidate for targeting breast tumors specifically when combined with chemotherapeutics.


Assuntos
Adenoviridae/genética , Neoplasias da Mama/terapia , Terapia de Alvo Molecular , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Animais , Linhagem Celular Tumoral , Proteínas de Drosophila , Feminino , Humanos , Camundongos , Camundongos Nus , Fosfotransferases (Aceptor do Grupo Álcool) , Regiões Promotoras Genéticas , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Oncol Rep ; 29(3): 960-6, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23254508

RESUMO

Gene transfer using different viral vectors has demonstrated different antitumor effects in suicide gene therapy. In the present study, in order to optimize the efficacy of replication-defective adenoviral and lentiviral vectors for gene therapy, RT-PCR was used to evaluate the expression of Drosophila melanogaster deoxyribonucleoside kinase (Dm-dNK) in the Bcap37 human breast cancer cell line, dThd was used to determine the activity of Dm-dNK, cell cytotoxicity was evaluated by MTT assay and cell proliferation was assessed using a hemocytometer. Moreover, apoptosis induction was evaluated by the Annexin V-FITC-labeled FACS method. Furthermore, BALB/C nude mice bearing tumors were treated with Dm-dNK mediated with the pyrimidine nucleoside analog, brivudine [BVDU, (E)-5-(2-bromovinyl)-2'-deoxyuridine]. Our results indicated that the gene expression of Dm-dNK transfected by adenoviral and lentiviral vectors may be detected and that its long-term activity may be retained. Both vectors containing the Dm-dNK gene revealed high cytotoxicity and sensitized cell apoptosis from the BVDU prodrug. In tumor models, lentivirus-mediated gene therapy significantly inhibited the growth of tumors compared with adenovirus-mediated gene therapy. Although adenovirus- and lentivirus-transduced Dm-dNK reveal strong treatment efficacy in vitro, the latter has great potential due to the long-term expression of the therapeutic gene in vivo.


Assuntos
Adenoviridae/genética , Neoplasias da Mama/terapia , Proteínas de Drosophila/biossíntese , Lentivirus/genética , Fosfotransferases (Aceptor do Grupo Álcool)/biossíntese , Animais , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Apoptose , Neoplasias da Mama/patologia , Bromodesoxiuridina/análogos & derivados , Bromodesoxiuridina/metabolismo , Bromodesoxiuridina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada , Proteínas de Drosophila/genética , Feminino , Expressão Gênica , Terapia Genética , Vetores Genéticos , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Pró-Fármacos/metabolismo , Pró-Fármacos/farmacologia , Proibitinas , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Chin J Cancer Res ; 24(4): 388-98, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23359451

RESUMO

Metastatic breast cancer (MBC) is characterized by a combination of tumor growth, proliferation and metastatic progression and is typically managed with palliative intent. The benefit of standard systemic therapies is relatively limited and the disease is considered incurable suggesting the need to investigate the biological drivers of the various phases of the metastatic process in order to improve the selection of molecularly driven therapies. The detection, enumeration and molecular analysis of circulating tumor cells (CTCs) provide an intriguing opportunity to advance this knowledge. CTCs enumerated by the Food and Drugs Administration-cleared CellSearch(®) system are an independent prognostic factor of progression-free survival (PFS) and overall survival (OS) in MBC patients. Several published papers demonstrated the poor prognosis for MBC patients that presented basal CTC count ≥5 in 7.5 mL of blood. Therefore, the enumeration of CTCs during treatment for MBC provides a tool with the ability to predict progression of disease earlier than standard timing of anatomical assessment using conventional radiological tests. During the metastatic process cancer cells exhibit morphological and phenotypic plasticity undergoing epithelial-mesenchymal transition (EMT). This important phenomenon is associated with down regulation of epithelial marker (e.g., EpCAM) with potential limitations in the applicability of current CTCs enrichment methods. Such observations translated in a number of investigations aimed at improving our capabilities to enumerate and perform molecular characterization of CTCs. Theoretically, the phenotypic analysis of CTCs can represent a "liquid" biopsy of breast tumor that is able to identify a new potential target against the metastatic disease and advanced the development and monitoring of personalized therapies.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...