Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Cell Mol Med ; 27(11): 1509-1522, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37082943

RESUMO

Human oral squamous cell carcinoma (OSCC) has been associated with a relatively low survival rate over the years and is characterized by a poor prognosis. C-X3-C motif chemokine ligand 1 (CX3CL1) has been involved in advanced migratory cells. Overexpressed CX3CL1 promotes several cellular responses related to cancer metastasis, including cell movement, migration and invasion in tumour cells. However, CX3CL1 controls the migration ability, and its molecular mechanism in OSCC remains unknown. The present study confirmed that CX3CL1 increased cell movement, migration and invasion. The CX3CL1-induced cell motility is upregulated through intercellular adhesion molecule-1 (ICAM-1) expression in OSCC cells. These effects were significantly suppressed when OSCC cells were pre-treated with CX3CR1 monoclonal antibody (mAb) and small-interfering RNA (siRNA). The CX3CL1-CX3CR1 axis activates promoted PLCß/PKCα/c-Src phosphorylation. Furthermore, CX3CL1 enhanced activator protein-1 (AP-1) activity. The CX3CR1 mAb and PLCß, PKCα, c-Src inhibitors reduced CX3CL1-induced c-Jun phosphorylation, c-Jun translocation into the nucleus and c-Jun binding to the ICAM-1 promoter. The present results reveal that CX3CL1 induces the migration of OSCC cells by promoting ICAM-1 expression through the CX3CR1 and the PLCß/PKCα/c-Src signal pathway, suggesting that CX3CL1-CX3CR1-mediated signalling is correlated with tumour motility and appealed to be a precursor for prognosis in human OSCC.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , Humanos , Carcinoma de Células Escamosas/patologia , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Proteína Quinase C-alfa , Carcinoma de Células Escamosas de Cabeça e Pescoço , Neoplasias Bucais/patologia , Movimento Celular , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Linhagem Celular Tumoral , Quimiocina CX3CL1/genética , Quimiocina CX3CL1/metabolismo
2.
J Biomed Sci ; 29(1): 92, 2022 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-36336681

RESUMO

BACKGROUND: Phosphatase and tensin homolog (PTEN) is a tumor suppressor. Low PTEN expression has been observed in pancreatic neuroendocrine tumors (pNETs) and is associated with increased liver metastasis and poor survival. Vascular endothelial growth factor receptor 3 (VEGFR3) is a receptor tyrosine kinase and is usually activated by binding with vascular endothelial growth factor C (VEGFC). VEGFR3 has been demonstrated with lymphangiogenesis and cancer invasiveness. PTEN is also a phosphatase to dephosphorylate both lipid and protein substrates and VEGFR3 is hypothesized to be a substrate of PTEN. Dual-specificity phosphatase 19 (DUSP19) is an atypical DUSP and can interact with VEGFR3. In this study, we investigated the function of PTEN on regulation of pNET invasiveness and its association with VEGFR3 and DUSP19. METHODS: PTEN was knocked down or overexpressed in pNET cells to evaluate its effect on invasiveness and its association with VEGFR3 phosphorylation. In vitro phosphatase assay was performed to identify the regulatory molecule on the regulation of VEGFR3 phosphorylation. In addition, immunoprecipitation, and immunofluorescence staining were performed to evaluate the molecule with direct interaction on VEGFR3 phosphorylation. The animal study was performed to validate the results of the in vitro study. RESULTS: The invasion and migration capabilities of pNETs were enhanced by PTEN knockdown accompanied with increased VEGFR3 phosphorylation, ERK phosphorylation, and increased expression of epithelial-mesenchymal transition molecules in the cells. The enhanced invasion and migration abilities of pNET cells with PTEN knockdown were suppressed by addition of the VEGFR3 inhibitor MAZ51, but not by the VEGFR3-Fc chimeric protein to neutralize VEGFC. VEGFR3 phosphorylation is responsible for pNET cell invasiveness and is VEGFC-independent. However, an in vitro phosphatase assay failed to show VEGFR3 as a substrate of PTEN. In contrast, DUSP19 was transcriptionally upregulated by PTEN and was shown to dephosphorylate VEGFR3 via direct interaction with VEGFR3 by an in vitro phosphatase assay, immunoprecipitation, and immunofluorescence staining. Increased tumor invasion into peripheral tissues was validated in xenograft mouse model. Tumor invasion was suppressed by treatment with VEGFR3 or MEK inhibitors. CONCLUSIONS: PTEN regulates pNET invasiveness via DUSP19-mediated VEGFR3 dephosphorylation. VEGFR3 and DUSP19 are potential therapeutic targets for pNET treatment.


Assuntos
Tumores Neuroectodérmicos Primitivos , Tumores Neuroendócrinos , Neoplasias Pancreáticas , Humanos , Camundongos , Animais , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo , Tumores Neuroendócrinos/genética , Fator A de Crescimento do Endotélio Vascular , PTEN Fosfo-Hidrolase/genética , Neoplasias Pancreáticas/genética , Invasividade Neoplásica/genética , Linhagem Celular Tumoral , Fosfatases de Especificidade Dupla
3.
J Cell Mol Med ; 25(19): 9128-9140, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34427969

RESUMO

The CXC chemokine ligand-13 (CXCL13) is a chemoattractant of B cells and has been implicated in the progression of many cancers. So far, CXCL13 and its related receptor CXCR5 have been proved to regulate cancer cell migration as well as tumour metastasis. However, the role of CXCL13-CXCR5 axis in metastasis of lung cancer is still poorly understood. In this study, we found that CXCL13 and CXCR5 were commonly up-regulated in lung cancer specimens compared with normal tissues among different cohorts. Our evidence showed that CXCL13 obviously promoted migration of lung cancer cells, and this effect was mediated by vascular cell adhesion molecule-1 (VCAM-1) expression. We also confirmed that CXCR5, the major receptor responsible for CXCL13 function, was required for CXCL13-promoted cell migration. We also test the candidate components which are activated after CXCL13 treatment and found that phospholipase C-ß (PLCß), protein kinase C-α (PKCα) and c-Src signalling pathways were involved in CXCL13-promoted cell migration and VCAM-1 expression in lung cancer cells. Finally, CXCL13 stimulated NF-κB transcription factor in lung cancer cells, contributing to VCAM-1 expression in translational level. These evidences propose a novel insight into lung cancer metastasis which is regulated by CXCL13.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Quimiocina CXCL13/metabolismo , Neoplasias Pulmonares/metabolismo , Receptores CXCR5/metabolismo , Transdução de Sinais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Quimiocina CXCL13/genética , Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Metanálise como Assunto , NF-kappa B/metabolismo , Metástase Neoplásica , Ligação Proteica , Proteína Quinase C-alfa/metabolismo , Receptores CXCR5/genética , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo , Quinases da Família src/metabolismo
4.
J Cell Physiol ; 236(3): 2205-2213, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32808296

RESUMO

Osteoarthritis (OA) is a progressive degenerative joint disorder characterized by synovial inflammation. Interleukin-6 (IL-6) is a key proinflammatory cytokine in OA progression. Particulate matter 2.5 (PM2.5) exposure increases the risk of different diseases, including OA. Up until now, no studies have described any association between PM2.5 and IL-6 expression in human OA synovial fibroblasts (OASFs). Here, our data show that PM2.5 concentration- and time-dependently promoted IL-6 synthesis in human OASFs. We also found that reactive oxygen species (ROS) generation potentiated the effects of PM2.5 on IL-6 production. ASK1, ERK, p38, and JNK inhibitors reduced PM2.5-induced increases of IL-6 expression. Treatment of OASFs with PM2.5 promoted phosphorylation of these signaling cascades. We also found that PM2.5 enhanced c-Jun phosphorylation and its translocation into the nucleus. Thus, PM2.5 increases IL-6 production in human OASFs via the ROS, ASK1, ERK, p38, JNK, and AP-1 signaling pathways. Our evidence links PM2.5 with OA progression.


Assuntos
Fibroblastos/patologia , Interleucina-6/biossíntese , MAP Quinase Quinase Quinase 5/metabolismo , Osteoartrite/enzimologia , Osteoartrite/patologia , Material Particulado/toxicidade , Membrana Sinovial/patologia , Ativação Enzimática/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Modelos Biológicos , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição AP-1/metabolismo , Regulação para Cima/efeitos dos fármacos
5.
Cancer Sci ; 112(1): 243-253, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33128283

RESUMO

Pancreatic neuroendocrine tumor (pNET) is a pancreatic neoplasm with neuroendocrine differentiation. pNET in early stage can be treated with surgical resection with long-term survival, whereas the prognosis of pNET with locoregional or distant metastasis is relatively poor. Lymphangiogenesis is essential for tumor metastasis via the lymphatic system and may overhead distant metastasis. c-Myc overexpression is involved in tumorigenesis. The role of c-Myc in lymphangiogenesis is unclear. In this study, we evaluated the mechanism and effect of c-Myc on lymphangiogenesis of pNET via interaction of lymphatic endothelial cells (LECs) and pNET cells. Lymph node metastasis was evaluated in pNET xenograft mice. Potential target agents to inhibit lymph node metastasis were evaluated in an animal model. We found that vascular endothelial growth factor C (VEGFC) expression and secretion was increased in pNET cell lines with c-Myc overexpression. c-Myc transcriptionally upregulates VEGFC expression and the secretion of pNET cells by directly binding to the E-box of the VEGFC promoter and enhances VEGF receptor 3 phosphorylation and the tube formation of LECs. c-Myc overexpression is associated with lymph node metastasis in pNET xenograft mice. Combinational treatment with an mTOR inhibitor and c-Myc inhibitor or VEGFC-neutralizing chimera protein reduced lymph node metastasis in the mice with c-Myc overexpression. The mTOR inhibitor acts on lymphangiogenesis by reducing VEGFC expression in pNET cells and inhibiting the tube formation of LECs. In conclusion, mTOR and c-Myc are important for lymphangiogenesis of pNET and are potential therapeutic targets for prevention and treatment of lymph node metastasis in pNET.


Assuntos
Metástase Linfática/patologia , Tumores Neuroendócrinos/patologia , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fator C de Crescimento do Endotélio Vascular/biossíntese , Animais , Linhagem Celular Tumoral , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Xenoenxertos , Humanos , Linfangiogênese/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Regulação para Cima
6.
J Cell Mol Med ; 24(21): 12826-12839, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33021341

RESUMO

Osteosarcoma is an extremely common primary bone malignancy that is highly metastatic, with most deaths resulting from pulmonary metastases. The extracellular matrix protein thrombospondin-2 (TSP-2) is key to many biological processes, such as inflammation, wound repair and tissue remodelling. However, it is unclear as to what biological role TSP-2 plays in human metastatic osteosarcoma. The immunochemistry analysis from osteosarcoma specimens identified marked up-regulation of TSP-2 in late-stage osteosarcoma. Furthermore, we found that TSP-2 increased the levels of matrix metallopeptidase 9 (MMP-9) expression and thereby increased the migratory potential of human osteosarcoma cells. Osteosarcoma cells pre-treated with an MMP-9 monoclonal antibody (mAb), an MMP-9 inhibitor, or transfected with MMP-9 small interfering RNA (siRNA) reduced the capacity of TSP-2 to potentiate cell migration. TSP-2 treatment activated the PLCß, PKCα, c-Src and nuclear kappa factor B (NF-κB) signalling pathways, while the specific siRNA, inhibitors and mutants of these cascades reduced TSP-2-induced stimulation of migration activity. Knockdown of TSP-2 expression markedly reduced cell metastasis in cellular and animal experiments. It appears that an interaction between TSP-2 and integrin αvß3 activates the PLCß, PKCα and c-Src signalling pathways and subsequently activates NF-κB signalling, increasing MMP-9 expression and stimulating migratory activity amongst human osteosarcoma cells.


Assuntos
Metaloproteinase 9 da Matriz/metabolismo , NF-kappa B/metabolismo , Osteossarcoma/patologia , Proteína Quinase C/metabolismo , Trombospondinas/metabolismo , Fosfolipases Tipo C/metabolismo , Quinases da Família src/metabolismo , Animais , Linhagem Celular , Movimento Celular , Técnicas de Silenciamento de Genes , Humanos , Integrina alfaVbeta3/metabolismo , Camundongos , Metástase Neoplásica , Transdução de Sinais
7.
Int J Mol Sci ; 21(17)2020 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-32847038

RESUMO

Osteosarcoma is the most common primary tumor of the skeletal system and is well-known to have an aggressive clinical outcome and high metastatic potential. The chemokine (C-X-C motif) ligand 13 (CXCL13) plays a vital role in the development of several cancers. However, the effect of CXCL13 in the motility of osteosarcoma cells remains uncertain. Here, we found that CXCL13 increases the migration and invasion potential of three osteosarcoma cell lines. In addition, CXCL13 expression was upregulated in migration-prone MG-63 cells. Vascular cell adhesion molecule 1 (VCAM-1) siRNA and antibody demonstrated that CXCL13 promotes migration via increasing VCAM-1 production. We also show that CXCR5 receptor controls CXCL13-mediated VCAM-1 expression and cell migration. Our study identified that CXCL13/CXCR5 axis facilitate VCAM-1 production and cell migration in human osteosarcoma via the phospholipase C beta (PLCß), protein kinase C α (PKCα), c-Src, and nuclear factor-κB (NF-κB) signaling pathways. CXCL13 and CXCR5 appear to be a novel therapeutic target in metastatic osteosarcoma.


Assuntos
Neoplasias Ósseas/patologia , Movimento Celular/genética , Quimiocina CXCL13/metabolismo , Osteossarcoma/patologia , Receptores CXCR5/metabolismo , Molécula 1 de Adesão de Célula Vascular/genética , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Quimiocina CXCL13/fisiologia , Humanos , Invasividade Neoplásica/genética , Osteossarcoma/genética , Osteossarcoma/metabolismo , Ligação Proteica , Receptores CXCR5/fisiologia , Transdução de Sinais/fisiologia , Células Tumorais Cultivadas , Molécula 1 de Adesão de Célula Vascular/metabolismo
8.
J Cell Physiol ; 234(8): 13157-13168, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30549031

RESUMO

Osteosarcoma is a malignant primary bone tumor that responds poorly to both chemotherapy and radiation therapy. However, because of side effects and drug resistance in chemotherapy and the insufficiency of an effective adjuvant therapy for osteosarcoma, it is necessary to research novel treatments. This study was the first to investigate the anticancer effects of the flavonoid derivative artocarpin in osteosarcoma. Artocarpin induced cell apoptosis in three human osteosarcoma cell lines-U2OS, MG63, and HOS. Artocarpin was also associated with increased intracellular reactive oxygen species (ROS). Mitochondrial dysfunction was followed by the release of cytochrome c from mitochondria and accompanied by decreased antiapoptotic Bcl-2 and Bcl-xL and increased proapoptotic protein Bak and Bax. Artocarpin triggered endoplasmic reticulum (ER) stress, as indicated by changes in cytosol calcium levels and increased glucose-regulated protein 78 and 94 expressions, and also increased calpains expression and activity. Animal studies revealed a dramatic 40% reduction in tumor volume after 18 days of treatment. This study demonstrated a novel anticancer activity of artocarpin against human osteosarcoma cells and in murine tumor models. In summary, artocarpin significantly induced cell apoptosis through ROS, ER stress, mitochondria, and the caspase pathway, and may thus be a novel anticancer treatment for osteosarcoma.


Assuntos
Neoplasias Ósseas/patologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Lectinas de Ligação a Manose/farmacologia , Osteossarcoma/patologia , Lectinas de Plantas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Apoptose/efeitos dos fármacos , Neoplasias Ósseas/metabolismo , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos SCID , Osteossarcoma/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Exp Cell Res ; 363(1): 65-72, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29305962

RESUMO

Triple-negative breast cancer (TNBC) is a subtype of cancer with aggressive behaviors (high recurrence and metastasis rate) and poor prognosis. Therefore, studying the determining factors that lead to malignant TNBCs is necessary to develop personalized therapy and improve survival rates. In this study, we first analyzed levels of chromodomain helicase DNA binding protein 4 (CHD4) in 60 TNBC patients by immunohistochemical staining. We then clarified the role of CHD4 in TNBC and non-TNBC cell lines. Our clinical data indicated that higher CHD4 expression is positively correlated with metastatic stage, tumor recurrence, and survival status. Consistent with the clinical analytical data, our in vitro data also indicated that high level of CHD4 is positively correlated with malignant behaviors in TNBC cells, such as cell motility and mortality. For further analyses, we found that E-cadherin, N-cadherin and fibronetin are involved in CHD4-mediated epithelial-mesenchymal transition (EMT). Silencing of CHD4 also increased drug sensitivity to cisplatin and PARP1 inhibitor, especially in TNBC cells. Altogether, our findings showed that CHD4 is not only a potential prognostic biomarker for TNBC patient survival, but is also a powerful candidate in the development of new anti-cancer agents in TNBC.


Assuntos
Caderinas/metabolismo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Metástase Neoplásica , Neoplasias de Mama Triplo Negativas/tratamento farmacológico
10.
Exp Cell Res ; 359(2): 458-465, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28842166

RESUMO

The Mi-2/nucleosome remodeling and deacetylase (NuRD) complex play a role in silencing gene expression. CHD4, the core component of the NuRD complex, which cooperates with histone deacetylase in reducing tumor suppressor genes (TSGs). To dissect the mechanisms underlying cancer promotion, we clarify the role of CHD4 in cyclin-dependent kinase inhibitor protein p21. Here, our data indicates that CHD4 deficiency impairs the recruitments of HDAC1 to the p21 promoter. ~ 300bp proximal promoter region is responsible for CHD4-HDAC1 axis-mediated p21 transcriptional activity. For identifying the role of anti-cancer drug response, knockdown of p21 overcomes cisplatin and poly-(ADP-ribose) polymerase (PARP) inhibitor-mediated growth suppression in CHD4-depleted cells. Consistent with in vitro data, tissue of patients and bioinformatics approach also showed positive correlation between CHD4 and p21. Overall, our findings not only identify that CHD4 deficiency preferentially impairs cell survival via increasing the level of p21, but also establishes targeting CHD4 as a potential therapeutic implication in BRCA-proficient breast cancer treatment.


Assuntos
Autoantígenos/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Reparo do DNA , DNA/genética , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica , Histona Desacetilase 1/genética , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Antineoplásicos/farmacologia , Autoantígenos/metabolismo , Linhagem Celular Tumoral , Montagem e Desmontagem da Cromatina , Cisplatino/farmacologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , DNA/metabolismo , Quebras de DNA de Cadeia Dupla , Bases de Dados de Proteínas , Feminino , Histona Desacetilase 1/metabolismo , Humanos , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Regiões Promotoras Genéticas , Análise Serial de Proteínas , Transdução de Sinais
11.
Int J Biol Sci ; 20(3): 1093-1109, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38322119

RESUMO

Background: As lung cancer is the leading cause of cancer death worldwide, the development of new medicines is a crucial endeavor. Naringenin, a flavanone derivative, possesses anti-cancer and anti-inflammatory properties and has been reported to have cytotoxic effects on various cancer cells. The current study investigated the underlying molecular mechanism by which naringenin induces cell death in lung cancer. Methods: The expression of apoptosis, cell cycle arrest, and autophagy markers in H1299 and A459 lung cancer cells was evaluated using a terminal deoxynucleotidyl transferase dUTP nick end labeling assay (TUNEL), Western blot, Annexin V/PI stain, PI stain, acridine orange staining, and transmission electron microscopy (TEM). Using fluorescence microscopy, DALGreen was used to observe the degradation of p62, a GFP-LC3 plasmid was used to evaluate puncta formation, and a pcDNA3-GFP-LC3-RFP-LC3ΔG plasmid was used to evaluate autophagy flux. Furthermore, the anti-cancer effect of naringenin was evaluated in a subcutaneous H1299 cell xenograft model. Results: Naringenin treatment of lung cancer cells (H1299 and A459) reduced cell viability and induced cell cycle arrest. Pretreatment of cells with ROS scavengers (N-acetylcysteine or catalase) suppressed the naringenin-induced cleavage of apoptotic protein and restored cyclin-dependent kinase activity. Naringenin also triggered autophagy by mediating ROS generation, thereby activating AMP-activated protein kinase (AMPK) signaling. ROS inhibition not only inhibited naringenin-induced autophagic puncta formation but also decreased the ratio of microtubule-associated proteins 1A/1B light chain 3 II (LC3II)/LC3I and activity of the AMPK signaling pathway. Furthermore, naringenin suppressed tumor growth and promoted apoptosis in the xenograft mouse model. Conclusion: This study demonstrated the potent anti-cancer effects of naringenin on lung cancer cells, thereby providing valuable insights for developing small-molecule drugs that can induce cell cycle arrest, apoptosis, and autophagic cell death.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Flavanonas , Neoplasias Pulmonares , Humanos , Animais , Camundongos , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Apoptose , Neoplasias Pulmonares/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Linhagem Celular Tumoral , Pontos de Checagem da Fase G2 do Ciclo Celular , Autofagia , Flavanonas/farmacologia
12.
Anticancer Res ; 44(3): 1173-1182, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38423664

RESUMO

BACKGROUND/AIM: Oral squamous cell carcinoma (OSCC), a major malignancy in Taiwan, is an invasive epithelial neoplasm resulting in a low survival rate. Current treatments do not prevent OSCC progression, and antitumor therapies should be improved. Plumbagin, a natural compound extracted from Plumbago zeylanica L., appears to have antitumor effects in various tumors. The antitumor mechanism of plumbagin in OSCC is still unclear. This study investigated the molecular mechanism through which plumbagin induces apoptosis. MATERIALS AND METHODS: To investigate the antiproliferative and pro-apoptotic effects of Plumbagin on OSCC cells and explore its underlying mechanism, cell counting kit-8, cell cycle analysis, and annexin V/PI assay were conducted. The functions of plumbagin on endoplasmic reticulum (ER) stress, reactive oxygen species (ROS) production, and mitochondrial membrane potential (MMP) deficiency were analyzed using flow cytometric analysis. Plumbagin-induced apoptosis-associated proteins were detected using western blotting. RESULTS: Plumbagin induced apoptosis in OSCC cells by suppressing tumor cell proliferation through ROS production, ER stress, mitochondrial dysfunction, and caspases activation. CONCLUSION: Plumbagin is a promising antitumor candidate targeting human OSCC.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , Naftoquinonas , Humanos , Espécies Reativas de Oxigênio/metabolismo , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/patologia , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/patologia , Carcinoma de Células Escamosas de Cabeça e Pescoço , Linhagem Celular Tumoral , Apoptose , Proliferação de Células , Estresse do Retículo Endoplasmático
13.
Crit Care Med ; 41(1): 120-32, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23222262

RESUMO

OBJECTIVE: The molecular mechanisms underlying lung inflammation in toxic smoke inhalation injury are unknown. We investigated the signaling pathway responsible for the induction of interleukin 8 by wood smoke extract in lung epithelial cells and lung inflammation induced by wood smoke exposure in mice. DESIGN: A randomized, controlled study. SETTING: A research laboratory. INTERVENTIONS AND MAIN RESULTS: Exposure of primary human bronchial epithelial cells to wood smoke extract sequentially activated NADPH oxidase and increased intracellular reactive oxygen species level; activated AMP-activated protein kinase, extracellular signal-regulated kinase and Jun N-terminal kinase (two mitogen-activated protein kinases), and nuclear factor-κB and signal transducer and activator of transcription protein 3 (two transcription factors); and induced interleukin-8. Inhibition of NADPH oxidase activation with apocynin or siRNA targeting p47(phox ) (a subunit of NADPH oxidase) attenuated the increased intracellular reactive oxygen species level, AMP-activated protein kinase activation, and interleukin-8 induction. Removal of intracellular reactive oxygen species by N-acetyl-cysteine reduced the activation of AMP-activated protein kinase, extracellular signal-regulated kinase and Jun N-terminal kinase, and interleukin-8 induction. Prevention of AMP-activated protein kinase activation by Compound C or AMP-activated protein kinase siRNA lessened the activation of Jun N-terminal kinase, extracellular signal-regulated kinase, nuclear factor-κB, signal transducer and activator of transcription protein 3 and interleukin-8 induction. Inhibition of Jun N-terminal kinase and extracellular signal-regulated kinase activation by inhibitors reduced the activation of nuclear factor-κB and signal transducer and activator of transcription protein 3 and interleukin-8 induction. Abrogation of nuclear factor-κB and signal transducer and activator of transcription protein 3 activation by inhibitors attenuated the interleukin-8 induction. Additionally, acute exposure of mice to wood smoke promoted AMP-activated protein kinase phosphorylation and expression of macrophage inflammatory protein 2 (an interleukin-8 homolog) in lung epithelial cells and lungs and lung inflammation, all of which were reduced by Compound C treatment. CONCLUSIONS: Interleukin-8 induction by wood smoke extract in lung epithelial cells is mediated by novel NADPH oxidase-dependent, reactive oxygen species-sensitive AMP-activated protein kinase signaling with Jun N-terminal kinase and extracellular signal-regulated kinase as the downstream kinases and nuclear factor-κB and signal transducer and activator of transcription protein 3 as the downstream transcription factors. This AMP-activated protein kinase signaling is likely important for inducing lung inflammation with toxic smoke exposure in mice.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Interleucina-8/metabolismo , Sistema de Sinalização das MAP Quinases , Lesão por Inalação de Fumaça/enzimologia , Lesão por Inalação de Fumaça/imunologia , Animais , Células Cultivadas , Humanos , Inflamação/enzimologia , Inflamação/imunologia , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NADPH Oxidases/metabolismo , Distribuição Aleatória , Espécies Reativas de Oxigênio/metabolismo , Mucosa Respiratória/imunologia , Lesão por Inalação de Fumaça/patologia
14.
Cancer Med ; 12(2): 1588-1601, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35757841

RESUMO

BACKGROUND: Tumor cells may aberrantly express metabolic enzymes to adapt to their environment for survival and growth. Targeting cancer-specific metabolic enzymes is a potential therapeutic strategy. Phosphoenolpyruvate carboxykinase (PEPCK) catalyzes the conversion of oxaloacetate to phosphoenolpyruvate and links the tricarboxylic acid cycle and glycolysis/gluconeogenesis. Mitochondrial PEPCK (PEPCK-M), encoded by PCK2, is an isozyme of PEPCK and is distributed in mitochondria. Overexpression of PCK2 has been identified in many human cancers and demonstrated to be important for the survival program initiated upon metabolic stress in cancer cells. We evaluated the expression status of PEPCK-M and investigated the function of PEPCK-M in breast cancer. METHODS: We checked the expression status of PEPCK-M in breast cancer samples by immunohistochemical staining. We knocked down or overexpressed PCK2 in breast cancer cell lines to investigate the function of PEPCK-M in breast cancer. RESULTS: PEPCK-M was highly expressed in estrogen receptor-positive (ER+ ) breast cancers. Decreased cell proliferation and G0 /G1 arrest were induced in ER+ breast cancer cell lines by knockdown of PCK2. PEPCK-M promoted the activation of mTORC1 downstream signaling molecules and the E2F1 pathways in ER+ breast cancer. In addition, glucose uptake, intracellular glutamine levels, and mTORC1 pathways activation by glucose and glutamine in ER+ breast cancer were attenuated by PCK2 knockdown. CONCLUSION: PEPCK-M promotes proliferation and cell cycle progression in ER+ breast cancer via upregulation of the mTORC1 and E2F1 pathways. PCK2 also regulates nutrient status-dependent mTORC1 pathway activation in ER+ breast cancer. Further studies are warranted to understand whether PEPCK-M is a potential therapeutic target for ER+ breast cancer.


Assuntos
Neoplasias da Mama , Receptores de Estrogênio , Humanos , Feminino , Fosfoenolpiruvato/metabolismo , Receptores de Estrogênio/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Glutamina/metabolismo , Fosfoenolpiruvato Carboxiquinase (ATP)/genética , Fosfoenolpiruvato Carboxiquinase (ATP)/metabolismo , Mitocôndrias/genética , Mitocôndrias/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo
15.
Int J Biol Sci ; 19(5): 1455-1470, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37056937

RESUMO

Background: Lung cancer is a malignant tumor with metastatic potential. Chemokine ligand 14 (CXCL14) has been reported to be associated with different cancer cell migration and invasion. However, few studies have explored the function of CXCL14 and its specific receptor in lung cancer metastasis. This study aims to determine the mechanism of CXCL14-promoted cancer metastasis. Methods: The expression of CXCL14, atypical chemokine receptor 2 (ACKR2), and epithelial mesenchymal transition (EMT) markers was evaluated by the public database of The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), Western blot, enzyme-linked immunosorbent assay (ELISA), quantitative real-time polymerase chain reaction (qPCR), immunohistochemistry (IHC), and immunofluorescence (IF). Migration and wound healing assays were used to observe the motility of cancer cells. A luciferase reporter assay was performed to analyze transcription factor activity. The metastasis of lung cancer cells was evaluated in an orthotopic model. Results: We have presented that overexpression of CXCL14 and ACKR2 was observed in lung cancer datasets, human lung tumor sections, and lung cancer cells. Furthermore, the migration of CXCL14-promoted lung cancer cells was determined in vitro and in vivo. In particular, ACKR2 knockdown abolished CXCL14-induced cancer cell motility. Additionally, ACKR2 was involved in CXCL14-triggered phospholipase Cß3 (PLCß3), protein kinase Cα (PKCα), and proto-oncogene c-Src signaling pathway and subsequently upregulated nuclear factor κB (NF-κB) transcription activity leading to EMT and migration of lung cancer cells. These results indicated that the CXCL14/ACKR2 axis played an important role in lung cancer metastasis. Conclusion: This study is the first to reveal the function of CXCL14 in promoting EMT and metastasis in lung cancer. As a specific receptor for CXCL14 in lung cancer, ACKR2 mediates CXCL14-induced signaling that leads to cell motility. Our findings can be used as a prognostic biomarker of lung cancer metastasis.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Linhagem Celular Tumoral , Transdução de Sinais/genética , Receptores de Quimiocinas , Quimiocinas CXC/genética
16.
Biochem Pharmacol ; 218: 115853, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37832794

RESUMO

Osteosarcoma is a malignant tumor with high metastatic potential, such that the overall 5-year survival rate of patients with metastatic osteosarcoma is only 20%. Therefore, it is necessary to unravel the mechanisms of osteosarcoma metastasis to identify predictors of metastasis by which to develop new therapies. Fibroblast growth factor 2 (FGF2) is a growth factor involved in embryonic development, cell migration, and proliferation. The overexpression of FGF2 and FGF receptors (FGFRs) has been shown to enhance cancer cell proliferation in lung, breast, gastric, and prostate cancers as well as melanoma. Nonetheless, the roles of FGF2 and FGFRs in human osteosarcoma cells remain unknown. In the present study, we found that FGF2 was overexpressed in human osteosarcoma sections and correlated with lung metastasis. Treatment of FGF2 induced migration activity, invasion activity, and intercellular adhesion molecule (ICAM)-1 expression in osteosarcoma cells. In particular, the downregulation or antagonism of FGFR1-4 suppressed FGF2-induced ICAM-1 expression and cancer cell migration. Furthermore, FGFR1, FGFR2, FGFR3, and FGFR4 were involved in FGF2-induced the phospholipase Cß/protein kinase Cα/proto-oncogene c-Src signaling pathway and triggered c-Jun nuclear translocation. Subsequent c-Jun upregulation of activator protein-1 transcription activity on the ICAM-1 promoter led to an increased migration of osteosarcoma cells. Moreover, the knockdown of endogenous FGF2 suppressed ICAM-1 expression and migration of osteosarcoma cells. These findings suggest that FGF2/FGFR1-4 signaling promotes metastasis via its direct downstream target gene ICAM-1, revealing a novel potential therapeutic target for osteosarcoma.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Humanos , Masculino , Neoplasias Ósseas/genética , Fator 2 de Crescimento de Fibroblastos/genética , Molécula 1 de Adesão Intercelular , Osteossarcoma/genética , Osteossarcoma/patologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Transdução de Sinais
17.
Aging (Albany NY) ; 15(24): 14900-14914, 2023 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-38126996

RESUMO

Despite advances in therapeutic strategies, lung cancer remains the leading cause of cancer-related death worldwide. Acetylshikonin is a derivative of the traditional Chinese medicine Zicao and presents a variety of anticancer properties. However, the effects of acetylshikonin on lung cancer have not been fully understood yet. This study explored the mechanisms underlying acetylshikonin-induced cell death in non-small cell lung cancer (NSCLC). Treating NSCLC cells with acetylshikonin significantly reduced cell viability, as evidenced by chromatin condensation and the appearance of cell debris. Acetylshikonin has also been shown to increase cell membrane permeability and induce cell swelling, leading to an increase in the population of necrotic cells. When investigating the mechanisms underlying acetylshikonin-induced cell death, we discovered that acetylshikonin promoted oxidative stress, decreased mitochondrial membrane potential, and promoted G2/M phase arrest in lung cancer cells. The damage to NSCLC cells induced by acetylshikonin resembled results involving alterations in the cell membrane and mitochondrial morphology. Our analysis of oxidative stress revealed that acetylshikonin induced lipid oxidation and down-regulated the expression of glutathione peroxidase 4 (GPX4), which has been associated with necroptosis. We also determined that acetylshikonin induces the phosphorylation of receptor-interacting serine/threonine-protein kinase 1 (RIPK1)/RIPK3 and mixed lineage kinase domain-like kinase (MLKL). Treatment with RIPK1 inhibitors (necrostatin-1 or 7-Cl-O-Nec-1) significantly reversed acetylshikonin-induced MLKL phosphorylation and NSCLC cell death. These results indicate that acetylshikonin activated the RIPK1/RIPK3/MLKL cascade, leading to necroptosis in NSCLC cells. Our findings indicate that acetylshikonin reduces lung cancer cells by promoting G2/M phase arrest and necroptosis.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Proteínas Quinases/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Necroptose , Apoptose , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo
18.
J Food Biochem ; 46(11): e14221, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35596593

RESUMO

Human oral squamous cell carcinoma (OSCC) has been one of the most common oral cancers owing to high percentage of betel nuts chewers, smokers, and alcohol consumption. With current treatment strategies in OSCC, more than half patients relapse and develop distant metastases with poor prognosis. To overcome the incident, OSCC poses a challenge in current therapies and treatments. Naringenin, a natural flavonoid, has been noted for antitumor effects on various types of cancers; however, the effects of naringenin on OSCC remain bias. In this study, naringenin demonstrated the potential multifunction in human OSCC cells not only leading to cell apoptosis, but also alternating the general function of autophagy, serving as pro-survival mechanism by inducing the endoplasmic reticulum (ER) stress signaling through intracellular reactive oxygen species (ROS) production. In the process of programmed cell death, naringenin induced apoptotic signaling through caspase-cascade, mitochondrial dysfunction, and ER stress by aberrance of Ca2+ release. In contrast, under the presence of naringenin, the pro-survival has been altered into pro-death to activate the caspases-mediated apoptosis achieving cell death. The cross-function of apoptosis and autophagy has demonstrated the effect of naringenin-induced intracellular ROS activity in OSCC cells. Therefore, this study found that the effect of naringenin induces intracellular ROS to trigger programmed cell death and ER stress through the mechanisms of apoptosis and autophagy in human oral squamous carcinoma. PRACTICAL APPLICATIONS: This study revealed that naringenin debilitated the OSCC cell viability via the intracellular ROS production, ER stress, and autophagy, leading to cell apoptosis. Based on these studies and findings, naringenin provided an antitumor effect as a novel natural compound to improve the current therapies in OSCC.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , Humanos , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Estresse do Retículo Endoplasmático , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Espécies Reativas de Oxigênio/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço , Apoptose , Autofagia
19.
J Cell Sci ; 122(Pt 18): 3358-64, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19706680

RESUMO

Prox1 is a master regulator for the development of lymphatic vasculature and the induction of lymphangiogenesis. In this study, we identified Prox1 as a new target for small ubiquitin-like modifier 1 (SUMO-1). Lysine 556 (K556) was found to be the major sumoylation site for Prox1 in vitro and in vivo. Mutation of this site (from lysine to arginine K556R) reduced DNA binding and the transcriptional activity of Prox1. Overexpression of Prox1 in EA.hy926 endothelial cells induced expression of lymphatic endothelial cell-specific genes including vascular endothelial growth factor receptor 3 (VEGFR3), fibroblast growth factor receptor 3 (FGFR3) and p57 while expression of K556R mutant Prox1 had little effect. The induction of VEGFR3 by Prox1 in EA.hy926 endothelial cells was an indication of their response to VEGF-C-induced lymphangiogenic signals, including the enhancement of proliferation, sprouting and tube formation and the inhibition of apoptosis. This effect is SUMO-dependent because ectopic expression of SUMO-specific protease 2 (SENP2) effectively reduced Prox1 sumoylation and Prox1-induced VEGFR3 expression. In addition, K556R mutant Prox1 could not induce lymphatic phenotypes. Taken together, our results indicate that Prox1 is a target for SUMO-1 and suggest that sumoylation of Prox1 controls its ability to induce VEGFR3 expression and lymphatic phenotypes in endothelial cells.


Assuntos
Células Endoteliais/metabolismo , Endotélio Linfático/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Apoptose , Linhagem Celular , Proliferação de Células , DNA/metabolismo , Células Endoteliais/citologia , Endotélio Linfático/citologia , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/genética , Humanos , Lisina/metabolismo , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Neovascularização Fisiológica , Fenótipo , Ligação Proteica , Transdução de Sinais , Transcrição Gênica , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/genética , Fator C de Crescimento do Endotélio Vascular/metabolismo
20.
J Exp Clin Cancer Res ; 39(1): 254, 2020 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-33228783

RESUMO

BACKGROUND: Osteosarcoma is generally reported among younger individuals and has a very poor prognosis, particularly for the development of metastasis. However, more effective metastatic biomarkers and therapeutic methods are absent. Monocyte chemoattractant protein-1 (MCP-1) is involved in cancer progression and inflammatory recruitment. Although previous studies have reported higher serum MCP-1 levels in patients with osteosarcoma, the role of MCP-1 in osteosarcoma progression remains to be addressed. METHODS: The osteosarcoma cell migratory ability was assessed by transwell migration assay. The MCP-1 and MMP-9 expression levels were analyzed by Western blot and qPCR. The signal activation was conducted by Western blot. The in vivo mouse experiment and tumor tissue array were performed to confirm our findings in vitro. RESULTS: The present study demonstrates that MCP-1 regulates cell mobility through matrix metalloproteinase (MMP)-9 expression in osteosarcoma cells. Moreover, MCP-1 promotes MMP-9 expression, cell migration, and cell invasion by mediating CCR2, c-Raf, MAPK, and AP-1 signal transduction. Using MCP-1 knockdown stable cell lines, we found that MCP-1 knockdown reduces MMP-9 expression and cell mobility. Finally, we found high MCP-1 expression levels in osteosarcoma specimens. CONCLUSIONS: Our results provide prognostic value of MCP-1 in osteosarcoma by promoting MMP-9 expression.


Assuntos
Neoplasias Ósseas/metabolismo , Quimiocina CCL2/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Osteossarcoma/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Células HEK293 , Xenoenxertos , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos SCID , Osteossarcoma/patologia , Prognóstico , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA