Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Int J Biol Sci ; 20(3): 818-830, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38250159

RESUMO

Insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2), with high affinity to a myriad of RNA transcripts, has been shown to elicit promotive effects on tumorigenesis and metastasis. Yet, the functional involvement of IGF2BP2 in the progression of oral squamous cell carcinoma (OSCC) remains poorly understood. In this study, we showed that IGF2BP2 was upregulated in head and neck cancer, and high levels of IGF2BP2 were associated with poor survival. In in vitro experiments, IGF2BP2 promoted migration and invasion responses of OSCC cells. Moreover, we identified an IGF2BP2-regulated gene, EREG, which functioned as a modulator of OSCC invasion downstream of IGF2BP2. In addition, EREG expression triggered the epithelia-mesenchymal transition (EMT) in OSCC, as evidenced by the observation that knockdown of EREG weakened the induction of EMT mediated by IFG2BP2, and replenishment of EREG favored the EMT in IGF2BP2-depleted cells. Such IGF2BP2-regulated EREG expression, EMT, and cell invasion were dependent on the activation of FAK/Src signaling pathway. Collectively, these findings suggest that EREG, serving as a functional mediator of IGF2BP2-regulated EMT and cell invasion in oral cancer, may be implicated as a potential target for antimetastatic therapies.


Assuntos
Neoplasias Bucais , Proteínas de Ligação a RNA , Carcinoma de Células Escamosas de Cabeça e Pescoço , Humanos , Epirregulina , Transição Epitelial-Mesenquimal/genética , Neoplasias Bucais/genética , Proteínas de Ligação a RNA/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Regulação para Cima/genética
2.
Front Cell Dev Biol ; 11: 1163314, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37091977

RESUMO

Targeting the tumor microenvironment is increasingly recognized as an effective treatment of advanced lung adenocarcinoma (LUAD). However, few studies have addressed the efficacy of immunotherapy for LUAD. Here, a novel method for predicting immunotherapy efficacy has been proposed, which combines single-cell and bulk sequencing to characterize the immune microenvironment and metabolic profile of LUAD. TCGA bulk dataset was used to cluster two immune subtypes: C1 with "cold" tumor characteristics and C2 with "hot" tumor characteristics, with different prognosis. The Scissor algorithm, which is based on these two immune subtypes, identified GSE131907 single cell dataset into two groups of epithelial cells, labeled as Scissor_C1 and Scissor_C2. The enrichment revealed that Scissor_C1 was characterized by hypoxia, and a hypoxic microenvironment is a potential inducing factor for tumor invasion, metastasis, and immune therapy non-response. Furthermore, single cell analysis was performed to investigate the molecular mechanism of hypoxic microenvironment-induced invasion, metastasis, and immune therapy non-response in LUAD. Notably, Scissor_C1 cells significantly interacted with T cells and cancer-associated fibroblasts (CAF), and exhibited epithelial-mesenchymal transition and immunosuppressive features. CellChat analysis revealed that a hypoxic microenvironment in Scissor_C1elevated TGFß signaling and induced ANGPTL4 and SEMA3C secretion. Interaction with endothelial cells with ANGPTL4, which increases vascular permeability and achieves distant metastasis across the vascular endothelium. Additionally, interaction of tumor-associated macrophages (TAM) and Scissor_C1 via the EREG/EFGR pathway induces tyrosine kinase inhibitor drug-resistance in patients with LAUD. Thereafter, a subgroup of CAF cells that exhibited same features as those of Scissor_C1 that exert immunosuppressive functions in the tumor microenvironment were identified. Moreover, the key genes (EPHB2 and COL1A1) in the Scissor_C1 gene network were explored and their expressions were verified using immunohistochemistry. Finally, the metabolism dysfunction in cells crosstalk was determined, which is characterized by glutamine secretion by TAM and uptake by Scissor_C1 via SLC38A2 transporter, which may induce glutamine addiction in LUAD cells. Overall, single-cell sequencing clarifies how the tumor microenvironment affects immunotherapy efficacy via molecular mechanisms and biological processes, whereas bulk sequencing explains immunotherapy efficacy based on clinical information.

3.
Pulm Pharmacol Ther ; 69: 102048, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34082109

RESUMO

Antizyme inhibitors 2 (AZIN2) was found to be associated with poor prognosis of patients with rectal cancer. However, no studies have reported whether AZIN2 functions in non-small cell lung cancer (NSCLC). This study aimed to investigate the role of AZIN2 in cisplatin (DDP) resistance in NSCLC. We established DDP resistant A549 and H1299 cell lines. The transcriptional and translational expression levels were examined using quantitative real-time polymerase chain reaction and western blot. Cell apoptosis was evaluated by caspase-3 activity and nucleosome ELISA assays. Luciferase reporter assay was employed to evaluate the impact of hypoxia-inducible factor (HIF-1α) on AZIN2 transcription. AZIN2 expression was found to be associated with DDP resistance and poor prognosis in patients with NSCLC. AZIN2 overexpression promoted cell viability, colony formation, and reduced cell apoptosis in H1299 cells and A549 upon DDP treatment. Correspondingly, AZIN2 knockdown significantly inhibited cell viability and colony formation, and increased cell apoptosis upon DDP treatment. Interestingly, AZIN2 expression in NSCLC cells was significantly induced by hypoxia condition. The occupancy of HIF-1α, an important regulator of the hypoxia response, remarkably enriched at the promoter region of AZIN2 under hypoxia condition. In addition, AZIN2 overexpression resulted in epithelial-mesenchymal transition (EMT). The results suggested that hypoxia-induced AZIN2 high expression may contribute to DDP resistance development by promoting the EMT.


Assuntos
Antineoplásicos , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Humanos , Hipóxia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética
4.
Open Med (Wars) ; 16(1): 410-418, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33748425

RESUMO

The aim of this study was to evaluate the association of the chromobox homologue 7 (CBX7) expression with the epithelial-mesenchymal transition in cervical cancer (CC), as well as with the disease prognosis. CBX7, E-cadherin (E-cad), and vimentin (VIM) expression levels were detected with immunohistochemistry. The relationship between the expression of CBX7, E-cad, and VIM expression and conventional clinicopathological characteristics of CC were evaluated. The positive expression rates of CBX7 and E-cad in the CC tissues were lower than the adjacent non-tumorous cervical tissues. Moreover, the VIM expression level was higher. The CBX7 expression was positively correlated with the E-cad expression, whereas was negatively correlated with the VIM expression. Furthermore, CBX7 was associated with the disease clinical staging, histological differentiation, lymph node metastasis, and vascular invasion. Patients with negative CBX7 expression showed decreased overall survival rates compared with those with low or high CBX7 expression. Multivariate Cox regression analysis indicated that the decreased CBX7 expression was an independent predictor for the poor prognosis of CC. In conclusion, the absence of CBX7 is associated with the histologic differentiation, lymphatic metastasis, vascular invasion, and poor prognosis of CC. CBX7 may be an independent prognostic factor for the prognosis of CC patients.

5.
Aging (Albany NY) ; 13(3): 3202-3217, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33539321

RESUMO

Prostate cell proliferation, driven by testosterone, is a major characteristic of benign prostatic hyperplasia (BPH). GV1001, a human telomerase reverse transcriptase catalytic subunit, is an injectable formulation used as a cancer vaccine. It functions as a cell penetrating peptide to regulate cell proliferation. Here, we found that GV1001 effectively suppressed proliferation of prostatic stromal myofibroblasts (WPMY-1) and prostatic epithelial cells (RWPE-1 and WPE-NA22) treated with dihydrotestosterone. Also, GV1001 bound to androgen receptors (ARs) in the cytosol of stromal and epithelial cells. In an experimental animal model implanted with an infusion pump for spontaneous and continuous release of testosterone, revealed that GV1001 reduced prostatic hypertrophy and inhibited the cell proliferation and the expression of Ki67, proliferating cell nuclear antigen, and prostate specific antigen. In addition, GV1001 prevented fibrosis of the prostate by downregulating expression of prostatic epithelial-mesenchymal transition (EMT)-related proteins such as transforming growth factor (TGF)-ß, Snail, Slug, N-cadherin, and Vimentin, and by up-regulating E-cadherin. Taken together, these results suggest that GV1001, which suppresses TGF-ß-mediated EMT by outcompeting testosterone for binding to AR, is a potential therapeutic drug for BPH accompanied by prostatic fibrosis.


Assuntos
Proliferação de Células/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Hiperplasia Prostática/metabolismo , Receptores Androgênicos , Telomerase/farmacologia , Animais , Linhagem Celular , Di-Hidrotestosterona/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Androgênicos/efeitos dos fármacos , Receptores Androgênicos/metabolismo
6.
Acta Pharmacol Sin ; 42(8): 1288-1297, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33159174

RESUMO

Recent evidence shows that the expression levels of histamine receptor H3 (Hrh3) are upregulated in several types of cancer. However, the role of Hrh3 in non-small cell lung cancer (NSCLC) has not been elucidated. In the present study, we showed that the expression levels of Hrh3 were significantly increased in NSCLC samples, and high levels of Hrh3 were associated with poor overall survival (OS) in NSCLC patients. In five human NSCLC cell lines tested, Hrh3 was significantly upregulated. In NSCLC cell lines H1975, H460, and A549, Hrh3 antagonist ciproxifan (CPX, 10-80 µM) exerted moderate and concentration-dependent inhibition on the cell growth and induced apoptosis, whereas its agonist RAMH (80 µM) reversed these effects. Furthermore, inhibition of Hrh3 by CPX or siRNA retarded the migration and invasion of NSCLC cells through inhibiting epithelial-mesenchymal transition (EMT) progression via reducing the phosphorylation of PI3K/Akt/mTOR and MEK/ERK signaling pathways. In nude mice bearing H1975 cell xenograft or A549 cell xenograft, administration of CPX (3 mg/kg every other day, intraperitoneal) significantly inhibited the tumor growth with increased E-cadherin and ZO-1 expression and decreased Fibronectin expression in tumor tissue. In conclusion, this study reveals that Hrh3 plays an important role in the growth and metastasis of NSCLC; it might be a potential therapeutic target against the lung cancer.


Assuntos
Antineoplásicos/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Antagonistas dos Receptores Histamínicos/farmacologia , Imidazóis/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Receptores Histamínicos H3/metabolismo , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Antagonistas dos Receptores Histamínicos/uso terapêutico , Humanos , Imidazóis/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
7.
FASEB J ; 34(11): 14892-14904, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32939891

RESUMO

Renal fibrosis is a common pathological hallmark of chronic kidney disease (CKD). Renal sympathetic nerve activity is elevated in patients and experimental animals with CKD and contributes to renal interstitial fibrosis in obstructive nephropathy. However, the mechanisms underlying sympathetic overactivation in renal fibrosis remain unknown. Norepinephrine (NE), the main sympathetic neurotransmitter, was found to promote TGF-ß1-induced epithelial-mesenchymal transition (EMT) and fibrotic gene expression in the human renal proximal epithelial cell line HK-2. Using both genetic and pharmacological approaches, we identified that NE binds Gαq-coupled α1-adrenoceptor (α1-AR) to enhance EMT of HK-2 cells by activating p38/Smad3 signaling. Inhibition of p38 diminished the NE-exaggerated EMT process and increased the fibrotic gene expression in TGF-ß1-treated HK-2 cells. Moreover, the pharmacological blockade of α1-AR reduced the kidney injury and renal fibrosis in a unilateral ureteral obstruction mouse model by suppressing EMT in the kidneys. Thus, sympathetic overactivation facilitates EMT of renal epithelial cells and fibrosis via the α1-AR/p38/Smad3 signaling pathway, and α1-AR inhibition may be a promising approach toward treating renal fibrosis.


Assuntos
Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Insuficiência Renal Crônica/metabolismo , Tansulosina/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 1/uso terapêutico , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Fibrose , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Norepinefrina/farmacologia , Receptores Adrenérgicos alfa 1/metabolismo , Insuficiência Renal Crônica/tratamento farmacológico , Insuficiência Renal Crônica/etiologia , Proteína Smad3/metabolismo , Tansulosina/uso terapêutico , Fator de Crescimento Transformador beta/farmacologia , Obstrução Uretral/complicações , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Drug Des Devel Ther ; 14: 1127-1141, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32214798

RESUMO

INTRODUCTION: Ulcerative colitis (UC) is a chronic and inflammatory bowel disease. UC-associated colorectal cancer (UC-CRC) is one of the most severe complications of long-standing UC. In the present study, we explored the effects of miR-370-3p on UC-CRC in vivo and investigated its underlying mechanisms in vivo and in vitro. METHODS: Azoxymethane (AOM) and dextran sodium sulfate (DSS) were used to induce UC-CRC in C57BL/6 mice. AOM/DSS-induced mice were treated with 5×108 pfu miR-370-3p overexpressing-adenovirus via tail-vein injection every two weeks. RESULTS: We found that miR-370-3p significantly improved the body weights and survival rates and inhibited the tumorigenesis of UC-CRC in AOM/DSS mice. Mechanically, miR-370-3p inhibited AOM/DSS-induced inflammatory response by decreasing tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and interleukin-6 (IL-6) through targeting toll-like receptor 4 (TLR4), as demonstrated by down-regulation of TLR4, cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), and phosphorylated epidermal growth factor receptor (pEGFR). miR-370-3p decreased the expression of tumor-associated proteins, including p53, ß-catenin, and ki67 in AOM/DSS-treated mice. Additionally, miR-370-3p remarkably inhibited epithelial-mesenchymal transition (EMT) via increasing E-cadherin expression and reducing N-cadherin and Vimentin expression in vivo. Further studies showed that miR-370-3p repressed proliferation and EMT of colon cancer cells in vitro. Moreover, we proved that miR-370-3p decreased the expression of tumor-associated proteins and reversed EMT by regulating ß-catenin in colon cancer cells. CONCLUSION: Taken together, miR-370-3p alleviated UC-CRC by inhibiting the inflammatory response and EMT in mice, which suggested miR-370-3p as a novel potential target for UC-CRC therapy.


Assuntos
Colite Ulcerativa/metabolismo , Neoplasias Colorretais/metabolismo , Transição Epitelial-Mesenquimal , Inflamação/metabolismo , MicroRNAs/metabolismo , Animais , Azoximetano , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/patologia , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Humanos , Inflamação/induzido quimicamente , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética
9.
Cells ; 9(3)2020 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-32188112

RESUMO

Understanding how heterogeneous cancer cell populations migrate collectively is of paramount importance to arrest metastasis. Here, we applied 3D culture-based approaches for in vitro modeling of the collective migration of squamous carcinoma cells and examine the impact of epithelial and mesenchymal cell interactions on this type of migration. We show that both mesenchymal N-cadherin-expressing cancer cells and cancer-associated fibroblasts cooperate in collective migration of epithelial cancer cells by leading their collective migration. This was consistent with the observed distribution of E-cadherin/N-cadherin in the human carcinoma tissues of head and neck. The presence of "leader" mesenchymal cancer cells or "leader" fibroblasts was significantly associated with metastasis development, recurrent disease and low overall disease survival in head and neck squamous cell carcinomas (HNSCC). In silico analysis of independent public datasets revealed that increased N-cadherin expression in the heterogeneous cancer tissues is associated with disease progression not only in HNSCC but also in other prevalent tumors, such as colorectal, breast and lung cancer. Collectively, our data highlight the importance of mesenchymal cells in collective cell migration and disease progression, findings that may have a broad significance in cancer, especially in those in which aberrant N-cadherin expression negatively impacts disease survival.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Comunicação Celular/fisiologia , Neoplasias Pulmonares/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Humanos
10.
Dig Liver Dis ; 52(6): 668-676, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31822388

RESUMO

BACKGROUND: Prognosis of hepatocellular carcinoma (HCC) remains poor despite significant recent improvement in therapy. Recent studies have reported that transglutaminase 3 (TGM3) plays an important role in several human cancer types. However, the role of TGM3 in HCC have not been previously elucidated. METHODS: We evaluated the role of TGM3 in regulating HCC cell proliferation, migration, and invasion. We also investigated the prognostic significance of TGM3 in an HCC cohort. Finally, we explored the signalling pathways that TGM3 regulates in HCC. RESULTS: We identified TGM3 to be overexpressed in HCC compared to normal tissues. Higher expression of TGM3 predicts poor prognosis in HCC patients. TGM3 knockdown led to decreased HCC cell proliferation, invasion, and xenograft tumour growth. TGM3 depletion inhibited AKT, extracellular signal-regulated kinase (ERK), p65, and glycogen synthase kinase 3ß (GSK3ß)/ß-catenin activation, but promoted levels of cleaved caspase 3. Moreover, TGM3 knockdown cells had increased E-cadherin levels and decreased vimentin levels, suggesting that TGM3 contributes to epithelial-mesenchymal transition (EMT) in HCC. CONCLUSION: Our results suggest that TGM3 controls multiple oncogenic pathways in HCC, thereby contributing to increased cell proliferation and EMT, and TGM3 potentially enhances HCC metastasis. TGM3 may serve as a novel therapeutic target in HCC.


Assuntos
Carcinoma Hepatocelular/metabolismo , Transição Epitelial-Mesenquimal , Neoplasias Hepáticas/metabolismo , Transdução de Sinais , Transglutaminases/metabolismo , Animais , Carcinogênese , Carcinoma Hepatocelular/mortalidade , Carcinoma Hepatocelular/cirurgia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/cirurgia , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Prognóstico , Transglutaminases/genética , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Eur J Clin Invest ; 49(6): e13098, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30838641

RESUMO

There is a complex oxidant and antioxidant system that maintains the redox homoeostasis in the liver. While suffering from exogenous or endogenous risk factors, the balance between oxidants and antioxidants is disturbed and excessive reactive oxygen species are generated, resulting in oxidative stress. Oxidative stress is prevalent in various liver diseases and is thought to be involved in their pathophysiology. Advanced oxidation protein products are generated under conditions of oxidative damage and are newly described protein markers of oxidative stress. Previous studies have underscored the universal pathogenic roles of oxidation protein products in various diseases. However, investigations into how these products participate in the development of liver diseases have been superficial and insufficient. In this review, we highlight the current understanding of the roles of advanced oxidation protein products in liver disease pathogenesis and the underlying mechanisms. Moreover, we summarize the current studies on advanced oxidation protein products in infectious and noninfectious, acute and chronic liver diseases. Different strategies for targeting these advanced oxidation protein products and future perspectives, which may pave the way for developing new therapeutic strategies, will also be discussed here.

12.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-849799

RESUMO

Objective: To explore the effect of Slit2/ROBO1 protein (Slit2/ROBO1) signaling pathway in high glucose-induced epithelial-mesenchymal transdifferentiation (EMT) and its mechanism. Methods: Human renal tubular epithelial cells (HK-2) were cultured in vitro and subjected to high glucose concentration and time gradient experiments. First, for concentration gradient experiment, the sample was randomly divided into normal group, control group 1, control group 2, high glucose group 1, high glucose group 2. While for high glucose time gradient experiment, the sample was randomly divided into normal group, control group, high glucose 24 h group, high glucose 36 h group and high glucose 48 h group. Western blotting was used to detect the expression changes of Slit2, ROBO1, α-smooth muscle actin (α-SMA) and fibronectin in HK-2 cells, and then the optimal high glucose stimulation concentration and time were screened out. Slit2 over-expressed plasmid and negative control plasmid were transfected into HK-2 cells to verify the successful transfection, the cells were then randomly divided into normal group, control group, high glucose group, high glucose empty group and high glucose Slit2 group. The total protein was extracted after stimulation with optimal high glucose concentration and time, and Western blotting was then performed to detect the change in expression of fibronectin and α-SMA. Results: In the high glucose concentration gradient experiment, the expression of Slit2 declined significantly in high glucose group 1(0.647±0.048) and high glucose group 2(0.210±0.023) than in the normal group (1.000±0.050); the expression of ROBO1 declined significantly in high glucose group 1(0.703±0.041) and high glucose group 2(0.303±0.022) than in the normal group (1.000±0.057); while the expression of fibronectin increased significantly in high glucose group 1(1.953±0.042) and high glucose group 2(2.997±0.078) than in the normal group (0.990±0.059), and the expression of α-SMA increased significantly in high glucose group 1(1.767±0.012) and high glucose group 2(2.427±0.059) than in the normal group (1.033±0.067), all the differences were of statistical significance(P<0.05). Compared with the high glucose group 1, the expressions of Slit2 and ROBO1 decreased, and of fibronectin and α-SMA increased significantly in the high glucose group 2(P<0.05). In the high glucose time gradient experiment, compared with the normal group, the expressions of Slit2 in high glucose 36 h group and high glucose 48 h group decreased (0.943±0.032 vs. 0.557±0.020, 0.450±0.055, respectively), and the expression of ROBO1 decreased (1.000±0.058 vs. 0.600±0.023, 0.227±0.028, respectively). Compared with the normal group, the expression of fibronectin increased significantly in high glucose 24 h group, high glucose 36 h group and high glucose 48 h group (0.970±0.040 vs. 1.247±0.052, 1.733±0.084, 2.780±0.090, respectively), and the expression of α-SMA increased significantly in high glucose 24 h group, high glucose 36 h group and high glucose 48 h group (1.033±0.067 vs. 1.277±0.041, 1.767±0.120, 2.537±0.078, respectively), and the difference was statistically significant (P<0.05). Compared with high glucose 24 h group, the expression of Slit2 declined significantly in high glucose 36 h group and high glucose 48 h group(0.893±0.034 vs. 0.557±0.020, 0.450±0.055, respectively), and the expression of ROBO1 declined significantly (0.930±0.025 vs. 0.600±0.023, 0.227±0.028, respectively), the expressions of fibronectin and α-SMA increased significantly with statistical significance (P<0.05). Compared with high glucose 36 h group, the expression of Slit2 and ROBO1 declined significantly, and the expression of fibronectin and α-SMA increased significantly in high glucose 48 h group (P<0.05). In the high glucose environment, and achieving Slit2 overexpression and negative control plasmid transfection, the expression of fibronectin increased significantly in high glucose group, high glucose+empty group and high glucose+Slit2 group (2.760±0.012, 2.667±0.027, 1.460±0.034, respectively) than in normal group (1.000±0.058); the expression of α-SMA increased also in high glucose group, high glucose+empty group and high glucose+Slit2 group (2.487±0.048, 2.557±0.037, 1.270±0.017, respectively) than in normal group (1.000±0.050) with statistical significance (P<0.05). Compared with the high glucose+empty group, the expression of fibronectin and α-SMA declined significantly in the high glucose+Slit2 group(P<0.05). Conclusion: The decreased expression of Slit2 and ROBO1 is involved in the high glucose-induced renal tubular EMT. Overexpression of Slit2 may significantly inhibit the high glucose-induced EMT.

13.
BMC Cancer ; 18(1): 1263, 2018 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-30563499

RESUMO

BACKGROUND: Glucose regulated protein 78 (GRP78) is a resident chaperone of the endoplasmic reticulum and a master regulator of the unfolded protein response under physiological and pathological cell stress conditions. GRP78 is overexpressed in many cancers, regulating a variety of signaling pathways associated with tumor initiation, proliferation, adhesion and invasion which contributes to metastatic spread. GRP78 can also regulate cell survival and apoptotic pathways to alter responsiveness to anticancer drugs. Tumors that reside in or metastasize to the bone and bone marrow (BM) space can develop pro-survival signals through their direct adhesive interactions with stromal elements of this niche thereby resisting the cytotoxic effects of drug treatment. In this study, we report a direct correlation between GRP78 and the adhesion molecule N-cadherin (N-cad), known to play a critical role in the adhesive interactions of multiple myeloma and metastatic prostate cancer with the bone microenvironment. METHODS: N-cad expression levels (transcription and protein) were evaluated upon siRNA mediated silencing of GRP78 in the MM.1S multiple myeloma and the PC3 metastatic prostate cancer cell lines. Furthermore, we evaluated the effects of GRP78 knockdown (KD) on epithelial-mesenchymal (EMT) transition markers, morphological changes and adhesion of PC3 cells. RESULTS: GRP78 KD led to concomitant downregulation of N-cad in both tumors types. In PC3 cells, GRP78 KD significantly decreased E-cadherin (E-cad) expression likely associated with the induction in TGF-ß1 expression. Furthermore, GRP78 KD also triggered drastic changes in PC3 cells morphology and decreased their adhesion to osteoblasts (OSB) dependent, in part, to the reduced N-cad expression. CONCLUSION: This work implicates GRP78 as a modulator of cell adhesion markers in MM and PCa. Our results may have clinical implications underscoring GRP78 as a potential therapeutic target to reduce the adhesive nature of metastatic tumors to the bone niche.


Assuntos
Neoplasias Ósseas/genética , Proteínas de Choque Térmico/genética , Mieloma Múltiplo/genética , Neoplasias da Próstata/genética , Apoptose/genética , Neoplasias Ósseas/patologia , Neoplasias Ósseas/secundário , Caderinas/genética , Adesão Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Chaperona BiP do Retículo Endoplasmático , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Choque Térmico/antagonistas & inibidores , Humanos , Masculino , Mieloma Múltiplo/patologia , Metástase Neoplásica , Osteoblastos/patologia , Células PC-3 , Neoplasias da Próstata/patologia , RNA Interferente Pequeno/genética , Fator de Crescimento Transformador beta1/genética
14.
J Biomed Mater Res A ; 105(8): 2252-2265, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28513985

RESUMO

Nanofibers are thought to enhance cell adhesion, growth, and function. We demonstrate that the choice of building blocks in self-assembling nanofiber systems can be used to control cell behavior. The use of 2 D-coated, self-assembled nanofibers in controlling lens epithelial cells, fibroblasts, and mesenchymal stem cells was investigated, focusing on gene and protein expression related to the fibrotic response. To this end, three nanofibers with different characteristics (morphology, topography, and wettability) were compared with two standard materials frequently used in culturing cells, TCPS, and a collagen type I coating. Cell metabolic activity, cell morphology, and gene and protein expression were analyzed. The most hydrophilic nanofiber with more compact network consisting of small fibers proved to provide a beneficial 2 D environment for cell proliferation and matrix formation while decreasing the fibrotic/stress behavior in all cell lines when compared with TCPS and the collagen type I coating. This nanofiber demonstrates the potential to be used as a biomimetic coating to study the development of fibrosis through epithelial-to-mesenchymal transition. This study also shows that nanofiber structures do not enhance cell function by definition, because the physico-chemical characteristics of the nanofibers influence cell behavior as well and actually can be used to regulate cell behavior toward suboptimal performance. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2252-2265, 2017.


Assuntos
Materiais Revestidos Biocompatíveis/química , Células Epiteliais/citologia , Fibroblastos/citologia , Células-Tronco Mesenquimais/citologia , Nanofibras/química , Alicerces Teciduais/química , Adesão Celular , Linhagem Celular , Proliferação de Células , Células Cultivadas , Materiais Revestidos Biocompatíveis/efeitos adversos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose/etiologia , Fibrose/metabolismo , Fibrose/patologia , Regulação da Expressão Gênica , Humanos , Interações Hidrofóbicas e Hidrofílicas , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Nanofibras/efeitos adversos , Nanofibras/ultraestrutura , Alicerces Teciduais/efeitos adversos
15.
Exp Cell Res ; 350(1): 218-225, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27914787

RESUMO

Tumor metastasis is the leading cause of death in cancer patients. Identifying metastatic biomarkers in tumor cells would help cancer diagnoses and the development of therapeutic targets. Yes-associated protein (YAP) plays an important role in organ development and has gained much attention in tumorigenesis. However, the role of YAP and the underlying mechanism in tumor metastasis of colorectal cancer (CRC) is still unclear. In this study, we generated metastatic 116-LM cells from the HCT116 CRC cell line. We found that the capacity for tumor aggressiveness was elevated in 116-LM cells and identified that YAP and its mRNA level were upregulated in 116-LM cells. Moreover, expression of YAP was found to correlate with epithelial-mesenchymal transition (EMT) marker expressions, whereas suppression of YAP decreased EMT marker expressions and impeded tumor migration and invasion. Additionally, upregulation of YAP was identified in colon cancer patients, and it was correlated with EMT gene expressions. Furthermore, we identified LBH589, a histone deacetylase inhibitor, that was capable of inhibiting tumor growth and aggressiveness in both HCT116 and 116-LM cells. LBH589 potentially inhibited YAP and its mRNA expression, accompanied by diminished expressions of YAP downstream genes and EMT markers. Together, YAP plays a crucial role in aggressiveness and metastasis of CRC, and YAP may be an attractive therapeutic target.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias Colorretais/metabolismo , Transição Epitelial-Mesenquimal , Fosfoproteínas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Células HCT116 , Humanos , Ácidos Hidroxâmicos/farmacologia , Indóis/farmacologia , Panobinostat , RNA Interferente Pequeno/genética , Fatores de Transcrição , Regulação para Cima , Proteínas de Sinalização YAP
16.
Bull Cancer ; 103(6 Suppl 1): S39-47, 2016 Jun.
Artigo em Francês | MEDLINE | ID: mdl-27494972

RESUMO

FUNDAMENTAL BASIS OF METASTATIC PROCESS: Metastatic process is described as a "dissemination of neoplastic cells in a distant secondary site, in which cells proliferate to develop a mass of cells partially differentiated". The vast majority of death in solid cancers is the consequence of metastasis development which lead to vital organ dysfunction. In the present review, either recent discoveries or controversial subjects associated with metastasis process will be discussed. Indeed epithelia-mesenchymal transition (EMT), circulating tumor cells, tumor dormancy, colonization in distant organ and cancer stem cells are tackled.


Assuntos
Transição Epitelial-Mesenquimal , Metástase Neoplásica , Células-Tronco Neoplásicas , Adesão Celular , Movimento Celular , Humanos , Invasividade Neoplásica , Metástase Neoplásica/patologia , Metástase Neoplásica/fisiopatologia , Células Neoplásicas Circulantes/patologia , Células-Tronco Neoplásicas/fisiologia , Especificidade de Órgãos , Microambiente Tumoral
17.
World J Surg Oncol ; 14(1): 195, 2016 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-27460906

RESUMO

BACKGROUND: Extracellular matrix protein 1 (ECM1) is a glycoprotein involved in many biologic processes. To determine the expression of ECM1 in hepatocellular carcinoma (HCC), and to study the role of ECM1 in inducing epithelia-mesenchymal transition (EMT) to analyze the effect of ECM1 on the migration and invasion of HCC cells. METHODS: The expression of ECM1 in HCC specimens was examined by immunohistochemistry staining, and the correlations were analyzed between the expression of ECM1 and the clinicopathological data. The ECM1 expression plasmid was transfected into Bel-7402 cells to induce exogenous overexpression of ECM1 protein. The changes of the expression of ECM1, EMT-related protein (E-cadherin, Vimentin), in Bel-7402 cells were detected by Western blot after transfection of ECM1; the wound healing and invasion assay in vitro were used to determine the role of ECM1 gene transfection on the ability of migration and invasive potential of Bel-7402 cells. RESULTS: Immumohistochemistry staining method displayed the ECM1 expression was positively associated with vascular invasion, TNM stage, and poor prognosis. A significant positive correlation was found between the expressions of ECM1 and Vimentin. After ECM1 overexpression, Western blot exhibited that the expression of E-cadherin was down-regulated and Vimentin expression was up-regulated in Bel-7402 cells; the wound healing and invasion assay showed that the migration and invasion potentials of Bel-7402 cells were significantly enhanced. CONCLUSIONS: ECM1, which displayed a high expression in HCC specimens, was closely associated with clinicopathologic data and may promote migration and invasion of HCC cells by inducing EMT.


Assuntos
Carcinoma Hepatocelular/patologia , Transição Epitelial-Mesenquimal , Proteínas da Matriz Extracelular/metabolismo , Neoplasias Hepáticas/patologia , Adulto , Idoso , Antígenos CD , Caderinas/metabolismo , Carcinoma Hepatocelular/cirurgia , Linhagem Celular Tumoral , Movimento Celular , Estudos de Coortes , Regulação para Baixo , Feminino , Seguimentos , Humanos , Imuno-Histoquímica , Neoplasias Hepáticas/cirurgia , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Estadiamento de Neoplasias , Prognóstico , Regulação para Cima , Vimentina/metabolismo , Adulto Jovem
18.
Int J Cancer ; 134(6): 1369-78, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24037692

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is known for its aggressive growth, and is characterized by early tissue invasion and metastasis with poor prognosis. Identifying prognostic markers and delineating the underlying mechanisms that promote progression of PDAC are important for the treatment of pancreatic cancer. TIP30, a newly identified tumor suppressor, appears to be involved in multiple processes during tumor development and metastasis. Here, we investigated the expression of TIP30 in PDAC and its prognostic value in PDAC patients. We examined the expression of TIP30 by immunohistochemistry in tissue microarrays containing 106 surgically resected PDAC. Kaplan-Meier analysis and Cox proportional hazards regression modeling analysis showed that TIP30 expression independently predicted better survival in pancreatectomy patients (p < 0.01). Moreover, decreased TIP30 expression was associated with lymph node metastasis (p < 0.05) and loss of E-cadherin expression (r = 0.329, p < 0.01). Suppression of TIP30 resulted in upregulation of Snail and subsequent downregulation of E-cadherin in SW1990 cells containing high-level of endogenous TIP30. However, in the PANC-1 cells containing low level of endogenous TIP30, suppressing TIP30 caused upregulation of Slug instead of Snail, followed by upregulation of MMP9 rather than E-cadherin. Taken together, our work reveals that decreased TIP30 expression is able to enhance invasion and metastasis of pancreatic cancer cells through upregulation of the Snail family members and may serve as an independent predictor for poor outcomes in PDAC patients.


Assuntos
Acetiltransferases/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma Ductal Pancreático/mortalidade , Neoplasias Pancreáticas/mortalidade , Fatores de Transcrição/metabolismo , Acetiltransferases/genética , Adulto , Western Blotting , Caderinas/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Proliferação de Células , Transição Epitelial-Mesenquimal , Feminino , Imunofluorescência , Seguimentos , Humanos , Técnicas Imunoenzimáticas , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Estadiamento de Neoplasias , Pâncreas/metabolismo , Pâncreas/patologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Prognóstico , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Estudos Retrospectivos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise Serial de Tecidos , Fatores de Transcrição/genética , Células Tumorais Cultivadas , Cicatrização
19.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-453611

RESUMO

Objective To study the relationship between the expression of Caveolin-1 (Cav-1) and epithelial-mesenchymal transition(EMT) or metastasis in human pancreatic cancer.Methods The Cav-1 protein and EMT markers in highly metastatic pancreatic cancer cell lines (L3.7,Panc02-H7) and poorly metastatic pancreatic cancer cell lines (COLO357,Panc02) was detected by Western blotting and immunofluorescence.The morphology of the pancreatic cancer cells were observed under phase-contrast photomicrographs.The plasmids pcDNA3.1-caveolin-1 and control vector pcDNA3.1 were transfected into COLO357 cells by Lipofectamine LTX.The expression of Cav-1 protein,E-cadherin and vimentin were detected,and the morphology of COLO357 cells observed.Results L3.7 and Panc02-H7 cells had strong positive Cav-1 staining and high expression of mesenchymal marker (vimentin,N-cadherin) and low epithelial marker (E-cadherin,β-catenin),whereas COLO357 and Panc02 cells with typical epithelial morphology were weak positive in Cav-1 staining and of low expression of vimentin,N-cadherin and high expression of E-cadherin,β-catenin.In Cav-1 transfected COLO357 cells vimentin expression increased,and E-cadherins decreased resulting in typical morphology changes of EMT.Conclusions Overexpression of caveolin-1 is correlated with epithelial-mesenchymal transition of pancreatic cancer cells and cancer metastasis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...