Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Cancer Immunol Immunother ; 73(5): 91, 2024 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-38554157

RESUMO

BACKGROUND: Accumulation studies found that tumor-associated macrophages (TAMs) are a predominant cell in tumor microenvironment (TME), which function essentially during tumor progression. By releasing bioactive molecules, including circRNA, small extracellular vesicles (sEV) modulate immune cell functions in the TME, thereby affecting non-small cell lung cancer (NSCLC) progression. Nevertheless, biology functions and molecular mechanisms of M2 macrophage-derived sEV circRNAs in NSCLC are unclear. METHODS: Cellular experiments were conducted to verify the M2 macrophage-derived sEV (M2-EV) roles in NSCLC. Differential circRNA expression in M0 and M2-EV was validated by RNA sequencing. circFTO expression in NSCLC patients and cells was investigated via real-time PCR and FISH. The biological mechanism of circFTO in NSCLC was validated by experiments. Our team isolated sEV from M2 macrophages (M2Ms) and found that M2-EV treatment promoted NSCLC CP, migration, and glycolysis. RESULTS: High-throughput sequencing found that circFTO was highly enriched in M2-EV. FISH and RT-qPCR confirmed that circFTO expression incremented in NSCLC tissues and cell lines. Clinical studies confirmed that high circFTO expression correlated negatively with NSCLC patient survival. Luciferase reporter analysis confirmed that miR-148a-3p and PDK4 were downstream targets of circFTO. circFTO knockdown inhibited NSCLC cell growth and metastasis in in vivo experiments. Downregulating miR-148a-3p or overexpressing PDK4 restored the malignancy of NSCLC, including proliferation, migration, and aerobic glycolysis after circFTO silencing. CONCLUSION: The study found that circFTO from M2-EV promoted NSCLC cell progression and glycolysis through miR-148a-3p/PDK4 axis. circFTO is a promising prognostic and diagnostic NSCLC biomarker and has the potential to be a candidate NSCLC therapy target.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Vesículas Extracelulares , Neoplasias Pulmonares , MicroRNAs , Humanos , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Vesículas Extracelulares/genética , Vesículas Extracelulares/patologia , Neoplasias Pulmonares/patologia , Macrófagos/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Circular/genética , RNA Circular/metabolismo , Microambiente Tumoral
2.
J Transl Med ; 22(1): 216, 2024 02 29.
Artigo em Inglês | MEDLINE | ID: mdl-38424632

RESUMO

Lung adenocarcinoma (LUAD) is the most common pathological type of lung cancer, but the early diagnosis rate is low. The RNA-binding ubiquitin ligase MEX3C promotes tumorigenesis in several cancers but its mechanism of action in LUAD is unclear. In this study, the biological activity of MEX3C was assessed in LUAD. MEX3C and RUNX3 mRNA levels in the tissues of LUAD patients were determined using reverse transcription­quantitative PCR. The involvement of MEX3C in the growth and metastasis of LUAD cells was measured by EdU assay, CCK-8, colony formation, Transwell assay, TUNEL, and flow cytometry. Expression of apoptosis and epithelial-mesenchymal transition related proteins were determined using western blotting analysis. LUAD cells transfected with si-MEX3C were administered to mice subcutaneously to monitor tumor progression and metastasis. We found that MEX3C is strongly upregulated in LUAD tissue sections, and involved in proliferation and migration. A549 and H1299 cells had significantly higher levels of MEX3C expression compared to control HBE cells. Knockdown of MEX3C dramatically decreased cell proliferation, migration, and invasion, and accelerated apoptosis. Mechanistically, we demonstrate MEX3C induces ubiquitylation and degradation of tumor suppressor RUNX3. Moreover, RUNX3 transcriptionally represses Suv39H1, as revealed by RNA pull-down and chromatin immunoprecipitation assays. The in vivo mice model demonstrated that knockdown of MEX3C reduced LUAD growth and metastasis significantly. Collectively, we reveal a novel MEX3C-RUNX3-Suv39H1 signaling axis driving LUAD pathogenesis. Targeting MEX3C may represent a promising therapeutic strategy against LUAD.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , MicroRNAs , Animais , Humanos , Camundongos , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Regulação Neoplásica da Expressão Gênica , Ligases/genética , Ligases/metabolismo , Neoplasias Pulmonares/patologia , MicroRNAs/genética , RNA/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitinação
3.
Odontology ; 111(2): 474-486, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36181561

RESUMO

It has been demonstrated that circular RNA (circRNA) is involved in the progression of tongue squamous cell carcinoma (TSCC). The aim of this study was to investigate the intrinsic mechanism of circ_0081069 in TSCC progression. The expression levels of circ_00081069, miR-634, and mitogen-activated protein kinase kinase 4 (MAP2K4) in TSCC tissues and cells were detected by quantitative real-time PCR (qRT-PCR). Cell counting kit 8 assay, Edu assay, and flow cytometry assay were used to detect cell proliferation and cell cycle distribution. Transwell assay was used to detect cell migration and invasion abilities. Western blot analysis was performed to detect the protein expression. Dual-luciferase reporter assay was used to detect the targeting relationships of circ_0081069, miR-634 and MAP2K4. Immunohistochemical staining was used to measure MAP2K4-positive cells in tissues. The effect of circ_0081069 silencing on tumor formation in TSCC in vivo was explored by xenograft tumor assay. Circ_0081069 was highly expressed in TSCC tissues and cells. Silencing of circ_0081069 inhibited cell proliferation, cell cycle progress, cell migration and invasion in vitro, as well as hindered tumor growth in vivo. Mechanistically, circ_0081069 targeted miR-634 to negatively regulate miR-634 expression, and inhibition of miR-634 was able to weaken the inhibitory effect of circ_0081069 knockdown on proliferation, migration, and invasion of TSCC cells. MiR-634 targeted MAP2K4 and negatively regulated MAP2K4 expression, and overexpression of miR-634 inhibited TSCC cell proliferation, migration, and invasion, while co-overexpression of MAP2K4 was able to reverse the effects of miR-634 in TSCC cells. Circ_0081069 is involved in the regulation of proliferation, cycle progress, migration, and invasion of TSCC cells through the miR-634/MAP2K4 axis and has the potential to serve as a diagnostic biomarker and therapeutic target.


Assuntos
Carcinoma de Células Escamosas , MicroRNAs , Neoplasias da Língua , Humanos , Carcinoma de Células Escamosas/genética , Neoplasias da Língua/genética , Movimento Celular , Proliferação de Células , Língua , MicroRNAs/genética , Linhagem Celular Tumoral , MAP Quinase Quinase 4
4.
J Transl Med ; 20(1): 318, 2022 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-35842667

RESUMO

Circular RNAs (circRNAs) belong to a novel class of noncoding RNA that gained more attention in human cancer pathogenesis. The role of circRNA in esophageal squamous cell carcinoma (ESCC) is largely unclear. Present investigation was to characterize new circRNAs regulating ESCC progression and explore the regulatory mechanisms in ESCC. In this study, circRNAs differentially expressed in ESCC and adjacent normal tissues were characterized via high-throughput sequencing. Then the differentially expressed circRNA between ESCC and adjacent normal tissues were investigated using Rt-qPCR. The role of circ-ARAP2 expression on tumor progression were detected in both in vivo and in vitro. Luciferase reporter assays were used to identify the relationships among circ-ARAP2, microRNA (miR)-761 and the cell cycle regulator Forkhead Box M1 (FOXM1). The result of the expression profile analyses regarding human circRNAs in ESCC demonstrated that circ-ARAP2 was up-regulated significantly in both ESCC tissues and cell lines. Downregulation circ-ARAP2 suppressed ESCC proliferation, tumor growth and metastasis in both in vivo and in vitro. The data also suggested that miR-761 and FOXM1 were circ-ARAP2 downstream targets which were confirmed through luciferase reporter analysis. Overexpression of FOXM1 or inhibiting miR-761 restored ESCC cell proliferation and invasion ability after silencing circ-ARAP2. The study also found that circ-ARAP2 influenced the endothelial-mesenchymal transition (EMT) and cancer stem cells differently by regulating miR-761/FOXM1. In one word, the results demonstrated that abnormal circ-ARAP2 expression promoted ESCC progression by regulating miR-761/FOXM1 axis-mediated stemness and EMT.


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , MicroRNAs , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/patologia , Proteína Forkhead Box M1/genética , Proteínas Ativadoras de GTPase/genética , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/genética , RNA Circular/genética
5.
Carcinogenesis ; 41(5): 711-721, 2020 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-31430374

RESUMO

Rho GTPase-activating proteins (RhoGAPs) have been reported to be of great importance in the initiation and development of many different cancers. However, their biological roles and regulatory mechanisms in lung cancer development and progression are poorly defined. Real-time PCR or western blotting analysis was used to detect Rho GTPase-activating protein 24 (ARHGAP24), WWP2, p27, p-STAT6 and STAT6 expression levels as well as the activity of RhoA and Rac1 in lung cancer. Cell proliferation, apoptosis and cell cycle were measured by CCK-8 and flow cytometry analysis. Tumor growth of lung cancer cells was measured using a nude mouse xenograft experiment model in vivo. The correlation between WWP2 and p27 was measured by co-immunoprecipitation and ubiquitination analysis. We found that ARHGAP24 expression was lower in lung cancer tissues collected from the The Cancer Genome Atlas and independent hospital database. Overexpression of ARHGAP24 significantly suppressed cell proliferation and the activity of RhoA and Rac1, induced cell apoptosis and arrested cell cycle at the G0-G1 phase. ARHGAP24 overexpression also inhibited tumor growth in nude mice, whereas knockdown of ARHGAP24 significantly promoted cell proliferation and WWP2 expression and inhibited cell cycle arrest at G1 phase through activating STAT6 signaling. ARHGAP24 overexpression inhibited WWP2 overexpression-induced cell proliferation, cell cycle progression and the decreased p27 expression. Moreover, WWP2 was found interacted with p27, and WWP2 overexpression promoted the ubiquitination of p27. In conclusion, our findings suggest that ARHGAP24 inhibits cell proliferation and cell cycle progression and induces cell apoptosis of lung cancer via a STAT6-WWP2-p27 axis.


Assuntos
Ciclo Celular , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Neoplasias Pulmonares/patologia , Fator de Transcrição STAT6/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Movimento Celular , Inibidor de Quinase Dependente de Ciclina p27/genética , Proteínas Ativadoras de GTPase/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Fator de Transcrição STAT6/genética , Células Tumorais Cultivadas , Ubiquitina-Proteína Ligases/genética , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Phys Chem Chem Phys ; 21(22): 11731-11739, 2019 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-31114818

RESUMO

A novel family of two-dimensional (2D) crystalline metal superhalogens V3X8 (X = F, Cl, Br, I) with intrinsic magnetism was predicted using first-principles calculations in the framework of density functional theory (DFT). The calculation results show that the V3Cl8 monolayer is an intrinsic anti-ferromagnetic semiconductor (AFS) with an indirect bandgap of 0.086 eV at the PBE functional level, while the V3X8 (X = F, Br, I) monolayer exhibits a fascinating ferromagnetic half-metal (FH) property with 100% spin-polarization at the Fermi level. Such 2D materials possess robust dynamical stability as well as thermal stability at room temperature except for V3Br8. In addition, Monte Carlo simulations based on the Ising model with the classical Heisenberg model estimate a Curie temperature of approximately 77 K and 103 K for the V3F8 and V3I8 systems, respectively. Furthermore, we predict an extraordinary phenomenon induced by biaxial compressive strain from the ferromagnetic half-metal (FH) to the ferromagnetic semiconductor (FS) at 2% strain and then to the antiferromagnetic metal (AM) with the biaxial strain increasing up to about 6.3% in the 2D V3I8 monolayer. In the case of 2D V3F8, as the strain varies from -10% to 8%, a series of electronic and magnetic phase transitions of AFS-AM-FH-AFS will occur. These tunable magnetic and electronic properties of 2D halides originate from the competition between AFM direct nearest-neighbor d-d exchange and FM superexchange via halogen p states, which leads to a variety of magnetic states. The explored controllable magnetic properties, electronic properties and the high Curie temperature render the 2D V3I8 monolayer a promising candidate for applications in magnetic logic devices and strain sensor devices.

7.
Med Sci Monit ; 25: 21-31, 2019 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-30599132

RESUMO

BACKGROUND Rho GTPase activating protein (RhoGAPs) is an important negative regulator of the Rho signaling pathway that is involved in tumorigenesis in liver, colon, and renal cancer. However, the mechanism by which Rho GTPase activating protein 24 (ARHGAP24) regulates cell invasion and migration of lung cancer has not been fully explained. MATERIAL AND METHODS In this study, ARHGAP24 expression in lung cancer tissues and cell lines was measured by immunohistochemical and Western blot analysis. Transwell or wound healing analysis was performed to detect the cell migration and invasion of ARHGAP24 modulated A549 and NCI-H1975 cells with ß-catenin inhibitor XAV-939 (10 µM) treatment, and the expression of MMP9, VEGF, and ß-catenin protein was measured by Western blotting. RESULTS Our results showed that ARHGAP24 expression was downregulated in lung cancer tissues and cell lines. pLVX-Puro-ARHGAP24 transfection in A549 cells significantly inhibited cell invasion and migration, along with increased E-cadherin and decreased MMP9, VEGF, Vimentin, and ß-catenin protein expression. pLKO.1-ARHGAP24-shRNA transfection in NCI-H1975 cells significantly promoted cell invasion and migration, accompanied with decreased E-cadherin and increased MMP9, VEGF, and ß-catenin protein expression. Moreover, NCI-H1975 cells with XAV-939 treatment showed decreased cell invasion and migration when compared with pLKO.1-ARHGAP24-shRNA transfection. ARHGAP24 silencing promoted the transcriptional activity of ß-catenin in NCI-H1975 cells. CONCLUSIONS Our findings indicate that ARHGAP24 silencing promotes lung cancer cell migration and invasion through activating ß-catenin signaling.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , beta Catenina/metabolismo , Células A549 , Adulto , Idoso , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Feminino , Proteínas Ativadoras de GTPase/genética , Humanos , Neoplasias Pulmonares/genética , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Transdução de Sinais
8.
Tumour Biol ; 37(9): 12181-12191, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27225290

RESUMO

Transmembrane protein 45B (TMEM45B) is a member of TMEMs. Altered expression of TMEMs is frequently observed in a variety of human cancers, but the expression and functional roles of TMEM45B in lung cancer is not reported. In the present study, levels of mRNA expression of TMEM45B in lung cancer tissues were assessed using re-analyzing expression data of The Cancer Genome Atlas (TCGA) lung cancer cohort and real-time PCR analysis on our own cohort. Lung cancer cells, A549 and NCI-H1975, infected with TMEM45B short hairpin RNA were examined in cell proliferation, cell cycle, cell apoptosis, wound-healing, and cell invasion assays as well as mouse xenograft models. Here, we demonstrated that TMEM45B was overexpressed in lung cancer and its expression correlated with overall survival of patients. In addition, silencing of TMEM45B expression reduced cell proliferation in vitro and in vivo, induced cell cycle arrest and cell apoptosis, and blocked cell migration and invasion. Moreover, knockdown of TMEM45B significantly suppressed G1/S transition, induced cell apoptosis, and inhibited cell invasion via regulating the expression of cell cycle-related proteins (CDK2, CDC25A, and PCNA), cell apoptosis-related proteins (Bcl2, Bax, and Cleaved Caspase 3), and metastasis-related proteins (MMP-9, Twist, and Snail), respectively. Thus, TMEM45B is a potential prognostic marker and cancer-selective therapeutic target in lung cancer.


Assuntos
Transformação Celular Neoplásica/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Proteínas de Membrana/genética , Regulação para Cima , Células A549 , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose/genética , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Western Blotting , Pontos de Checagem do Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Estudos de Coortes , Feminino , Células HEK293 , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sobrevida
9.
Biochem Biophys Res Commun ; 460(3): 703-8, 2015 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-25817787

RESUMO

The histone ubiquitin hydrolase ubiquitin-specific protease 22 (USP22) is an epigenetic modifier and an oncogene that is upregulated in many types of cancer. In non-small cell lung cancer (NSCLC), aberrant expression of USP22 is a predictor of poor survival, as is high expression of cyclooxygenase-2 (COX-2). Despite its oncogenic role, few substrates of USP22 have been identified and its mechanism of action in cancer remains unclear. Here, we identified COX-2 as a direct substrate of USP22 and showed that its levels are modulated by USP22 mediated deubiquitination. Silencing of USP22 downregulated COX-2, decreased its half-life, and inhibited lung carcinoma cell proliferation by directly interacting with and modulating the stability and activity of COX-2 through the regulation of its ubiquitination status. The findings of the present study suggest a potential mechanism underlying the oncogenic role of USP22 mediated by the modulation of the stability and activity of COX-2.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/enzimologia , Ciclo-Oxigenase 2/metabolismo , Neoplasias Pulmonares/enzimologia , Oncogenes , Tioléster Hidrolases/fisiologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Neoplasias Pulmonares/patologia , Estabilidade Proteica , Especificidade por Substrato , Tioléster Hidrolases/genética , Tioléster Hidrolases/metabolismo , Ubiquitina Tiolesterase
10.
Tumour Biol ; 36(7): 5031-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25663460

RESUMO

MicroRNAs (miRNAs) play critical roles in cancer development and progression. In this study, we examined the roles and molecular mechanisms of miR-342-3p in human non-small cell lung cancer (NSCLC). The results showed that miR-342-3p is downregulated in NSCLC cell lines and tissues, and its overexpression induces significant inhibition of NSCLC cell proliferation, invasion, and tumor growth in nude mice. In addition, miR-342-3p repressed RAP2B expression through interactions with its 3'-UTR region. Restoration of RAP2B expression reversed miR-342-3p-mediated inhibitory activity in NSCLC cells. Finally, analyses of miR-342-3p and RAP2B levels in NSCLCs revealed that miR-342-3p inversely correlated with RAP2B mRNA expression. Our collective findings provide preliminary evidence that miR-342-3p acts as a tumor suppressor in NSCLC through repression of RAP2B.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Proliferação de Células/genética , MicroRNAs/genética , Invasividade Neoplásica/genética , Proteínas rap de Ligação ao GTP/biossíntese , Regiões 3' não Traduzidas/genética , Animais , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas rap de Ligação ao GTP/genética
11.
Int J Mol Sci ; 16(1): 307-20, 2014 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-25547493

RESUMO

Increasing evidence suggests that ubiquitin-specific protease 22 (USP22) has great clinicopathologic significance in oncology. In this study, we investigated the role of USP22 in human NSCLC tumorigenesis along with the underlying mechanisms of action. First, we determined the expression of USP22 in human NSCLC, as well as normal tissues and cell lines. We then studied the effects of USP22 silencing by shRNA on NSCLC cell growth in vitro and tumorigenesis in vivo, along with the effect on the p53 pathway. We found that USP22 is overexpressed in human NSCLC tissues and cell lines. USP22 silencing by shRNA inhibits proliferation, induces apoptosis and arrests cells at the G0/G1 phases in NSCLC cells and curbs human NSCLC tumor growth in a mouse xenograft model. Additionally, USP22 silencing downregulates MDMX protein expression and activates the p53 pathway. Our co-immunoprecipitation analysis shows that USP22 interacts with MDMX in NSCLC cells. Furthermore, MDMX silencing leads to growth arrest and apoptosis in NSCLC cells, and over-expression of MDMX reverses the USP22 silencing-induced effects. Taken together, our results suggest that USP22 promotes NSCLC tumorigenesis in vitro and in vivo through MDMX upregulation and subsequent p53 inhibition. USP22 may represent a novel target for NSCLC treatment.


Assuntos
Carcinogênese/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Tioléster Hidrolases/metabolismo , Animais , Apoptose , Carcinogênese/patologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Nucleares/genética , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Tioléster Hidrolases/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina Tiolesterase , Regulação para Cima
12.
Spectrochim Acta A Mol Biomol Spectrosc ; 316: 124342, 2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-38676981

RESUMO

Two spirobifluene-based fluorescent probes SPF1 and SPF2, were designed and synthesized. The probes displayed "turn-on" fluorescence response for Cysteine. One of the challenges in developing a Cysteine probe is to secure high selectivity. SPF1/SPF2 can discriminate Cysteine from GSH as well as Hcy, and showed high substrate selectivity. The detection limit of SPF1 is 36 nM, which is excellent comparing with other optical sensors for Cysteine. The sensing mechanism of SPF1/SPF2 was verified by experimental data and theoretical calculations. There was a good linear relationship between the fluorescence intensity of SPF1/SPF2 and the concentration of Cysteine. The MTT tests indicated that SPF1/SPF2 had low cytotoxicity and good biocompatibility. Theoretical calculations demonstrated that SPF1, SPF2, and their related reaction products with Cysteine exhibited good two-photon absorption properties. Finally, SPF1/SPF2 had been successfully applied to the imaging of Cysteine in living cells under two-photon excitation.


Assuntos
Cisteína , Corantes Fluorescentes , Compostos de Espiro , Corantes Fluorescentes/química , Corantes Fluorescentes/síntese química , Cisteína/análise , Humanos , Compostos de Espiro/química , Células HeLa , Imagem Óptica/métodos , Limite de Detecção , Fótons , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Espectrometria de Fluorescência/métodos
13.
Spectrochim Acta A Mol Biomol Spectrosc ; 317: 124461, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-38759393

RESUMO

Esophageal cancer is one of the leading causes of cancer-related deaths worldwide. The identification of residual tumor tissues in the surgical margin of esophageal cancer is essential for the treatment and prognosis of cancer patients. But the current diagnostic methods, either pathological frozen section or paraffin section examination, are laborious, time-consuming, and inconvenient. Raman spectroscopy is a label-free and non-invasive analytical technique that provides molecular information with high specificity. Here, we report the use of a portable Raman system and machine learning algorithms to achieve accurate diagnosis of esophageal tumor tissue in surgically resected specimens. We tested five machine learning-based classification methods, including k-Nearest Neighbors, Adaptive Boosting, Random Forest, Principal Component Analysis-Linear Discriminant Analysis, and Support Vector Machine (SVM). Among them, SVM shows the highest accuracy (88.61 %) in classifying the esophageal tumor and normal tissues. The portable Raman system demonstrates robust measurements with an acceptable focal plane shift of up to 3 mm, which enables large-area Raman mapping on resected tissues. Based on this, we finally achieve successful Raman visualization of tumor boundaries on surgical margin specimens, and the Raman measurement time is less than 5 min. This work provides a robust, convenient, accurate, and cost-effective tool for the diagnosis of esophageal cancer tumors, advancing toward Raman-based clinical intraoperative applications.


Assuntos
Neoplasias Esofágicas , Aprendizado de Máquina , Análise Espectral Raman , Máquina de Vetores de Suporte , Análise Espectral Raman/métodos , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/patologia , Humanos , Análise Discriminante , Análise de Componente Principal , Algoritmos
14.
Tumour Biol ; 34(6): 3893-9, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23893384

RESUMO

Dysregulation of hedgehog signaling has been involved in esophageal squamous cell carcinoma (ESCC) by the mechanisms that are not fully understood. The receptor for activated protein kinase C (RACK1) is involved in the progression of multiple cancers. However, its expression and function in ESCC have not been investigated. Here, we found that the expression of RACK1 was upregulated in ESCC clinical samples. Moreover, over-expression of RACK1 in ESCC cells promoted cell proliferation and migration, while downregulation of RACK1 impaired the proliferation and migration of ESCC cells in vitro and in vivo. Mechanistically, RACK1 promoted the proliferation and migration of ESCC cells by activating hedgehog signaling. Taken together, our study suggested RACK1 might be an important therapeutic target in ESCC.


Assuntos
Carcinoma de Células Escamosas/genética , Movimento Celular/genética , Neoplasias Esofágicas/genética , Proteínas de Ligação ao GTP/genética , Proteínas de Neoplasias/genética , Receptores de Superfície Celular/genética , Animais , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Proteínas de Ligação ao GTP/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Luciferases/genética , Luciferases/metabolismo , Camundongos , Camundongos Nus , Proteínas de Neoplasias/metabolismo , Interferência de RNA , Receptores de Quinase C Ativada , Receptores de Superfície Celular/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
15.
Life Sci ; 323: 121592, 2023 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-36934972

RESUMO

AIMS: DEAD-box helicase 1 (DDX1) has oncogenic properties in several human cancers. However, the clinical significance and biological role of DDX1 in non-small cell lung cancer (NSCLC) remain elusive. Here, we examined the chemotherapeutic relevance of DDX1 in NSCLC. MAIN METHODS: We used the UALCAN database, Western blot analysis, and immunohistochemical and RT-qPCR assays to assess DDX1 expression in NSCLC cell lines (H1650 and A549) and patient tissues. The role of DDX1 in the chemosensitivity of NSCLC cells and the underlying mechanisms were determined using colony formation, CCK-8, flow cytometry, wound healing, Transwell, tumor sphere formation, and immunostaining assays, together with a xenograft tumor model in nude mice. KEY FINDINGS: Our study revealed that DDX1 was overexpressed in NSCLC cell lines and tissues. We further found that depleting DDX1 increased the sensitivity of NSCLC cells to the chemotherapy drug cisplatin, increased cell apoptosis, and inhibited cell migration and invasion. Co-immunoprecipitation assays revealed that DDX1 bound to ADAR1, and increased ADAR1 protein expression. Furthermore, we found that ADAR1 mediated cancer-promoting effects, independent of deaminase activity, by binding to RAC3 mRNA. Our findings not only show that DDX1 mediates chemosensitivity to cisplatin via the ADAR1/RAC3 axis but also highlight the importance of ADARs as essential RNA-binding proteins for cell homeostasis, as well as cancer progression. SIGNIFICANCE: Our results suggest that DDX1 plays an important role in the development and progression of human NSCLC and that DDX1 may serve as a therapeutic target in NSCLC patients.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , MicroRNAs , Animais , Humanos , Camundongos , Adenosina Desaminase/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Cisplatino/farmacologia , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Neoplasias Pulmonares/patologia , Camundongos Nus , MicroRNAs/genética , Células-Tronco Neoplásicas/metabolismo
16.
Quant Imaging Med Surg ; 13(10): 7052-7064, 2023 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-37869325

RESUMO

Background: The early surgical intervention for pulmonary ground-glass nodules (GGNs) has become increasingly important, but accurate identification of these nodules during thoracoscopic surgery poses challenges due to the need for sublobar resections and reliance on visual and tactile perception alone. The prognosis of the procedure is closely tied to the use of precise positioning technology. Thus, it is crucial to develop an accurate positioning technology that can improve patient prognosis. Methods: Clinical data from the cardiothoracic department of a tertiary hospital in Shanghai were collected and analyzed between January 2020 and December 2021. The patients were categorized into 2 groups: an indocyanine green (ICG) group and a hook-wire group. Outcome measures including success rate, complications, procedure time, localization-related pain, and interval time were assessed. Adverse events and reactions were reported and compared between the 2 groups. Results: A total of 62 patients (17 males and 45 females, aged 50.5±13.2 years) were in the ICG group, while 66 patients (23 males and 43 females, aged 48.4±12.9 years) were localized in the hook-wire group. The success rate was comparable between the 2 groups. However, the ICG group showed significant advantages over the hook-wire group in terms of procedure time (22.6±4.4 vs. 24.1±4.9 min; P=0.012), localization-related pain (P<0.001), and interval time [median and interquartile range (IQR): 3 (0.7, 104.9) vs. 1.2 (0.5, 3.3) h; P<0.001]. In the ICG group, there were 11 cases of pneumothorax, 4 cases of hemothorax, and 2 cases of ICG diffusion. In the hook-wire group, there were 24 cases of pneumothorax, 25 cases of hemothorax, and 2 cases of dislodgement. The ICG group had fewer complications, including pneumothorax (P=0.018) and hemothorax (P=0.007), compared to the hook-wire group. Conclusions: Computed tomography (CT)-guided intrapulmonary injection of ICG for preoperative localization of peripheral pulmonary GGNs is a practical and safe technique. It offers advantages in terms of reduced procedure time, localization-related pain, and interval time compared to the hook-wire method. Moreover, the ICG technique results in fewer complications, making it a valuable preoperative localization technique worthy of popularization.

17.
Exp Eye Res ; 97(1): 154-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21933673

RESUMO

Conbercept(KH902), a recombinant fusion protein in clinical trial II/III, shows good potential to treat the neovascular age-related macular degeneration (AMD). This investigation evaluated its ocular pharmacokinetics and pharmacodynamic profile in rabbits following intravitreal administration (IVT). Rabbits (n = 120) received single bilateral conbercept IVT administration or single IV administration. Conbercept concentrations in ocular tissues and serum were measured after dosing. VEGF concentration was also measured simultaneously. The results showed that conbercept rapidly distributed from vitreous into targeted tissues and lasted over 81 days. Clearance in ocular tissues was parallel and exhibited a terminal half of 2.5-4.2 days. The drug exposure in the retina was 1/4 to 1/5 of that in vitreous. Serum conbercept concentrations after IVT dosing were low and bioavailability was approximately 44%. And single intravitreal injection induced that ocular VEGF concentration declined over 60 days and serum VEGF concentration decreased for a short time but rebounded to higher level than baseline later. All these indicated conbercept good pharmacokinetic profile in rabbits, with good ocular tropism and systemic tolerance. Combined with the efficacy data from our earlier in vitro and in vivo studies, it should have a promising clinical application for AMD treatment.


Assuntos
Inibidores da Angiogênese/farmacocinética , Proteínas Recombinantes de Fusão/farmacocinética , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Corpo Vítreo/metabolismo , Inibidores da Angiogênese/imunologia , Animais , Autoanticorpos/sangue , Disponibilidade Biológica , Ensaio de Imunoadsorção Enzimática , Meia-Vida , Injeções Intravenosas , Injeções Intravítreas , Masculino , Coelhos , Proteínas Recombinantes de Fusão/imunologia , Distribuição Tecidual
18.
Guang Pu Xue Yu Guang Pu Fen Xi ; 32(8): 2147-51, 2012 Aug.
Artigo em Zh | MEDLINE | ID: mdl-23156770

RESUMO

Self-assembled monolayer film of gold nanoparticles (55 nm) was formed at air-water interface by the driving force of wettability-shift of gold nanoparticles from hydrophilic property to hydrophobic property when encapsulated with 1-dodecanethiol. SEM image shows that the structure of the surface is nearly monolayer with closed arrays of uniform particle size when the film is transferred onto Si wafer. The substrate can be used for SERS substrate to realize semiquantitative analysis of melamine and the detection limit can reach l x 10(-9) g x L(-1). This SERS substrate is of wide application, not only for melamine but also for nonpolar molecule such as polycyclic aromatic hydrocarbons.


Assuntos
Ouro , Nanopartículas Metálicas , Triazinas/análise , Interações Hidrofóbicas e Hidrofílicas , Tamanho da Partícula , Compostos de Sulfidrila , Propriedades de Superfície , Água , Molhabilidade
19.
Laryngoscope Investig Otolaryngol ; 7(2): 425-436, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35434335

RESUMO

Background: Increasing evidence demonstrated the important roles of circular RNAs (circRNAs) in human cancer progression, including oral squamous cell carcinoma (OSCC). The study intentions were to explore the role and molecular mechanism of hsa_circ_0004390 (circLPAR3) in OSCC progression. Methods: Expression of circLPAR3 in collected samples and cultured cell lines was detected with real-time quantitative reverse transcription-polymerase chain reaction (RT-qPCR). Loss-of-function experiments were performed to determine the effect of circLPAR3 silencing on OSCC cell proliferation, migration, invasion, apoptosis, angiopoiesis, and glycolysis. The sponge function of circLPAR3 was predicted by bioinformatics analysis and validated by the dual-luciferase reporter and RNA pull-down assays. In vivo experiments were conducted to validate the function of circLPAR3. Results: A marked increase in circLPAR3 expression was observed in OSCC samples and cell lines. Furthermore, circLPAR3 could distinguish OSCC samples from paired non-tumor samples, and patients with high circLPAR3 expression had a poor prognosis. Furthermore, circLPAR3 inhibition decreased OSCC growth in xenograft mouse models. Moreover, circLPAR3 silencing repressed cell proliferation, migration, invasion, angiopoiesis, glycolysis, and induced cell apoptosis in OSCC cells in vitro. Mechanically, circLPAR3 sponged miR-144-3p to prohibit the inhibiting effect of miR-144-3p on LPCAT1, thus promoting OSCC progression. Conclusion: CircLPAR3 exerted a tumor-promoting effect on OSCC growth through elevating LPCAT1 expression via functioning as a miR-144-3p sponge. This study supports the possible role of circLPAR3 in the diagnosis, prognosis, and treatment of OSCC.

20.
Front Oncol ; 12: 985827, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36818671

RESUMO

Non-small cell lung cancer (NSCLC) is the most common pathological type of lung cancer, which is a severer threaten to human health because of its extremely high morbidity and mortality. In this study, the role of Notchless homolog 1 (NLE1) in the development of NSCLC was investigated and the underlying mechanism was explored. The outcomes showed that NLE1 expression is significantly higher in tumor tissues than normal tissues, and is correlated with the pathological stage. The regulation of NSCLC development by NLE1 was also visualized by the in vitro and in vivo loss-of-function studies, which indicated the inhibition of cell growth and migration, as well as enhancement of cell apoptosis on condition of NLE1 knockdown. As for the mechanism, it was demonstrated that NLE1 may execute its tumor-regulating function through activating E2F1-mediated transcription of CDK1, and PI3K/Akt signaling pathway was also supposed as a downstream of NLE1 in the regulation of NSCLC. Both CDK1 overexpression and treatment of Akt pathway activator could reverse the NLE1 knockdown induced NSCLC inhibition to some extent. In conclusion, this study identified NLE1 as a novel tumor promotor in the development and progression of NSCLC, which may be a potential therapeutic target in the treatment of NSCLC.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA