Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cancer Gene Ther ; 30(12): 1636-1648, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37726387

RESUMO

Glioblastoma (GBM) is a highly malignant type of brain tumor with limited treatment options. Recent research has focused on epigenetic regulatory factors, such as Enhancer of Zeste Homolog 2 (EZH2), which plays a role in gene expression through epigenetic modifications. EZH2 inhibitors have been developed as potential therapeutic agents for GBM, but resistance to these inhibitors remains a considerable challenge. This study aimed to investigate the role of ribosomal S6 protein kinase 4 (RSK4) in GBM and its association with resistance to EZH2 inhibitors. We first induced drug resistance in primary GBM cell lines by treatment with an EZH2 inhibitor and observed increases in the expression of stemness markers associated with glioblastoma stem cells (GSCs) in the drug-resistant cells. We also found high expression of RSK4 in GBM patient samples and identified the correlation of high RSK4 expression with poor prognosis and GSC marker expression. Further experiments showed that knocking down RSK4 in drug-resistant GBM cells restored their sensitivity to EZH2 inhibitors and decreased the expression of GSC markers, thus reducing their self-renewal capacity. From a mechanistic perspective, we discovered that RSK4 directly phosphorylates EZH2, activating the EZH2/STAT3 pathway and promoting resistance to EZH2 inhibitors in GBM. We also found that combining EZH2 inhibitors with an RSK4 inhibitor called BI-D1870 had better inhibitory effects on GBM occurrence and progression in both in vitro and in vivo experiments. In conclusion, this study demonstrates that RSK4 enhances cancer stemness and mediates resistance to EZH2 inhibitors in GBM. Combination treatment with EZH2 inhibitors and RSK4 inhibitors is a promising potential therapeutic strategy for GBM. Collectively, our results strongly demonstrate that RSK4 regulates the EZH2/STAT3 pathway to promote GSC maintenance and EZH2i resistance in a PRC2-independent manner, indicating that RSK4 is a promising therapeutic target for GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Epigênese Genética , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Quinases/farmacologia , Linhagem Celular Tumoral , Células-Tronco Neoplásicas/patologia , Regulação Neoplásica da Expressão Gênica , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo
2.
Pathol Res Pract ; 240: 154236, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36434855

RESUMO

BACKGROUND: PRKCH is a member of the PKC family with the potential to regulate cell proliferation and differentiation. Glioma is the most common primary tumor of the central nervous system, with a high recurrence rate and poor prognosis. Recent studies have demonstrated that PRKCH can promote the proliferation of glioma cells. The purpose of this study was to investigate the promoting effect and possible mechanism of PRKCH on glioma development. METHODS: Tumor tissue and paracancerous tissue were collected from 160 glioma patients treated at the General Hospital of Northern Theater Command. The expression level of PRKCH was detected by immunohistochemistry and immunoblotting. Univariate/multivariate analysis and log-rank analysis, as well as prognosis and survival analysis, were performed using SPSS26 software. The PRKCH overexpression model was constructed in vitro to study the effect of PRKCH expression on the characteristics of glioma stem cells. RESULTS: The expression of PRKCH in glioma tissues was higher than that in adjacent tissues. PRKCH expression level is an independent prognostic factor in glioma patients, promoting poor prognosis and shorter survival in glioma patients. Furthermore, overexpression of PRKCH in glioma stem cells significantly increased stem cell properties and enhanced cell viability. Downregulation of PRKCH significantly inhibited the progression of glioma stem cells. CONCLUSION: PRKCH promotes the development of gliomas and may be a therapeutic target for gliomas.


Assuntos
Transformação Celular Neoplásica , Glioma , Humanos , Carcinogênese , Diferenciação Celular , Células-Tronco Neoplásicas
3.
Mol Pharm ; 19(11): 4382-4389, 2022 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-36268880

RESUMO

Programmed death-1 (PD-1) and programmed death ligand-1 (PD-L1), the research focus in immune checkpoint regulation, play an important role in tumor immunotherapy. Inhibitors of this pathway are also the focus of tumor immunotherapy research. The PD-1/PD-L1 pathway can be blocked by selective binding to PD-L1. Clinical trials have been conducted in a variety of patients with advanced solid tumors. CS1001 is a high-affinity humanized full-length anti-PD-L1 monoclonal antibody with great clinical significance. We constructed a PD-L1-targeted radioactive molecular probe, 124/125I-labeled full-length antibody CS1001, and evaluated its binding specificity and targeting ability to PD-L1 in tumor cells and tumor models. Additionally, a comparison study with 68Ga-WL12, a PD-L1 targeting peptide, was conducted. The binding potency of 125I-CS1001 to human PD-L1 was evaluated by enzyme-linked immunosorbent assay (ELISA), and the Kd value was 52.1 ± 19.3 nM. The cellular uptake of 125I-CS1001 was examined in Chinese hamster ovary cells (CHO) and CHO expressing human PD-L1 (CHO-hPD-L1). At 2 h, the uptake values of 125I-CS1001 in CHO-hPD-L1 without blocking and in the presence of 0.1 mg non-radiolabeled CS1001 were 3.60 ± 0.08 and 0.09 ± 0.005 (%AD/2 × 105 cells, p < 0.001). Micro-PET imaging was performed between 8 to 192 h after injection of 124I-CS1001 into normal KM mice and CHO-hPD-L1 and HeLa tumor models. The standard uptake value (SUV) of relevant organs in PET images was calculated by drawing regions of interest (ROI). SUVmean of CHO-hPD-L1 tumors was significantly higher than that of HeLa tumors at 48 h (1.98 ± 0.04 vs 0.73 ± 0.14, p = 0.005). The SUVmean of 124I-CS1001 in CHO-hPD-L1 tumors at 48 h was higher than that of 68Ga-WL12 in CHO-hPD-L1 tumors at 0.5 h (1.98 ± 0.04 vs 1.09 ± 0.1 SUVmean, p = 0.007). In conclusion, this work provides a new method for monitoring and evaluating the in vivo expression of PD-L1 in tumors.


Assuntos
Iodo , Neoplasias , Cricetinae , Animais , Humanos , Camundongos , Células CHO , Radioisótopos de Gálio/química , Receptor de Morte Celular Programada 1 , Cricetulus , Anticorpos Monoclonais , Peptídeos/química , Tomografia por Emissão de Pósitrons/métodos , Linhagem Celular Tumoral
4.
J Nucl Med ; 63(4): 536-542, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34326125

RESUMO

68Ga-NOTA-WL12 is a peptide-based PET imaging agent. We conducted a first-in-human study of 68Ga-NOTA-WL12 for PET to study the in vivo biodistribution, metabolism, radiation dosimetry, safety, and potential for quantifying programmed death ligand-1 (PD-L1) expression levels in patients with advanced non-small cell lung cancer (NSCLC). Methods: In vitro assessment of the PD-L1 expression and cellular uptake of 68Ga-NOTA-WL12 was performed, followed by in vivo evaluation of 68Ga-NOTA-WL12 uptake in mouse models with tumors. Nine patients with NSCLC with lesions expressing PD-L1 were enrolled and monitored for adverse events during the study. 68Ga-NOTA-WL12 and paired 18F-FDG PET/CT imaging were performed. Uptake (SUV, SUL [SUVlean], and kBq/mL) values of tumors and normal organs were obtained. Radiopharmaceutical biodistribution, radiation dosimetry, and the relationship of tumor uptake to PD-L1 expression were evaluated. Follow-up 18F-FDG PET/CT was performed in patients who had undergone treatment with a combination of pembrolizumab with chemotherapy. Results:68Ga-NOTA-WL12 exhibited PD-L1-specific uptake in vitro and in PD-L1-positive tumors in vivo. 68Ga-NOTA-WL12 PET imaging proved safe with acceptable radiation dosimetry. Physiologic tracer uptake was mainly visible in the liver, spleen, small intestine, and kidney. Tumors were clearly visible, particularly in the lungs, with a tumor-to-lung ratio of 4.45 ± 1.89 at 1 h. One hour was a suitable time point for image acquisition because no significant differences were noted in tumor-to-background ratios between 1 and 2 h. A strong, positive correlation was found between tumor uptake (SUVpeak) and PD-L1 immunohistochemistry results (r = 0.9349; P = 0.002). 68Ga-NOTA-WL12 and 18F-FDG PET studies suggest that PD-L1 PET before therapy may indicate the therapeutic efficacy of pembrolizumab plus chemotherapy combination treatment. Conclusion: Our first-in-human findings demonstrate the safety and feasibility of 68Ga-NOTA-WL12 for noninvasive, in vivo detection of tumor PD-L1 expression levels, indicating potential benefits for clinical PD-L1 therapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Animais , Antígeno B7-H1/metabolismo , Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Humanos , Neoplasias Pulmonares/diagnóstico por imagem , Camundongos , Peptídeos/metabolismo , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Distribuição Tecidual
5.
Eur J Nucl Med Mol Imaging ; 48(13): 4508-4516, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34170361

RESUMO

PURPOSE: Develop a 64Cu labeled radiopharmaceutical targeting prostate specific membrane antigen (PSMA) and investigate its application for prostate cancer imaging. METHODS: 64Cu-PSMA-BCH was prepared and investigated for stability, PSMA specificity, and micro-PET imaging. With the approval of Ethics Committee of Beijing Cancer Hospital (No. 2017KT97), PET/CT imaging in 4 patients with suspected prostate cancer was performed and the radiation dosimetry was estimated. Then, PSMA PET-ultrasound image-guided biopsies were performed on 3 patients and the fine needle aspirates were further performed for autoradiography and immunohistochemistry analysis. RESULTS: 64Cu-PSMA-BCH was prepared with high radiochemical yield and stability. In vivo study showed higher uptake in PSMA ( +) 22Rv1 cells than PSMA ( -) PC-3 cells (5.59 ± 0.36 and 1.97 ± 0.22 IA%/106 cells at 1 h). It accumulated in 22Rv1 tumor with increasing radioactivity uptake and T/N ratios from 1 to 24 h post-injection. In patients with suspected prostate cancer, SUVmax and T/N ratios increased within 24 h post-injection. Compared with image at 1 h post-injection, more tumor lesions were detected at 6 h and 24 h post-injection. The human organ radiation dosimetry showed gallbladder wall was most critical, liver and kidneys were followed, and the whole-body effective dose was 0.0292 mSv/MBq. Two fine needle aspirates obtained by PET-ultrasound-guided targeted biopsy showed high radioactive signal by autoradiography, with 100% PSMA expression in cytoplasm and 30% expression in nucleus. CONCLUSION: 64Cu-PSMA-BCH was PSMA specific and showed high stability in vivo with lower uptake in liver than 64Cu-PSMA-617. Biodistribution in mice and PCa patients showed similar profile compared with other PSMA ligands and it was safe with moderate effective dosimetry. The increased tumor uptake and T/N ratios by delayed imaging may facilitate the detection of small lesions and guiding targeted biopsies.


Assuntos
Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Neoplasias da Próstata , Amidas , Animais , Hidrocarbonetos Aromáticos com Pontes , Humanos , Masculino , Camundongos , Tomografia por Emissão de Pósitrons , Neoplasias da Próstata/diagnóstico por imagem , Distribuição Tecidual
6.
Adv Sci (Weinh) ; 8(16): e2100965, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34174177

RESUMO

Rapid progress has been made to identify and study the causative agent leading to coronavirus disease 2019 (COVID-19) but many questions including who is most susceptible and what determines severity remain unanswered. Angiotensin-converting enzyme 2 (ACE2) is a key factor in the infection process of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). In this study, molecularly specific positron emission tomography imaging agents for targeting ACE2 are first developed, and these novel agents are evaluated in vitro, in preclinical model systems, and in a first-in-human translational ACE2 imaging of healthy volunteers and a SARS-CoV-2 recovered patient (NCT04422457). ACE2 expression levels in different organs in live subjects are quantitatively delineated and observable differences are measured in the patient recovered from COVID-19. Surprising sites of uptake in the breast, reproductive system and very low uptake in pulmonary tissues are reported. This novel method can add a unique tool to facilitate SARS-CoV-2 related research and improve understanding of this enigmatic disease. Molecular imaging provides quantitative annotation of ACE2, the SARS-CoV-2 entry receptor, to noninvasively monitor organs impacted by the COVID-19.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/metabolismo , COVID-19/virologia , Peptídeos/farmacocinética , SARS-CoV-2/metabolismo , Animais , COVID-19/patologia , Células Cultivadas , Feminino , Radioisótopos de Gálio/farmacocinética , Humanos , Masculino , Camundongos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Ligação Proteica , SARS-CoV-2/isolamento & purificação , SARS-CoV-2/patogenicidade , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Bioorg Med Chem Lett ; 40: 127901, 2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-33705912

RESUMO

As an indicative biomarker for immunotherapy, PD-L1 plays an important role in the clinical decision-making of the immune checkpoint blockade therapy. PET imaging through radiotracer can real-timely, quantitatively, and non-invasively assess the expression of PD-L1 in tumors. Here, we reported a copper-64 radiolabeled NOTA-WL12, 64Cu-NOTA-WL12, and preliminarily evaluated its application in non-invasively detecting the PD-L1 expression.64Cu-NOTA-WL12 was produced with high radiochemical yield (>90%), radiochemical purity (>98%), and specific activity (20 MBq/nmol). 64Cu-NOTA-WL12 showed high in vitro stability and high binding affinity to the PD-L1 (KD ≈ 3.012 nM). The micro-positron emission tomography/computerized tomography (micro-PET/CT) imaging indicated that 64Cu-NOTA-WL12 was specifically accumulated in the tumor with PD-L1 expression. All results demonstrated that 64Cu-NOTA-WL12 holds great potential for noninvasive evaluation of PD-L1 expression levels.


Assuntos
Antígeno B7-H1/análise , Antígeno B7-H1/genética , Radioisótopos de Cobre/economia , Peptídeos/química , Compostos Radiofarmacêuticos/química , Animais , Células CHO , Cricetulus , Expressão Gênica , Humanos , Neoplasias Experimentais , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Ligação Proteica , Coloração e Rotulagem , Relação Estrutura-Atividade
8.
Front Oncol ; 11: 651005, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33777814

RESUMO

This study aims to further explore dynamic 68Ga-FAPI-04 PET/CT imaging of healthy Chinese subjects and lung cancer patients. Moreover, the variability of 68Ga-FAPI-04 uptake in normal organs was measured to provide a basis for analyzing its biological distribution, interpreting auxiliary images, determining the reliability of image quantification, and monitoring treatment. Six patients (3 subjects without tumors and 3 lung cancer patients) separately underwent 68Ga-FAPI-04 and 2-[18F]FDG PET/CT imaging within 1 week. The biodistribution and internal radiation dosimetry were reported and compared with data previously obtained from Caucasian patients. Moreover, the mean SUV (standardized uptake value) was normalized to body mass or to lean body mass (SUL), and the coefficients of variation (CVs) were calculated and compared for each volume of interest. The average whole-body effective dose was calculated to be 1.27E-02 mSv/MBq, which was comparable with previously reported results of 68Ga-FAPI-04 probes. Furthermore, the SUVmean was slightly higher than the SULmean in most organs; however, the CV of the SULmean for most organs was higher than that of the SUVmean at later time points. In the liver, the CV of the SUVmean was lower (12.7%) than that of the SULmean and was similar to the CV for corresponding 2-[18F]FDG PET/CT value (11.8%). In addition, 68Ga-FAPI-04 PET/CT showed good efficacy for diagnosing lung cancer patients in this study. A comparison of the radiation dosimetry obtained before from a Caucasian population demonstrated no clinically significant differences between these two populations after 68Ga-FAPI-04 injection. The variability in most organs was slightly lower for SUVmean than for SULmean, suggesting that SUVmean may be the preferable parameter for quantifying images obtained with 68Ga-FAPI-04. In addition, 68Ga-FAPI-04 PET/CT imaging is expected to be a promising tool for diagnosing lung cancer.

9.
Bioorg Med Chem Lett ; 30(4): 126915, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31926788

RESUMO

Recently, we selected a novel anti-hPD-L1-specific HCAb named Nb6 with high affinity (EC50 = 0.65 ng/mL) for potential hPD-L1 targeted non-invasive PET imaging. In this research, Nb6 was conjugated with the bifunctional chelator NCS-Bz-NOTA ((2-[(4-Isothiocyanophenyl) methyl]-1,4,7-triazacy-clononane-1,4,7-triacetic acid)) and further labeled with radio-nuclide 64Cu. 64Cu-NOTA-Nb6 was prepared with over 95% labeling yield, over 99% radiochemical purity and 14-16 GBq/µmol specific activity after PD-10 column purification. It shows good stability in 0.01 M PBS and 5% HSA solutions. 64Cu-NOTA-Nb6 has a high binding affinity to 3.60 nM which was tested by humanlungadenocarcinoma A549 cell lines. Tumor lesion can be clearly observed from 20 h to 38 h by Micro-PET equipment after 64Cu-NOTA-Nb6 administration. The study revealed that 64Cu-NOTA-Nb6 has good lesion detection ability, high ratios between tumor and non-tumor signal and can specifically target A549 xenografted tumor model. Taken together of good stability, high binding affinity, and tumor detection ability, 64Cu labeled Nb6 is a promising radio-tracer in diagnosing of hPD-L1 overexpression tumor, supposed to monitor PD-L1overexpression tumor progression and guide targeted therapy with PET molecular imaging.


Assuntos
Anticorpos Monoclonais/química , Antígeno B7-H1/imunologia , Neoplasias Pulmonares/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/química , Animais , Anticorpos Monoclonais/imunologia , Linhagem Celular Tumoral , Radioisótopos de Cobre/química , Humanos , Marcação por Isótopo , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Front Mol Biosci ; 7: 585024, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33392253

RESUMO

Prostate-specific membrane antigen (PSMA) is an attractive target for the diagnosis and therapy of prostate cancer as it is specifically overexpressed in prostate cancer cells. Improving the circulation of radioligands in the blood is considered as an effective strategy that can improve tumor burden, which benefits detection of small lesions and improves the effect of PSMA radioligand therapy (PRLT). In this study, we introduced maleimidopropionic acid (MPA) to a PSMA-targeted tracer and developed Al18F-PSMA-CM, which targets human serum albumin (HSA) binding and PSMA. Al18F-PSMA-CM is evaluated in vitro and in vivo for stability, PSMA specificity, and biodistribution in 22Rv1 tumor-bearing mice. Al18F-PSMA-CM was prepared with a radiochemical purity of >99% and specific activity of 11.22-18.70 MBq/nmol. Al18F-PSMA-CM was stable in vitro and in vivo and prolonged circulation in blood with a binding ratio of 47 ± 3.2% and Kd value of 3.08 ± 0.45 nM to HSA. The uptake of Al18F-PSMA-CM in PSMA(+) 22Rv1 cells was increased in 2 h, and the uptake was blocked by a PSMA inhibitor, ZJ-43. The Kd value of Al18F-PSMA-CM to PSMA was 8.46 ± 0.24 nM. Al18F-PSMA-CM was accumulated in kidneys and 22Rv1 tumors [74.76 ± 15.42 and 6.16 ± 0.74 ID%/g at 2 h post injection (p.i.)], which were decreased by -80.0 and -84.3% when co-injected with ZJ-43. Al18F-PSMA-CM showed high PSMA specificity and accumulated in 22Rv1 tumors with increasing uptake in 4 h. MPA moiety showed the ability to prolong the half-life of tracers, and the MPA-conjugated tracer showed the potential to improve tumor uptake. MPA may be a choice to develop radiopharmaceuticals for PRLT of prostate cancer.

11.
Nanoscale ; 11(30): 14400-14409, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-31334537

RESUMO

Somatostatin receptor subtype 2 (SSTR2) is highly expressed in pulmonary neuroendocrine tumors, which account for approximately 25% of all lung cancers including small-cell lung cancer (SCLC). It is possible to establish SCLC-specific imaging agents for multimodal imaging to obtain tumor integrity information. Herein, we constructed novel multifunctional organic melanin nanoparticles (MNPs) as a carrier and surface-loaded somatostatin analog octreotide to produce a human small-cell lung cancer-targeted nanoprobe OCT-PEG-MNPs. MNPs have an excellent photoacoustic imaging (PAI) function and can be directly chelated with the magnetic resonance contrast agent Mn2+, and N-bromo succinimide (NBS) can be used as an oxidant to label the nanoparticles with the long half-life radionuclide 124I by an electrophilic substitution reaction. Therefore, (124I, Mn) OCT-PEG-MNPs can not only be used for PAI but also be used for positron emission tomography (PET) and magnetic resonance imaging (MRI). The NCI-H69 SCLC tumor xenograft model with high SSTR2 expression was constructed to evaluate the multimodal imaging ability of (124I, Mn) OCT-PEG-MNPs. This nanoprobe showed good imaging abilities in PAI, MRI and PET. The PA images showed that the photoacoustic signal in the NCI-H69 tumor site gradually increased with time, and the NCI-H69 xenograft showed a clear increase in the T1-weighted signal intensity after injection of Mn-OCT-PEG-MNPs at 24 h compared to that in the prescan. MicroPET and biodistribution studies showed that the uptake of NCI-H69 tumors (8.03 ± 0.37% ID g-1) was significantly higher than that in the control A549 model (3.35 ± 0.54% ID g-1) after injection of (124I, Mn) OCT-PEG-MNPs at 24 h. The (124I, Mn) OCT-PEG-MNPs were successfully applied to multimodal imaging in a small-cell lung cancer model with high SSTR2 expression. This nanoprobe may be considered for clinical trials since it combines the numerous advantages of organic nanoparticles.


Assuntos
Carcinoma de Células Pequenas/diagnóstico , Neoplasias Pulmonares/diagnóstico , Melaninas/química , Nanopartículas/química , Receptores de Somatostatina/química , Animais , Carcinoma de Células Pequenas/diagnóstico por imagem , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Radioisótopos do Iodo/química , Neoplasias Pulmonares/diagnóstico por imagem , Imageamento por Ressonância Magnética , Manganês/química , Melaninas/metabolismo , Camundongos , Camundongos Nus , Nanopartículas/toxicidade , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/química , Receptores de Somatostatina/metabolismo , Transplante Heterólogo
12.
Oncol Rep ; 40(2): 783-792, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29901114

RESUMO

Lung cancer is the most common cause of cancer­related deaths, and early diagnosis and targeted therapy are extremely important in the treatment of this disease. Sperm­associated antigen 4 (SPAG4) was recently found to be a novel cancer biomarker. In the present study, the expression of SPAG4 was revealed to be high in lung adenocarcinoma tissues as determined by western blotting and immunohistochemistry. SPAG4 knockdown by RNAi efficiently reduced the migration of the lung cancer A549 cells in vitro. Mechanistically, SPAG4 exerted its tumor promoting functions by interacting with Nesprin3 as determined by co­immunoprecipitation (Co­IP) and bimolecular fluorescence complementation (BiFC) techniques. In addition, immunofluorescence revealed that the level of SPAG4 in lung cancer cells could affect the location and expression of Nesprin3. Furthermore, silencing of Nesprin3 reduced the migration of A549 cells and we provided evidence to demonstrate that SPAG4 acted as a positive regulator of Nesprin3. The results revealed that SPAG4, in cooperation with Nesprin3, has a fundamental pathological function in the migration of lung carcinoma cells, and the SPAG4 gene may be useful for the clinical diagnosis and new treatment strategies in patients with lung cancer.


Assuntos
Biomarcadores Tumorais/genética , Proteínas de Transporte/metabolismo , Movimento Celular/genética , Neoplasias Pulmonares/metabolismo , Proteínas dos Microfilamentos/metabolismo , Células A549 , Adulto , Idoso , Linhagem Celular Tumoral , Feminino , Humanos , Imuno-Histoquímica/métodos , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Espermatozoides/metabolismo , Espermatozoides/patologia
13.
FASEB J ; 32(9): 5162-5173, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29694248

RESUMO

ADAR1 (adenosine deaminase acting on double-stranded RNA 1) is an RNA-editing enzyme that mediates adenosine-to-inosine RNA editing events, an important post-transcriptional modification mechanism that can alter the coding properties of mRNA or regulate microRNA biogenesis. ADAR1 also regulates the innate immune response. Here, we have demonstrated that ADAR1 expression increased in LPS-stimulated macrophages. Silencing ADAR1 by using small interfering RNA in macrophages resulted in the pronounced polarization of macrophages to M1, whereas ADAR1 overexpression promoted M2 polarization, which indicated that ADAR1 can inhibit macrophage hyperpolarization and prevent immune hyperactivity. The RNA-RNP immunoprecipitation binding assay demonstrated a direct interaction between ADAR1 and miR-21 precursor. Significant up-regulation in IL-10 and down-regulation in miR-21 were observed in ADAR1-overexpressing macrophages. We evaluated miR-21 target mRNAs and macrophage polarization signaling pathways and found that forkhead box protein O1 (Foxo1) was up-regulated in cells that overexpressed ADAR1. In a mouse allogeneic skin transplantation model, grafts in the ADAR1-overexpressed group survived longer and suffered less immune cell infiltration. In ADAR1-overexpressed recipients, splenic macrophages were significantly polarized to M2, and levels of sera IL-10 were markedly higher than those in the control group. In summary, ADAR1 modulates macrophage M2 polarization via the ADAR1-miR-21-Foxo1-IL-10 axis, thereby suppressing allogeneic graft rejection.-Li, J., Xie, J., Liu, S., Li, X., Zhang, D., Wang, X., Jiang, J., Hu, W., Zhang, Y., Jin, B., Zhuang, R., Yin, W. ADAR1 attenuates allogeneic graft rejection by suppressing miR-21 biogenesis in macrophages and promoting M2 polarization.


Assuntos
Adenosina Desaminase/metabolismo , Aloenxertos/metabolismo , Rejeição de Enxerto/metabolismo , Macrófagos/metabolismo , MicroRNAs/metabolismo , Animais , Regulação para Baixo/fisiologia , Feminino , Proteína Forkhead Box O1/metabolismo , Imunidade Inata/fisiologia , Interleucina-10/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia
14.
Cytokine ; 104: 30-37, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29414324

RESUMO

Adenosine deaminase acting on RNA 1 (ADAR1), a double-stranded RNA-editing enzyme that converts adenosine (A) to inosine (I), has been identified as a modulator of immune responses. However, the role of ADAR1 in small intestinal homeostasis during sepsis remains unclear. In this study, we examined the role of ADAR1 on intestinal inflammation in a murine model of sepsis. We found that ADAR1 was highly expressed in "septic" macrophages and small intestinal tissue of septic mice. Deletion of ADAR1 in "septic" macrophages led to rapid apoptosis. In addition, suppression of ADAR1 in "septic" macrophages significantly enhanced inflammation, while over-expression of ADAR1 significantly suppressed the level of inflammatory cytokines. Furthermore, suppression of ADAR1 in septic mice significantly enhanced inflammation and intestinal damage, while enhanced ADAR1 expression resulted in reduced damage and inflammation. Finally, over-expression of ADAR1 improved survival of septic mice. In conclusion, we have identified a novel ADAR1 protective effect for maintaining intestinal homeostasis. Our findings may provide a new targeted therapy for sepsis treatment.


Assuntos
Adenosina Desaminase/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Intestino Delgado/lesões , Intestino Delgado/metabolismo , Sepse/metabolismo , Sepse/patologia , Animais , Apoptose , Citocinas/metabolismo , Modelos Animais de Doenças , Deleção de Genes , Mediadores da Inflamação/metabolismo , Intestino Delgado/patologia , Estimativa de Kaplan-Meier , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células RAW 264.7
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA