RESUMO
ETHNOPHARMACOLOGICAL RELEVANCE: Metastasis is the leading cause of death in lung cancer worldwide, and immune escape plays a vital role in the process of metastasis. Clinical studies have proven that Jinfukang (JFK) can effectively treat lung cancer metastasis by regulating T lymphocytes. However, it is still unknown whether JFK plays a role in treating lung cancer metastasis by regulating T-cell receptors (TCRs). AIM OF THE STUDY: To explore the effect of JFK in inhibiting lung cancer metastasis by regulating TCR. MATERIALS AND METHODS: A lung metastasis model was established in C57BL/6J and BALB/c-nude mice by tail vein injection of Lewis lung cancer cells. JFK was given by continuous intragastric administration. Anatomical observation combined with hematoxylin-eosin staining was used to evaluate lung metastasis. T cells, MDSCs, and macrophages in the peripheral blood were detected by flow cytometry, and the proliferation and immune cell infiltration of lung metastases were observed by immunohistochemistry and immunofluorescence. The diversity and gene expression of TCR in peripheral blood and lung tissues were detected by immune repertoire sequencing, and bioinformatics analysis was carried out. RESULTS: Compared with the control group, the number of pulmonary metastatic nodules in JFK-treated mice showed a decreasing trend, and it significantly reduced the burden of lung tumor metastasis in mice. We found that the expression level of Ki-67 protein in lung metastatic tumor tissues of mice treated with JFK was significantly reduced, while the infiltration level of CD8+ T lymphocytes and NK cells was significantly increased. In addition, we also found that JFK could significantly increase the proportion of CD4+ T, CD8+ T and NKT cells in the peripheral blood of mice. Moreover, JFK reduced the ratio of M-MDSCs and increased the ratio of PMN-MDSCs in the peripheral blood of mice. JFK increased the ratio of M1 macrophages in the peripheral blood of Lewis tumor-bearing mice. The sequencing of TCR in the peripheral blood and lung tissue of mice indicated that there was no notable difference in TCR diversity as the tumor progressed and JFK treatment was administered. However, the downregulation of TRBV16, TRBV17, TRBV1 and the upregulation of the TRBV12-2 gene in the TCR caused by tumor progression can be reversed by JFK. CONCLUSION: These results suggest that JFK may upregulate the proportion of CD4+ T, CD8+ T and NKT cells in peripheral blood, reverse the TCR changes caused by tumor metastasis, and promote the infiltration of CD8+ T and NK cells in tumor tissues, thereby inhibiting the growth of tumors and ultimately reducing the burden of lung cancer metastasis. This will provide new strategies for developing Chinese herbal medicine to treat metastasis by regulating TCR.
Assuntos
Medicamentos de Ervas Chinesas , Neoplasias Pulmonares , Animais , Camundongos , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Camundongos Nus , Camundongos Endogâmicos C57BL , Neoplasias Pulmonares/patologia , Receptores de Antígenos de Linfócitos T/uso terapêutico , Linfócitos T CD8-PositivosRESUMO
Mitophagy is an essential physiological process that eliminates damaged mitochondria via lysosomes. It is reported that hypoxia, inflammatory stimuli or other stress conditions could lead to mitochondrial damage and mitochondrial dysfunction, which induces the process of mitophagy. Herein, we report a novel fluorescent probe PC-NTR for imaging hypoxia-induced mitophagy by monitoring the change of nitroreductase and viscosity simultaneously. To our delight, PC-NTR could respond simultaneously to nitroreductase and viscosity at different fluorescence channels with no mutual interference under the same excitation wavelength. The fluorescence emission around 535 nm was enhanced dramatically after addition of nitroreductase while the fluorescence emission around 635 nm heightened as the viscosity increased. The probe would be able to selectively targeting of mitochondria in cells because of the positively charged pyridine salt structure of PC-NTR. The probe was successfully applied to assess the different levels of hypoxia and real-time imaging of mitochondrial autophagy in live cells. More importantly, using dual channel imaging, PC-NTR could be used to distinguish cancer cells from normal cells and was successfully applied to imaging experiments in HeLa-derived tumor-bearing nude mice. Therefore, PC-NTR would be an important molecular tool for hypoxia imaging and detecting solid tumors in vivo.
Assuntos
Hipóxia , Mitofagia , Camundongos , Animais , Viscosidade , Camundongos Nus , Corantes Fluorescentes/química , Microscopia de Fluorescência , Imagem Óptica/métodos , NitrorredutasesRESUMO
BACKGROUND: Bioinformatics analysis suggests an association between lysyl oxidase like 1 (LOXL1) and forkhead box F2 (FOXF2), both of which are found to be dysregulated in thyroid cancer. This study aims to elucidate their specific roles in thyroid cancer. METHODS: The correlation of LOXL1 expression with thyroid cancer staging and the overall survival was analyzed. LOXL1 levels were determined in several thyroid cancer cells, and its effects on poorly differentiated BCPAP cell proliferation, colony formation, malignant phenotypes, epithelial-mesenchymal transition (EMT) progression, and angiogenesis were evaluated. The relationship between LOXL1 and FOXF2 was confirmed using Luciferase reporter and ChIP assays. The impacts of FOXF2 on LOXL1 regulation along with the Wnt/ß-catenin signaling were assessed, followed by the verification of transplanted tumor in nude mice. RESULTS: Elevated LOXL1 expression was associated with advanced clinical staging and poorer overall survival. Reduced LOXL1 suppressed cell proliferation, colony formation, migration, invasion, EMT, and angiogenesis. FOXF2 was found to be down-regulated in thyroid cancer, acting as a transcription factor that recognizes the LOXL1 promoter and modulates its transcriptional expression. Moreover, the regulatory outcome of LOXL1 knockdown was partially reversed upon FOXF2 knockdown, including the modulation of the Wnt/ß-catenin signaling and tumor growth in vivo. CONCLUSION: Our findings indicate that LOXL1 is transcriptionally regulated by FOXF2 and activates the Wnt/ß-catenin to promote malignant phenotypes, EMT progression, and angiogenesis in BCPAP cells.
Assuntos
Neoplasias da Glândula Tireoide , beta Catenina , Animais , Camundongos , beta Catenina/metabolismo , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/genética , Proteína-Lisina 6-Oxidase/genética , Camundongos Nus , Regulação Neoplásica da Expressão Gênica , Via de Sinalização Wnt/genética , Proliferação de Células/genética , Neoplasias da Glândula Tireoide/genética , Movimento Celular/genéticaRESUMO
Bladder cancer, the most common malignant tumor in the urinary system, exhibits significantly up-regulated expression of P3H4, which is associated with pathological factors. The objective of this study was to elucidate the underlying mechanism of P3H4 in bladder cancer. Initially, we analyzed P3H4 gene expression using the TCGA database and evaluated P3H4 levels in clinical samples and various bladder cell lines. P3H4 was found to be markedly overexpressed in bladder cancer samples. Subsequently, bladder cancer cells were transfected with shRNA targeting P3H4 (sh-P3H4), sh-METTL3, and P3H4 overexpression vectors (P3H4 OE). Viability, migration, and invasion of bladder cancer cells were assessed using CCK-8, wound healing, and transwell assays. Western blot analysis was performed to determine the levels of EMT-associated proteins, while RNA stability assays determined the half-life of P3H4. Knockdown of P3H4 resulted in inhibition of bladder cancer cell proliferation, migration, invasion, and EMT progression. Mechanistically, METTL3 was found to regulate the mRNA stability of P3H4 in bladder cancer. Moreover, overexpression of P3H4 reversed the inhibitory effects of METTL3 knockdown on bladder cancer cell behaviors. Stable cell lines were established by infecting EJ cells with lentiviral vectors containing sh-METTL3 or P3H4 OE. These cells were then implanted into the skin of BALB/c nude mice, and IHC analysis was used to analyze the expression levels of EMT-associated proteins. In vivo studies demonstrated that inhibition of METTL3 suppressed bladder cancer growth and EMT through P3H4. In conclusion, our findings suggest that METTL3 regulates the proliferation, metastasis, and EMT progression of bladder cancer through P3H4, highlighting its potential as a therapeutic target.
Assuntos
Neoplasias da Bexiga Urinária , Animais , Camundongos , Linhagem Celular Tumoral , Camundongos Nus , Proliferação de Células/genética , Neoplasias da Bexiga Urinária/patologia , Movimento Celular/genética , Regulação Neoplásica da Expressão GênicaRESUMO
Sphingolipids are important for the physicochemical properties of cellular membranes and deregulated in tumors. In human colon cancer tissue ceramide synthase (CerS) 4 and CerS5 are reduced which correlates with a reduced survival probability of late-stage colon cancer patients. Both enzymes are reduced after hypoxia in advanced colorectal cancer (CRC) cells (HCT-116, SW620) but not in non-metastatic CRC cells (SW480, Caco-2). Downregulation of CerS4 or CerS5 in advanced CRC cells enhanced tumor formation in nude mice and organoid growth in vitro. This was accompanied by an enhanced proliferation rate and metabolic changes leading to a shift towards the Warburg effect. In contrast, CerS4 or CerS5 depletion in Caco-2 cells reduced tumor growth in vivo. Lipidomic and proteomic analysis of membrane fractions revealed significant changes in tumor-promoting cellular pathways and cellular transporters. This study identifies CerS4 and CerS5 as prognostic markers for advanced colon cancer patients and provides a comprehensive overview about the associated cellular metabolic changes. We propose that the expression level of CerS4 and CerS5 in colon tumors could serve as a basis for decision-making for personalized treatment of advanced colon cancer patients. Trial registration: The study was accredited by the study board of the Deutsche Krebsgesellschaft (Registration No: St-D203, 2017/06/30, retrospectively registered).
Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Animais , Camundongos , Humanos , Esfingolipídeos/metabolismo , Células CACO-2 , Camundongos Nus , Prognóstico , Proteômica , Neoplasias do Colo/metabolismo , Neoplasias Colorretais/patologia , HipóxiaRESUMO
BACKGROUND: Ferroptosis is an emerging iron-dependent programmed cell death mode characterized by lipid peroxidation and iron accumulation, closely associated with Hepatocellular Carcinoma (HCC) progression. Although the impact of Polyphyllin I (PPI), a prominent bioactive constituent derived from Paris polyphylla, on diverse malignancies has been established, the specific role and potential mechanistic pathways through which PPI modulates ferroptosis in HCC remain elusive. PURPOSE: This study aimed to elucidate the anti-cancer properties and potential mechanisms of PPI in inducing ferroptosis and triggering mitochondrial injury in HCC. METHODS: Cell viability was assessed using CCK-8 assays. EdU proliferation and colony formation assays were employed to evaluate cell proliferation. A wound-healing assay was performed to assess cell migration. Transwell assay was utilized to evaluate cell invasion. Ferroptosis was evaluated through the utilization of a FerroOrange fluorescent probe, malondialdehyde (MDA) and reduced glutathione (GSH) assay kits, DCFH-DA fluorescent probe, western blotting, and transmission electron microscopy (TEM) analysis. Molecular docking, immunofluorescence, and western blotting were employed to predict and validate the binding and interaction of PPI with Nrf2, HO-1, xCT, and GPX4. Mitochondrial structure and membrane potential changes were evaluated using JC-1 and Mito Tracker Green fluorescent probes. A nude mice xenograft model was constructed to determine the inhibitory effects and the levels of ferroptosis of PPI on HCC through hematoxylin and eosin (H&E), Prussian blue reaction, immunofluorescence staining, immunohistochemistry, and western blotting analysis, in vivo. RESULTS: PPI exhibited dose-dependent inhibitory effects on the proliferation, invasion, and metastasis of HCC cells mediated by increasing reactive oxygen species (ROS) and MDA levels, promoting Fe2+ accumulation, depleting GSH, and suppressing the expression of xCT and GPX4, thereby inducing ferroptosis in HCC. The induction of ferroptosis by PPI was associated with the binding of PPI to Nrf2, HO-1, and GPX4 proteins, modulating the Nrf2/HO-1/GPX4 antioxidant axis. PPI also induced mitochondrial structural damage and decreased mitochondrial membrane potential (MMP). Inhibition of ferroptosis by ferrostatin-1 (Fer-1) mitigated the mitochondrial disruption induced by PPI. In vivo, PPI inhibited Nrf2/HO-1/GPX4 axis-induced ferroptosis, impeding HCC growth similar to the effects of sorafenib. CONCLUSION: These results demonstrated that PPI intervention can suppress the proliferation, invasion, and metastasis of HCC cells by enhancing mitochondrial disruption and inducing ferroptosis via the Nrf2/HO-1/GPX4 axis. Consequently, our research advances the frontiers of pharmacodynamics and deepens our comprehension of the intricate mechanisms underpinning PPI. Furthermore, it has yielded an innovative treatment stratagem rooted in the tenets of Traditional Chinese Medicine (TCM), thereby furnishing a novel therapeutic avenue for addressing HCC.
Assuntos
Carcinoma Hepatocelular , Ferroptose , Neoplasias Hepáticas , Animais , Camundongos , Humanos , Carcinoma Hepatocelular/tratamento farmacológico , Fator 2 Relacionado a NF-E2 , Corantes Fluorescentes , Camundongos Nus , Simulação de Acoplamento Molecular , Neoplasias Hepáticas/tratamento farmacológico , Ferro , Espécies Reativas de OxigênioRESUMO
BACKGROUND AND OBJECTIVES: Targeted drug is often engulfed and cleared by the reticuloendothelial system in vivo, resulting in reduced treatment efficacy. This study aimed to explore the biodistribution and HER-2-targeted antitumor effects of trastuzumab-modified gold nanorods (Tra-AuNRs) in a gastric cancer animal model. METHODS: Gold nanorods were synthesized using a seed-mediated growth method, and then subjected to trastuzumab-targeted modification. Elemental analysis, Fourier transform infrared spectroscopy, and Xray photoelectron spectroscopy were performed; UV-visible absorption peak, photothermal effects, morphology, and size distribution of Tra-AuNRs were characterized. The targeted killing effect of Tra- AuNRs on gastric cancer cells was assessed in vitro. Tra-AuNRs were injected intravenously and intratumorally into gastric cancer-bearing nude mice in vivo and their distribution was detected. Tumor growth inhibition rate and tumor apoptosis-related protein expression were compared between groups. RESULTS: Tra-AuNRs presented a relatively uniform morphology with an average particle size of 59.9 nm and a longitudinal plasmon resonance absorption peak of 790 nm. The targeted killing rate of gastric cancer cells in vitro by Tra-AuNRs was 87.9%. After intravenous injection, Tra-AuNRs were mainly distributed in the liver, tumor, spleen, and lungs. Comparatively, Tra-AuNRs were mainly distributed in the tumor when intratumorally injected, with a tumor concentration of 6.42 µg/g after 24 h. The tumor growth inhibition rate reached 78.3% in the intratumoral injection group, with significantly higher BAX, BAD, and CASPASE-3 expression than that in the intravenous injection group. CONCLUSION: The findings suggest that Tra-AuNRs can be used for HER-2-positive gastric cancer treatment. Intratumoral injection of Tra-AuNRs significantly increased the local tumor drug concentration and improved the molecular targeted antitumor growth effect in gastric cancer-bearing nude mice.
Assuntos
Nanotubos , Neoplasias Gástricas , Animais , Camundongos , Neoplasias Gástricas/tratamento farmacológico , Camundongos Nus , Distribuição Tecidual , Trastuzumab , Ouro/química , Linhagem Celular Tumoral , Nanotubos/químicaRESUMO
The immune system plays a crucial role in cancer development and progression. More than a century ago, mouse models showed that primary tumors suppressed the growth of newly implanted secondary tumors. This phenomenon, in which tumor-primed T cells mediate the rejection of tumor growth at a distant site, is known as concomitant tumor immunity. Here, we investigated the role of concomitant immunity in the development of breast cancer bone metastases using newly developed syngeneic immunocompetent mouse models. The presence of primary breast tumors developed by tumor cell injection into the mammary fat pads (MFPs) significantly reduced bone metastases of mouse breast cancer 4T1 and EMT6 cells induced by cell injection through the caudal artery (CA). Similar results were obtained when primary tumors were surgically resected prior to CA injection of tumor cells. In contrast, no inhibition was found when MFP and CA injections were performed using different cell combinations. Immunohistochemical studies revealed that the number of CD8+ T cells in bone metastases of 4T1 and EMT6 cells was significantly increased in the presence of primary tumors. The primary tumor-induced inhibition of bone metastases was not reproduced in T cell-deficient athymic nude mice. Furthermore, depletion of CD8+ T cells using an anti-CD8α antibody also abolished the primary tumor-induced inhibition of bone metastases. Taken together, these results suggest that immune cell priming by orthotopic breast tumors inhibits the development of breast cancer bone metastases, which is predominantly mediated by CD8+ cytotoxic T lymphocytes.
Assuntos
Neoplasias Ósseas , Neoplasias da Mama , Camundongos , Animais , Humanos , Feminino , Linfócitos T CD8-Positivos/patologia , Camundongos Nus , Linhagem Celular Tumoral , Neoplasias Ósseas/secundário , Modelos Animais de Doenças , Neoplasias da Mama/patologiaRESUMO
ETHNOPHARMACOLOGICAL RELEVANCE: Yiqi Jiedu formula (YQJDF), rooted in the traditional Chinese medicinal principle of "tonifying qi and detoxifying", is remarkably efficacious in the clinical treatment of nasopharyngeal carcinoma (NPC). Previous studies have shed light on some of its anti-NPC effects and mechanisms, but the responsible pharmacological substances and their precise mechanisms of action remain unclear. AIM OF THE STUDY: The purpose of this study was to identify components of YQJDF that entered the bloodstream and to investigate their mechanisms of action against NPC through network pharmacology and serum metabolomics. MATERIAL AND METHODS: Components of YQJDF in serum were identified using liquid chromatography-tandem mass spectrometry. With these serum species as the focus of our research, network pharmacology analysis was used to identify active compounds and target genes that might mediate the efficacy of YQJDF in the treatment of NPC. Following establishment of an NPC xenograft model in nude mice, a non-targeted metabolomics approach was adopted to identify significant serum metabolites and metabolic pathways influenced by YQJDF. RESULTS: Thirty-six components of YQJDF were identified, primarily consisting of alkaloids, phenylpropanoids, and flavonoids. Notably, pathways such as PI3K/AKT, factors associated with Epstein-Barr virus infection, IL-17 signaling, and lipid metabolism, were highlighted as potential therapeutic targets of YQJDF during NPC treatment. Additionally, our findings suggested that YQJDF modified the metabolism of arginine and proline in the serum of mice bearing nasopharyngeal tumor grafts. CONCLUSIONS: This study identified the primary active components of YQJDF, highlighting its holistic role in the treatment of NPC through multiple targets and pathways. Furthermore, our findings provided a roadmap for future research into the mechanism of YQJDF in the therapy of NPC, setting the stage for its clinical application.
Assuntos
Medicamentos de Ervas Chinesas , Infecções por Vírus Epstein-Barr , Neoplasias Nasofaríngeas , Humanos , Animais , Camundongos , Carcinoma Nasofaríngeo/tratamento farmacológico , Neoplasias Nasofaríngeas/tratamento farmacológico , Camundongos Nus , Farmacologia em Rede , Fosfatidilinositol 3-Quinases , Herpesvirus Humano 4 , Metabolômica , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Simulação de Acoplamento MolecularRESUMO
Lenvatinib is the first-line treatment for patients with advanced HCC, however, drug resistance cannot be avoided during the treatment process, limiting the efficacy of Lenvatinib. We constructed drug-resistant HCC cells by gradually increasing the dose of Lenvatinib. The study found for the first time that USP22 and JMJD8 are upregulated in Lenvatinib resistant HCC cells. In addition, the expression level of stemness related proteins (CD133, C-MYC, BMI1, ß-CATENIN) in drug-resistant cells was higher than that in wild-type HCC cells. Knockdown of USP22 in drug-resistant HCC cells could reduce the invasion, migration and stemness of cells. Next, we explored the mechanism of USP22 in Lenvatinib resistance of HCC cells. Under the treatment of Lenvatinib, USP22 knockdown inhibited the cell viability of drug-resistant HCC cells and promoted the apoptosis of drug-resistant cells. Animal experiments in nude mice further demonstrated the important role of USP22 in inducing the resistance of HCC to Lenvatinib in vivo. More importantly, we found that USP22 and JMJD8 constitute a functional axis regulating the drug resistance of Lenvatinib in HCC. In the rescue experiment, the overexpression of JMJD8 could reduce the apoptosis induced by USP22 knockdown. In general, this study shows that USP22-JMJD8 is a drug design target for the mechanism of Lenvatinib resistance in HCC, which may improve the long-term efficacy of Lenvatinib.
Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Humanos , Camundongos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Camundongos NusRESUMO
INTRODUCTION: Gastric cancer poses a major therapeutic challenge. Improved visualization of tumor margins at the time of gastrectomy with fluorescent tumor-specific antibodies could improve outcomes. The present report demonstrates the potential of targeting gastric cancer with a humanized anti-carcinoembryonic antigen (CEA) antibody in orthotopic mouse models. METHODS: MKN45 cells were injected subcutaneously into nude mice to establish xenograft models. Tumor fragments collected from subcutaneous models were then implanted into the greater curvature of the stomach to establish orthotopic models. For tumor labeling, a humanized anti-CEA antibody (M5A) and IgG as a control, were conjugated with the near-infrared dye IRDye800CW. Time (24-72 h) and dose (50-100 µg) response curves were performed in subcutaneous models. Orthotopic models received 50 µg of M5A-IR800 or 50 µg IgG-IR800 as a control and were imaged after 72 h. Fluorescence imaging was performed on the mice using the LI-COR Pearl Imaging System. RESULTS: In subcutaneous models, tumor to background ratios (TBRs) reached 8.85 at 72 h. Median TBRs of orthotopic model primary tumors were 6.25 (interquartile range [IQR] 6.03-7.12) for M5A-IR800 compared to 0.42 (IQR 0.38-0.54) for control. Abdominal wall metastasis median TBRs were 13.52 (IQR 12.79-13.76) for M5A-IR800 and 3.19 (IQR 2.65-3.73) for the control. Immunohistochemistry confirmed CEA expression within tumors. CONCLUSIONS: Humanized anti-CEA antibodies conjugated to near-infrared dyes provide specific labeling of gastric cancers in mouse models. Orthotopic models demonstrated bright and specific labeling with TBRs greater than ten times that of control. This tumor-specific fluorescent antibody is a promising potential clinical tool for improving visualization of gastric cancer margins at time of surgical resection.
Assuntos
Neoplasias Gástricas , Humanos , Animais , Camundongos , Camundongos Nus , Antígeno Carcinoembrionário , Anticorpos Monoclonais , Modelos Animais de Doenças , Imunoglobulina G , Corantes Fluorescentes , Linhagem Celular TumoralRESUMO
Radiotherapy (RT) is a highly valuable method in cancer therapy, but its therapeutic efficacy is limited by its side effects and tumour radiation resistance. The resistance is mainly induced by hypoxia in the tumour microenvironment (TME). As a nano-oxygen carrier, Haemoglobin-based oxygen carriers (HBOCs) administration is a promising strategy to alleviate tumour hypoxia which may remodel TME to ameliorate radiation resistance and enable RT more effective. In this study, we administered fractionated RT combined with HBOC to treat Miapaca-2 cell and Hela cell xenografts on nude mice. The study found that HBOC relieved hypoxic environment and down-regulate expression of hypoxia-inducible factor-1α (Hif-1α) both in regular (100 mm3) and large (360/400 mm3) tumours. The proliferation and metastasis of tumour tissue also decreased after HBOC application. Nevertheless, in vivo RT combined with HBOC performed more effectively to suppress tumour growth in large tumours than in regular tumours. This is due to more severe hypoxic regions exist in the large solid tumours compared to the regular counterparts, and HBOC administration may be more effective in alleviating hypoxia in large tumours. Thus, HBOC sensitization therapy is more suitable for large solid tumours.
Assuntos
Hipóxia , Hipóxia Tumoral , Animais , Camundongos , Humanos , Células HeLa , Camundongos Nus , Oxigênio , Hipóxia Celular , Linhagem Celular TumoralRESUMO
ADAMTS8 (A Disintegrin-like and Metalloproteinase with Thrombospondin motifs 8) is a secreted zinc-dependent metalloproteinase whose expression is downregulated in a variety of solid tumors. Xenografts expressing high levels of ADAMTS8 have a poor capacity to invade and migrate in nude mice. While this data highlights a beneficial, anti-cancerogenic role of ADAMTS8, the mechanism behind this activity is still not fully elucidated. So far, the only reported substrate for ADAMTS8 is osteopontin (OPN), an extracellular matrix protein widely implicated in multiple steps of cancer progression, albeit, similar to other ADAMTS family members, it is very likely that ADAMTS8 cleaves a variety of substrates. The availability of purified ADAMTS8 may enlighten the biological role of this metalloproteinase.Here we describe methods for expression and purification of recombinant ADAMTS8 in HEK293T cells as well as a convenient assay to test ADAMTS8 proteolytic activity using OPN as a substrate.
Assuntos
Proteínas ADAMTS , Neoplasias , Camundongos , Animais , Humanos , Proteínas ADAMTS/genética , Proteínas ADAMTS/metabolismo , Camundongos Nus , Células HEK293 , Proteínas da Matriz ExtracelularRESUMO
Drug resistance is an obstacle in therapy of esophageal cancers (ECs), and the role of ferroptosis in progression ECs is still not clearly clarified. In the present study, we investigated the role of Apolipoprotein C1 (Apoc1) in regulating the sorafenib resistance in EC cells. Apoc1 was knock down after infection with Apoc1 shRNA lentivirus and stable cell lines for Apoc1 knockdown were screened. Cell viabilities were tested by MTT assay. ROS, MDA, and GSH tested by specific kits. In vivo experiment in nude mice were performed to test the correlation of Apoc1 and ferroptosis. The expression of Apoc1 and GPX4 was tested by western blotting. The results showed that Apoc1 was highly expressed in EC tissues and associated with poor overall survival rate of EC. Knockdown Apoc1 overcame resistance of sorafenib in EC cells and promoted erastin and sorafenib induced ferroptosis by upregulating the levels of ROS and MDA and downregulating the level of GSH in OE19/Sora and EC109/Sora cells. Rescue experiments proved that Apoc1 regulated sorafenib induced ferroptosis via GPX4. Furthermore, knockdown of Apoc1 inhibited the tumor progression by promoting ferroptosis in nude mice. In conclusion, knockdown Apoc1 overcome resistance of sorafenib in EC cells and in vivo by promoting sorafenib induced ferroptosis via GPX4. Targeting Apoc1 might be an effective way to reverse the drug resistance of sorafenib via inducing ferroptosis in EC progression.
Assuntos
Neoplasias Esofágicas , Ferroptose , Animais , Camundongos , Apolipoproteína C-I , Camundongos Nus , Sorafenibe/farmacologia , Espécies Reativas de Oxigênio , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/genéticaRESUMO
ETHNOPHARMACOLOGICAL RELEVANCE: Ludwigia hyssopifolia (LH), an ethnopharmacological herb used in Guangxi Zhuang medicine, is known for its extensive therapeutic use in treating throat disorders. The anti-laryngeal-cancer benefits of the ethyl acetate and petroleum ether fractions of the ethanolic extracts of LH have been shown in our prior cell-based research. Nevertheless, the specific impacts and underlying processes by which LH combats throat cancer effects have not been fully understood. AIM OF THE STUDY: This study involved the extraction of a composition containing two derivatives of ursolic acid from LH (LH-CUAD). The present study aimed to assess the anti-throat-cancer effects of these derivatives and the underlying mechanisms through in vitro and in vivo experiments. MATERIALS AND METHODS: Solvent extraction, fractionation, chromatography, and semipreparative high-performance liquid chromatography were used for the extraction, purification, and analysis of LH-CUAD. The in vitro and in vivo anti-throat-cancer effects of LH-CUAD were investigated using the throat cancer cell lines Hep-2 and FaDu as well as Hep-2 tumor-bearing nude mice. RESULTS: LH-CUAD significantly inhibited the proliferation and migration of throat cancer cells without any prominent toxicity. The Hoechst 33258 staining, Annexin V-FITC/PI double-staining assays, and flow cytometry confirmed that LH-CUAD could induce throat cancer cell death from early to late apoptosis in vitro. LH-CUAD exhibited significant antitumor activity and low toxicity in a xenograft model, and induced throat cancer cells apoptosis in vivo. The apoptotic effects of LH-CUAD therapy were validated using Western blotting, which demonstrated the activation of a caspase cascade response triggered by an imbalance between the endoplasmic reticulum and mitochondria. In addition, it was observed that LH-CUAD exhibited inhibitory effects on Akt and mTOR phosphorylation, hence promoting apoptosis. CONCLUSIONS: LH-CUAD induces apoptosis in both in vivo and in vitro models of throat cancer. This effect is achieved by activating the mitochondrial pathway, inhibiting the Akt/mTOR pathway and initiating endoplasmic reticulum stress. The findings of this study suggest that LH-CUAD has the potential to offer a novel approach to the clinical management of throat cancer.
Assuntos
Neoplasias , Faringe , Animais , Camundongos , Humanos , Proteínas Proto-Oncogênicas c-akt , Camundongos Nus , China , Transdução de Sinais , Serina-Treonina Quinases TOR , ApoptoseRESUMO
Thyroid cancer is one of the most common endocrine neoplasms. Treatment methods include surgical resection, radioactive iodine therapy, inhibition of thyroid-stimulating hormone, and inhibition of kinase-based target therapies. These treatments induced adverse effects. Lithospermum officinale possesses antioxidant, anticancer, burn-healing, and anti-inflammatory activities, and Shikonin is the main ingredient. Antithyroid cancer studies of Shikonin discovered that it inhibited thyroid cancer cell migration and invasion by suppressing the epithelial-mesenchymal transition; induced cell cycle arrest; induced DNA damage and apoptosis by producing excessive reactive oxygen species; upregulated Bax; increased the stability of p53; decreased the expression of Mdm2; downregulated Slug and MMP-2, MMP-9, and MMP-14; repressed the phosphorylation of Erk and Akt; activated the p16/retinoblastoma protein pathway, leading to apoptosis; suppressed the expression of DNMT1; reduced the PTEN gene methylation; increased the expression of PTEN, leading to the inhibition of migration; increased LC3-II to induce autophagy and apoptosis of medullary thyroid carcinoma; and upregulated ßII-tubulin in the cell to produce less resistance to cisplatin and paclitaxel, without cross-resistance to other anticancer agents. In vivo studies showed that it is safe in Sprague-Dawley rats, Beagle dogs, and nude mice.
Assuntos
Neoplasias da Glândula Tireoide , Ratos , Camundongos , Animais , Cães , Ratos Sprague-Dawley , Neoplasias da Glândula Tireoide/tratamento farmacológico , Radioisótopos do Iodo , Camundongos NusRESUMO
ETHNOPHARMACOLOGICAL RELEVANCE: The YangzhengXiaoji capsule (YXC) has a wide range of applications as effective traditional Chinese medicine (TCM) preparation for hepatocellular carcinoma (HCC) in China. However, the potential bioactive components and the mechanisms are yet unclear. AIM OF THE STUDY: The treatment mechanism of YXC on HCC using a network pharmacology integrated serum pharmacochemistry strategy to investigate associated targets and pathways. MATERIALS AND METHODS: We utilised HPLC-Q-TOF-MS/MS technology to identify components of the serum samples from both the model group and the YXC (H) group serum, which were collected from nude mice with orthotopic liver tumours. Following this, we conducted compound-target prediction and identified the overlap between the target genes in the YXC group and the oncogenes associated with HCC. The anticancer mechanisms of YXC were investigated by creating a compound-target-pathway network using the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and Gene Ontology (GO) analysis. The anticancer efficacy was evaluated in vitro and in vivo. Also, potential predictive targets and pathways associated with YXC in HCC treatment were assessed by western blotting. RESULTS: The YXC (H) serum had 47 bioactive compounds compared to other models, and identified 173 specific target genes. Using the compound-target-disease network, 141 possible target genes were identified. The KEGG pathway analysis revealed vital enrichment of pathways associated with HCC, including regulating Oncology related pathways of inflammation, immunity, apoptosis, and necrosis biological processes. YXC significantly inhibited HCC cell growth in vitro and in vivo. After YXC treatment, western blotting detected alterations in the p53/Bcl-2/Bax/Caspase-3 and PI3K/Akt pathways. CONCLUSIONS: YXC can inhibit HCC development and advancement by a variety of components, targets and pathways, especially apoptosis-induction.
Assuntos
Carcinoma Hepatocelular , Medicamentos de Ervas Chinesas , Neoplasias Hepáticas , Animais , Camundongos , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Farmacologia em Rede , Camundongos Nus , Fosfatidilinositol 3-Quinases , Espectrometria de Massas em Tandem , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Simulação de Acoplamento MolecularRESUMO
PURPOSE: There is no report about the direct relationship between m6A modification and androgen receptor (AR)-related genes in prostate cancer (PC). We aimed to study the mechanisms of m6A methylation in regulating the pathogenesis of PC from the perspective of AR-related genes. METHODS: qRT-PCR was applied to detect the expression of m6A-related genes in PC cell with or without AR inhibitor. The effects of YTHDF1 knockdown on PC cell viability, apoptosis, migration and invasion were investigated using flow cytometry, wound healing and transwell assays, respectively. The mechanism of YTHDF1 action was investigated using m6A RNA immunoprecipitation (MeRIP) sequencing. The biological functions of YTHDF1 were also explored through in vivo experiments. RESULTS: YTHDF1 was significantly down-regulated in AR inhibitor group. YTHDF1 knockdown significantly decreased AR level, viability and m6A methylation level of PC cells. TRIM68 was identified as a direct target of YTHDF1. Both YTHDF1 and TRIM68 knockdown increased apoptosis, and decreased cell viability, migration, and invasion of PC cells, while TRIM68 overexpression reversed the effects of YTHDF1 knockdown on PC cells. In addition, knockdown of YTHDF1 or TRIM68 significantly decreased the m6A methylation level, and mRNA and protein levels of YTHDF1, TRIM68 and AR in PC cells, while TRIM68 overexpression increased the expression levels above. Furthermore, subcutaneous xenografts of nude mice also revealed that TRIM68 could reverse the effects of YTHDF1 knockdown in PC in vivo. CONCLUSION: This study suggested the key role of YTHDF1-mediated m6A modification in PC progression by regulating androgen function-related gene TRIM68 in PC.
Assuntos
Androgênios , Neoplasias da Próstata , Animais , Camundongos , Masculino , Humanos , RNA , Camundongos Nus , Neoplasias da Próstata/genética , Proteínas de Ligação a RNA/genética , Proteínas com Motivo Tripartido , Autoantígenos , Ubiquitina-Proteína LigasesRESUMO
Cervical cancer (CC) is the fourth most common cancer amongst females worldwide. Histone deacetylase (HDAC) 1 plays a vital role in several tumors. Nevertheless, its potential and mechanism in radiotherapy sensitivity underlying CC remains obscure. Hence, the objective of this research was to probe the potential of HDAC1 in CC radiotherapy sensitivity and its mechanism of action. The expression HDACs and survival analysis of HDAC1 were investigated based on the GEPIA database. Immunohistochemical staining was implemented to detect HDAC1 and Ki-67 expression in tumor tissues. RT-qPCR and Western blot were conducted to assess HDAC1, HIF-1α, VEGFA, along with VEGFR expressions in CC cells and tumor tissues. Cell viability, apoptosis, invasion, migration, along with cell cycle were analyzed by functional assays. Tumor-bearing nude mice model was established, and the tumor weight and volume were determined. HDAC1 was high-expressed in the tumor tissue and CC cells. In vitro, overexpression of HDAC1 suppressed radiotherapy sensitivity in C33A cells, while knockdown of HDAC1 promoted radiotherapy sensitivity in SiHa cells. In vivo, we found that HDAC1 silence hindered tumor growth and cell proliferation and promoted tumor cell apoptosis in nude mice after radiotherapy. In addition, we found that HDAC1 impacted radiotherapy sensitivity by modulating the HIF-1α/VEGF signaling pathway. In conclusion, HDAC1 suppressed the radiotherapy sensitivity of CC via regulating HIF-1α/VEGF signaling pathway, suggesting that HDAC1 may act as a crucial participant in regulating CC radiosensitivity, which may provide a novel method for treating CC.
Assuntos
Histona Desacetilase 1 , Neoplasias do Colo do Útero , Animais , Feminino , Humanos , Camundongos , Apoptose/genética , Histona Desacetilase 1/genética , Camundongos Nus , Transdução de Sinais , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/radioterapia , Fator A de Crescimento do Endotélio Vascular/genética , Tolerância a Radiação/genéticaRESUMO
Protein degradation in eukaryotic cells is mainly carried out by the 26S proteasome, a macromolecular complex not only present in the cytosol and nucleus but also associated with various membranes. How proteasomes are anchored to the membrane and the biological meaning thereof have been largely unknown in higher organisms. Here, we show that N-myristoylation of the Rpt2 subunit is a general mechanism for proteasome-membrane interaction. Loss of this modification in the Rpt2-G2A mutant cells leads to profound changes in the membrane-associated proteome, perturbs the endomembrane system, and undermines critical cellular processes such as cell adhesion, endoplasmic reticulum-associated degradation and membrane protein trafficking. Rpt2G2A/G2A homozygous mutation is embryonic lethal in mice and is sufficient to abolish tumor growth in a nude mice xenograft model. These findings have defined an evolutionarily conserved mechanism for maintaining membrane protein homeostasis and underscored the significance of compartmentalized protein degradation by myristoyl-anchored proteasomes in health and disease.