Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Cancer Lett ; 581: 216495, 2024 01 28.
Article in English | MEDLINE | ID: mdl-37993085

ABSTRACT

Immunity-related GTPase M (IRGM), an Interferon-inducible protein, functions as a pivotal immunoregulator in multiple autoimmune diseases and infection. However, the role of IRGM in hepatocellular carcinoma (HCC) development remains unveiled. Here, we found interferon-γ (IFN-γ) treatment in HCC drastically triggered the expression of IRGM, and the high level of IRGM indicated poor prognosis in HCC patients. Functionally, IRGM promoted the malignant progression of HCC. Single-cell sequencing revealed that IRGM inhibition promoted the infiltration of CD8+ cytotoxic T lymphocytes (CTLs) with significant downregulation of PD-L1 expression in HCC. Furthermore, Immunoprecipitation-Mass Spectrometry assay revealed that IRGM interacted with transcription factor YBX1, which facilitated PD-L1 transcription. Mechanistically, IRGM promoted the interaction of YBX1 and phosphokinase S6K1, increasing phosphorylation and nuclear localization of YBX1, transcription of PD-L1. Additionally, the combination of IRGM inhibition with α-PD1 demonstrated a stronger anti-tumor effect compared to the single application of α-PD1. In summary, IRGM is a novel regulator of PD-L1, which suppresses CD8+ CTLs infiltration and function in HCC, resulting in cancer progression. This study may raise a novel therapeutic strategy combined with immune checkpoint inhibitors (ICIs) against HCC.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Carcinoma, Hepatocellular/drug therapy , CD8-Positive T-Lymphocytes , GTP-Binding Proteins/metabolism , Liver Neoplasms/drug therapy , Phosphorylation , Tumor Microenvironment , Y-Box-Binding Protein 1/genetics , Y-Box-Binding Protein 1/metabolism , Ribosomal Protein S6 Kinases
2.
Small ; 20(21): e2307273, 2024 May.
Article in English | MEDLINE | ID: mdl-38102096

ABSTRACT

In the course of antitumor therapy, the complex tumor microenvironment and drug-mediated changes in cell signaling and biological processes lead to drug resistance. The effect of sorafenib is greatly limited by the specific tumor microenvironment induced by antiangiogenic therapy and ferroptosis resistance induced by the upregulation of nuclear factor erythroid-2 related factor 2 (NRF2). In this study, a pH responsive and amphiphilic hyperbranched polyglycerol, HDP, is synthesized based on a co-graft click chemistry pathway. This nano-scale carrier provides excellent drug-loading capacity, storing stability and pH responsibility, and effectively co-delivery of sorafenib and siRNA. Sorafenib and siNRF2 plays a greatly synergistic effect in inducing reactive oxygen species (ROS), iron overloading, depleting glutathione (GSH), and promoting lipid peroxidation. Importantly, verified in two different animal experiments, HDP-ss (HDP loaded with both siNRF2 and sorafenib) presents a superior anti-tumor effect, by achieving a tumor inhibition rate of ≈94%. Thus, HDP can serve as an excellent targeted delivery nanocarrier with good biocompatibility in antitumor therapy, and combined application of siNRF2 effectively improves the antitumor effect of sorafenib by overcoming NRF2-mediated ferroptosis resistance. Taken together, this study provides a novel therapeutic strategy to combat the drug resistance in antiangiogenic therapy and ferroptosis.


Subject(s)
Carcinoma, Hepatocellular , Ferroptosis , Liver Neoplasms , NF-E2-Related Factor 2 , Sorafenib , Sorafenib/pharmacology , Sorafenib/chemistry , Ferroptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Humans , Animals , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , NF-E2-Related Factor 2/metabolism , Drug Carriers/chemistry , Reactive Oxygen Species/metabolism , Nanoparticles/chemistry , Cell Line, Tumor , Mice , Glutathione/metabolism
4.
J Transl Med ; 21(1): 420, 2023 06 29.
Article in English | MEDLINE | ID: mdl-37381011

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most lethal tumor types worldwide. Glycosylation has shown promise in the study of tumor mechanisms and treatment. The glycosylation status of HCC and the underlying molecular mechanisms are still not fully elucidated. Using bioinformatic analysis we obtained a more comprehensive characterization of glycosylation of HCC. Our analysis presented that high glycosylation levels might correlate with tumor progression and poor prognosis. Subsequent Experiments identified key molecular mechanisms for ST6GALNAC4 promoting malignant progression by inducing abnormal glycosylation. We confirmed the contribution of ST6GALNAC4 to proliferation, migration, and invasion in vitro and in vivo. Mechanistic studies revealed that ST6GALNAC4 may be induced abnormal TGFBR2 glycosylation, resulting in the higher protein levels of TGFBR2 and TGF[Formula: see text] pathway increased activation. Our study also provided a further understand of immunosuppressive function of ST6GALNAC4 through T antigen-galectin3+ TAMs axis. This study has provided one such possibility that galectin3 inhibitors might be an acceptable treatment choice for HCC patients with high T antigen expression.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Sialyltransferases , Humans , Antigens, Viral, Tumor , Carcinogenesis , Carcinoma, Hepatocellular/genetics , Glycosylation , Liver Neoplasms/genetics , Receptor, Transforming Growth Factor-beta Type II , Sialyltransferases/genetics
6.
Epigenetics ; 18(1): 2204772, 2023 12.
Article in English | MEDLINE | ID: mdl-37092305

ABSTRACT

Background: Circular RNA (circRNA) plays a critical role in tumour progression. Circ-CCT3, a particularly abundant circRNA, was proposed to be involved in tumorigenesis. However, the role of circ-CCT3 in hepatocellular carcinoma remains elusive.Methods: Here, circ-CCT3 (a circRNA derived from exons 3, 4 and 5 of the CCT3 gene, hsa_circ_0004680) was identified by circRNA microarray and validated by qRT-PCR. RNA immunoprecipitation (RIP) was performed to confirm the binding between ALKBH5 along with METTL3 and circ-CCT3. Methylated RNA Immunoprecipitation (MeRIP) was used to detect the N6-methyladenosine (m 2A) levels of circ-CCT3. CircRNAs in vivo precipitation, luciferase reporter assay, biotin-coupled microRNA capture, and fluorescence in situ hybridization were conducted to assess the interaction between circ-CCT3 and miR-378a-3p. The functions of circ-CCT3 in HCC were evaluated both in vitro and in vivo.Results: We demonstrated that circ-CCT3 was highly expressed in HCC which indicated the poor prognosis. Circ-CCT3 expression served as an independent risk factor for overall survival in patients with HCC. Knocking-down of circ-CCT3 inhibited the proliferation, invasion and migration of HCC cells, and angiogenesis of HUVEC. Mechanistically, ALKBH5 and METTL3 could bind and regulate m A-modification of circ-CCT3. Further, circ-CCT3 upregulated the expression of FLT-1 by sponging miR-378a-3p.Conclusions: Circ-CCT3 was significantly up-regulated in HCC and promoted liver cancer development via miR-378a-3p-FLT1 axis. It was also found that circ-CCT3 was under m A-modification mediated by ALKBH5 and METTL3. Our study highlights circ-CCT3 as a potential therapeutic target of HCC treatment, which provides a novel understanding on mechanisms of circRNAs in HCC progression.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , Humans , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Chaperonin Containing TCP-1/genetics , Chaperonin Containing TCP-1/metabolism , DNA Methylation , Gene Expression Regulation, Neoplastic , In Situ Hybridization, Fluorescence , Liver Neoplasms/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics
7.
Adv Sci (Weinh) ; 10(15): e2206669, 2023 05.
Article in English | MEDLINE | ID: mdl-36994647

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common malignancies with poor prognosis, and novel treatment strategies are urgently needed. Mitochondria are key regulators of cellular homeostasis and potential targets for tumor therapy. Here, the role of mitochondrial translocator protein (TSPO) in the regulation of ferroptosis and antitumor immunity is investigated and the potential therapeutic implications for HCC are assessed. TSPO is highly expressed in HCC and associated with poor prognosis. Gain- and loss-of-function experiments present that TSPO promotes HCC cell growth, migration, and invasion in vitro and in vivo. In addition, TSPO inhibits ferroptosis in HCC cells via enhancing the Nrf2-dependent antioxidant defense system. Mechanistically, TSPO directly interacts with P62 and interferes with autophagy, leading to the accumulation of P62. The P62 accumulation competes with KEAP1, preventing it from targeting Nrf2 for proteasomal degradation. Furthermore, TSPO promotes HCC immune escape by upregulating PD-L1 expression through Nrf2-mediated transcription. Notably, TSPO inhibitor PK11195 combines with anti-PD-1 antibody showing a synergistic anti-tumor effect in a mouse model. Overall, the results demonstrated that mitochondrial TSPO promotes HCC progression by inhibiting ferroptosis and antitumor immunity. Targeting TSPO can be a promising new strategy for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular , Ferroptosis , Liver Neoplasms , Animals , Mice , Carcinoma, Hepatocellular/metabolism , Carrier Proteins , Immune Evasion , Kelch-Like ECH-Associated Protein 1/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Mitochondrial Proteins/metabolism , NF-E2-Related Factor 2/metabolism
8.
Biochem Biophys Res Commun ; 640: 1-11, 2023 01 15.
Article in English | MEDLINE | ID: mdl-36495604

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) was one of the most prevalent life-threatening cancers. Metastasis is the leading cause of cancer-related death in HCC. MiRNAs play essential roles in cancer metastasis. METHODS: Expression of miR-652-3p in HCC was assessed. Function experiments of miR-652-3p and trinucleotide repeat-containing gene 6A protein (TNRC6A) were performed both in vitro and in vivo. mRNA sequencing, PCR, and western blot were performed to verify the target genes and pathway of miR-652-3p. The lung metastasis and xenograft cancer model in nude mice was established to investigate the effects of the miR-652-3p and TRNC6A on tumor metastasis in vivo. The relationship between the expression of the miR-652-3p, TNRC6A and the prognosis of HCC patients was analyzed. RESULTS: Upregulated miR-652-3p was found in the tumor tissues of HCC, especially in metastatic HCC patients. Overexpression of miR-652-3p promoted and knockdown of miR-652-3p suppressed HCC metastasis both in vitro and in vivo. MiR-652-3p promoted HCC metastasis via regulating the EMT pathway. TNRC6A was identified as a direct target of miR-652-3p, and the knockdown of TNRC6A restored repressed EMT and HCC metastasis caused by the inhibition of miR-652-3p. Clinical results revealed that high expression of miR-652-3p and low expression of TNRC6A were positively correlated to shortened overall survival and disease-free survival in HCC patients. CONCLUSIONS: MiR-652-3p promotes EMT and HCC metastasis by inhibiting TNRC6A expression in HCC. MiR-652-3p and TNRC6A may serve as potential biomarkers to predict prognosis in HCC patients with metastasis.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , Animals , Humans , Mice , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/pathology , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Metastasis
10.
BMC Gastroenterol ; 21(1): 195, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33931011

ABSTRACT

BACKGROUND: Donor age is an important predictor for liver transplant recipients. Studies have not fully explored its impact on transplant outcomes in hepatocellular carcinoma (HCC) patients as well as its involvement in tumor recurrence. METHODS: HCC patients who received liver transplants during 2010-2017 from the Scientific Registry of Transplant Recipients database were included. The recipients were divided into four groups based on donor age: I (≤ 34 years), II (35-49 years), III (50-64 years), and IV (≥ 65 years). Transplant outcomes, including the overall survival (OS), tumor recurrence, and risks, were evaluated. RESULTS: A total of 13,276 HCC recipients were included in this study. Statistical significant differences were observed in OS among the four groups. The best 5-year survival was 76.0% in group I, followed by 73.5% in group II, 72.8% in group III, and 69.2% in group IV (P < 0.001). However, the liver-specific survival did not differ among these groups (P = 0.260). Donor age was found to be the independent predictor of OS after adjusting for other variables (P < 0.001, ref. group I; 1.087 (0.979-1.208) for group II, P = 0.119; 1.124 (1.015-1.246) for group III, P = 0.025; 1.395 (1.215-1.602) for group IV, P < 0.001). In subgroup analysis, OS was significantly different in recipients with hepatitis C virus (HCV), but there was no significant difference for recipients with hepatitis B virus (HBV), alcoholic liver diseases and nonalcoholic steatohepatitis (NASH). The post-transplant cumulative tumor recurrence rates were similar among the four groups (P = 0.382). CONCLUSIONS: Older donor age was associated with decreased OS but not liver-specific survival as well as post-transplant tumor recurrence in HCC recipients. Donor age also had different effects in patients with different underlying liver diseases.


Subject(s)
Carcinoma, Hepatocellular , Hepatitis C , Liver Neoplasms , Liver Transplantation , Adult , Carcinoma, Hepatocellular/surgery , Humans , Liver Neoplasms/surgery , Neoplasm Recurrence, Local , Retrospective Studies
11.
Langmuir ; 37(2): 969-977, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33393784

ABSTRACT

Although with high antioxidant activity, epigallocatechin-3-gallate (EGCG) was restricted by its poor chemical stability in practical applications. One of EGCG derivatives, EGCG palmitate, was synthesized with EGCG and palmitoyl chloride to overcome instability of EGCG. However, uncertainties still exist in chemical stability and cytotoxicity of EGCG palmitate, which are essential for further exploration in anticancer therapy. Our work aims to analyze the resistance of EGCG palmitate to oxidation and summarize its targeted inhibition efficiency on cancerous cells and normal cells. High-performance liquid chromatography analysis confirmed that EGCG palmitate remained stable in air and Dulbecco's modified eagle medium (DMEM) for a longer time than EGCG. Antioxidative and pro-oxidative effects of EGCG palmitate on treated cells are proposed through reactive oxygen species (ROS) detection, respectively. It reveals that pro-oxidants by H2O2 production can exert antiproliferative and proapoptotic effects on cancerous cells and stimulate autophagy, while an antioxidant relieves oxidative stress caused by superoxide as compared to normal cells. Consequently, targeted cytotoxicity is adopted by EGCG palmitate-treated cancerous cells. Results above manifest that EGCG palmitate possesses potential to serve as a promising prodrug in anticancer treatment.


Subject(s)
Catechin , Hydrogen Peroxide , Antioxidants/pharmacology , Catechin/analogs & derivatives , Catechin/pharmacology , Palmitates/toxicity , Reactive Oxygen Species
12.
Biomater Sci ; 9(3): 590-625, 2021 Feb 09.
Article in English | MEDLINE | ID: mdl-33305765

ABSTRACT

Nanoparticle-based therapeutic and detectable modalities can augment anticancer efficiency, holding potential in capable target and suppressive metastases post administration. However, the individual discrepancies of the current "one-size-fits-all" strategies for anticancer nanotherapeutics have heralded the need for "personalized therapy". Benefiting from the special inherency of various cells, diverse cell membrane-coated nanoparticles (CMCNs) were established on a patient-by-patient basis, which would facilitate the personalized treatment of individual cancer patients. CMCNs in a complex microenvironment can evade the immune system and target homologous tumors with a suppressed immune response, as well as a prolonged circulation time, consequently increasing the drug accumulation at the tumor site and anticancer therapeutic efficacy. This review focuses on the emerging strategies and advances of CMCNs to synergistically integrate the merit of source cells with nanoparticulate delivery systems for the orchestration of personalized anticancer nanotherapeutics, thus discussing their rationalities in facilitating chemotherapy, imaging, immunotherapy, phototherapy, radiotherapy, sonodynamic, magnetocaloric, chemodynamic and gene therapy. Furthermore, the mechanism, challenges and opportunities of CMCNs in personalized anticancer therapy were highlighted to further boost cooperation from different fields, including materials science, chemistry, medicine, pharmacy and biology for the lab-to-clinic translation of CMCNs combined with the individual advantages of source cells and nanotherapeutics.


Subject(s)
Nanoparticles , Neoplasms , Biomimetics , Cell Membrane , Drug Delivery Systems , Humans , Neoplasms/drug therapy , Tumor Microenvironment
13.
Oncogene ; 39(42): 6529-6543, 2020 10.
Article in English | MEDLINE | ID: mdl-32917956

ABSTRACT

Exosomes play an important role in intercellular communication and metastatic progression of hepatocellular carcinoma (HCC). However, cellular communication between heterogeneous HCC cells with different metastatic potentials and the resultant cancer progression are not fully understood in HCC. Here, HCC cells with high-metastatic capacity (97hm and Huhm) were constructed by continually exerting selective pressure on primary HCC cells (MHCC-97H and Huh7). Through performing exosomal miRNA sequencing in HCC cells with different metastatic potentials (MHCC-97H and 97hm), many significantly different miRNA candidates were found. Among these miRNAs, miR-92a-3p was the most abundant miRNA in the exosomes of highly metastatic HCC cells. Exosomal miR92a-3p was also found enriched in the plasma of HCC patient-derived xenograft mice (PDX) model with high-metastatic potential. Exosomal miR-92a-3p promotes epithelial-mesenchymal transition (EMT) in recipient cancer cells via targeting PTEN and regulating its downstream Akt/Snail signaling. Furthermore, through mRNA sequencing in HCC cells with different metastatic potentials and predicting potential transcription factors of miR92a-3p, upregulated transcript factors E2F1 and c-Myc were found in high-metastatic HCC cells promote the expression of cellular and exosomal miR-92a-3p in HCC by directly binding the promoter of its host gene, miR17HG. Clinical data showed that a high plasma exosomal miR92a-3p level was correlated with shortened overall survival and disease-free survival, indicating poor prognosis in HCC patients. In conclusion, hepatoma-derived exosomal miR92a-3p plays a critical role in the EMT progression and promoting metastasis by inhibiting PTEN and activating Akt/Snail signaling. Exosomal miR92a-3p is a potential predictive biomarker for HCC metastasis, and this may provoke the development of novel therapeutic and preventing strategies against metastasis of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Exosomes/metabolism , Liver Neoplasms/genetics , MicroRNAs/metabolism , PTEN Phosphohydrolase/genetics , Animals , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/secondary , Cell Line, Tumor , Cell Movement/genetics , Disease-Free Survival , E2F1 Transcription Factor/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/blood , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Mice , MicroRNAs/genetics , Middle Aged , Neoplasm Metastasis/genetics , PTEN Phosphohydrolase/metabolism , Prognosis , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/genetics , Up-Regulation , Xenograft Model Antitumor Assays
14.
Mol Cancer ; 19(1): 123, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32772918

ABSTRACT

BACKGROUND: N6-methyladenosine (m6A) modification is an emerging layer of epigenetic regulation which is widely implicated in the tumorigenicity of hepatocellular carcinoma (HCC), offering a novel perspective for investigating molecular pathogenesis of this disease. The role of AlkB homolog 5 (ALKBH5), one of the m6A demethylases, has not been fully explored in HCC. Here we clarify the biological profile and potential mechanisms of ALKBH5 in HCC. METHODS: Expression of ALKBH5 and its correlation with clinicopathological characteristics of HCC were evaluated using tissue microarrays and online datasets. And biological effects of ALKBH5 in HCC were determined in vitro and in vivo. Subsequently, methylated RNA immunoprecipitation sequencing (MeRIP-seq) combined with RNA sequencing (RNA-seq), and following m6A dot blot, MeRIP-qPCR, RIP-qPCR or dual luciferase reporter assays were employed to screen and validate the candidate targets of ALKBH5. RESULTS: We demonstrated that ALKBH5 was down-regulated in HCC, and decreased ALKBH5 expression was an independent prognostic factor of worse survival in HCC patients. Functionally, ALKBH5 suppressed the proliferation and invasion capabilities of HCC cells in vitro and in vivo. Mechanistically, ALKBH5-mediated m6A demethylation led to a post-transcriptional inhibition of LY6/PLAUR Domain Containing 1 (LYPD1), which could be recognized and stabilized by the m6A effector IGF2BP1. In addition, we identified that LYPD1 induced oncogenic behaviors of tumors in contrast to ALKBH5. Dysregulation of ALKBH5/LYPD1 axis impelled the progression of HCC. CONCLUSION: Our study reveals that ALKBH5, characterized as a tumor suppressor, attenuates the expression of LYPD1 via an m6A-dependent manner in HCC cells. Our findings enrich the landscape of m6A-modulated tumor malignancy, and provide new insights into potential biomarkers and therapeutic targets of HCC treatment.


Subject(s)
Adenosine/analogs & derivatives , AlkB Homolog 5, RNA Demethylase/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Epigenesis, Genetic , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Tumor Suppressor Proteins/genetics , Adenosine/metabolism , Animals , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Carrier Proteins , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Liver Neoplasms/pathology , Mice , Models, Biological , Prognosis , Protein Binding , Signal Transduction
15.
Biomater Sci ; 8(5): 1380-1393, 2020 Mar 07.
Article in English | MEDLINE | ID: mdl-31916560

ABSTRACT

To achieve highly systemic therapeutic efficacy, chemotherapy is combined with photothermal therapy for chemo-photothermal synergistic therapy; however, this strategy suffers from high toxicity and unsatisfactory sensitivity for cancer cells. Herein, we developed a pH- and photothermal-responsive zeolitic imidazolate framework (ZIF-8) compound for loading a dual-drug in the tumor site and improving their curative effects. Since autophagy always accompanies tumor progression and metastasis, there is an unmet need for an anticancer treatment related to the regulation of autophagy. Green tea polyphenols, namely, (-)-epigallocatechin-3-gallate (EGCG) and doxorubicin (DOX), both of which exhibit anticancer activity, were dual-loaded via polydopamine (PDA) coating ZIF-8 (EGCG@ZIF-PDA-PEG-DOX, EZPPD for short) through hierarchical self-assembly. PDA could transfer photothermal energy to increase the temperature under near-infrared (NIR) laser irradiation. Due to its pH-response, EZPPD released EGCG and DOX in the tumor microenvironment, wherein the temperature increased with the help of PDA and NIR laser irradiation. The duo of DOX and EGCG induced autophagic flux and accelerated the formation of autophagosomes. In a mouse HeLa tumor model, photothermal-chemotherapy could ablate the tumor with a significant synergistic effect and potentiate the anticancer efficacy. Thus, the results indicate that EZPPD renders the key traits of a clinically promising candidate to address the challenges associated with synergistic chemotherapy and photothermal utilization in antitumor therapy.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antioxidants/pharmacology , Catechin/analogs & derivatives , Doxorubicin/pharmacology , Indoles/chemistry , Polymers/chemistry , Uterine Cervical Neoplasms/therapy , Zeolites/chemistry , Animals , Antibiotics, Antineoplastic/chemical synthesis , Antibiotics, Antineoplastic/chemistry , Antioxidants/chemical synthesis , Antioxidants/chemistry , Catechin/chemical synthesis , Catechin/chemistry , Catechin/pharmacology , Doxorubicin/chemical synthesis , Doxorubicin/chemistry , Female , HeLa Cells , Humans , Mice , Mice, Nude , Phototherapy , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
16.
Medicine (Baltimore) ; 98(44): e17554, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31689759

ABSTRACT

INTRODUCTION: Solid pseudopapillary tumors (SPT) account for 1% to 3% of all pancreatic tumors. They have low malignant potential with a favorable prognosis, and predominantly occur in young women. The pathogenesis and clinical behavior of SPT are still uncertain. In addition, most ruptures of SPT were associated with blunt abdominal trauma, while spontaneous ruptures seemed to be quite rare. Up to now, there have been only 3 spontaneous ruptured SPT cases reported worldwide. PATIENT CONCERNS: Here, we reported a 22-year-old female patient with left lower abdominal pain. Computed tomography (CT) showed that a hemorrhagic complex solid cystic mass located in the lesser omentum sac. DIAGNOSIS: According to pathological findings of tumor specimen, the diagnosis of solid pseudopapillary tumor (SPT) of the pancreas was made. INTERVENTIONS: Distal pancreatectomy and splenectomy was carried out. OUTCOMES: The patient recovered to normal status within 10 days after surgery. CONCLUSION: Besides, we reviewed about 50 cases in literatures to find out the clinical characteristics and differential diagnostic strategies of SPT.


Subject(s)
Pancreatic Neoplasms/pathology , Female , Humans , Pancreatectomy , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/surgery , Rupture, Spontaneous , Splenectomy , Young Adult
17.
Am J Transl Res ; 11(8): 4713-4725, 2019.
Article in English | MEDLINE | ID: mdl-31497193

ABSTRACT

Dimethyl fumarate (DMF) is generally used to treat psoriasis and multiple sclerosis. In the present study, we aimed to investigate the effects of DMF on hepatocellular carcinoma progression and its mechanism of action. In vitro, cell viability was examined using CCK-8 assay; cell cycle was analyzed by flow cytometry; angiogenesis was detected using tube formation assay; and autophagic flux assay results were examined using fluorescence microscopy. We also used western blotting to explore the potential mechanisms. In vivo, tumor xenograft experiment was performed with nude mice, and liver function, renal function, and routine blood counts were assessed using biochemical tests. Dimethyl fumarate inhibited tumor growth and angiogenesis in hepatocellular carcinoma, both in vitro and in vivo. Dimethyl fumarate decreased autophagy in hepatocellular carcinoma cells. Treatment with DMF activated SOCS3, which led to repression of JAK1 and STAT3 phosphorylation. DMF inhibited cell proliferation, angiogenesis, and autophagy via activation of the SOCS3/JAK1/STAT3 signaling pathway. This finding may provide a novel approach for the treatment of hepatocellular carcinoma.

18.
J Mater Chem B ; 7(42): 6476-6487, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31465082

ABSTRACT

Liver cancer is a kind of lethal and aggressive malignant neoplasm with a high rate of relapse and metastasis after therapy. An important cause for the relapse and metastasis is the existence of liver cancer stem cells (CSCs), which have high resistance to chemotherapy and high tumorigenic potential. Therefore, it is crucial to develop new methods to eradicate CSCs in tumors. Herein, we develop a photodynamic therapy (PDT) that features bimodal metallacage-loaded nanoparticles (MNPs) for integrated chemotherapy. This platform achieves chemo-photodynamic combinational therapy. Organoplatinum(ii) metallacage-loaded nanoparticles show excellent ability to kill liver CSCs, decreasing their mobility and sphenoid formation ability under near-infrared laser irradiation. Importantly, MNPs can successfully penetrate into 3D tumor spheroids, which display higher drug resistance compared to traditional 2D cultured cells. This destroys CSCs and prevents subsequent tumor formation in vivo. With the excellent combinational therapeutic results in hand, the working mechanisms of MNPs were then studied. MNPs under NIR light irradiation can generate reactive oxygen species (ROS), resulting in damage of mitochondrial membrane and subsequent cell apoptosis with chemotherapeutic platinum. This study proves the great potential of MNPs for combinational cancer therapy, providing a new insight for the next generation of nanomedicines.


Subject(s)
Antineoplastic Agents/therapeutic use , Liver Neoplasms/drug therapy , Metal Nanoparticles/therapeutic use , Neoplastic Stem Cells/drug effects , Photosensitizing Agents/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/radiation effects , Apoptosis/drug effects , Cell Line, Tumor , Coordination Complexes/chemistry , Coordination Complexes/radiation effects , Coordination Complexes/therapeutic use , Humans , Infrared Rays , Liver/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Metal Nanoparticles/chemistry , Metal Nanoparticles/radiation effects , Mice, Nude , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/radiation effects , Platinum/chemistry , Platinum/radiation effects , Porphyrins/chemistry , Porphyrins/radiation effects , Porphyrins/therapeutic use , Reactive Oxygen Species/metabolism , Spheroids, Cellular/drug effects , Xenograft Model Antitumor Assays
19.
EBioMedicine ; 44: 225-236, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31109829

ABSTRACT

BACKGROUND: Myb-binding protein 1A (Mybbp1a) is a nucleolar protein that can regulate rRNA metabolism, the stress response and carcinogenesis. However, the function of Mybbp1a in the progression of hepatocellular carcinoma (HCC) is unclear. We aimed to determine the role of Mybbp1a in HCC and the underlying mechanism. METHODS: We investigated the function of Mybbp1a in HCC cell models and the xenograft mouse model. The relationship between Mybbp1a and IGFBP5 was found through expression profile chip. The molecular mechanism of Mybbp1a regulating IGFBP5 was proved through CO-IP, CHIP, Bisulfite Sequencing and Pyrosequencing. FINDINGS: In this study, we observed that Mybbp1a was overexpressed in HCC tissues and associated with the poor prognosis of HCC patients. Suppression of Mybbp1a led to a reduction in the proliferation and migration ability of HCC cells through inhibiting the IGF1/AKT signaling pathway. Further study found that Mybbp1a could form a complex with DNMT1 and induce aberrant hyper-methylation of CpG islands of IGFBP5, which inhibits secretion of IGFBP5 and then activates IGF1/AKT signaling pathway. INTERPRETATION: These findings extend our understanding of the function of Mybbp1a in the progression of HCC. The newly identified Mybbp1a may provide a novel biomarker for developing potential therapeutic targets of HCC. FUND: Science Technology Department of Zhejiang Province (No. 2015C03034), National Health and Family Planning Commission of China (No. 2016138643), Innovative Research Groups of National Natural Science Foundation of China (No. 81721091), Major program of National Natural Science Foundation of China (No. 91542205).


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Insulin-Like Growth Factor I/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Nuclear Proteins/antagonists & inhibitors , Nucleocytoplasmic Transport Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Biomarkers, Tumor , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/mortality , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Movement , Cell Proliferation , CpG Islands , DNA Methylation , DNA-Binding Proteins , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Insulin-Like Growth Factor Binding Protein 5 , Liver Neoplasms/drug therapy , Liver Neoplasms/mortality , Mice , Models, Biological , Prognosis , RNA-Binding Proteins , Transcription Factors , Xenograft Model Antitumor Assays
20.
Int J Oncol ; 52(4): 1235-1245, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29532867

ABSTRACT

Intrahepatic cholangiocarcinoma (ICC) is a fatal, malignant tumor of the liver; effective diagnostic biomarkers and therapeutic targets for ICC have not been identified yet. High expression of H2A histone family member Z (H2A.Z) is a high-risk factor for poor prognosis in patients with breast cancer and primary hepatocellular cancer. However, the significance of H2A.Z and its expression in ICC remains unknown. The present study demonstrated that H2A.Z is overexpressed in ICC and expression of H2A.Z correlated with poor prognosis in patients with ICC. H2A.Z regulated cell proliferation in vitro and in vivo via H2A.Z/S-phase kinase-associated protein 2/p27/p21 signaling. Inhibition of H2A.Z reduced cell proliferation and induced apoptosis in ICC. In addition, downregulation of H2AZ reduced tumor metastasis by repressing epithelial-mesenchymal transition and enhanced the antitumor effects of cisplatin in the treatment of ICC. Overall, H2A.Z promoted cell proliferation and epithelial-mesenchymal transition in ICC, suggesting that H2A.Z may be a novel biomarker and therapeutic target for ICC.


Subject(s)
Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/metabolism , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/metabolism , Cisplatin/pharmacology , Histones/biosynthesis , Animals , Antineoplastic Agents/pharmacology , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Carcinogenesis , Cell Line, Tumor , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Drug Resistance, Neoplasm , Heterografts , Histones/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL
...