Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Hepatol Int ; 2024 Jan 06.
Article in English | MEDLINE | ID: mdl-38184503

ABSTRACT

BACKGROUND AND AIMS: Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease worldwide. In this study, we aimed to investigate the role and regulatory mechanism of Annexin A2 (ANXA2) in the pathogenesis of NAFLD. METHODS: Histological analyses and ELISA were used to illuminate the expression of ANXA2 in NAFLD and healthy subjects. The role of ANXA2 was evaluated using high-fat diet (HFD)-fed mice via vein injection of adeno-associated viruses (AAV) knocking down ANXA2 or non-targeting control (NC) shRNAs. Moreover, HepG2 and LO2 cells were employed as in vitro hepatocyte models to investigate the expression and function of ANXA2. RESULTS: ANXA2 was confirmed to be one of three hub genes in liver injury, and its expression was positively correlated with NAFLD activity score (NAS) and macrophage infiltration in NAFLD. Moreover, ANXA2 was significantly upregulated in NAFLD patients and HFD-fed mice. LPS/TLR4 pathway strongly upregulated ANXA2 expression, which is mediated by direct ANXA2 promoter binding by TLR4 downstream NF-κB p65 and c-Jun transcription factors. Increased ANXA2 expression was correlated with decreased autophagy flux and autophagy was activated by the depletion of ANXA2 in the models of NAFLD. Furthermore, ANXA2 interference led to the activation of AMPK/mTOR signaling axis, which may play a causal role in autophagy flux and the amelioration of steatosis. CONCLUSIONS: ANXA2 is a pathological predictor and promising therapeutic target for NAFLD. ANXA2 plays a crucial role in linking inflammation to hepatic metabolic disorder and injury, mainly through the blockage of AMPK/mTOR-mediated lipophagy.

3.
Cells ; 11(19)2022 09 27.
Article in English | MEDLINE | ID: mdl-36230979

ABSTRACT

Besides the canonical function in ribosome biogenesis, there have been significant recent advances towards the fascinating roles of the nucleolus in stress response, cell destiny decision and disease progression. Nucleolar stress, an emerging concept describing aberrant nucleolar structure and function as a result of impaired rRNA synthesis and ribosome biogenesis under stress conditions, has been linked to a variety of signaling transductions, including but not limited to Mdm2-p53, NF-κB and HIF-1α pathways. Studies have uncovered that nucleolus is a stress sensor and signaling hub when cells encounter various stress conditions, such as nutrient deprivation, DNA damage and oxidative and thermal stress. Consequently, nucleolar stress plays a pivotal role in the determination of cell fate, such as apoptosis, senescence, autophagy and differentiation, in response to stress-induced damage. Nucleolar homeostasis has been involved in the pathogenesis of various chronic diseases, particularly tumorigenesis, neurodegenerative diseases and metabolic disorders. Mechanistic insights have revealed the indispensable role of nucleolus-initiated signaling in the progression of these diseases. Accordingly, the intervention of nucleolar stress may pave the path for developing novel therapies against these diseases. In this review, we systemically summarize recent findings linking the nucleolus to stress responses, signaling transduction and cell-fate decision, set the spotlight on the mechanisms by which nucleolar stress drives disease progression, and highlight the merit of the intervening nucleolus in disease treatment.


Subject(s)
NF-kappa B , Tumor Suppressor Protein p53 , Cell Nucleolus/metabolism , Disease Progression , Humans , NF-kappa B/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
4.
Cancers (Basel) ; 14(15)2022 Jul 30.
Article in English | MEDLINE | ID: mdl-35954387

ABSTRACT

Our understanding of the fascinating connection between nervous system and gastrointestinal (GI) tumorigenesis has expanded greatly in recent years. Recent studies revealed that neurogenesis plays an active part in GI tumor initiation and progression. Tumor-driven neurogenesis, as well as neurite outgrowth of the pre-existing peripheral nervous system (PNS), may fuel GI tumor progression via facilitating cancer cell proliferation, chemoresistance, invasion and immune escape. Neurotransmitters and neuropeptides drive the activation of various oncogenic pathways downstream of neural receptors within cancer cells, underscoring the importance of neural signaling pathways in GI tumor malignancy. In addition, neural infiltration also plays an integral role in tumor microenvironments, and contributes to an environment in favor of tumor angiogenesis, immune evasion and invasion. Blockade of tumor innervation via denervation or pharmacological agents may serve as a promising therapeutic strategy against GI tumors. In this review, we summarize recent findings linking the nervous system to GI tumor progression, set the spotlight on the molecular mechanisms by which neural signaling fuels cancer aggressiveness, and highlight the importance of targeting neural mechanisms in GI tumor therapy.

5.
J Mol Histol ; 53(4): 645-655, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35717490

ABSTRACT

Recent investigations indicate that ß2-adrenergic receptor (ß2-AR) signaling may facilitate the progression of various tumors, whose underlying mechanisms remain largely elusive. In the present study, we showed that ß2-AR recruited Cdc42 in response to isoproterenol (ISO, a ß-AR selective agonist) exposure in pancreatic ductal adenocarcinoma (PDAC) cells. The association of ß2-AR and Cdc42 promoted the activation of Cdc42, as revealed by increased levels of Cdc42-GTP, and co-incubation with ß2-AR antagonist abrogated ISO-induced activation of Cdc42. ß2-AR-mediated Cdc42 activation further led to the phosphorylation of downstream PAK1, LIMK1 and Merlin. Furthermore, we showed that the activation of ß2-AR/Cdc42 signaling facilitated the migration and invasion of PDAC cells. In addition, ß2-AR and Cdc42 were overexpressed in PDAC specimens, compared with adjacent non-tumor tissues. High expression of ß2-AR and Cdc42 were correlated with lymph node metastasis and TNM stage in PDAC patients. Finally, we showed that overexpression of ß2-AR and Cdc42 were indicative of unfavorable prognosis in PDAC patients. Taken together, our findings suggested that ß2-AR might facilitate Cdc42 signaling to drive the migration and invasion of PDAC cells, consequently resulting in the metastasis and dismal prognosis of PDAC. These studies highlight targeting ß2-AR/Cdc42 signaling as a therapeutic strategy against PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Lim Kinases/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Receptors, Adrenergic, beta-2 , Signal Transduction , Pancreatic Neoplasms
6.
Front Oncol ; 12: 850767, 2022.
Article in English | MEDLINE | ID: mdl-35444953

ABSTRACT

Background: Colony stimulating factor 1 receptor (CSF-1R) is a single channel III transmembrane receptor tyrosine kinase (RTK) and plays an important role in immune regulation and the development of various cancer types. The expression of CSF-1R in colon adenocarcinoma (COAD) and its prognostic value remain incompletely understood. Therefore, we aim to explore the prognostic value of CSF-1R in COAD and its relationship with tumor immunity. Methods: CSF-1R expression in a COAD cohort containing 103 patients was examined using immunohistochemistry (IHC). The relationship between CSF-1R expression and clinicopathological parameters and prognosis was evaluated. Dual immunofluorescence staining was conducted to determine the localization of CSF-1R in COAD tissues. Univariate and multivariate Cox regression analysis were performed to evaluate independent prognostic factors. Transcriptomic profiles of CSF-1Rhigh and CSF-1Rlow tumor-associated macrophages (TAMs) were investigated. Gene enrichment analysis was used to explore the signal pathways related to CSF-1R. In addition, the relationship between CSF-1R in tumor microenvironment (TME) and tumor immunity was also studied. Results: IHC analysis showed that CSF-1R was overexpressed in COAD, and higher expression was associated with shorter overall survival (OS). Immunofluorescence staining showed that CSF-1R was co-localized with macrophage marker CD68. Univariate and multivariate Cox regression analysis showed that CSF-1R was an independent prognostic factor for COAD. The results of gene enrichment analysis showed that CSF-1R was involved in tumor immune response and regulation of TME. In addition, CSF-1R was significantly correlated with TME, immune cell infiltration, TMB, MSI, Neoantigen, and immune checkpoint molecules. Conclusion: CSF-1R can serve as an independent prognostic factor of COAD and promising immunotherapeutic target of COAD.

7.
Urol Oncol ; 40(4): 166.e15-166.e25, 2022 04.
Article in English | MEDLINE | ID: mdl-34998671

ABSTRACT

BACKGROUND: CD47 has been identified as a phagocytosis checkpoint conferring poor clinical outcomes in various cancer types. A flurry of clinical trials designed to evaluate agents that block CD47 have been initiated. We aimed to explore the clinical significance of CD47 and its correlation with immune infiltration and molecular features in clear cell renal cell carcinoma (ccRCC). METHODS: 235 tumor tissue microarray specimens of ccRCC patients from Zhongshan Hospital, 530 ccRCC patients from The Cancer Genome Atlas and 726 ccRCC patients from JAVELIN Renal 101 study were analyzed. CD47 expression and immune contexture were examined by immunohistochemistry and CIBERSORT algorithm. Survival analyses were conducted through Kaplan-Meier curves and Cox regression model. RESULTS: We demonstrated that ccRCC patients with high CD47 expression exhibited inferior overall survival and recurrence-free survival. CD47 expression associated with heavily immune infiltrated but immunosuppressed microenvironment. CD8+ T cells infiltration had discordant prognostic value based on CD47 expression, where high CD8+ T cell infiltration was associated with worse clinical outcome in CD47hi patients and with favorable prognosis in CD47lo patients. Patients with mutated PBRM1 and SETD2 correlated with decreased CD47 mRNA expression. Patients with higher CD47 expression possessed improved PFS in ICI + VEGFR TKI combination therapy. CONCLUSIONS: CD47 expression was an independent prognosticator of clinical outcome for ccRCC patients. CD47 expression correlated with ccRCC molecular classification and response to combination therapy. The phagocytosis checkpoint CD47 could be applied as an attractive candidate for immunotherapeutic approach in ccRCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Biomarkers, Tumor/metabolism , CD47 Antigen/genetics , CD47 Antigen/therapeutic use , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Female , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Prognosis , Tumor Microenvironment
8.
J Appl Toxicol ; 42(5): 806-817, 2022 05.
Article in English | MEDLINE | ID: mdl-34687223

ABSTRACT

Perfluorooctane sulfonate (PFOS) is a fluorinated organic pollutant with substantial accumulation in mammalian liver tissues. However, the impact of chronic PFOS exposure on liver disease progression and the underlying molecular mechanisms remain elusive. Herein, we for the first time revealed that micromolar range of PFOS exposure initiates the activation of NLR pyrin domain containing 3 (NLRP3) inflammasome to drive hepatocyte pyroptosis. We showed that 5 mg/kg/day PFOS exposure may exacerbated liver inflammation and steatosis in high-fat diet (HFD)-fed mice with concurrently elevated expression of NLRP3 and caspase-1. PFOS exposure resulted in viability impairment and LDH release in BRL-3A rat liver cells. 25-100 µM concentrations of PFOS exposure activated the NLRP3 inflammasome, leading to consequent GSDMD cleavage, IL-1ß release and the initiation of pyroptosis in a dose-dependent manner, whereas treatment with 10 µM NLRP3 inhibitor MCC950 abrogated this effect. Moreover, pretreatment of 5 mM ROS scavenger N-acetyl-L-cysteine (NAC) ameliorated PFOS-induced NLRP3 inflammasome activation and pyroptosis. Collectively, our data highlight a pivotal role of pyroptotic death in PFOS-mediated liver inflammation and metabolic disorder.


Subject(s)
Inflammasomes , Pyroptosis , Alkanesulfonic Acids , Animals , Fluorocarbons , Hepatocytes , Inflammasomes/metabolism , Inflammation/chemically induced , Liver/metabolism , Mammals/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Rats , Reactive Oxygen Species/metabolism
9.
Cancer Immunol Immunother ; 71(6): 1497-1506, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34716763

ABSTRACT

BACKGROUND: Muscle-invasive bladder cancer (MIBC) is an aggressive and heterogeneous malignancy. Tumor-associated macrophages (TAMs) are key infiltrating cell populations in the inflammatory microenvironment of malignant tumors including MIBC. It intrigues us to explore the clinical significance and immunoregulatory role of TAMs infiltration and polarization in MIBC. METHODS: A total of 141 patients with MIBC from Zhongshan Hospital and 391 patients with MIBC from The Cancer Genome Atlas (TCGA) database were included in this study. Moreover, 195 patients who received anti-PD-L1 therapy from the IMvigor210 trial were enrolled. Patients were categorized into three subtypes considering the infiltration level and polarization status of TAMs, denoted as TAMlow (Subtype I), TAMhigh&M2/M1low (Subtype II), and TAMhigh&M2/M1high (Subtype III). RESULTS: Subtype III suffered inferior prognosis, and Subtype II could benefit more from adjuvant chemotherapy (ACT). Subtype III was featured with increased pro-tumor cells and immunosuppressive cytokines, while Subtype II possessed more immunogenic cells infiltration with activated and tumoricidal properties. Subtype II and Subtype III presented basal/squamous-like characterization and showed additional prognostic merit beyond molecular classification. Subtype I exhibited elevated level of FGFR3 signature, while Subtype II had EGFR signaling activation and immunotherapeutic indication. Additionally, Subtype II patients were indeed highly sensitive to PD-L1 blockade therapy in IMvigor210 trial. CONCLUSION: The infiltration and polarization status of TAMs shaped distinct immune microenvironment with predictive significance for survival outcome, ACT benefit, and PD-L1 blockade therapy sensitivity in MIBC. Immune classification based on TAMs polarization and infiltration might provide tools to tailor chemotherapy and immunotherapy.


Subject(s)
Urinary Bladder Neoplasms , B7-H1 Antigen/therapeutic use , Humans , Muscles/pathology , Prognosis , Tumor Microenvironment , Tumor-Associated Macrophages , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics
10.
Eur J Cancer ; 157: 114-123, 2021 11.
Article in English | MEDLINE | ID: mdl-34508993

ABSTRACT

AIM: CD73 overexpression has been reported in several malignancies and is considered to be a novel immune checkpoint. However, the role and significance of CD73 in gastric cancer (GC) still remain obscure. We aim to investigate the role of CD73 expression in predicting prognosis, shaping immune contexture and guiding therapeutic strategy in GC. METHODS: The study enrolled four independent cohorts with a total of 902 patients with GC. CD73 expression and immune contexture were examined by immunohistochemistry, single-sample gene set enrichment analysis and flow cytometry. Clinical outcomes of patient subgroups were evaluated using the Kaplan-Meier curves and Cox proportional hazard analysis. All statistical tests were two-sided. RESULTS: CD73 was identified as an independent adverse prognostic factor for survival in GC. CD73high tumours showed a specific microenvironment with more CD8+ T cell infiltration, but these CD8+ T cells displayed a dysfunctional phenotype. Furthermore, the CD73 (NT5E) mRNA level was associated with the Cancer Genome Atlas molecular subtypes, and NT5E high tumours showed significant fibroblast growth factor receptor 2 activation and vascular endothelial growth factor and receptor enrichment. In addition, CD73high tumours indicated better chemotherapeutic responsiveness to fluorouracil yet a worse objective response rate to pembrolizumab in GC. CONCLUSIONS: High CD73 expression indicated an immunoevasive contexture with CD8+ T cell dysfunction and represented an independent predictor for adverse clinical outcomes. As a potential immunotherapeutic target, CD73 could potentially be a novel biomarker for adjuvant chemotherapy, targeted therapies and immunotherapy. The crucial role of CD73 in the therapeutic landscape of GC needs further validation retrospectively and prospectively.


Subject(s)
5'-Nucleotidase/physiology , Stomach Neoplasms/immunology , 5'-Nucleotidase/analysis , CD8-Positive T-Lymphocytes/immunology , Disease Progression , GPI-Linked Proteins/analysis , GPI-Linked Proteins/physiology , Humans , Immunotherapy , Prognosis , Proportional Hazards Models , Stomach Neoplasms/drug therapy , Stomach Neoplasms/mortality , Tumor Escape , Tumor Microenvironment
11.
Front Oncol ; 11: 731942, 2021.
Article in English | MEDLINE | ID: mdl-34368004

ABSTRACT

[This corrects the article DOI: 10.3389/fonc.2021.666418.].

12.
Toxicology ; 441: 152502, 2020 08.
Article in English | MEDLINE | ID: mdl-32473187

ABSTRACT

Cigarette smoking is a well-recognized risk factor for type 2 diabetes (T2DM), and may result in islet ß cell damage and impaired insulin secretion. However, the underlying mechanisms remain largely elusive. In the present study, we demonstrated that nicotine induced premature senescence of pancreatic ß cells in vitro and in vivo. The senescence-associated ß-galactosidase (SA-ß-Gal) assay showed that nicotine exposure induced apparent senescence phenotype of ß-TC-6 cells at an initiating dose of 100 µM and starting from 12 h. In addition, 100 and 500 µM of nicotine exposure altered the expression of senescence marker proteins, such as p16, p19 and p21. Furthermore, we uncovered that the levels of intracellular Ca2+ and reactive oxygen species (ROS) were significantly elevated in ß-TC-6 cells following exposure to 100 and 500 µM nicotine, while calcium channel blocker can reverse this effect. Furthermore, the senescence-inducing phenotype was confirmed in rat insulinoma INS-1 cells at a similar dose range, whereas blockade of nAChRs, calcium and ROS led to apparent impairment of senescence. Finally, we found that administration with 100 and 200 µg/mL nicotine in drinking water for 28 days significantly exacerbated aberrant glucose homeostasis in a mouse model of fat-induced T2DM. Of great intrigue, pancreatic ß cells exhibited significantly enhanced senescence following nicotine administration. Taken together, this study suggests that premature senescence plays a pivotal role in nicotine-triggered ß cell destruction and glucose intolerance, providing a theoretical basis for targeted prevention and treatment of smoking-induced T2DM.


Subject(s)
Cellular Senescence/drug effects , Diabetes Mellitus, Type 2/chemically induced , Insulin-Secreting Cells/drug effects , Nicotine/toxicity , Animals , Blotting, Western , Calcium/metabolism , Disease Progression , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , beta-Galactosidase/metabolism
13.
Cancer Res ; 80(8): 1707-1719, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32060149

ABSTRACT

Tumor-associated macrophages (TAM) play an indispensable role in the modulation of the cancer immune microenvironment. Despite the fact that TAMs may exert both antitumor and protumor activities, the molecular mechanisms involved remain poorly understood. Here, we characterized a subpopulation of TAMs expressing dendritic cell-specific C-type lectin (DC-SIGN) and investigated its relevance to the prognosis and immune microenvironment of muscle-invasive bladder cancer (MIBC). DC-SIGN+ TAMs were abundant in a significant proportion of human MIBC specimens. High levels of DC-SIGN+ TAMs were associated with dismal prognosis and unresponsiveness to adjuvant chemotherapy in MIBC. Notably, multiple anti-inflammatory cytokines were enriched in DC-SIGN+ TAMs. RNA-seq analysis revealed that multiple M2-like signaling pathways were significantly upregulated in DC-SIGN+ TAMs. High infiltration of DC-SIGN+ TAMs was associated with CD8+ T-cell tolerance in MIBC. Moreover, abrogating DC-SIGN function using a neutralizing antibody led to impaired expression of anti-inflammatory cytokines and augmented PD-1 inhibitor pembrolizumab-mediated cytotoxic effects of CD8+T cells toward MIBC cells. In summary, these results suggest that DC-SIGN+ TAM infiltration is closely linked to a protumor immune microenvironment and may serve as a promising therapeutic target in the immunotherapy of MIBC. SIGNIFICANCE: DC-SIGN+ TAMs have an immunosuppressive and tumor-promoting function and may serve as a prognostic indicator and therapeutic target in MIBC.


Subject(s)
Cell Adhesion Molecules/antagonists & inhibitors , Immunotherapy/methods , Lectins, C-Type/antagonists & inhibitors , Macrophages/immunology , Receptors, Cell Surface/antagonists & inhibitors , Tumor Escape/immunology , Urinary Bladder Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Neutralizing , Antineoplastic Agents, Immunological/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion Molecules/metabolism , Cytokines/metabolism , Disease Progression , Female , Humans , Lectins, C-Type/metabolism , Macrophage Inflammatory Proteins/metabolism , Macrophages/metabolism , Male , Middle Aged , Neoplasm Proteins/metabolism , Prognosis , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Receptors, Cell Surface/metabolism , Sequence Analysis, RNA , Tumor Microenvironment/immunology , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology , Young Adult
14.
Eur Urol ; 75(5): 752-763, 2019 05.
Article in English | MEDLINE | ID: mdl-30293904

ABSTRACT

BACKGROUND: Glutamine addiction is a hallmark of clear cell renal cell carcinoma (ccRCC); yet whether glutamine metabolism impacts local immune surveillance is unclear. This knowledge may yield novel immunotherapeutic opportunities. OBJECTIVE: To seek a potential therapeutic target in glutamine-addicted ccRCC. DESIGN, SETTING, AND PARTICIPANTS: Tumors from ccRCC patients from a Shanghai cohort and ccRCC tumor data from The Cancer Genome Atlas (TCGA) cohort were analyzed. In vivo and in vitro studies were conducted with fresh human ccRCC tumors and murine tumor cells. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Immune cell numbers and functions were analyzed by flow cytometry. Glutamine and cytokine concentrations were determined. Survival was compared between different subpopulations of patients using Kaplan-Meier and Cox regression analyses. RESULTS AND LIMITATIONS: We found that in ccRCC, high interleukin (IL)-23 expression was significantly associated with poor survival in both TCGA (overall survival [OS] hazard ratio [HR]=2.04, cancer-specific survival [CSS] HR=2.95; all p<0.001) and Shanghai (OS HR=2.07, CSS HR=3.92; all p<0.001) cohorts. IL-23 blockade prolongs the survival of tumor-bearing mice, promotes T-cell cytotoxicity in in vitro cultures of human ccRCC tumors, and augments the therapeutic benefits of anti-PD-1 antibodies. Mechanistically, glutamine consumption by ccRCC tumor cells results in the local deprivation of extracellular glutamine, which induces IL-23 secretion by tumor-infiltrating macrophages via the activation of hypoxia-inducible factor 1α (HIF1α). IL-23 activates regulatory T-cell proliferation and promotes IL-10 and transforming growth factor ß expression, thereby suppressing tumor cell killing by cytotoxic lymphocytes. The positive correlations between glutamine metabolism, IL-23 levels, and Treg responses are confirmed in both TCGA cohort and tumors from Shanghai ccRCC patients. Study limitations include the unclear impacts of glutamine deprivation and IL-23 on other immune cells. CONCLUSIONS: Macrophage-secreted IL-23 enhanced Treg functions in glutamine-addicted tumors; thus, IL-23 is a promising target for immunotherapy in ccRCC. PATIENT SUMMARY: In this study, we analyzed the immune components in glutamine-addicted clear cell renal cell carcinoma (ccRCC) tumors from two patient cohorts and conducted both in vitro and in vivo studies. We found that ccRCC tumor cell-intrinsic glutamine metabolism orchestrates immune evasion via interleukin (IL)-23, and IL-23-high patients had significantly poorer survival than IL-23-low patients. IL-23 should thus be considered a therapeutic target in ccRCC, either alone or in combination with immune checkpoint inhibitors.


Subject(s)
Carcinoma, Renal Cell/immunology , Glutamine/metabolism , Interleukin-23/metabolism , Kidney Neoplasms/immunology , Macrophages/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies/therapeutic use , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/therapy , Cell Proliferation/drug effects , Cells, Cultured , Gene Ontology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immune Evasion , Immune Tolerance/drug effects , Interleukin-23/antagonists & inhibitors , Interleukin-23/immunology , Interleukin-23/pharmacology , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/therapy , Lymphocyte Activation , Mice , Oncogene Addiction , Survival Rate , T-Lymphocytes, Regulatory/physiology , Tumor Escape
15.
Hum Pathol ; 72: 80-90, 2018 02.
Article in English | MEDLINE | ID: mdl-29126834

ABSTRACT

NKD2 is a member of the Naked cuticle (Nkd) protein family and functions as a negative regulator of the Wnt signaling pathway. We investigated the prognostic value of NKD2 expression in hepatocellular carcinoma (HCC). Western blot and immunohistochemical analyses revealed that NKD2 was significantly downregulated in HCC specimens compared with adjacent nontumorous tissues. Next, we found that NKD2 expression correlated significantly with several clinicopathologic features, such as tumor grade, tumor size, and Ki-67 expression. Univariate and multivariate analyses demonstrated that NKD2 was an independent prognostic factor for the survival of HCC patients. In particular, Kaplan-Meier survival curves suggested that low NKD2 was statistically associated with poor overall survival. Furthermore, serum refeeding of cultured HCC cells led to impaired amounts of NKD2 and induced HepG2 and Huh7 cells to transition from the G1 to the S phase. Small interfering RNA-mediated depletion of NKD2 in LO2 hepatocytes caused accelerated cell growth. To further clarify the role of NKD2 in cell cycle progression, a Flag-tagged NKD2 construct was used to overexpress NKD2 exogenously in Huh7 cells. These results showed that overexpression of NKD2 induced G1-phase cell cycle arrest. Reduced expression of NKD2 correlated with hyperactivation of the Wnt/ß-catenin pathway and doxorubicin resistance in HCC cells. On the basis of these findings, we conclude that NKD2 may be a novel prognostic marker and therapeutic target in HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Carrier Proteins/metabolism , Cell Proliferation/physiology , Liver Neoplasms/pathology , Adaptor Proteins, Signal Transducing , Aged , Biomarkers, Tumor/metabolism , Calcium-Binding Proteins , Carcinoma, Hepatocellular/diagnosis , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Liver Neoplasms/diagnosis , Male , Middle Aged , Neoplasm Grading/methods , Wnt Signaling Pathway/physiology
16.
Biochem Biophys Res Commun ; 495(1): 1136-1143, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29175326

ABSTRACT

Accumulating evidence indicates that the neural precursor cell-expressed, developmentally downregulated 4-like (Nedd4L) related with some tumor progression pathways and was found abnormally expressed in several kinds of solid cancers. However, the role and mechanism of Nedd4L in HCC remain unknown. This study was to assess the role of Nedd4L in HCC tumorigenesis and prognosis. The real-time quantitative RT-PCR and immunohistochemistry results revealed that Nedd4L was downregulated in HCC tissues compared to corresponding peri-noncancerous tissue, and HCC patients with low expression of Nedd4L exhibited poor prognosis assessed by Kaplan-Meier and Cox regression analysis in 78 HCC patients. Furthermore, knockdown of Nedd4L could significantly promote proliferation of HCC cells by CCK-8 and colony formation assays in vitro; whereas ectopic expression of Nedd4L resulted in attenuating proliferation in vitro and tumor growth in vivc determined by nude mice xenografts model. Mechanically, Nedd4L could phosphorylate ERK1/2 and regulate genes related with apoptosis. Collectively, Nedd4L plays a tumor suppressive role in HCC, possibly through triggering MAPK/ERK-mediated apoptosis, and Nedd4L downregulation may be a potential prognostic biomarker as well as a therapeutic target for HCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Cell Proliferation , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Nedd4 Ubiquitin Protein Ligases/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/pathology , China/epidemiology , Down-Regulation , Female , Humans , Liver Neoplasms/pathology , MAP Kinase Signaling System , Male , Middle Aged , Neoplasm Invasiveness , Prevalence , Prognosis , Risk Factors , Survival Rate , Tumor Cells, Cultured
17.
Pathol Res Pract ; 213(6): 688-697, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28476378

ABSTRACT

Nucleostemin (NS)/GNL3 protein has been recently documented to be a nucleolar protein that was abundantly expressed in stem cells and cancer cells. Herein, we showed that NS was upregulated in HCC tissues and the expression of NS was inversely correlated with that of p53. Overexpression of NS predicted significantly worsened prognosis in HCC patients, suggesting that NS might serve as a prognostic marker of HCC. In addition, we found that depletion of NS sensitized HCC cells to sorafenib-induced apoptosis. Moreover, we found that the mechanism underlying NS-mediated sorafenib resistance involved dysregulated expression of p53, and downstream Bax and Bcl-2 proteins. NS interacted with p53 in HCC cells. Depletion of NS increased the expression of p53 and Bax, whereas impaired the level of cellular Bcl-2. Interference of NS enhanced the cytotoxic effects of sorafenib in HCC cells. Furthermore, ectopic expression of NS impaired the apoptosis of HCC cells following sorafenib exposure. Therefore, NS may contribute to sorafenib resistance in HCC cells through the modulation of p53 pathway and Bcl-2 proteins. These findings indicated that the combination of silencing NS expression and sorafenib treatment is a promising therapeutic strategy in treatment of HCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/metabolism , Drug Resistance, Neoplasm/physiology , GTP-Binding Proteins/metabolism , Liver Neoplasms/metabolism , Niacinamide/analogs & derivatives , Nuclear Proteins/metabolism , Phenylurea Compounds/therapeutic use , Adult , Aged , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Female , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Middle Aged , Niacinamide/pharmacology , Niacinamide/therapeutic use , Phenylurea Compounds/pharmacology , Prognosis , Proto-Oncogene Proteins c-bcl-2/metabolism , Sorafenib , Survival Rate , Up-Regulation , Young Adult , bcl-2-Associated X Protein/metabolism
18.
Hum Pathol ; 64: 61-68, 2017 06.
Article in English | MEDLINE | ID: mdl-28412208

ABSTRACT

The purinergic P2X7 receptor (P2X7R) is a master regulator of inflammation and inflammation-related diseases. Recently, P2X7R has been reportedly involved in carcinogenesis and tumor progression. In this study, we investigated the expression pattern and prognostic merit of P2X7R in human colorectal cancer (CRC). The expression profile of P2X7R in 12 pairs of CRC and non-tumorous specimens was evaluated using Western blotting analysis. Additionally, we performed immunohistochemistry (IHC) on 116 paraffin-embedded CRC specimens, and evaluated the correlation between P2X7R expression and clinicopathological factors. P2X7R was overexpressed in CRC samples, compared with adjacent non-tumorous ones. High P2X7R expression significantly correlated with tumor size (P = .0177), Lymph node metastasis (P = .0128), and TNM stage (P = .0081). Furthermore, univariate and multivariate Cox regression analyses revealed that P2X7R expression could serve as an independent prognostic factor for poor overall survival (P = .0197). Treatment with P2X7R agonist BzATP led to the activation of Akt and NF-κB pathways. Consequently, we revealed that BzATP accelerated the proliferation of CRC cells, whereas co-incubation with PI3K/Akt inhibitor LY294002 significantly impaired BzATP-induced proliferation of CRC cells. Our findings implied that P2X7R may serve as a valuable prognostic indicator and promising therapeutic target of CRC.


Subject(s)
Biomarkers, Tumor/analysis , Colorectal Neoplasms/metabolism , Receptors, Purinergic P2X7/metabolism , Biopsy , Blotting, Western , Cell Proliferation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Female , HCT116 Cells , Humans , Immunohistochemistry , Lymphatic Metastasis , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging , Predictive Value of Tests , Proportional Hazards Models , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Purinergic P2X Receptor Agonists/pharmacology , Receptors, Purinergic P2X7/drug effects , Risk Factors , Signal Transduction , Time Factors , Transcription Factor RelA/metabolism , Tumor Burden
19.
Cancer Lett ; 388: 220-229, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27998760

ABSTRACT

p27kip, as a cyclin dependent kinase inhibitor (CDKI), plays a pivotal role in the regulation of cell cycle progression and hepatocarcinogenesis. Herein, we revealed that p27 exhibited apparent nucleolar distribution and interacted with nucleolar protein nucleostemin (NS) in Hepatocellular carcinoma (HCC) cells. Furthermore, subcellular fractionation experiments demonstrated that nucleolar p27 had significantly higher level of polyubiquitylation, compared with nucleoplasmic fraction. Depletion of NS inhibited nucleolar polyubiquitylation of p27, indicating an involvement of NS in triggering p27 ubiquitylation and inactivation during HCC development. Moreover, we found that knockdown of NS promoted p27 to bind to CDK2-Cyclin E complex and inhibited the activity of CDK2, resulting in consequent cell cycle arrest in HCC cells. Furthermore, silencing NS expression reduced in vitro colony formation and in vivo tumor growth of HCC cells. Finally, we found that NS was upregulated in HCC tissues, compared with adjacent non-tumorous tissues. Kaplan-Meier analysis indicated patients with high expression of NS and low expression of p27 had significantly worsened prognosis. Our results suggested NS mediated p27-dependent cell cycle control via inducing nucleolar sequestration and polyubiquitylation of p27 in HCC. These findings help gain an insightful view into the mechanism underlying aberrant cell cycle progression during hepatocarcinogenesis, and thus benefit the development of molecular-targeted therapies in HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , GTP-Binding Proteins/metabolism , Liver Neoplasms/genetics , Nuclear Proteins/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Female , Humans , Liver Neoplasms/pathology , Mice , Mice, Nude , Transfection
20.
Anticancer Agents Med Chem ; 17(7): 948-954, 2017.
Article in English | MEDLINE | ID: mdl-27671301

ABSTRACT

BACKGROUND: Recent investigations have implicated that Chitosan-nucleotide nanoparticles might be useful non-viral carriers in gene therapy. Polo-like kinase 1 (PLK1) has been reported to be an important oncogene that exerted considerable therapeutic merit in hepatocellular carcinoma (HCC). OBJECTIVE: We explored whether Galactosylated chitosan-graft-poly(ethylene glycol) (GCP) nanoparticlemediated delivery of PLK1 siRNA nucleotides could serve as an effective anti-cancer agent for HCC therapy. METHOD: GCP nanoparticles were prepared to deliver PLK1 siRNA oligos into HCC cells and tissues. Real-time fluorescence quantitative PCR (RFQ-PCR) and western blotting analyses were used to examine the efficiency of nanoparticle-mediated depletion of PLK1 in HepG2 cells. Cell proliferation and apoptotic death were also examined using flow cytometric, MTT and TUNEL assays. Xenograft mouse model was conducted to assess the impact of GCP/siRNA nanoparticles on the in vivo growth of HCC cells. RESULTS: GCP nanoparticles bind to PLK1 siRNA efficiently. The particle size and zeta potential of GCP/siRNA nanoparticles are suitable for cellular delivery. PLK1-targeting nanoparticles inhibited cell proliferation through inducing G2/M phase arrest with a higher efficacy than a selective and potent PLK1 inhibitor BI 2536. Moreover, TUNEL assay revealed that PLK1-siRNA nanoparticles induced apparent apoptosis in HepG2 cells. In addition, PLK1-targeting nanoparticles induced significant upregulation of cellular p53, Bax and p21, whereas the level of Bcl-2 was impaired in HCC cells. Moreover, PLK1-targeting nanoparticles impaired the tumorigenicity of HepG2 cells in vivo. CONCLUSION: These findings indicate that PLK1-targeting nanoparticles exert considerable therapeutic merit and GCP/siRNA nanoparticles would be a valuable therapeutic carrier for HCC.


Subject(s)
Carcinoma, Hepatocellular/therapy , Cell Cycle Proteins/genetics , Chitosan/analogs & derivatives , Liver Neoplasms/therapy , Nanoparticles/chemistry , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/administration & dosage , RNAi Therapeutics/methods , Animals , Apoptosis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Hep G2 Cells , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Small Interfering/therapeutic use , Polo-Like Kinase 1
SELECTION OF CITATIONS
SEARCH DETAIL
...