Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Oncogene ; 2024 May 22.
Article in English | MEDLINE | ID: mdl-38778160

ABSTRACT

Angiogenesis is one of the characteristics of malignant tumors, and persistent generation of abnormal tumor blood vessels is an important factor contributing to tumor treatment resistance. Epstein-Barr virus (EBV) is a highly prevalent DNA oncogenic virus that is associated with the development of various epithelial malignancies. However, the relationship between EBV infection and tumor vascular abnormalities as well as its underlying mechanisms is still unclear. In this study, we found that compared to EBV-uninfected tumors, EBV-infected tumors were more angiogenic, but the neovascularization was mostly immature vessels without pericyte attachment in both clinical patient tumor samples and mouse xenograft models; These immature vessels exhibited aberrant functionality, characterized by poor blood perfusion and increased vascular permeability. The vascular abnormalities caused by EBV infection exacerbated tumor hypoxia and was responsible for accelerated tumor growth. Mechanistically, EBV infection upregulated ANXA3-HIF-1α-VEGF pathway. Silencing the ANXA3 gene or neutralizing ANXA3 with an antibody can diminish vascular abnormalities, thereby increasing immune cell infiltration and alleviating treatment resistance. Finally, a new therapy combining ANXA3 blockade and NK cell + PD1 antibody significantly inhibited the growth of EBV-infected xenografts in mice. In conclusion, our study identified a previously unrecognized role for EBV infection in tumor vascular abnormalities and revealed its underlying mechanism that upregulated the ANXA3-HIF-1α-VEGF pathway. ANXA3 is a potential therapeutic target for EBV-infected tumors and ANXA3 blockade to improve vascular conditions, in combination with NK cell + PD1 antibody therapy, holds promise as an effective treatment strategy for EBV-associated epithelial malignancies.

2.
Cancer Res ; 84(10): 1613-1629, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38381538

ABSTRACT

Neutrophil extracellular traps (NET), formed by the extracellular release of decondensed chromatin and granules, have been shown to promote tumor progression and metastasis. Tumor-associated neutrophils in hepatocellular carcinoma (HCC) are prone to NET formation, highlighting the need for a more comprehensive understanding of the mechanisms of action of NETs in liver cancer. Here, we showed that DNA of NETs (NET-DNA) binds transmembrane and coiled-coil domains 6 (TMCO6) on CD8+ T cells to impair antitumor immunity and thereby promote HCC progression. TGFß1 induced NET formation, which recruited CD8+ T cells. Binding to NET-DNA inhibited CD8+ T cells function while increasing apoptosis and TGFß1 secretion, forming a positive feedback loop to further stimulate NET formation and immunosuppression. Mechanistically, the N-terminus of TMCO6 interacted with NET-DNA and suppressed T-cell receptor signaling and NFκB p65 nuclear translocation. Blocking NET formation by inhibiting PAD4 induced potent antitumor effects in wild-type mice but not TMCO6-/- mice. In clinical samples, CD8+ T cells expressing TMCO6 had an exhausted phenotype. TGFß1 signaling inhibition or TMCO6 deficiency combined with anti-PD-1 abolished NET-driven HCC progression in vivo. Collectively, this study unveils the role of NET-DNA in impairing CD8+ T-cell immunity by binding TMCO6 and identifies targeting this axis as an immunotherapeutic strategy for blocking HCC progression. SIGNIFICANCE: TMCO6 is a receptor for DNA of NETs that mediates CD8+ T-cell dysfunction in HCC, indicating that the NET-TMCO6 axis is a promising target for overcoming immunosuppression in liver cancer.


Subject(s)
CD8-Positive T-Lymphocytes , Carcinoma, Hepatocellular , Extracellular Traps , Liver Neoplasms , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Animals , Humans , Mice , Extracellular Traps/immunology , Extracellular Traps/metabolism , Transforming Growth Factor beta1/metabolism , Neutrophils/immunology , Neutrophils/metabolism , DNA/immunology , DNA/metabolism , Mice, Inbred C57BL , Mice, Knockout , Cell Line, Tumor , Male
4.
BMC Med ; 21(1): 327, 2023 08 28.
Article in English | MEDLINE | ID: mdl-37635247

ABSTRACT

BACKGROUND: CD133 is considered a marker for cancer stem cells (CSCs) in several types of tumours, including hepatocellular carcinoma (HCC). Chimeric antigen receptor-specific T (CAR-T) cells targeting CD133-positive CSCs have emerged as a tool for the clinical treatment of HCC, but immunogenicity, the high cost of clinical-grade recombinant viral vectors and potential insertional mutagenesis limit their clinical application. METHODS: CD133-specific CAR-T cells secreting PD-1 blocking scFv (CD133 CAR-T and PD-1 s cells) were constructed using a sleeping beauty transposon system from minicircle technology, and the antitumour efficacy of CD133 CAR-T and PD-1 s cells was analysed in vitro and in vivo. RESULTS: A univariate analysis showed that CD133 expression in male patients at the late stage (II and III) was significantly associated with worse progression-free survival (PFS) (P = 0.0057) and overall survival (OS) (P = 0.015), and a multivariate analysis showed a trend toward worse OS (P = 0.041). Male patients with advanced HCC exhibited an approximately 20-fold higher PD-L1 combined positive score (CPS) compared with those with HCC at an early stage. We successfully generated CD133 CAR-T and PD-1 s cells that could secrete PD-1 blocking scFv based on a sleeping beauty system involving minicircle vectors. CD133 CAR-T and PD-1 s cells exhibited significant antitumour activity against HCC in vitro and in xenograft mouse models. Thus, CD133 CAR-T and PD-1 s cells may be a therapeutically tractable strategy for targeting CD133-positive CSCs in male patients with advanced HCC. CONCLUSIONS: Our study provides a nonviral strategy for constructing CAR-T cells that could also secrete checkpoint blockade inhibitors based on a Sleeping Beauty system from minicircle vectors and revealed a potential benefit of this strategy for male patients with advanced HCC and high CD133 expression (median immunohistochemistry score > 2.284).


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Receptors, Chimeric Antigen , Humans , Male , Animals , Mice , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/therapy , Programmed Cell Death 1 Receptor , Receptors, Chimeric Antigen/genetics , Liver Neoplasms/genetics , Liver Neoplasms/therapy , Disease Models, Animal , T-Lymphocytes
5.
Front Immunol ; 14: 1212577, 2023.
Article in English | MEDLINE | ID: mdl-37545530

ABSTRACT

Introduction: The limited response to immune checkpoint blockades (ICBs) in patients with hepatocellular carcinoma (HCC) highlights the urgent need for broadening the scope of current immunotherapy approaches. Lenvatinib has been shown a potential synergistic effect with ICBs. This study investigated the optimal method for combining these two therapeutic agents and the underlying mechanisms. Methods: The effect of lenvatinib at three different doses on promoting tissue perfusion and vascular normalization was evaluated in both immunodeficient and immunocompetent mouse models. The underlying mechanisms were investigated by analyzing the vascular morphology of endothelial cells and pericytes. The enhanced immune infiltration of optimal-dose lenvatinib and its synergistic effect of lenvatinib and anti-PD-1 antibody was further evaluated by flow cytometry and immunofluorescence imaging. Results: There was an optimal dose that superiorly normalized tumor vasculature and increased immune cell infiltration in both immunodeficient and immunocompetent mouse models. An adequate concentration of lenvatinib strengthened the integrity of human umbilical vein endothelial cells by inducing the formation of the NRP-1-PDGFRß complex and activating the Crkl-C3G-Rap1 signaling pathway in endothelial cells. Additionally, it promoted the interaction between endothelial cells and pericytes by inducing tyrosine-phosphorylation in pericytes. Furthermore, the combination of an optimal dose of lenvatinib and an anti-PD-1 antibody robustly suppressed tumor growth. Conclusions: Our study proposes a mechanism that explains how the optimal dose of lenvatinib induces vascular normalization and confirms its enhanced synergistic effect with ICBs.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Mice , Humans , Carcinoma, Hepatocellular/pathology , Antineoplastic Agents/therapeutic use , Liver Neoplasms/pathology , Endothelial Cells/metabolism , Disease Models, Animal
7.
Clin Transl Med ; 13(5): e1247, 2023 05.
Article in English | MEDLINE | ID: mdl-37132170

ABSTRACT

BACKGROUND: In the past decade, the field of tumour immunotherapy has made a great progress. However, the efficacy of immune checkpoint blocking (ICB) in the treatment of hepatocellular carcinoma (HCC) remains limited. Cytotoxic lymphocyte trafficking into tumours is critical for the success of ICB. Therefore, additional strategies that increase cytotoxic lymphocyte trafficking into tumours are urgently needed to improve patient immune responses. METHODS: Paired adjacent tissue and cancerous lesions with HBV-associated HCC were subjected to RNA-seq analysis. Bone morphogenetic protein (BMP9), which reflects vessel normalisation, was identified through Cytoscape software, clinical specimens and Gene Expression Omnibus (GEO) datasets for HCC. The functional effects and mechanism of BMP9 on the tumour vasculature were evaluated in cells and animals. An ultrasound-targeted microbubble destruction (UTMD)-mediated BMP9 delivery strategy was used to normalise the vasculature and evaluate therapeutic efficacy mediated by cytotoxic lymphocytes (NK cells) in combination with a PD-L1 antibody in human cancer xenografts of immune-deficient mice. RESULTS: We discovered that hepatitis B virus (HBV) infection-induced downregulation of BMP9 expression correlated with a poor prognosis and pathological vascular abnormalities in patients with HCC. BMP9 overexpression in HBV-infected HCC cells promoted intra-tumoural cytotoxic lymphocyte infiltration via vascular normalisation by inhibiting the Rho-ROCK-myosin light chain (MLC) signalling cascade, resulting in enhanced efficacy of immunotherapy. Furthermore, UTMD-mediated BMP9 delivery restored the anti-tumour function of cytotoxic lymphocytes (NK cells) and showed therapeutic efficacy in combination with a PD-L1 antibody in human cancer xenografts of immune-deficient mice. CONCLUSIONS: HBV-induced BMP9 downregulation causes vascular abnormalities that inhibit intra-tumoural cytotoxic lymphocyte infiltration, providing a rationale for developing and combining immunotherapy with BMP9-based therapy to treat HBV-associated HCC.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Hepatitis B , Liver Neoplasms , Animals , Humans , Mice , Antineoplastic Agents/therapeutic use , B7-H1 Antigen , Bone Morphogenetic Proteins/therapeutic use , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/drug therapy , Hepatitis B/complications , Hepatitis B virus/genetics , Immunotherapy/methods , Liver Neoplasms/therapy , Liver Neoplasms/drug therapy
8.
Theranostics ; 13(5): 1649-1668, 2023.
Article in English | MEDLINE | ID: mdl-37056569

ABSTRACT

Rationale: Resistance to 5-fluorouracil (5-FU) chemotherapy remains the main barrier to effective clinical outcomes for patients with colorectal cancer (CRC). A better understanding of the detailed mechanisms underlying 5-FU resistance is needed to increase survival. Interleukin (IL)-33 is a newly discovered alarmin-like molecule that exerts pro- and anti-tumorigenic effects in various cancers. However, the precise role of IL-33 in CRC progression, as well as in the development of 5-FU resistance, remains unclear. Methods: High-quality RNA-sequencing analyses were performed on matched samples from patients with 5-FU-sensitive and 5-FU-resistant CRC. The clinical and biological significance of IL-33, including its effects on both T cells and tumor cells, as well as its relationship with 5-FU chemotherapeutic activity were examined in ex vivo, in vitro and in vivo models of CRC. The molecular mechanisms underlying these processes were explored. Results: IL-33 expressed by tumor cells was a dominant mediator of antitumoral immunity in 5-FU-sensitive patients with CRC. By binding to its ST2 receptor, IL-33 triggered CD4+ (Th1 and Th2) and CD8+ T cell responses by activating annexin A1 downstream signaling cascades. Mechanistically, IL-33 enhanced the sensitivity of CRC cells to 5-FU only in the presence of T cells, which led to the activation of both tumor cell-intrinsic apoptotic and immune killing-related signals, thereby synergizing with 5-FU to induce apoptosis of CRC cells. Moreover, injured CRC cells released more IL-33 and the T cell chemokines CXCL10 and CXCL13, forming a positive feedback loop to further augment T cell responses. Conclusions: Our results identified a previously unrecognized connection between IL-33 and enhanced sensitivity to 5-FU. IL-33 created an immune-active tumor microenvironment by orchestrating antitumoral T cell responses. Thus, IL-33 is a potential predictive biomarker for 5-FU chemosensitivity and favorable prognosis and has potential as a promising adjuvant immunotherapy to improve the clinical benefits of 5-FU-based therapies in the treatment of CRC.


Subject(s)
Colorectal Neoplasms , Fluorouracil , Humans , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Alarmins/therapeutic use , Colorectal Neoplasms/pathology , Interleukin-33 , Cell Line, Tumor , Drug Resistance, Neoplasm , Tumor Microenvironment
9.
Int J Biol Sci ; 18(14): 5241-5259, 2022.
Article in English | MEDLINE | ID: mdl-36147467

ABSTRACT

The imbalance of kinetochore-microtubule attachment during cell mitosis is a response to the initiation and progression of human cancers. Spindle component 25 (SPC25) is indispensable for spindle apparatus organization and chromosome segregation. SPC25 plays an important role in the development of malignant tumors, but its role in hepatocellular carcinoma (HCC) is yet to be determined. In this study, we aimed to preliminarily investigate the role of SPC25 in HCC progression and the molecular mechanisms underlying the process. We identified SPC25 as a clinically notable molecule significantly correlated with the grade of malignancy and poor survival in both The Cancer Genome Atlas (TCGA) cohort and the HCC patient cohort from our center. Mechanistically, SPC25 promoted the incidence of DNA damage and activated the DNA-PK/Akt/Notch1 signaling cascade in HCC cells; the NICD/ RBP-Jκ complex directly targeted SOX2 and NANOG in a transcriptional manner to regulate the proliferation and self-renewal of HCC cells. Our study suggests that HCC-intrinsic SPC25/DNA-PK/Akt/Notch1 signaling is an important mechanism to promote carcinogenesis by regulating the proliferation and stemness program, which provides possible biomarkers for predicting HCC progression and poor survival, as well as potential therapeutic targets for HCC patients.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Microtubule-Associated Proteins , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , DNA , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/metabolism , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Signal Transduction/genetics
10.
J Immunother Cancer ; 10(3)2022 03.
Article in English | MEDLINE | ID: mdl-35277458

ABSTRACT

BACKGROUND: Platinum resistance is a major challenge in the clinical treatment of advanced ovarian cancer (OC). Accumulating evidence shows that the tumor-promotive M2 macrophage is linked to the limiting chemotherapy efficacy of multiple malignancies including OC. Circular RNAs (circRNAs) are a novel class of non-coding RNAs which function as the critical regulator in biological process of cancer. However, their impact on macrophage polarization and chemoresistance of OC remain unclear. METHODS: Platinum-resistant circRNAs were screened using circRNA deep sequencing and validated using in situ hybridization in OC tissues with or without platinum resistance. The role of circITGB6 in inducing cisplatin (CDDP) resistance was evaluated by clone formation, immunofluorescence and annexin V assays in vitro, and by intraperitoneal tumor model in vivo. The mechanism underlying circITGB6-mediated tumor-associated macrophage (TAM) polarization into M2 phenotype was investigated using RNA pull-down, luciferase reporter, electrophoretic mobility shift, RNA binding protein immunoprecipitation (RIP), ELISA and immunofluorescence assays. RESULTS: We identified that a novel circRNA, circITGB6, robustly elevated in tumor tissues and serums from patients with OC with platinum resistance, was correlated with poor prognosis. circITGB6 overexpression promoted an M2 macrophage-dependent CDDP resistance in both vivo and vitro. Mechanistic research determined that circITGB6 directly interacted with IGF2BP2 and FGF9 mRNA to form a circITGB6/IGF2BP2/FGF9 RNA-protein ternary complex in the cytoplasm, thereby stabilizing FGF9 mRNA and inducing polarization of TAMs toward M2 phenotype. Importantly, blocking M2 macrophage polarization with an antisense oligonucleotide targeting circITGB6 markedly reversed the circITGB6-induced CDDP resistance of OC in vivo. CONCLUSIONS: This study reveals a novel mechanism for platinum resistance in OC and demonstrates that circITGB6 may serve as a potential prognostic marker and a therapeutic target for patients with OC.


Subject(s)
Ovarian Neoplasms , Tumor-Associated Macrophages , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cisplatin/pharmacology , Cisplatin/therapeutic use , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Phenotype , RNA , RNA, Circular/genetics , RNA, Messenger , RNA-Binding Proteins
11.
Cell Death Dis ; 13(3): 251, 2022 03 19.
Article in English | MEDLINE | ID: mdl-35304440

ABSTRACT

Hepatocellular carcinoma is one of the most common malignancies and has a poor prognosis. The ubiquitin-proteasome pathway is required for the degradation of most short-lived proteins. CMTM6 has been implicated in the progression of various tumors, but its biological function and the underlying molecular mechanisms in HCC are still unknown. In this study, we found that the expression of CMTM6 was significantly reduced in HCC and predicted better prognosis of HCC patients. Through in vitro and in vivo experiments, CMTM6 was shown to inhibit the proliferation of HCC cells by blocking the G1/S phase transition. Mechanistically, CMTM6 interacted with p21 and prevented its ubiquitination mediated by SCFSKP2, CRL4CDT2 and APC/CCDC20 in a cell-cycle-independent manner. As a result, CMTM6 stabilized p21 protein, leading to the inactivation of pRB/E2F pathway. Additionally, CMTM6 sensitized HCC cells to doxorubicin and cisplatin, positively correlated with better clinical outcomes of the transarterial chemoembolization (TACE) treatment for postoperative recurrence. Taken together, our study reports a novel mechanism by which p21 can be stabilized by CMTM6 and pinpoints a crucial role of the CMTM6-p21 axis in suppressing the progression of HCC and sensitizing patients with postoperative recurrence to TACE treatment.


Subject(s)
Carcinoma, Hepatocellular , Chemoembolization, Therapeutic , Liver Neoplasms , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Ubiquitination
12.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-929611

ABSTRACT

@#[Abstract] Objective: To analyze tumor-associated neutrophils (TANs) infiltration in EBV-positive nasopharyngeal carcinoma (NPC) tumor microenvironment (TME) and its correlation with the prognosis and clinicopathological features of patients with NPC, and to preliminarily investigate the role of TANs in activating CD8+ T cells in EBV-positive NPC. Methods: Tumor tissue specimens from 118 patients with EBV-positive NPC without metastases admitted to Sun Yat-sen University Cancer Center from 2008 to 2012 were collected. Immunohistochemistry was performed to analyze CD8+ T cells infiltration and TANs infiltration in NPC and their correlation with EBV infection, and to evaluate the infiltration of TANs in 118 human EBV-positive NPC tissue samples and analyze its correlation with the prognosis and clinicopathological features of NPC patients; flow cytometry was used to detect the expression of N2 marker (CD182 and CD206) of neutrophils stimulated by HK1-EBV-conditioned medium and the expression of CD69 and PD-1 of CD8+ T cells co-cultured with TANs. Results: There were more CD8+ T cells and neutrophils infiltrating in EBV-positive NPC where they were negatively correlated(P=0.0052); densely infiltrated TANs in EBV-positive NPC tissues were positively correlated with poor prognosis of patients (P=0.025 in OS; P=0.027 in PFS); the infiltration of TANs was proved to be an independent risk factor for overall survival (P=0.035) in patients with NPC; compared with Control group, freshly isolated neutrophils cultured in HK1-EBV-conditioned medium showed an increased expression of N2 markers (P<0.001 in CD182; P<0.01 in CD206) which could suppress the expression of CD69 and promote the expression of PD-1 of CD8+ T cells. Conclusion: More TANs infiltrate in EBV-positive NPC where they polarize into N2 neutrophils, exerting an immunosuppressive effect by suppressing the activation of CD8+ T cells and having a positive correlation with the poor prognosis of NPC patients.

13.
Cell Death Dis ; 12(12): 1093, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34795209

ABSTRACT

Most patients with hepatocellular carcinoma (HCC) are in the middle or advanced stage at the time of diagnosis, and the therapeutic effect is limited. Therefore, this study aimed to verify whether deoxythymidylate kinase (DTYMK) increased in HCC and was an effective therapeutic target in HCC. The findings revealed that the DTYMK level significantly increased and correlated with poor prognosis in HCC. However, nothing else is known, except that DTYMK could catalyze the phosphorylation of deoxythymidine monophosphate (dTMP) to form deoxythymidine diphosphate (dTDP). A number of experiments were performed to study the function of DTYMK in vitro and in vivo to resolve this knowledge gap. The knockdown of DTYMK was found to significantly inhibit the growth of HCC and increase the sensitivity to oxaliplatin, which is commonly used in HCC treatment. Moreover, DTYMK was found to competitively combine with miR-378a-3p to maintain the expression of MAPK activated protein kinase 2 (MAPKAPK2) and thus activate the phospho-heat shock protein 27 (phospho-HSP27)/nuclear factor NF-kappaB (NF-κB) axis, which mediated the drug resistance, proliferation of tumor cells, and infiltration of tumor-associated macrophages by inducing the expression of C-C motif chemokine ligand 5 (CCL5). Thus, this study demonstrated a new mechanism and provided a new insight into the role of mRNA in not only encoding proteins to regulate the process of life but also regulating the expression of other genes and tumor microenvironment through the competing endogenous RNA (ceRNA) mechanism.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Nucleoside-Phosphate Kinase/metabolism , Oxaliplatin/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Humans , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Mice , Oxaliplatin/pharmacology , Survival Analysis
14.
Hepatology ; 73(5): 1717-1735, 2021 05.
Article in English | MEDLINE | ID: mdl-33682185

ABSTRACT

BACKGROUND AND AIMS: Cancer-associated fibroblasts (CAFs) are key players in multicellular, stromal-dependent alterations leading to HCC pathogenesis. However, the intricate crosstalk between CAFs and other components in the tumor microenvironment (TME) remains unclear. This study aimed to investigate the cellular crosstalk among CAFs, tumor cells, and tumor-associated neutrophils (TANs) during different stages of HCC pathogenesis. APPROACH AND RESULTS: In the HCC-TME, CAF-derived cardiotrophin-like cytokine factor 1 (CLCF1) increased chemokine (C-X-C motif) ligand 6 (CXCL6) and TGF-ß secretion in tumor cells, which subsequently promoted tumor cell stemness in an autocrine manner and TAN infiltration and polarization in a paracrine manner. Moreover, CXCL6 and TGF-ß secreted by HCC cells activated extracellular signal-regulated kinase (ERK) 1/2 signaling of CAFs to produce more CLCF1, thus forming a positive feedback loop to accelerate HCC progression. Inhibition of ERK1/2 or CLCF1/ciliary neurotrophic factor receptor signaling efficiently impaired CLCF1-mediated crosstalk among CAFs, tumor cells, and TANs both in vitro and in vivo. In clinical samples, up-regulation of the CLCF1-CXCL6/TGF-ß axis exhibited a marked correlation with increased cancer stem cells, "N2"-polarized TANs, tumor stage, and poor prognosis. CONCLUSIONS: This study reveals a cytokine-mediated cellular crosstalk and clinical network involving the CLCF1-CXCL6/TGF-ß axis, which regulates the positive feedback loop among CAFs, tumor stemness, and TANs, HCC progression, and patient prognosis. These results may support the CLCF1 cascade as a potential prognostic biomarker and suggest that selective blockade of CLCF1/ciliary neurotrophic factor receptor or ERK1/2 signaling could provide an effective therapeutic target for patients with HCC.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Hepatocellular/metabolism , Chemokine CXCL6/metabolism , Cytokines/metabolism , Disease Progression , Female , Humans , Liver Neoplasms/metabolism , MAP Kinase Signaling System , Male , Middle Aged , Signal Transduction , Transforming Growth Factor beta/metabolism , Tumor Microenvironment
15.
Clin Transl Immunology ; 10(3): e1257, 2021.
Article in English | MEDLINE | ID: mdl-33717483

ABSTRACT

OBJECTIVES: Although axitinib has achieved a preferable response rate for advanced renal cell carcinoma (RCC), patient survival remains unsatisfactory. In this study, we evaluated the efficacy and safety of a combination treatment of axitinib and a low dose of pembrolizumab-activated autologous dendritic cells-co-cultured cytokine-induced killer cells in patients with advanced RCC. METHODS: All adult patients, including treatment-naive or pretreated with VEGF-targeted agents, were enrolled from May 2016 to March 2019. Patients received axitinib 5 mg twice daily and pembrolizumab-activated dendritic cells-co-cultured cytokine-induced killer cells intravenously weekly for the first four cycles, every 2 weeks for the next four cycles, and every month thereafter. RESULTS: The 43 patients (22 untreated and 21 previously treated) showed a median progression-free survival (mPFS) of 14.7 months (95% CI, 11.16-18.30). mPFS in treatment-naive patients was 18.2 months, as compared with 14.4 months in pretreated patients (log-rank P-value = 0.07). Overall response rates were 25.6% (95% CI, 13.5-41.2%). Grade 3 or higher adverse events occurred in 5% of patients included hypertension (11.6%) and palmar-plantar erythrodysesthesia (7.0%). Peripheral blood lymphocyte immunophenotype and serum cytokine profile analyses demonstrated increased antitumor immunity after combination treatment particularly in patients with a long-term survival benefit, while those with a minimal survival benefit demonstrated an elevated proportion of peripheral CD8+TIM3+ T cells and lower serum-level immunostimulatory cytokine profile. CONCLUSIONS: The combination therapy was active and well tolerated for treatment of advanced RCC, either as first- or second-line treatment following other targeted agents. Changes in immunophenotype and serum cytokine profile may be used as prognostic biomarkers.

16.
Oncol Lett ; 21(3): 205, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33574944

ABSTRACT

Bladder cancer (BC) is the ninth most common lethal malignancy worldwide. Great efforts have been devoted to clarify the pathogenesis of BC, but the underlying molecular mechanisms remain unclear. To screen for the genes associated with the progression and carcinogenesis of BC, three datasets were obtained from the Gene Expression Omnibus. A total of 37 tumor and 16 non-cancerous samples were analyzed to identify differentially expressed genes (DEGs). Subsequently, 141 genes were identified, including 55 upregulated and 86 downregulated genes. The protein-protein interaction network was established using the Search Tool for Retrieval of Interacting Genes database. Hub gene identification and module analysis were performed using Cytoscape software. Hierarchical clustering of hub genes was conducted using the University of California, Santa Cruz Cancer Genomics Browser. Among the hub genes, kinesin family member 11 (KIF11) was identified as one of the most significant prognostic biomarkers among all the candidates. The Kaplan Meier Plotter database was used for survival analysis of KIF11. The expression profile of KIF11 was analyzed using the ONCOMINE database. The expression levels of KIF11 in BC samples and bladder cells were measured using reverse transcription-quantitative pCR, immunohistochemistry and western blotting. In summary, KIF11 was significantly upregulated in BC and might act as a potential prognostic biomarker. The present identification of DEGs and hub genes in BC may provide novel insight for investigating the molecular mechanisms of BC.

17.
Br J Cancer ; 123(10): 1521-1534, 2020 11.
Article in English | MEDLINE | ID: mdl-32801345

ABSTRACT

BACKGROUND: High probability of metastasis limited the long-term survival of patients with hepatocellular carcinoma (HCC). Our previous study revealed that Galectin-3 was closely associated with poor prognosis in HCC patients. METHODS: The effects of Galectin-3 on tumour metastasis were investigated in vitro and in vivo, and the underlying biological and molecular mechanisms involved in this process were evaluated. RESULTS: Galectin-3 showed a close correlation with vascular invasion and poor survival in a large-scale study in HCC patients from multiple sets. Galectin-3 was significantly involved in diverse metastasis-related processes in HCC cells, such as angiogenesis and epithelial-to-mesenchymal transition (EMT). Mechanistically, Galectin-3 activated the PI3K-Akt-GSK-3ß-ß-catenin signalling cascade; the ß-catenin/TCF4 transcriptional complex directly targeted IGFBP3 and vimentin to regulate angiogenesis and EMT, respectively. In animal models, Galectin-3 enhanced the tumorigenesis and metastasis of HCC cells via ß-catenin signalling. Moreover, molecular deletion of Galectin-3-ß-catenin signalling synergistically improved the antitumour effect of sorafenib. CONCLUSIONS: The Galectin-3-ß-catenin-IGFBP3/vimentin signalling cascade was determined as a central mechanism controlling HCC metastasis, providing possible biomarkers for predicating vascular metastasis and sorafenib resistance, as well as potential therapeutic targets for the treatment of HCC patients.


Subject(s)
Carcinoma, Hepatocellular/pathology , Galectin 3/physiology , Liver Neoplasms/pathology , beta Catenin/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neoplasms, Vascular Tissue/genetics , Neoplasms, Vascular Tissue/mortality , Neoplasms, Vascular Tissue/secondary , Survival Analysis , Tissue Array Analysis , Wnt Signaling Pathway/genetics , beta Catenin/metabolism
18.
Oncoimmunology ; 9(1): 1752563, 2020.
Article in English | MEDLINE | ID: mdl-32363125

ABSTRACT

Adjuvant chemotherapy after surgery is the standard treatment modality for stage III and part of stage II or stage IV colorectal cancer (CRC) patients. However, the 5-year overall survival (OS) rate remains unsatisfactory. Thus, developing combination therapies is essential to improve the prognosis of patients with CRC. The present study aimed to determine the effect of a sequential combination of cytokine-induced killer cell (CIK) infusion and chemotherapy for patients with CRC. 122 patients with CRC treated with postoperative adjuvant chemotherapy were retrospectively included in this study. Among them, 62 patients received adjuvant chemotherapy only (control group), while the other 60 patients, with similar demographic and clinical characteristics, received adjuvant chemotherapy and sequential CIK cell immunotherapy (CIK group). Survival analysis showed significantly improved disease free survival (DFS) and OS rates in the CIK group compared with the control group (log-rank test, P = .0024; P = .008, respectively). Univariate and multivariate analyses indicated that sequential CIK cell treatment was an independent prognostic factor for patients' DFS and OS. Subgroup analyses showed that sequential CIK cell treatment significantly improved the DFS and OS of patients with high-risk T4 stage and insufficient chemotherapy duration. In conclusion, these data indicate that sequential adjuvant CIK cell treatment combined with chemotherapy is an effective therapeutic strategy to prevent disease recurrence and prolong survival of patients with CRC, particularly for patients with high-risk T4 stage and insufficient chemotherapy duration.


Subject(s)
Colorectal Neoplasms , Cytokine-Induced Killer Cells , Chemotherapy, Adjuvant , Colorectal Neoplasms/therapy , Combined Modality Therapy , Digestive System Surgical Procedures , Female , Humans , Immunotherapy, Adoptive , Male , Middle Aged , Neoplasm Recurrence, Local , Retrospective Studies
19.
Physiol Meas ; 41(2): 025004, 2020 03 06.
Article in English | MEDLINE | ID: mdl-31962305

ABSTRACT

OBJECTIVE: The objectives of this study were to investigate the response of plantar skin blood flow (SBF) to different frequencies of local vibration (LV) and investigate the mechanisms of blood flow control in response to the different frequencies of LV using wavelet analysis. APPROACH: Twelve healthy participants were recruited to test the effects of three frequencies [0 Hz (sham control), 35 Hz and 100 Hz at 1 mm amplitude] of 10 min LV on the skin of the right first metatarsal head. A repeated measures design was used in this study and the order of vibration frequencies was randomly assigned to participants while they lay on a mat table. Laser Doppler flowmetry was used to measure SBF on the first metatarsal head before and after LV for 10 min. SBF after vibration was expressed as a ratio of SBF before vibration. Wavelet analysis was used to characterize changes in SBF control mechanisms. MAIN RESULTS: Our results showed that the SBF with the 100 Hz protocol (1.56 ± 0.19) was significantly higher than for the 35 Hz (1.29 ± 0.18, p  < 0.05) and 0 Hz (0.85 ± 0.11, p  < 0.01) protocols. Wavelet analysis demonstrated that metabolic endothelial control (SBF oscillations of 0.0095-0.02 Hz) and neurogenic control (SBF oscillations of 0.02-0.05 Hz) were associated with the increase in SBF after LV at 100 Hz. SIGNIFICANCE: LV at 100 Hz results in a significant increase in SBF compared with 35 Hz and 0 Hz vibrations. Such an increase in SBF is related to the regulation of metabolic endothelial (0.0095-0.02 Hz) and neurogenic (0.02-0.05 Hz) controls. This is the first study to demonstrate that 100 Hz LV stimulates an increase in plantar SBF compared with 35 Hz and 0 Hz vibrations through the frequency intervals of 0.0095-0.02 and 0.02-0.05 Hz of SBF oscillations.


Subject(s)
Regional Blood Flow , Skin/blood supply , Vibration , Wavelet Analysis , Healthy Volunteers , Humans
20.
J Hematol Oncol ; 13(1): 2, 2020 01 03.
Article in English | MEDLINE | ID: mdl-31900208

ABSTRACT

BACKGROUND: Clinically, the median survival in patients with metastatic renal cell carcinoma (RCC) was only 6-12 months and a 5-year survival rate of less than 20%. Therefore, an in-depth study of the molecular mechanisms involved in RCC is of great significance for improving the survival of patients with advanced RCC. Acylglycerol kinase (AGK) is a newly discovered lipid kinase that has been reported to be a potent oncogene that may be involved in the regulation of malignant progression in a variety of tumours. However, the expression and biological characteristics of the AGK gene in RCC remain unclear. METHODS: AGK expression was quantified by quantitative real-time PCR, Western blotting and immunohistochemistry in RCC cell lines and paired patient tissues. Kaplan-Meier method and Cox proportional hazards models were used to evaluate the prognostic value of AGK in human RCC tissue samples. Chi-squared test was performed to analyse the correlation between AGK expression and the clinicopathological features. Stable overexpression and knockdown of AGK in RCC cells was constructed with lentivirus. The oncogenic effects of AGK in human RCC progression were investigated using assays of colony formation, anchorage-independent growth, EdU assay, cell cycle analysis, wound-healing, trans-well analysis and xenograft tumour model. GSEA and KEGG analysis were conducted to detect the potential pathway of AGK involved in RCC. These results were further confirmed using the luciferase reporter assays, immunofluorescence and in vivo experiments. RESULTS: AGK expression is significantly elevated in RCC and closely related to the malignant development and poor prognosis in RCC patients. By in vitro and in vivo experiments, AGK was shown to enhance the proliferation of RCC cells by promoting the transition from the G1 phase to the S phase in the cell cycle and to enhance the migration and invasion by promoting epithelial-mesenchymal transition. By activating the PI3K/AKT/GSK3ß signalling pathway in RCC, AGK can increase nuclear accumulation of ß-catenin, which further upregulated TCF/LEF transcription factor activity. CONCLUSIONS: AGK promotes the progression of RCC via activating the PI3K/AKT/GSK3ß signalling pathway and might be a potential target for the further research of RCC.


Subject(s)
Carcinoma, Renal Cell/pathology , Glycogen Synthase Kinase 3 beta/metabolism , Kidney Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Disease Progression , Enzyme Activation , Female , Humans , Kidney Neoplasms/metabolism , Male , Middle Aged , Neoplasm Metastasis/pathology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...