Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunother Cancer ; 11(2)2023 02.
Article in English | MEDLINE | ID: mdl-36849198

ABSTRACT

BACKGROUND: Effects of immune checkpoint blockade (ICB) treatment in hepatocellular carcinoma (HCC) are limited. The current study explored the possibility of exploiting tumor metabolic switches to enhance HCC sensitivity to immune therapies. METHODS: Levels of one-carbon (1C) metabolism and the expression of phosphoserine phosphatase (PSPH), an upstream enzyme of 1C pathway, were evaluated in paired non-tumor and tumor tissues from HCC. Underlying mechanisms mediating the role of PSPH in regulating the infiltration of monocytes/macrophages and CD8+ T lymphocytes were studied through both in vitro and in vivo experiments. RESULTS: PSPH was significantly upregulated in tumor tissues of HCC and its levels were positively correlated with disease progression. PSPH knockdown inhibited tumor growth in immunocompetent mice, but not in those with macrophage or T lymphocyte deficiencies, indicating the pro-tumor effects of PSPH were dependent on both immune components. Mechanistically, PSPH facilitated monocytes/macrophages infiltration by inducing the production of C-C motif chemokine 2 (CCL2), while at the same time reduced CD8+ T lymphocytes recruitment through inhibiting the production of C-X-C Motif Chemokine 10 (CXCL10) in tumor necrosis factor alpha (TNF-α)-conditioned cancer cells. Glutathione and S-adenosyl-methionine were partially involved in regulating the production of CCL2 and CXCL10, respectively. shPSPH (short hairpin RNA) transfection of cancer cells enhanced tumor sensitivity to anti-programmed cell death protein 1 (PD-1) therapy in vivo, and interestingly, metformin could inhibit PSPH expression in cancer cells and mimic the effects of shPSPH in sensitizing tumors to anti-PD-1 treatment. CONCLUSIONS: By tilting the immune balance towards a tumor-friendly composition, PSPH might be useful both as a marker in stratifying patients for ICB therapy, and as an attractive therapeutic target in the treatment of human HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Animals , Mice , Carcinoma, Hepatocellular/drug therapy , Down-Regulation , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Liver Neoplasms/drug therapy
2.
J Clin Invest ; 132(7)2022 04 01.
Article in English | MEDLINE | ID: mdl-35362480

ABSTRACT

Macrophages constitute a major immune component in tumor tissues, but how these cells adapt to and survive in the nutrient-depleted and lactic acid-induced acidic tumor microenvironments is not yet fully understood. Here, we found that levels of carbonic anhydrase XII (CA12) expression were significantly and selectively upregulated on macrophages in human hepatocellular carcinoma (HCC). Transient glycolytic activation of peritumoral monocytes induced sustained expression of CA12 on tumor-infiltrating macrophages via autocrine cytokines and HIF1α pathways. On the one hand, CA12 mediated the survival of macrophages in relatively acidic tumor microenvironments, while on the other hand, it induced macrophage production of large amounts of C-C motif chemokine ligand 8 (CCL8), which enhanced cancer cell epithelial-mesenchymal transition (EMT) and facilitated tumor metastasis. Consistently, the accumulation of CA12+ macrophages in tumor tissues was associated with increased tumor metastatic potential and reduced survival of patients with HCC. Selective targeting of tumor-infiltrating macrophages with a CA12 inhibitor reduced tumor growth in mice and was sufficient to synergistically enhance the therapeutic efficacy of immune-checkpoint blockade. We suggest that CA12 activity is a previously unappreciated mechanism regulating the accumulation and functions of macrophages in tumor microenvironments and therefore represents a selective vulnerability that could be exploited in future designs for antitumor immunotherapeutic strategies.


Subject(s)
Carbonic Anhydrases , Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Carbonic Anhydrases/genetics , Carbonic Anhydrases/metabolism , Carcinoma, Hepatocellular/metabolism , Humans , Liver Neoplasms/metabolism , Macrophages/metabolism , Mice , Tumor Microenvironment
3.
Oncoimmunology ; 11(1): 2052418, 2022.
Article in English | MEDLINE | ID: mdl-35309732

ABSTRACT

Neutrophils constitute a major component in human hepatocellular carcinoma (HCC) and can facilitate disease progression via poorly understood mechanisms. Here, we show that neutrophil extracellular traps (NETs) formation was increased in human HCC tumor tissues than in paired non-tumor liver tissues. Mechanism study revealed that tumor-induced metabolic switch toward glycolysis and pentose phosphate pathway in tumor infiltrating neutrophils promoted NETs formation in a reactive oxygen species dependent-manner. NETs subsequently induced the migration of cancer cells and down-regulation of tight junction molecules on adjacent endothelial cells, thus facilitating tumor intravasation and metastasis. Accordingly, NETs depletion could inhibit tumor metastasis in mice in vivo, and the infiltration levels of NETs-releasing neutrophils were negatively associated with patient survival and positively correlated with tumor metastasis potential of HCC patients. Our results unveiled a pro-metastatic role of NETs in the milieu of human HCC, and pointed to the importance of metabolic reprogramming in shaping their characteristics, thus providing an applicable efficient target for anti-cancer therapies.


Subject(s)
Carcinoma, Hepatocellular , Extracellular Traps , Liver Neoplasms , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Extracellular Traps/metabolism , Humans , Liver Neoplasms/secondary , Mice , Neutrophils
4.
J Hepatol ; 73(4): 906-917, 2020 10.
Article in English | MEDLINE | ID: mdl-32407813

ABSTRACT

BACKGROUND & AIMS: Neutrophils are one of the most abundant components in human hepatocellular carcinoma (HCC) and have been shown to play important roles in regulating disease progression. However, neutrophils are very short-lived cells in circulation, and mechanisms regulating their accumulation and functions in HCC are not yet fully understood. METHODS: Monocytes were purified from non-tumor or paired tumor tissues of patients with HCC, and their production of neutrophil-attracting chemokines was evaluated. Mechanisms regulating the expression of CXCL2/8 by tumor monocytes, and the role of tumor monocyte-derived chemokines and cytokines in modulating neutrophil accumulation and functions were studied with both ex vivo analyses and in vitro experiments. RESULTS: Monocyte-derived CXCL2 and CXCL8 were major factors in regulating the recruitment of neutrophils into tumor milieus. These chemokines, in addition to tumor-derived soluble factors, could inhibit apoptosis and sustain survival of neutrophils, thus leading to neutrophil accumulation in tumor tissues. Moreover, monocyte-derived TNF-α acted synergistically with tumor-derived soluble factors to induce the production of the pro-metastasis factor OSM by neutrophils. Further, the glycolytic switch in tumor-infiltrating monocytes mediated their production of CXCL2 and CXCL8 via the PFKFB3-NF-κB signaling pathway. Accordingly, levels of PFKFB3, CXCL2/CXCL8 production in monocytes and infiltration of OSM-producing neutrophils were positively correlated in human HCC tissues. CONCLUSIONS: Our results unveiled a previously unappreciated link between monocytes and neutrophils in human HCC, identifying possible targets that could be therapeutically exploited in the future. LAY SUMMARY: Neutrophils constitute a major but poorly understood component of human hepatocellular carcinoma (HCC). Herein, we unveil a novel mechanism by which metabolic switching in monocytes promotes the accumulation of neutrophils in the tumors of patients with HCC. Both monocyte-produced chemokines and signals from the tumor microenvironment promote the production of the pro-metastatic factor OSM by neutrophils. These data identify potential targets for immune-based anticancer therapies for HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Glycolysis/physiology , Liver Neoplasms/metabolism , Monocytes/metabolism , Neutrophils/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Disease Progression , Female , Humans , Liver Neoplasms/pathology , Male , Monocytes/pathology , Neutrophils/pathology , Retrospective Studies , Signal Transduction
5.
J Hepatol ; 71(2): 333-343, 2019 08.
Article in English | MEDLINE | ID: mdl-31071366

ABSTRACT

BACKGROUND & AIMS: Programmed cell death 1 ligand 1 (PD-L1) expression on antigen-presenting cells is essential for T cell impairment, and PD-L1-expressing macrophages may mechanistically shape and therapeutically predict the clinical efficacy of PD-L1 or programmed cell death 1 blockade. We aimed to elucidate the mechanisms underlying PD-L1 upregulation in human tumor microenvironments, which remain poorly understood despite the clinical success of immune checkpoint inhibitors. METHODS: Monocytes/macrophages were purified from peripheral blood, non-tumor, or paired tumor tissues of patients with hepatocellular carcinoma (HCC), and their possible glycolytic switch was evaluated. The underlying regulatory mechanisms and clinical significance of metabolic switching were studied with both ex vivo analyses and in vitro experiments. RESULTS: We found that monocytes significantly enhanced the levels of glycolysis at the peritumoral region of human HCC. The activation of glycolysis induced PD-L1 expression on these cells and subsequently attenuated cytotoxic T lymphocyte responses in tumor tissues. Mechanistically, tumor-derived soluble factors, including hyaluronan fragments, induced the upregulation of a key glycolytic enzyme, PFKFB3, in tumor-associated monocytes. This enzyme not only modulated the cellular metabolic switch but also mediated the increased expression of PD-L1 by activating the nuclear factor kappa B signaling pathway in these cells. Consistently, the levels of PFKFB3+CD68+ cell infiltration in peritumoral tissues were negatively correlated with overall survival and could serve as an independent prognostic factor for survival in patients with HCC. CONCLUSIONS: Our results reveal a mechanism by which the cellular metabolic switch regulates the pro-tumor functions of monocytes in a specific human tumor microenvironment. PFKFB3 in both cancer cells and tumor-associated monocytes is a potential therapeutic target in human HCC. LAY SUMMARY: Programmed cell death 1 ligand 1 (PD-L1) expressed on antigen-presenting cells, rather than tumor cells, has been reported to play an essential role in checkpoint blockade therapy. A fundamental understanding of mechanisms that regulate the expression of PD-L1 on tumor-infiltrating monocytes/macrophages will undoubtedly lead to the possibility of developing novel PD-L1 blockade strategies with high specificity and efficiency. The current study unveils a novel mechanism by which metabolic switching links immune activation responses to immune tolerance in the tumor milieu, identifying potential targets for future immune-based anti-cancer therapies.


Subject(s)
B7-H1 Antigen/metabolism , Carcinoma, Hepatocellular/immunology , Glycolysis , Immune Privilege , Liver Neoplasms/immunology , Monocytes/metabolism , Phosphofructokinase-2/metabolism , Adult , Aged , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/pathology , Disease Progression , Female , Follow-Up Studies , Hep G2 Cells , Humans , Liver Neoplasms/blood , Liver Neoplasms/pathology , Macrophages/metabolism , Male , Middle Aged , Monocytes/immunology , Tumor Microenvironment/immunology , Young Adult
6.
Autophagy ; 14(8): 1335-1346, 2018.
Article in English | MEDLINE | ID: mdl-29940792

ABSTRACT

Macroautophagy/autophagy is an important catabolic process mediating cellular homeostasis and plays critical roles in cancer development. Whereas autophagy has been widely studied in various pathological models, little is known about the distribution, clinical significance and regulatory mechanism of this process in human hepatocellular carcinoma (HCC). In the present study, we found that tumor tissues exhibited significantly increased levels of autophagy compared with non-tumor tissues, and cancer cells with higher levels of autophagy were predominantly enriched in the invading edge regions of human HCC. Increased MAP1LC3B/LC3B expression in the invading edge regions was significantly correlated with a higher density of closely located monocytes, and TNF and IL1B derived from tumor-activated monocytes synergistically induced cancer cell autophagy in the invading edge regions of HCC. Monocyte-elicited autophagy induced the epithelial-mesenchymal transition (EMT) of cancer cells and promoted tumor metastasis by activating the NFKB-SNAI1 signaling pathway. Moreover, the increase of LC3B+ cancer cells in the invading edge areas was associated with high mortality and reduced survival of patients with HCC. These findings indicated that cancer cell autophagy is regulated by a collaborative interaction between tumor and immune cell components in distinct HCC microenvironments, thus allowing the inflammatory monocytes to be rerouted in a tumor-promoting direction.


Subject(s)
Autophagy , Carcinoma, Hepatocellular/pathology , Disease Progression , Liver Neoplasms/pathology , Monocytes/pathology , Autophagy/drug effects , Carcinoma, Hepatocellular/ultrastructure , Cell Line, Tumor , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Interleukin-1beta/pharmacology , Liver Neoplasms/ultrastructure , Male , Middle Aged , Monocytes/drug effects , Monocytes/ultrastructure , Multivariate Analysis , NF-kappa B/metabolism , Neoplasm Recurrence, Local/pathology , Signal Transduction/drug effects , Snail Family Transcription Factors/metabolism , Solubility , Survival Analysis , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
7.
Zootaxa ; 4179(2): 209-224, 2016 Oct 31.
Article in English | MEDLINE | ID: mdl-27811669

ABSTRACT

Two new species of turbellarians, Bothrioplana sinensis n. sp., and Pentacoelum sinensis n. sp., were described based on their reproductive behavior, ontogenic development, morphology in whole-mounted and sectioned specimens, and 18S rDNA phylogenetic classification. Bothrioplana sinensis n. sp. represents a newly recorded order in China and the second identified species in the genus Bothrioplana. It is characterized by a pair of spherical-shaped well-developed testes located dorsally behind the pharynx, and the common vas deference located ventrally to the gonopore. Phylogenetic analysis showed that this new species is closely related to parasitic flatworms. Pentacoelum sinensis n. sp. is characterized by two uteri instead of lateral bursae located laterally near the tail end and a lack of connection between the posterior intestinal branches. Each uterus has a ventral receptaculum seminalis (also functioning as bursa copulatrix) which has a muscular vagina externa opening obliquely to the ventral side at the tail end.


Subject(s)
Platyhelminths/anatomy & histology , Platyhelminths/classification , Animals , China , DNA, Ribosomal/genetics , Hermaphroditic Organisms , Phylogeny , Platyhelminths/genetics , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...