4
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Roles of hypoxia-inducible factor in hepatocellular carcinoma under local ablation therapies

      review-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Hepatocellular carcinoma (HCC) is one of the most common digestive malignancies. HCC It ranges as the fifth most common cause of cancer mortality worldwide. While The prognosis of metastatic or advanced HCC is still quite poor. Recently, locoregional treatment, especially local ablation therapies, plays an important role in the treatment of HCC. Radiofrequency ablation (RFA) and high-intensity focused ultrasound (HIFU) ablation are the most common-used methods effective and feasible for treating HCC. However, the molecular mechanisms underlying the actions of ablation in the treatments for HCC and the HCC recurrence after ablation still are poorly understood. Hypoxia-inducible factor (HIF), the key gene switch for adaptive responses to hypoxia, has been found to play an essential role in the rapid aggressive recurrence of HCC after ablation treatment. In this review, we summarized the current evidence of the roles of HIF in the treatment of HCC with ablation. Fifteen relevant studies were included and further analyzed. Among them, three clinical studies suggested that HIF-1α might serve as a crucial role in the RAF treatment of HCC or the local recurrence of HCC after RFA. The remainder included experimental studies demonstrated that HIF-1, 2α might target the different molecules (e.g., BNIP3, CA-IX, and arginase-1) and signaling cascades (e.g., VEGFA/EphA2 pathway), constituting a complex network that promoted HCC invasion and metastasis after ablation. Currently, the inhibitors of HIF have been developed, providing important proof of targeting HIF for the prevention of HCC recurrence after IRFA and HIFU ablation. Further confirmation by prospective clinical and in-depth experimental studies is still warranted to illustrate the effects of HIF in HCC recurrence followed ablation treatment in the future.

          Related collections

          Most cited references136

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy

          Hypoxia is a non-physiological level of oxygen tension, a phenomenon common in a majority of malignant tumors. Tumor-hypoxia leads to advanced but dysfunctional vascularization and acquisition of epithelial-to-mesenchymal transition phenotype resulting in cell mobility and metastasis. Hypoxia alters cancer cell metabolism and contributes to therapy resistance by inducing cell quiescence. Hypoxia stimulates a complex cell signaling network in cancer cells, including the HIF, PI3K, MAPK, and NFĸB pathways, which interact with each other causing positive and negative feedback loops and enhancing or diminishing hypoxic effects. This review provides background knowledge on the role of tumor hypoxia and the role of the HIF cell signaling involved in tumor blood vessel formation, metastasis, and development of the resistance to therapy. Better understanding of the role of hypoxia in cancer progression will open new windows for the discovery of new therapeutics targeting hypoxic tumor cells and hypoxic microenvironment.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway.

            HIF plays a central role in the transcriptional response to changes in oxygen availability. The PHD family of oxygen-dependent prolyl hydroxylases plays a pivotal role in regulating HIF stability. The biochemical properties of these enzymes make them well suited to act as oxygen sensors. They also respond to other intracellular signals, including reactive oxygen species, nitric oxide, and certain metabolites, that can modulate the hypoxic response. HIF transcriptional activity is further tuned by FIH1-mediated asparagine hydroxylation. HIF affects signaling pathways that influence development, metabolism, inflammation, and integrative physiology. Accordingly, HIF-modulatory drugs are now being developed for diverse diseases.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I

              Immune evasion is a major obstacle for cancer treatment. Common mechanisms include impaired antigen presentation through mutations or loss of heterozygosity (LOH) of the major histocompatibility complex class I (MHC-I), which has been implicated in resistance to immune checkpoint blockade (ICB) therapy 1–3 . However, in pancreatic ductal adenocarcinoma (PDAC), a malignancy refractory to most therapies including ICB 4 , mutations causing MHC-I loss are rarely found 5 despite the frequent downregulation of MHC-I expression 6–8 . Here we find that, in PDAC, MHC-I molecules are selectively targeted for lysosomal degradation through an autophagy-dependent mechanism that involves the autophagy cargo receptor NBR1. PDAC cells display reduced MHC-I cell surface expression and instead demonstrate predominant localization within autophagosomes and lysosomes. Notably, autophagy inhibition restores surface MHC-I levels, leading to improved antigen presentation, enhanced anti-tumour T cell response and reduced tumour growth in syngeneic hosts. Accordingly, anti-tumour effects of autophagy inhibition are reversed by depleting CD8+ T cells or reducing surface MHC-I expression. Autophagy inhibition, either genetically or pharmacologically with Chloroquine (CQ), synergizes with dual ICB (anti-PD1 and anti-CTLA4), and leads to an enhanced anti-tumour immune response. Our findings uncover a role for enhanced autophagy/lysosome function in immune evasion through selective targeting of MHC-I molecules for degradation, and provide a rationale for the combination of autophagy inhibition and dual ICB as a therapeutic strategy against PDAC.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Pharmacol
                Front Pharmacol
                Front. Pharmacol.
                Frontiers in Pharmacology
                Frontiers Media S.A.
                1663-9812
                06 February 2023
                2023
                : 14
                : 1086813
                Affiliations
                [1] 1 Department of Ultrasound , Taizhou Central Hospital (Taizhou University , Hospital) , Taizhou, Zhejiang, China
                [2] 2 Department of Hepatobiliary Surgery , Hubei Cancer Hospital , Tongji Medical College , Huazhong University of Science and Technology , Wuhan, China
                [3] 3 Department of General Surgery , Taizhou Central Hospital (Taizhou University , Hospital) , Taizhou, Zhejiang, China
                [4] 4 Precision Medicine Center , Taizhou Central Hospital (Taizhou University Hospital) , Taizhou, Zhejiang, China
                [5] 5 Liyuan Hospital , Tongji Medical College , Huazhong University of Science and Technology , Wuhan, China
                Author notes

                Edited by: Sarah El-Nakeep, Ain Shams University, Egypt

                Reviewed by: Anuhya Kommalapati, Mayo Clinic, United States

                Antonio Facciorusso, University of Foggia, Italy

                *Correspondence: Qianqian Zhang, Zhangqq20221130@ 123456126.com ; Jianyu Zhou, zhoujy9980@ 123456126.com
                [ † ]

                These authors have contributed equally to this work and share first authorship

                This article was submitted to Gastrointestinal and Hepatic Pharmacology, a section of the journal Frontiers in Pharmacology

                Article
                1086813
                10.3389/fphar.2023.1086813
                9939531
                36814489
                9428c5e1-9138-4c41-9bdf-adced2eeb4fe
                Copyright © 2023 Xiao, Liu, Ge, Jiang, Wang, Chen, Jin, Mo, Li, Wang, Zhang and Zhou.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 01 November 2022
                : 18 January 2023
                Funding
                This project was supported by the grants from Zhejiang Province Public Welfare Technology Application Research Project (No. LGF21H160022), the Natural Science Foundation of Zhejiang Province (No. LQ22H160055), Science and Technology Plan Project of Taizhou (No. 21ywb26, 21ywb29, and 22ywa14), Medical Science and Technology Project of Zhejiang Province (No. 2017KY711, 2023KY403, 2023KY404, and 2023KY1340).
                Categories
                Pharmacology
                Review

                Pharmacology & Pharmaceutical medicine
                hepatocellular carcinoma,hypoxia-inducible factor,ablation,therapy,mechanism

                Comments

                Comment on this article