簡易檢索 / 詳目顯示

研究生: 林原禾
Lin, Yuan-Ho
論文名稱: 抑制長鏈非編碼核糖核酸RPPH1的活性能下調發炎相關的致癌基因以減少腫瘤的生長與轉移
Inhibition of lncRNA RPPH1 activity decreases tumor proliferation and metastasis through down-regulation of inflammation-related oncogenes
指導教授: 張文粲
Chang, Wen-Tsan
學位類別: 博士
Doctor
系所名稱: 醫學院 - 基礎醫學研究所
Institute of Basic Medical Sciences
論文出版年: 2020
畢業學年度: 108
語文別: 英文
論文頁數: 103
中文關鍵詞: 長鏈非編碼核糖核酸RPPH1轉運核糖核酸癌症發炎白细胞介素6
外文關鍵詞: LncRNA, RPPH1, tRNA, HeLa, MDA-MB-231, inflammation, cancer, IL-6
相關次數: 點閱:83下載:0
分享至:
查詢本校圖書館目錄 查詢臺灣博碩士論文知識加值系統 勘誤回報
  • 核糖核酸酶 P是由十個相異的蛋白單元以及一個核糖核酸所組成的酵素,在細胞中負責切割 tRNA 5’端並使其轉變為成熟的tRNA。早期的研究中發現,過度表現核糖核酸酶 P中的核醣核酸單元-RPPH1會促進癌細胞的生長與發展,然而其詳細機制仍尚未明瞭。為了調查RPPH1在腫瘤發展中所扮演的角色,我們設計出能高專一性與RPPH1結合的shRNA以作為研究手段。藉由次世代定序與生物資訊的方式分析RPPH1抑制細胞(HeLa與MDA-MB-231)與控制組細胞後一共列出40個顯著被下調節的基因。這些被下調的基因主要參與tRNA路徑、免疫反應與癌症相關發炎反應(IL-6路徑)。為了證實生物資訊分析的結果,我們分析了RPPH1抑制細胞與控制組細胞的生長與轉移。實驗的結果呈現出RPPH1抑制細胞比起控制組細胞其生長與轉移能力均顯著的降低。我們證明了抑制RPPH1後所導致的G1細胞週期延遲主要是透過下調細胞週期蛋白D1所導致。此外,RPPH1抑制細胞內的糖解酵素(HK1與HK2)也會被下調。總結來說,這些結果證實抑制RPPH1能下調發炎相關的致癌基因並減少腫瘤的發展。這項研究為RPPH1在腫瘤的發展中所扮演的角色提供了新的觀點。

    Ribonuclease P RNA component H1 (RPPH1) is an RNA subunit of RNase P that plays a central role in RNase P-mediated pre-tRNA cleavage. Previous studies showed that overexpression of RPPH1 promotes tumor growth and progression, but the molecular mechanism is still unclear. To investigate the role and function of RPPH1 in cancer malignancy, short hairpin RNAs were designed to specifically target RPPH1. A total of 40 significantly down-regulated genes between RPPH1 knockdown (HeLa-shRPPH1-1-52 and MDA-MB-231-shRPPH1-1-72) and mock cells were identified using next-generation sequencing and bioinformatics analysis. The results indicate that these down-regulated genes are the major genes involved in tRNA pathways, immune response, and cancer-related inflammation (IL-6 pathway). To confirm the results of the bioinformatics analysis, RPPH1-silenced cells were analyzed using various experiments. A significant down-regulation of tumor proliferation and migration was observed in RPPH1 knockdown cells compared to that of mock- and vector-transfected cells. We demonstrate that the inhibition of RPPH1 promotes cell cycle arrest in the G1 phase mainly through the down-regulation of cyclin D1. In addition, the glycolytic enzyme (HK1 and HK2) is also down-regulated in RPPH1-silenced HeLa cells. Taken together, these results clearly reveal that the knockdown of RPPH1 expression decreases tumor progression via the down-regulation of inflammation-related oncogene. This study provides insight into the role of RPPH1 in tumor development.

    一、中文摘要 I 二、Abstract II 三、誌謝 III 四、Table of Contents IV~VIII 五、Main text Chapter 1. Introduction 1~8 1.1 Inflammation and Cancer 2 1.2 Hallmarks of cancer 3 1.3 Reprogramming of energy metabolism 4 1.4 Translational alteration in cancer 5 1.5 Ribonuclease P 5~6 1.6 Metabolic reprogramming of SDH-deficient cancers 6~7 1.7 Study aim 7~8 Chapter 2. Materials and Methods 9~18 Materials 10~13 Methods 14~18 2.1 Cell culture 14 2.2 Cell transfection 14 2.3 Design and construction of the shRNA expression vector 14~15 2.4 Next-Generation Sequencing 15 2.5 MTT cell growth assay 15 2.6 Colony formation assay 16 2.7 Wound healing migration assay 16 2.8 Boyden chamber migration assay 16 2.9 Cell synchronization by double thymidine block 17 2.10 Western blot analysis 17 2.11 pH measurement in conditioned medium 18 2.12 Immunohistochemical staining 18 Chapter 3. Results 19~27 Project 1 3.1 RPPH1 expression in various cancers and affect on prognosis 20 3.2 Construction and identification of differentially expressed genes in RPPH1 knockdown cancer cells 20~21 3.3 Top 40 potential down-regulated genes in RPPH1 knockdown cells 21 3.4 Inhibition of RPPH1 expression decreases mature tRNA generation and several tRNA pathways 21~22 3.5 Down-regulation of RPPH1 expression decreases tumor cell proliferation and migration 22~23 3.6 RPPH1 knockdown cells promote G1 arrest through down-regulation of cyclin D1 23~24 3.7 Down-regulated RPPH1 affects glycolytic pathway in cervical cancer 24 Project 2 3.8 High SDHC expression in various cancers as compared to SDHB subunits 25 3.9 SDHC overexpression correlates with poor prognosis in lung cancers 25~26 3.10 Inhibition of SDHC decreases lung cancer cell proliferation and migration 26~27 3.11 Inhibition of SDHC expression promotes the up-regulation of SDHA protein expression 27 Chapter 4. Discussion 28~34 Project 1 4.1 The role of RPPH1 in tumor protein synthesis 29 4.2 Link between RPPH1 and cancer-related inflammation 30 4.3 The role of RPPH1 in the glycolytic pathway 31 4.4 Inhibition of RPPH1 affects the function of RNase P 31~32 4.5 Conclusion 32 Project 2 4.6 High SDHC expression is associated with the malignancy of lung cancer cells and poor prognosis 32~33 4.7 Up-regulation of SDHA reduces the accumulation of succinate 33 4.8 Conclusion 34 Chapter 5. References 35~45 六、Table and Figures 46~74 Table 47 Table 1. Enrichment by pathways maps 47 Figures 48~74 Project 1 Figure 1. High RPPH1 expression in various human cancers 48 Figure 2. Tumors with high RPPH1 expression exhibit a poor prognosis 49 Figure 3. Flowchart of study design 50 Figure 4. Differentially expressed RNA between RPPH1 knockdown cells and mock cells 51 Figure 5. Potential pathways of down-regulated gene in RPPH1 silencing cells 52 Figure 6. Top 40 potential down-regulated genes in RPPH1 knockdown cells 53 Figure 7. Several amino acid pathways and tRNA production were down-regulated in RPPH1 inhibited cells 54 Figure 8. RPPH1 inhibited cells decrease IL-6 protein expression 55 Figure 9. Inhibition of RPPH1 decreases cancer cell proliferation and migration...56~57 Figure 10. Knockdown of RPPH1 decreases cell cycle progression 58 Figure 11. RPPH1 inhibited HeLa cell down-regulates the expression of glucose pathway-related genes 59 Figure 12. RPPH1 inhibited cells decrease glucose dependence and uptake 60 Figure 13. Potential pathway of RPPH1 in cervical cancer 61 Project 2 Figure 14. Gene expression of SDH subunits in various tumors 62 Figure 15. SDHC expression is related to the lung cancer malignancy 63 Figure 16. Lung cancers with high SDHC expression exhibit a poor prognosis 64 Figure 17. Construction of SDHC knockdown A549 and H1299 cells 65 Figure 18. Knockdown of SDHC decreases cancer cell proliferation and colony formation 66 Figure 19. Inhibition of SDHC decreases tumor migration 67 Figure 20. Knockdown of SDHC decreases mobility of lung cancer cells 68 Figure 21. Inhibition of SDHC increases SDHA protein expression 69 Supplemental Figures 70~74 Figure S1. Tumors with high Rpp40 expression exhibit a poor prognosis 70 Figure S2. Inhibition of Rpp40 decreases cancer cell proliferation and migration …..71~72 Figure S3. Inhibition of Rpp40 decreases cell cycle progression 73 Figure S4. Rpp40 inhibited cells decrease glucose dependence and uptake 74 六、Publication List 75~103

    [1] Cronkite DA and Strutt TM. The Regulation of Inflammation by Innate and Adaptive Lymphocytes. J Immunol Res 2018; 2018: 1467538.
    [2] Agita A and Alsagaff MT. Inflammation, Immunity, and Hypertension. Acta Med Indones 2017; 49: 158-165.
    [3] Netea MG, Joosten LA, Latz E, Mills KH, Natoli G, Stunnenberg HG, O'Neill LA and Xavier RJ. Trained immunity: A program of innate immune memory in health and disease. Science 2016; 352: aaf1098.
    [4] Singh N, Baby D, Rajguru JP, Patil PB, Thakkannavar SS and Pujari VB. Inflammation and cancer. Ann Afr Med 2019; 18: 121-126.
    [5] Diakos CI, Charles KA, McMillan DC and Clarke SJ. Cancer-related inflammation and treatment effectiveness. Lancet Oncol 2014; 15: e493-503.
    [6] Elinav E, Nowarski R, Thaiss CA, Hu B, Jin C and Flavell RA. Inflammation-induced cancer: crosstalk between tumours, immune cells and microorganisms. Nat Rev Cancer 2013; 13: 759-771.
    [7] Ma Y, Ren Y, Dai ZJ, Wu CJ, Ji YH and Xu J. IL-6, IL-8 and TNF-α levels correlate with disease stage in breast cancer patients. Adv Clin Exp Med 2017; 26: 421-426.
    [8] Coşkun Ö, Öztopuz Ö and Özkan Ö F. Determination of IL-6, TNF-α and VEGF levels in the serums of patients with colorectal cancer. Cell Mol Biol (Noisy-le-grand) 2017; 63: 97-101.
    [9] De Simone V, Franzè E, Ronchetti G, Colantoni A, Fantini MC, Di Fusco D, Sica GS, Sileri P, MacDonald TT, Pallone F, Monteleone G and Stolfi C. Th17-type cytokines, IL-6 and TNF-α synergistically activate STAT3 and NF-kB to promote colorectal cancer cell growth. Oncogene 2015; 34: 3493-3503.
    [10] Jourdan M, Tarte K, Legouffe E, Brochier J, Rossi JF and Klein B. Tumor necrosis factor is a survival and proliferation factor for human myeloma cells. Eur Cytokine Netw 1999; 10: 65-70.
    [11] Hagi T, Nakamura T, Iino T, Matsubara T, Asanuma K, Matsumine A and Sudo A. The diagnostic and prognostic value of interleukin-6 in patients with soft tissue sarcomas. Sci Rep 2017; 7: 9640.
    [12] Zhao G, Wang Z, Huang Y, Ye L, Yang K, Huang Q, Chen X, Li G, Chen Y, Wang J and Zhou Y. Effects of Benzoapyrene on migration and invasion of lung cancer cells functioning by TNF-α. J Cell Biochem 2018; 119: 6492-6500.
    [13] Gupta M, Babic A, Beck AH and Terry K. TNF-α expression, risk factors, and inflammatory exposures in ovarian cancer: evidence for an inflammatory pathway of ovarian carcinogenesis? Hum Pathol 2016; 54: 82-91.
    [14] Wu Y and Zhou BP. TNF-alpha/NF-kappaB/Snail pathway in cancer cell migration and invasion. Br J Cancer 2010; 102: 639-644.
    [15] Xia L, Tan S, Zhou Y, Lin J, Wang H, Oyang L, Tian Y, Liu L, Su M, Wang H, Cao D and Liao Q. Role of the NFκB-signaling pathway in cancer. Onco Targets Ther 2018; 11: 2063-2073.
    [16] Taniguchi K and Karin M. NF-κB, inflammation, immunity and cancer: coming of age. Nat Rev Immunol 2018; 18: 309-324.
    [17] Chonov DC, Ignatova MMK, Ananiev JR and Gulubova MV. IL-6 Activities in the Tumour Microenvironment. Part 1. Open Access Maced J Med Sci 2019; 7: 2391-2398.
    [18] Tang X, Ding Q, Chen C, Chen F, Zhou X, Hong CJ and Pan W. Micheliolide inhibits gastric cancer growth in vitro and in vivo via blockade of the IL-6/STAT3 pathway. Pharmazie 2019; 74: 175-178.
    [19] Kampan NC, Xiang SD, McNally OM, Stephens AN, Quinn MA and Plebanski M. Immunotherapeutic Interleukin-6 or Interleukin-6 Receptor Blockade in Cancer: Challenges and Opportunities. Curr Med Chem 2018; 25: 4785-4806.
    [20] Josephs SF, Ichim TE, Prince SM, Kesari S, Marincola FM, Escobedo AR and Jafri A. Unleashing endogenous TNF-alpha as a cancer immunotherapeutic. J Transl Med 2018; 16: 242.
    [21] Garbers C, Heink S, Korn T and Rose-John S. Interleukin-6: designing specific therapeutics for a complex cytokine. Nat Rev Drug Discov 2018; 17: 395-412.
    [22] Wang LH, Wu CF, Rajasekaran N and Shin YK. Loss of Tumor Suppressor Gene Function in Human Cancer: An Overview. Cell Physiol Biochem 2018; 51: 2647-2693.
    [23] Jia P and Zhao Z. Characterization of Tumor-Suppressor Gene Inactivation Events in 33 Cancer Types. Cell Rep 2019; 26: 496-506.e493.
    [24] Pavlova NN and Thompson CB. The Emerging Hallmarks of Cancer Metabolism. Cell Metab 2016; 23: 27-47.
    [25] DeBerardinis RJ and Chandel NS. Fundamentals of cancer metabolism. Sci Adv 2016; 2: e1600200.
    [26] Akram M. Mini-review on glycolysis and cancer. J Cancer Educ 2013; 28: 454-457.
    [27] Ganapathy-Kanniappan S and Geschwind JF. Tumor glycolysis as a target for cancer therapy: progress and prospects. Mol Cancer 2013; 12: 152.
    [28] Vaupel P, Schmidberger H and Mayer A. The Warburg effect: essential part of metabolic reprogramming and central contributor to cancer progression. Int J Radiat Biol 2019; 95: 912-919.
    [29] Schwartz L, Supuran CT and Alfarouk KO. The Warburg Effect and the Hallmarks of Cancer. Anticancer Agents Med Chem 2017; 17: 164-170.
    [30] Baig MH, Adil M, Khan R, Dhadi S, Ahmad K, Rabbani G, Bashir T, Imran MA, Husain FM, Lee EJ, Kamal MA and Choi I. Enzyme targeting strategies for prevention and treatment of cancer: Implications for cancer therapy. Semin Cancer Biol 2019; 56: 1-11.
    [31] Xintaropoulou C, Ward C, Wise A, Queckborner S, Turnbull A, Michie CO, Williams ARW, Rye T, Gourley C and Langdon SP. Expression of glycolytic enzymes in ovarian cancers and evaluation of the glycolytic pathway as a strategy for ovarian cancer treatment. BMC Cancer 2018; 18: 636.
    [32] Sun RF, Zhao CY, Chen S, Yu W, Zhou MM and Gao CR. Androgen Receptor Stimulates Hexokinase 2 and Induces Glycolysis by PKA/CREB Signaling in Hepatocellular Carcinoma. Dig Dis Sci 2020;
    [33] Zhang J, Chen G, Gao Y and Liang H. HOTAIR/miR-125 axis-mediated Hexokinase 2 expression promotes chemoresistance in human glioblastoma. J Cell Mol Med 2020; 24: 5707-5717.
    [34] Li WC, Huang CH, Hsieh YT, Chen TY, Cheng LH, Chen CY, Liu CJ, Chen HM, Huang CL, Lo JF and Chang KW. Regulatory Role of Hexokinase 2 in Modulating Head and Neck Tumorigenesis. Front Oncol 2020; 10: 176.
    [35] Gallo M, Sapio L, Spina A, Naviglio D, Calogero A and Naviglio S. Lactic dehydrogenase and cancer: an overview. Front Biosci (Landmark Ed) 2015; 20: 1234-1249.
    [36] Zhang X, Guo M, Fan J, Lv Z, Huang Q, Han J, Wu F, Hu G, Xu J and Jin Y. Prognostic significance of serum LDH in small cell lung cancer: A systematic review with meta-analysis. Cancer Biomark 2016; 16: 415-423.
    [37] Serganova I, Cohen IJ, Vemuri K, Shindo M, Maeda M, Mane M, Moroz E, Khanin R, Satagopan J, Koutcher JA and Blasberg R. LDH-A regulates the tumor microenvironment via HIF-signaling and modulates the immune response. PLoS One 2018; 13: e0203965.
    [38] Dong T, Liu Z, Xuan Q, Wang Z, Ma W and Zhang Q. Tumor LDH-A expression and serum LDH status are two metabolic predictors for triple negative breast cancer brain metastasis. Sci Rep 2017; 7: 6069.
    [39] Jiang F, Ma S, Xue Y, Hou J and Zhang Y. LDH-A promotes malignant progression via activation of epithelial-to-mesenchymal transition and conferring stemness in muscle-invasive bladder cancer. Biochem Biophys Res Commun 2016; 469: 985-992.
    [40] Han ZJ, Feng YH, Gu BH, Li YM and Chen H. The post-translational modification, SUMOylation, and cancer (Review). Int J Oncol 2018; 52: 1081-1094.
    [41] Pal I, Safari M, Jovanovic M, Bates SE and Deng C. Targeting Translation of mRNA as a Therapeutic Strategy in Cancer. Curr Hematol Malig Rep 2019; 14: 219-227.
    [42] Kim SY. Cancer Energy Metabolism: Shutting Power off Cancer Factory. Biomol Ther (Seoul) 2018; 26: 39-44.
    [43] Dell' Antone P. Energy metabolism in cancer cells: how to explain the Warburg and Crabtree effects? Med Hypotheses 2012; 79: 388-392.
    [44] Santos M, Fidalgo A, Varanda AS, Oliveira C and Santos MAS. tRNA Deregulation and Its Consequences in Cancer. Trends Mol Med 2019; 25: 853-865.
    [45] Hyeon DY, Kim JH, Ahn TJ, Cho Y, Hwang D and Kim S. Evolution of the multi-tRNA synthetase complex and its role in cancer. J Biol Chem 2019; 294: 5340-5351.
    [46] Huang SQ, Sun B, Xiong ZP, Shu Y, Zhou HH, Zhang W, Xiong J and Li Q. The dysregulation of tRNAs and tRNA derivatives in cancer. J Exp Clin Cancer Res 2018; 37: 101.
    [47] Torres AG, Reina O, Stephan-Otto Attolini C and Ribas de Pouplana L. Differential expression of human tRNA genes drives the abundance of tRNA-derived fragments. Proc Natl Acad Sci U S A 2019; 116: 8451-8456.
    [48] Pan T. Modifications and functional genomics of human transfer RNA. Cell Res 2018; 28: 395-404.
    [49] Pavon-Eternod M, Gomes S, Geslain R, Dai Q, Rosner MR and Pan T. tRNA over-expression in breast cancer and functional consequences. Nucleic Acids Res 2009; 37: 7268-7280.
    [50] Goodarzi H, Nguyen HCB, Zhang S, Dill BD, Molina H and Tavazoie SF. Modulated Expression of Specific tRNAs Drives Gene Expression and Cancer Progression. Cell 2016; 165: 1416-1427.
    [51] Birch J, Clarke CJ, Campbell AD, Campbell K, Mitchell L, Liko D, Kalna G, Strathdee D, Sansom OJ, Neilson M, Blyth K and Norman JC. The initiator methionine tRNA drives cell migration and invasion leading to increased metastatic potential in melanoma. Biol Open 2016; 5: 1371-1379.
    [52] Karasik A, Fierke CA and Koutmos M. Interplay between substrate recognition, 5' end tRNA processing and methylation activity of human mitochondrial RNase P. Rna 2019; 25: 1646-1660.
    [53] Jarrous N. Roles of RNase P and Its Subunits. Trends Genet 2017; 33: 594-603.
    [54] Klemm BP, Wu N, Chen Y, Liu X, Kaitany KJ, Howard MJ and Fierke CA. The Diversity of Ribonuclease P: Protein and RNA Catalysts with Analogous Biological Functions. Biomolecules 2016; 6:
    [55] Brillante N, Gossringer M, Lindenhofer D, Toth U, Rossmanith W and Hartmann RK. Substrate recognition and cleavage-site selection by a single-subunit protein-only RNase P. Nucleic Acids Res 2016; 44: 2323-2336.
    [56] Mann H, Ben-Asouli Y, Schein A, Moussa S and Jarrous N. Eukaryotic RNase P: role of RNA and protein subunits of a primordial catalytic ribonucleoprotein in RNA-based catalysis. Mol Cell 2003; 12: 925-935.
    [57] Kikovska E, Svard SG and Kirsebom LA. Eukaryotic RNase P RNA mediates cleavage in the absence of protein. Proc Natl Acad Sci U S A 2007; 104: 2062-2067.
    [58] Liang ZX, Liu HS, Wang FW, Xiong L, Zhou C, Hu T, He XW, Wu XJ, Xie D, Wu XR and Lan P. LncRNA RPPH1 promotes colorectal cancer metastasis by interacting with TUBB3 and by promoting exosomes-mediated macrophage M2 polarization. Cell Death Dis 2019; 10: 829.
    [59] Zhang Y and Tang L. Inhibition of breast cancer cell proliferation and tumorigenesis by long non-coding RNA RPPH1 down-regulation of miR-122 expression. Cancer Cell Int 2017; 17: 109.
    [60] Cai Y, Sun Z, Jia H, Luo H, Ye X, Wu Q, Xiong Y, Zhang W and Wan J. Rpph1 Upregulates CDC42 Expression and Promotes Hippocampal Neuron Dendritic Spine Formation by Competing with miR-330-5p. Front Mol Neurosci 2017; 10: 27.
    [61] Xia T, Liao Q, Jiang X, Shao Y, Xiao B, Xi Y and Guo J. Long noncoding RNA associated-competing endogenous RNAs in gastric cancer. Sci Rep 2014; 4: 6088.
    [62] Moosavi B, Zhu XL, Yang WC and Yang GF. Genetic, epigenetic and biochemical regulation of succinate dehydrogenase function. Biol Chem 2020; 401: 319-330.
    [63] Moosavi B, Berry EA, Zhu XL, Yang WC and Yang GF. The assembly of succinate dehydrogenase: a key enzyme in bioenergetics. Cell Mol Life Sci 2019; 76: 4023-4042.
    [64] Huang S and Millar AH. Succinate dehydrogenase: the complex roles of a simple enzyme. Curr Opin Plant Biol 2013; 16: 344-349.
    [65] Jardim-Messeder D, Caverzan A, Rauber R, de Souza Ferreira E, Margis-Pinheiro M and Galina A. Succinate dehydrogenase (mitochondrial complex II) is a source of reactive oxygen species in plants and regulates development and stress responses. New Phytol 2015; 208: 776-789.
    [66] Rutter J, Winge DR and Schiffman JD. Succinate dehydrogenase - Assembly, regulation and role in human disease. Mitochondrion 2010; 10: 393-401.
    [67] Gill AJ. Succinate dehydrogenase (SDH)-deficient neoplasia. Histopathology 2018; 72: 106-116.
    [68] Tsai TH and Lee WY. Succinate Dehydrogenase-Deficient Renal Cell Carcinoma. Arch Pathol Lab Med 2019; 143: 643-647.
    [69] Settas N, Faucz FR and Stratakis CA. Succinate dehydrogenase (SDH) deficiency, Carney triad and the epigenome. Mol Cell Endocrinol 2018; 469: 107-111.
    [70] Nazar E, Khatami F, Saffar H and Tavangar SM. The Emerging Role of Succinate Dehyrogenase Genes (SDHx) in Tumorigenesis. Int J Hematol Oncol Stem Cell Res 2019; 13: 72-82.
    [71] Zhao T, Mu X and You Q. Succinate: An initiator in tumorigenesis and progression. Oncotarget 2017; 8: 53819-53828.
    [72] Tretter L, Patocs A and Chinopoulos C. Succinate, an intermediate in metabolism, signal transduction, ROS, hypoxia, and tumorigenesis. Biochim Biophys Acta 2016; 1857: 1086-1101.
    [73] Tannahill GM, Curtis AM, Adamik J, Palsson-McDermott EM, McGettrick AF, Goel G, Frezza C, Bernard NJ, Kelly B, Foley NH, Zheng L, Gardet A, Tong Z, Jany SS, Corr SC, Haneklaus M, Caffrey BE, Pierce K, Walmsley S, Beasley FC, Cummins E, Nizet V, Whyte M, Taylor CT, Lin H, Masters SL, Gottlieb E, Kelly VP, Clish C, Auron PE, Xavier RJ and O'Neill LA. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature 2013; 496: 238-242.
    [74] Dalla Pozza E, Dando I, Pacchiana R, Liboi E, Scupoli MT, Donadelli M and Palmieri M. Regulation of succinate dehydrogenase and role of succinate in cancer. Semin Cell Dev Biol 2020; 98: 4-14.
    [75] King A, Selak MA and Gottlieb E. Succinate dehydrogenase and fumarate hydratase: linking mitochondrial dysfunction and cancer. Oncogene 2006; 25: 4675-4682.
    [76] Huang Y, Wang LA, Xie Q, Pang J, Wang L, Yi Y, Zhang J, Zhang Y, Chen R, Lan W, Zhang D and Jiang J. Germline SDHB and SDHD mutations in pheochromocytoma and paraganglioma patients. Endocr Connect 2018; 7: 1217-1225.
    [77] Elston MS, Sehgal S, Dray M, Phillips E, Conaglen JV, Clifton-Bligh RJ and Gill AJ. A Duodenal SDH-Deficient Gastrointestinal Stromal Tumor in a Patient With a Germline SDHB Mutation. J Clin Endocrinol Metab 2017; 102: 1447-1450.
    [78] Assadipour Y, Sadowski SM, Alimchandani M, Quezado M, Steinberg SM, Nilubol N, Patel D, Prodanov T, Pacak K and Kebebew E. SDHB mutation status and tumor size but not tumor grade are important predictors of clinical outcome in pheochromocytoma and abdominal paraganglioma. Surgery 2017; 161: 230-239.
    [79] Gimenez-Roqueplo AP, Favier J, Rustin P, Rieubland C, Crespin M, Nau V, Khau Van Kien P, Corvol P, Plouin PF and Jeunemaitre X. Mutations in the SDHB gene are associated with extra-adrenal and/or malignant phaeochromocytomas. Cancer Res 2003; 63: 5615-5621.
    [80] Tseng PL, Wu WH, Hu TH, Chen CW, Cheng HC, Li CF, Tsai WH, Tsai HJ, Hsieh MC, Chuang JH and Chang WT. Decreased succinate dehydrogenase B in human hepatocellular carcinoma accelerates tumor malignancy by inducing the Warburg effect. Sci Rep 2018; 8: 3081.
    [81] Xiao Z, Liu S, Ai F, Chen X, Li X, Liu R, Ren W, Zhang X, Shu P and Zhang D. SDHB downregulation facilitates the proliferation and invasion of colorectal cancer through AMPK functions excluding those involved in the modulation of aerobic glycolysis. Exp Ther Med 2018; 15: 864-872.
    [82] Cervera AM, Apostolova N, Crespo FL, Mata M and McCreath KJ. Cells silenced for SDHB expression display characteristic features of the tumor phenotype. Cancer Res 2008; 68: 4058-4067.
    [83] Burnichon N, Brière JJ, Libé R, Vescovo L, Rivière J, Tissier F, Jouanno E, Jeunemaitre X, Bénit P, Tzagoloff A, Rustin P, Bertherat J, Favier J and Gimenez-Roqueplo AP. SDHA is a tumor suppressor gene causing paraganglioma. Hum Mol Genet 2010; 19: 3011-3020.
    [84] Ong RKS, Flores SK, Reddick RL, Dahia PLM and Shawa H. A Unique Case of Metastatic, Functional, Hereditary Paraganglioma Associated With an SDHC Germline Mutation. J Clin Endocrinol Metab 2018; 103: 2802-2806.
    [85] Shulskaya MV, Shadrina MI, Bakilina NA, Zolotova SV and Slominsky PA. The spectrum of SDHD mutations in Russian patients with head and neck paraganglioma. Int J Neurosci 2018; 128: 1174-1179.
    [86] Bayley JP, Bausch B, Rijken JA, van Hulsteijn LT, Jansen JC, Ascher D, Pires DEV, Hes FJ, Hensen EF, Corssmit EPM, Devilee P and Neumann HPH. Variant type is associated with disease characteristics in SDHB, SDHC and SDHD-linked phaeochromocytoma-paraganglioma. J Med Genet 2020; 57: 96-103.
    [87] Ham IH, Oh HJ, Jin H, Bae CA, Jeon SM, Choi KS, Son SY, Han SU, Brekken RA, Lee D and Hur H. Targeting interleukin-6 as a strategy to overcome stroma-induced resistance to chemotherapy in gastric cancer. Mol Cancer 2019; 18: 68.
    [88] Masjedi A, Hashemi V, Hojjat-Farsangi M, Ghalamfarsa G, Azizi G, Yousefi M and Jadidi-Niaragh F. The significant role of interleukin-6 and its signaling pathway in the immunopathogenesis and treatment of breast cancer. Biomed Pharmacother 2018; 108: 1415-1424.
    [89] Cordier-Bussat M, Thibert C, Sujobert P, Genestier L, Fontaine E and Billaud M. [Even the Warburg effect can be oxidized: metabolic cooperation and tumor development]. Med Sci (Paris) 2018; 34: 701-708.
    [90] Schwartz L, Seyfried T, Alfarouk KO, Da Veiga Moreira J and Fais S. Out of Warburg effect: An effective cancer treatment targeting the tumor specific metabolism and dysregulated pH. Semin Cancer Biol 2017; 43: 134-138.
    [91] Tekade RK and Sun X. The Warburg effect and glucose-derived cancer theranostics. Drug Discov Today 2017; 22: 1637-1653.
    [92] Liberti MV and Locasale JW. The Warburg Effect: How Does it Benefit Cancer Cells? Trends Biochem Sci 2016; 41: 211-218.
    [93] Clarke CJ, Berg TJ, Birch J, Ennis D, Mitchell L, Cloix C, Campbell A, Sumpton D, Nixon C, Campbell K, Bridgeman VL, Vermeulen PB, Foo S, Kostaras E, Jones JL, Haywood L, Pulleine E, Yin H, Strathdee D, Sansom O, Blyth K, McNeish I, Zanivan S, Reynolds AR and Norman JC. The Initiator Methionine tRNA Drives Secretion of Type II Collagen from Stromal Fibroblasts to Promote Tumor Growth and Angiogenesis. Curr Biol 2016; 26: 755-765.
    [94] Pavon-Eternod M, Gomes S, Rosner MR and Pan T. Overexpression of initiator methionine tRNA leads to global reprogramming of tRNA expression and increased proliferation in human epithelial cells. Rna 2013; 19: 461-466.
    [95] Murata M. Inflammation and cancer. Environ Health Prev Med 2018; 23: 50.
    [96] Coussens LM and Werb Z. Inflammation and cancer. Nature 2002; 420: 860-867.
    [97] Zhang P, Sun Y, Peng R, Chen W, Fu X, Zhang L, Peng H and Zhang Z. Long non-coding RNA Rpph1 promotes inflammation and proliferation of mesangial cells in diabetic nephropathy via an interaction with Gal-3. Cell Death Dis 2019; 10: 526.
    [98] Kondratova M, Czerwinska U, Sompairac N, Amigorena SD, Soumelis V, Barillot E, Zinovyev A and Kuperstein I. A multiscale signalling network map of innate immune response in cancer reveals cell heterogeneity signatures. Nat Commun 2019; 10: 4808.
    [99] Chakraborty C, Sharma AR, Sharma G and Lee SS. The Interplay among miRNAs, Major Cytokines, and Cancer-Related Inflammation. Mol Ther Nucleic Acids 2020; 20: 606-620.
    [100] Du GL, Chen WY, Li XN, He R and Feng PF. Induction of MMP1 and 3 by cyclical mechanical stretch is mediated by IL6 in cultured fibroblasts of keratoconus. Mol Med Rep 2017; 15: 3885-3892.
    [101] Li Y, Samuvel DJ, Sundararaj KP, Lopes-Virella MF and Huang Y. IL-6 and high glucose synergistically upregulate MMP-1 expression by U937 mononuclear phagocytes via ERK1/2 and JNK pathways and c-Jun. J Cell Biochem 2010; 110: 248-259.
    [102] Sundararaj KP, Samuvel DJ, Li Y, Sanders JJ, Lopes-Virella MF and Huang Y. Interleukin-6 released from fibroblasts is essential for up-regulation of matrix metalloproteinase-1 expression by U937 macrophages in coculture: cross-talking between fibroblasts and U937 macrophages exposed to high glucose. J Biol Chem 2009; 284: 13714-13724.
    [103] Topacio BR, Zatulovskiy E, Cristea S, Xie S, Tambo CS, Rubin SM, Sage J, Koivomagi M and Skotheim JM. Cyclin D-Cdk4,6 Drives Cell-Cycle Progression via the Retinoblastoma Protein's C-Terminal Helix. Mol Cell 2019; 74: 758-770.e754.
    [104] Dong P, Zhang C, Parker BT, You L and Mathey-Prevot B. Cyclin D/CDK4/6 activity controls G1 length in mammalian cells. PLoS One 2018; 13: e0185637.
    [105] Qie S and Diehl JA. Cyclin D1, cancer progression, and opportunities in cancer treatment. J Mol Med (Berl) 2016; 94: 1313-1326.
    [106] Shan YS, Hsu HP, Lai MD, Hung YH, Wang CY, Yen MC and Chen YL. Cyclin D1 overexpression correlates with poor tumor differentiation and prognosis in gastric cancer. Oncol Lett 2017; 14: 4517-4526.
    [107] Luo J, Yan R, He X and He J. Constitutive activation of STAT3 and cyclin D1 overexpression contribute to proliferation, migration and invasion in gastric cancer cells. Am J Transl Res 2017; 9: 5671-5677.
    [108] Kurosaka M and Machida S. Interleukin-6-induced satellite cell proliferation is regulated by induction of the JAK2/STAT3 signalling pathway through cyclin D1 targeting. Cell Prolif 2013; 46: 365-373.
    [109] Won C, Lee CS, Lee JK, Kim TJ, Lee KH, Yang YM, Kim YN, Ye SK and Chung MH. CADPE suppresses cyclin D1 expression in hepatocellular carcinoma by blocking IL-6-induced STAT3 activation. Anticancer Res 2010; 30: 481-488.
    [110] Lin J, Xia L, Liang J, Han Y, Wang H, Oyang L, Tan S, Tian Y, Rao S, Chen X, Tang Y, Su M, Luo X, Wang Y, Wang H, Zhou Y and Liao Q. The roles of glucose metabolic reprogramming in chemo- and radio-resistance. J Exp Clin Cancer Res 2019; 38: 218.
    [111] Bose S and Le A. Glucose Metabolism in Cancer. Adv Exp Med Biol 2018; 1063: 3-12.
    [112] Jiang SH, Dong FY, Da LT, Yang XM, Wang XX, Weng JY, Feng L, Zhu LL, Zhang YL, Zhang ZG, Sun YW, Li J and Xu MJ. Ikarugamycin inhibits pancreatic cancer cell glycolysis by targeting hexokinase 2. Faseb j 2020; 34: 3943-3955.
    [113] Liu C, Wang X and Zhang Y. The Roles of HK2 on Tumorigenesis of Cervical Cancer. Technol Cancer Res Treat 2019; 18: 1533033819871306.
    [114] Chen X, Tian J, Su GH and Lin J. Blocking IL-6/GP130 Signaling Inhibits Cell Viability/Proliferation, Glycolysis, and Colony Forming Activity in Human Pancreatic Cancer Cells. Curr Cancer Drug Targets 2019; 19: 417-427.
    [115] Ando M, Uehara I, Kogure K, Asano Y, Nakajima W, Abe Y, Kawauchi K and Tanaka N. Interleukin 6 enhances glycolysis through expression of the glycolytic enzymes hexokinase 2 and 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3. J Nippon Med Sch 2010; 77: 97-105.
    [116] Kitazawa S, Ebara S, Ando A, Baba Y, Satomi Y, Soga T and Hara T. Succinate dehydrogenase B-deficient cancer cells are highly sensitive to bromodomain and extra-terminal inhibitors. Oncotarget 2017; 8: 28922-28938.
    [117] Xiao M, Yang H, Xu W, Ma S, Lin H, Zhu H, Liu L, Liu Y, Yang C, Xu Y, Zhao S, Ye D, Xiong Y and Guan KL. Inhibition of α-KG-dependent histone and DNA demethylases by fumarate and succinate that are accumulated in mutations of FH and SDH tumor suppressors. Genes Dev 2012; 26: 1326-1338.
    [118] Loriot C, Domingues M, Berger A, Menara M, Ruel M, Morin A, Castro-Vega LJ, Letouzé É, Martinelli C, Bemelmans AP, Larue L, Gimenez-Roqueplo AP and Favier J. Deciphering the molecular basis of invasiveness in Sdhb-deficient cells. Oncotarget 2015; 6: 32955-32965.

    下載圖示 校內:2025-07-21公開
    校外:2025-07-21公開
    QR CODE