[en] [en] BACKGROUND & AIMS: Rapid deconditioning, also called cachexia, and metabolic reprogramming are two hallmarks of pancreatic cancer. Acetyl-coenzyme A synthetase short-chain family member 2 (ACSS2) is an acetyl-enzyme A synthetase that contributes to lipid synthesis and epigenetic reprogramming. However, the role of ACSS2 on the nonselective macropinocytosis and cancer cachexia in pancreatic cancer remains elusive. In this study, we demonstrate that ACSS2 potentiates macropinocytosis and muscle wasting through metabolic reprogramming in pancreatic cancer.
METHODS: Clinical significance of ACSS2 was analyzed using samples from patients with pancreatic cancer. ACSS2-knockout cells were established using the clustered regularly interspaced short palindromic repeats-associated protein 9 system. Single-cell RNA sequencing data from genetically engineered mouse models was analyzed. The macropinocytotic index was evaluated by dextran uptake assay. Chromatin immunoprecipitation assay was performed to validate transcriptional activation. ACSS2-mediated tumor progression and muscle wasting were examined in orthotopic xenograft models.
RESULTS: Metabolic stress induced ACSS2 expression, which is associated with worse prognosis in pancreatic cancer. ACSS2 knockout significantly suppressed cell proliferation in 2-dimensional and 3-dimensional models. Macropinocytosis-associated genes are upregulated in tumor tissues and are correlated with worse prognosis. ACSS2 knockout inhibited macropinocytosis. We identified Zrt- and Irt-like protein 4 (ZIP4) as a downstream target of ACSS2, and knockdown of ZIP4 reversed ACSS2-induced macropinocytosis. ACSS2 upregulated ZIP4 through ETV4-mediated transcriptional activation. ZIP4 induces macropinocytosis through cyclic adenosine monophosphate response element-binding protein-activated syndecan 1 (SDC1) and dynamin 2 (DNM2). Meanwhile, ZIP4 drives muscle wasting and cachexia via glycogen synthase kinase-β (GSK3β)-mediated secretion of tumor necrosis factor superfamily member 10 (TRAIL or TNFSF10). ACSS2 knockout attenuated muscle wasting and extended survival in orthotopic mouse models.
CONCLUSIONS: ACSS2-mediated metabolic reprogramming activates the ZIP4 pathway, and promotes macropinocytosis via SDC1/DNM2 and drives muscle wasting through the GSK3β/TRAIL axis, which potentially provides additional nutrients for macropinocytosis in pancreatic cancer.
Disciplines :
Biochemistry, biophysics & molecular biology
Author, co-author :
Zhou, Zhijun; Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
Ren, Yu; Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
Yang, Jingxuan; Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
Liu, Mingyang; Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
Shi, Xiuhui ; Université de Liège - ULiège > Département de pharmacie ; Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
Luo, Wenyi; Department of Pathology, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
Fung, Kar-Ming; Department of Pathology, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
Xu, Chao; Department of Biostatistics and Epidemiology, Hudson College of Public Health, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
Bronze, Michael S; Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
Zhang, Yuqing; Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma. Electronic address: Yuqing-Zhang@ouhsc.edu
Houchen, Courtney W; Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma. Electronic address: courtney-houchen@ouhsc.edu
Li, Min ; Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma. Electronic address: Min-Li@ouhsc.edu
Language :
English
Title :
Acetyl-Coenzyme A Synthetase 2 Potentiates Macropinocytosis and Muscle Wasting Through Metabolic Reprogramming in Pancreatic Cancer.
William and Ella Owens Medical Research Foundation NIH - National Institutes of Health
Funding text :
Funding This work was supported in part by National Institutes of Health National Cancer Institute grants R01 CA186338 , R01 CA203108 , R01 CA247234, and award P30 CA225520 , and by the William and Ella Owens Medical Research Foundation (Min Li).
Baracos, V.E., Martin, L., Korc, M., et al. Cancer-associated cachexia. Nat Rev Dis Primers, 4, 2018, 17105.
Argiles, J.M., Stemmler, B., Lopez-Soriano, F.J., et al. Inter-tissue communication in cancer cachexia. Nat Rev Endocrinol 15 (2018), 9–20.
Huot, J.R., Pin, F., Narasimhan, A., et al. ACVR2B antagonism as a countermeasure to multi-organ perturbations in metastatic colorectal cancer cachexia. J Cachexia Sarcopenia Muscle 11 (2020), 1779–1798.
Roeland, E.J., Bohlke, K., Baracos, V.E., et al. Management of cancer cachexia: ASCO Guideline. J Clin Oncol 38 (2020), 2438–2453.
Wood, L.D., Canto, M.I., Jaffee, E.M., et al. Pancreatic cancer: pathogenesis, screening, diagnosis, and treatment. Gastroenterology 163 (2022), 386–402.e1.
Kamphorst, J.J., Nofal, M., Commisso, C., et al. Human pancreatic cancer tumors are nutrient poor and tumor cells actively scavenge extracellular protein. Cancer Res 75 (2015), 544–553.
Hosein, A.N., Brekken, R.A., Maitra, A., Pancreatic cancer stroma: an update on therapeutic targeting strategies. Nat Rev Gastroenterol Hepatol 17 (2020), 487–505.
Schug, Z.T., Peck, B., Jones, D.T., et al. Acetyl-CoA synthetase 2 promotes acetate utilization and maintains cancer cell growth under metabolic stress. Cancer Cell 27 (2015), 57–71.
Mashimo, T., Pichumani, K., Vemireddy, V., et al. Acetate is a bioenergetic substrate for human glioblastoma and brain metastases. Cell 159 (2014), 1603–1614.
Luong, A., Hannah, V.C., Brown, M.S., et al. Molecular characterization of human acetyl-CoA synthetase, an enzyme regulated by sterol regulatory element-binding proteins. J Biol Chem 275 (2000), 26458–26466.
Carrer, A., Trefely, S., Zhao, S., et al. Acetyl-CoA metabolism supports multistep pancreatic tumorigenesis. Cancer Discov 9 (2019), 416–435.
Slaymaker, I.M., Gao, L., Zetsche, B., et al. Rationally engineered Cas9 nucleases with improved specificity. Science 351 (2016), 84–88.
Joung, J., Konermann, S., Gootenberg, J.S., et al. Genome-scale CRISPR-Cas9 knockout and transcriptional activation screening. Nat Protoc 12 (2017), 828–863.
Liu, M., Zhang, Y., Yang, J., et al. Zinc-dependent regulation of ZEB1 and YAP1 coactivation promotes epithelial-mesenchymal transition plasticity and metastasis in pancreatic cancer. Gastroenterology 160 (2021), 1771–1783.e1.
Shi, X., Yang, J., Liu, M., et al. Circular RNA ANAPC7 inhibits tumor growth and muscle wasting via PHLPP2-AKT-TGF-β signaling axis in pancreatic cancer. Gastroenterology 162 (2022), 2004–2017.e2.
Zhou, Z., Xia, G., Xiang, Z., et al. A C-X-C chemokine receptor type 2-dominated cross-talk between tumor cells and macrophages drives gastric cancer metastasis. Clin Cancer Res 25 (2019), 3317–3328.
Commisso, C., Flinn, R.J., Bar-Sagi, D., Determining the macropinocytic index of cells through a quantitative image-based assay. Nat Protoc 9 (2014), 182–192.
Durbin, A.D., Wang, T., Wimalasena, V.K., et al. EP300 selectively controls the enhancer landscape of MYCN-amplified neuroblastoma. Cancer Discov 12 (2022), 730–751.
Mews, P., Donahue, G., Drake, A.M., et al. Acetyl-CoA synthetase regulates histone acetylation and hippocampal memory. Nature 546 (2017), 381–386.
Zhang, Y., Yang, J., Cui, X., et al. A novel epigenetic CREB-miR-373 axis mediates ZIP4-induced pancreatic cancer growth. EMBO Mol Med 5 (2013), 1322–1334.
Kim, C., Wilcox-Adelman, S., Sano, Y., et al. Antiinflammatory cAMP signaling and cell migration genes co-opted by the anthrax bacillus. Proc Natl Acad Sci U S A 105 (2008), 6150–6155.
Li, X., Yu, W., Qian, X., et al. Nucleus-translocated ACSS2 promotes gene transcription for lysosomal biogenesis and autophagy. Mol Cell 66 (2017), 684–697.e9.
Comerford, S.A., Huang, Z., Du, X., et al. Acetate dependence of tumors. Cell 159 (2014), 1591–1602.
Kondo, A., Yamamoto, S., Nakaki, R., et al. Extracellular acidic pH activates the sterol regulatory element-binding protein 2 to promote tumor progression. Cell Rep 18 (2017), 2228–2242.
Su, H., Yang, F., Fu, R., et al. Cancer cells escape autophagy inhibition via NRF2-induced macropinocytosis. Cancer Cell 39 (2021), 678–693.e11.
Lee, S.W., Zhang, Y., Jung, M., et al. EGFR-Pak signaling selectively regulates glutamine deprivation-induced macropinocytosis. Dev Cell 50 (2019), 381–392.e5.
Liu, M., Yang, J., Zhang, Y., et al. ZIP4 promotes pancreatic cancer progression by repressing ZO-1 and claudin-1 through a ZEB1-dependent transcriptional mechanism. Clin Cancer Res 24 (2018), 3186–3196.
Liu, M., Zhang, Y., Yang, J., et al. ZIP4 increases expression of transcription factor ZEB1 to promote integrin α3β1 signaling and inhibit expression of the gemcitabine transporter ENT1 in Pancreatic Cancer Cells. Gastroenterology 158 (2020), 679–692.e1.
Yang, J., Zhang, Z., Zhang, Y., et al. ZIP4 promotes muscle wasting and cachexia in mice with orthotopic pancreatic tumors by stimulating RAB27B-regulated release of extracellular vesicles from cancer cells. Gastroenterology 156 (2019), 722–734.e6.
Yao, W., Rose, J.L., Wang, W., et al. Syndecan 1 is a critical mediator of macropinocytosis in pancreatic cancer. Nature 568 (2019), 410–414.
Amyere, M., Payrastre, B., Krause, U., et al. Constitutive macropinocytosis in oncogene-transformed fibroblasts depends on sequential permanent activation of phosphoinositide 3-kinase and phospholipase C. Mol Biol Cell 11 (2000), 3453–3467.
Reis, C.R., Chen, P.H., Srinivasan, S., et al. Crosstalk between Akt/GSK3β signaling and dynamin-1 regulates clathrin-mediated endocytosis. EMBO J 34 (2015), 2132–2146.
Suriben, R., Chen, M., Higbee, J., et al. Antibody-mediated inhibition of GDF15-GFRAL activity reverses cancer cachexia in mice. Nat Med 26 (2020), 1264–1270.
Loumaye, A., de Barsy, M., Nachit, M., et al. Circulating activin A predicts survival in cancer patients. J Cachexia Sarcopenia Muscle 8 (2017), 768–777.
Johnston, A.J., Murphy, K.T., Jenkinson, L., et al. Targeting of Fn14 prevents cancer-induced cachexia and prolongs survival. Cell 162 (2015), 1365–1378.
Rupert, J.E., Narasimhan, A., Jengelley, D.H.A., et al. Tumor-derived IL-6 and trans-signaling among tumor, fat, and muscle mediate pancreatic cancer cachexia. J Exp Med, 218, 2021, e20190450.
Kandarian, S.C., Nosacka, R.L., Delitto, A.E., et al. Tumour-derived leukaemia inhibitory factor is a major driver of cancer cachexia and morbidity in C26 tumour-bearing mice. J Cachexia Sarcopenia Muscle 9 (2018), 1109–1120.
Freire, P.P., Fernandez, G.J., de Moraes, D., et al. The expression landscape of cachexia-inducing factors in human cancers. J Cachexia Sarcopenia Muscle 11 (2020), 947–961.
Michalopoulou, E., Auciello, F.R., Bulusu, V., et al. Macropinocytosis renders a subset of pancreatic tumor cells resistant to mTOR inhibition. Cell Rep 30 (2020), 2729–2742.e4.
Hosein, A.N., Huang, H., Wang, Z., et al. Cellular heterogeneity during mouse pancreatic ductal adenocarcinoma progression at single-cell resolution. JCI Insight, 5, 2019, e129212.
Hao, Y., Hao, S., Andersen-Nissen, E., et al. Integrated analysis of multimodal single-cell data. Cell 184 (2021), 3573–3587.e29.
Zhou, Z., Zhang, J., Xu, C., et al. An integrated model of N6-methyladenosine regulators to predict tumor aggressiveness and immune evasion in pancreatic cancer. EBioMedicine, 65, 2021, 103271.