AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (2.4 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Original Article | Open Access

Glycolytic potential enhanced by blockade of pyruvate influx into mitochondria sensitizes prostate cancer to detection and radiotherapy

Huan Xu1,2,*Junyi Chen3,*Zhi Cao1Xi Chen1,4Caihong Huang5Jin Ji1Yalong Xu1Junfeng Jiang6Yue Wang6Guowang Xu7Lina Zhou7Jingyi He8Xuedong Wei8Jason Boyang Wu9Zhong Wang2Shancheng Ren1 ( )Fubo Wang5 ( )
Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai 200433, China
Department of Urology, Shanghai Ninth People’s Hospital, Shanghai 200011, China
Department of Urology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
Department of Urology, No. 971 Hospital of the People’s Liberation Army Navy, Qingdao 266000, China
Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning 530021, China
Research Center of Developmental Biology, Second Military Medical University, Shanghai 200433, China
Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
Department of Urology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA

* These authors contributed equally to this article.

Show Author Information

Abstract

Objective

This study aimed to evaluate the effects of mitochondrial pyruvate carrier (MPC) blockade on the sensitivity of detection and radiotherapy of prostate cancer (PCa).

Methods

We investigated glycolysis reprogramming and MPC changes in patients with PCa by using metabolic profiling, RNA-Seq, and tissue microarrays. Transient blockade of pyruvate influx into mitochondria was observed in cellular studies to detect its different effects on prostate carcinoma cells and benign prostate cells. Xenograft mouse models were injected with an MPC inhibitor to evaluate the sensitivity of 18F-fluorodeoxyglucose positron emission tomography with computed tomography and radiotherapy of PCa. Furthermore, the molecular mechanism of this different effect of transient blockage towards benign prostate cells and prostate cancer cells was studied in vitro.

Results

MPC was elevated in PCa tissue compared with benign prostate tissue, but decreased during cancer progression. The transient blockade increased PCa cell proliferation while decreasing benign prostate cell proliferation, thus increasing the sensitivity of PCa cells to 18F-PET/CT (SUVavg, P = 0.016; SUVmax, P = 0.03) and radiotherapy (P < 0.01). This differential effect of MPC on PCa and benign prostate cells was dependent on regulation by a VDAC1-MPC-mitochondrial homeostasis-glycolysis pathway.

Conclusions

Blockade of pyruvate influx into mitochondria increased glycolysis levels in PCa but not in non-carcinoma prostate tissue. This transient blockage sensitized PCa to both detection and radiotherapy, thus indicating that glycolytic potential is a novel mechanism underlying PCa progression. The change in the mitochondrial pyruvate influx caused by transient MPC blockade provides a critical target for PCa diagnosis and treatment.

Electronic Supplementary Material

Download File(s)
cbm-19-9-1315_ESM.pdf (3.7 MB)

References

1

Hainaut P, Plymoth A. Cancer as a metabolic disease. Curr Opin Oncol. 2012; 24: 56-7.

2

Pavlova NN, Thompson CB. The emerging hallmarks of cancer metabolism. Cell Metab. 2016; 23: 27-47.

3

Warburg O, Wind F, Negelein E. The metabolism of tumors in the body. J Gen Physiol. 1927; 8: 519-30.

4

Martinez-Outschoorn UE, Peiris-Pages M, Pestell RG, Sotgia F, Lisanti MP. Cancer metabolism: a therapeutic perspective. Nat Rev Clin Oncol. 2017; 14: 11-31.

5

DeBerardinis RJ, Chandel NS. Fundamentals of cancer metabolism. Sci Adv. 2016; 2: e1600200.

6

Eidelman E, Twum-Ampofo J, Ansari J, Siddiqui MM. The metabolic phenotype of prostate cancer. Front Oncol. 2017; 7: 131.

7

Elia I, Schmieder R, Christen S, Fendt SM. Organ-specific cancer metabolism and its potential for therapy. Handb Exp Pharmacol. 2016; 233: 321-53.

8

Xu H, Chen J, He J, Ji J, Cao Z, Chen X, et al. Serum metabolic profiling identifies a biomarker panel for improvement of prostate cancer diagnosis. Front Oncol. 2021; 11: 666320.

9

Xu H, Chen Y, Gu M, Liu C, Chen Q, Zhan M, et al. Fatty acid metabolism reprogramming in advanced prostate cancer. Metabolites. 2021; 11: 765.

10

Herzig S, Raemy E, Montessuit S, Veuthey JL, Zamboni N, Westermann B, et al. Identification and functional expression of the mitochondrial pyruvate carrier. Science. 2012; 337: 93-6.

11

Bricker DK, Taylor EB, Schell JC, Orsak T, Boutron A, Chen YC, et al. A mitochondrial pyruvate carrier required for pyruvate uptake in yeast, drosophila, and humans. Science. 2012; 337: 96-100.

12

Bader DA, Hartig SM, Putluri V, Foley C, Hamilton MP, Smith EA, et al. Mitochondrial pyruvate import is a metabolic vulnerability in androgen receptor-driven prostate cancer. Nat Metab. 2019; 1: 70-85.

13

Corbet C, Bastien E, Draoui N, Doix B, Mignion L, Jordan BF, et al. Interruption of lactate uptake by inhibiting mitochondrial pyruvate transport unravels direct antitumor and radiosensitizing effects. Nat Commun. 2018; 9: 1208.

14

McCommis KS, Chen Z, Fu X, McDonald WG, Colca JR, Kletzien RF, et al. Loss of mitochondrial pyruvate carrier 2 in the liver leads to defects in gluconeogenesis and compensation via pyruvate-alanine cycling. Cell Metab. 2015; 22: 682-94.

15

Yang C, Ko B, Hensley CT, Jiang L, Wasti AT, Kim J, et al. Glutamine oxidation maintains the TCA cycle and cell survival during impaired mitochondrial pyruvate transport. Mol Cell. 2014; 56: 414-24.

16

Schell JC, Olson KA, Jiang L, Hawkins AJ, van Vranken JG, Xie J, et al. A role for the mitochondrial pyruvate carrier as a repressor of the warburg effect and colon cancer cell growth. Mol Cell. 2014; 56: 400-13.

17

Mishra P, Chan DC. Metabolic regulation of mitochondrial dynamics. J Cell Biol. 2016; 212: 379-87.

18

Li J, Ren S, Piao HL, Wang F, Yin P, Xu C, et al. Integration of lipidomics and transcriptomics unravels aberrant lipid metabolism and defines cholesteryl oleate as potential biomarker of prostate cancer. Sci Rep. 2016; 6: 20984.

19

Ren S, Shao Y, Zhao X, Hong CS, Wang F, Lu X, et al. Integration of metabolomics and transcriptomics reveals major metabolic pathways and potential biomarker involved in prostate cancer. Mol Cell Proteomics. 2016; 15: 154-63.

20

Ren S, Wei GH, Liu D, Wang L, Hou Y, Zhu S, et al. Whole-genome and transcriptome sequencing of prostate cancer identify new genetic alterations driving disease progression. Eur Urol. 2017; 73: 322-39.

21

Cao Z, Ji J, Zhang C, Wang F, Xu H, Yu Y, et al. The preoperative neutrophil-to-lymphocyte ratio is not a marker of prostate cancer characteristics but is an independent predictor of biochemical recurrence in patients receiving radical prostatectomy. Cancer Med. 2019; 8: 1004-12.

22

Cookson MS, Aus G, Burnett AL, Canby-Hagino ED, D’Amico AV, Dmochowski RR, et al. Variation in the definition of biochemical recurrence in patients treated for localized prostate cancer: The American urological association prostate guidelines for localized prostate cancer update panel report and recommendations for a standard in the reporting of surgical outcomes. J Urol. 2007; 177: 540-5.

23

Cao K, Lei X, Liu H, Zhao H, Guo J, Chen Y, et al. Polydatin alleviated radiation-induced lung injury through activation of Sirt3 and inhibition of epithelial-mesenchymal transition. J Cell Mol Med. 2017; 21: 3264-76.

24

Rogatzki MJ, Ferguson BS, Goodwin ML, Gladden LB. Lactate is always the end product of glycolysis. Front Neurosci. 2015; 9: 22.

25

Shoshan-Barmatz V, Mizrachi D. VDAC1: from structure to cancer therapy. Front Oncol. 2012; 2: 164.

26

Bravo-Sagua R, Parra V, Lopez-Crisosto C, Diaz P, Quest AF, Lavandero S. Calcium transport and signaling in mitochondria. Compr Physiol. 2017; 7: 623-34.

27

Fulop L, Szanda G, Enyedi B, Varnai P, Spat A. The effect of OPA1 on mitochondrial CA2+ signaling. PLoS One. 2011; 6: e25199.

28

Weisthal S, Keinan N, Ben-Hail D, Arif T, Shoshan-Barmatz V. Ca(2+)-mediated regulation of VDAC1 expression levels is associated with cell death induction. Biochim Biophys Acta. 2014; 1843: 2270-81.

29

Pfeiffer T, Schuster S, Bonhoeffer S. Cooperation and competition in the evolution of ATP-producing pathways. Science. 2001; 292: 504-7.

30

Young CD, Anderson SM. Sugar and fat - that’s where it’s at: metabolic changes in tumors. Breast Cancer Res. 2008; 10: 202.

31

DeBerardinis RJ, Lum JJ, Hatzivassiliou G, Thompson CB. The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 2008; 7: 11-20.

32

Vander Heiden MG, Cantley LC, Thompson CB. Understanding the warburg effect: the metabolic requirements of cell proliferation. Science. 2009; 324: 1029-33.

33

Cui J, Quan M, Xie D, Gao Y, Guha S, Fallon MB, et al. A novel KDM5A/MPC-1 signaling pathway promotes pancreatic cancer progression via redirecting mitochondrial pyruvate metabolism. Oncogene. 2020; 39: 1140-51.

34

Papandreou I, Cairns RA, Fontana L, Lim AL, Denko NC. HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab. 2006; 3: 187-97.

35

Tang XP, Chen Q, Li Y, Wang Y, Zou HB, Fu WJ, et al. Mitochondrial pyruvate carrier 1 functions as a tumor suppressor and predicts the prognosis of human renal cell carcinoma. Lab Invest. 2019; 99: 191-9.

36

Hsu WK, Virk MS, Feeley BT, Stout DB, Chatziioannou AF, Lieberman JR. Characterization of osteolytic, osteoblastic, and mixed lesions in a prostate cancer mouse model using 18F-FDG and 18F-fluoride PET/CT. J Nucl Med. 2008; 49: 414-21.

37

Morris MJ, Akhurst T, Larson SM, Ditullio M, Chu E, Siedlecki K, et al. Fluorodeoxyglucose positron emission tomography as an outcome measure for castrate metastatic prostate cancer treated with antimicrotubule chemotherapy. Clin Cancer Res. 2005; 11: 3210-6.

38

Hara T, Bansal A, DeGrado TR. Effect of hypoxia on the uptake of [methyl-3H]choline, [1-14C] acetate and [18F]FDG in cultured prostate cancer cells. Nucl Med Biol. 2006; 33: 977-84.

39

Schlaepfer IR, Glode LM, Hitz CA, Pac CT, Boyle KE, Maroni P, et al. Inhibition of lipid oxidation increases glucose metabolism and enhances 2-Deoxy-2-[(18)F]Fluoro-D-Glucose uptake in prostate cancer mouse Xenografts. Mol Imaging Biol. 2015; 17: 529-38.

40

Rauckhorst AJ, Gray LR, Sheldon RD, Fu X, Pewa AD, Feddersen CR, et al. The mitochondrial pyruvate carrier mediates high fat diet-induced increases in hepatic TCA cycle capacity. Mol Metab. 2017; 6: 1468-79.

41

Xu H, Li S, Sun Y, Xu L, Hong X, Wang Z, et al. ELOVL5-mediated long chain fatty acid elongation contributes to enzalutamide resistance of prostate cancer. Cancers. 2021; 13: 3957.

42

Vasileiou PVS, Evangelou K, Vlasis K, Fildisis G, Panayiotidis MI, Chronopoulos E, et al. Mitochondrial homeostasis and cellular senescence. Cells. 2019; 8: 686.

43

Magri A, Di Rosa MC, Orlandi I, Guarino F, Reina S, Guarnaccia M, et al. Deletion of voltage-dependent anion channel 1 knocks mitochondria down triggering metabolic rewiring in yeast. Cell Mol Life Sci. 2020; 77: 3195-213.

44

Mazure NM. VDAC in cancer. Biochim Biophys Acta Bioenerg. 2017; 1858: 665-73.

45

Pedersen PL. Voltage dependent anion channels (VDACS): a brief introduction with a focus on the outer mitochondrial compartment’s roles together with hexokinase-2 in the “Warburg effect” in cancer. J Bioenerg Biomembr. 2008; 40: 123-6.

46

Shoshan-Barmatz V, Israelson A, Brdiczka D, Sheu SS. The voltage-dependent anion channel (VDAC): function in intracellular signalling, cell life and cell death. Curr Pharm Des. 2006; 12: 2249-70.

47

Shoshan-Barmatz V, De Pinto V, Zweckstetter M, Raviv Z, Keinan N, Arbel N. VDAC, a multi-functional mitochondrial protein regulating cell life and death. Mol Aspects Med. 2010; 31: 227-85.

48

Song Z, Chen H, Fiket M, Alexander C, Chan DC. OPA1 processing controls mitochondrial fusion and is regulated by mRNA splicing, membrane potential, and Yme1L. J Cell Biol. 2007; 178: 749-55.

49

Baricault L, Segui B, Guegand L, Olichon A, Valette A, Larminat F, et al. OPA1 cleavage depends on decreased mitochondrial ATP level and bivalent metals. Exp Cell Res. 2007; 313: 3800-8.

50

Cipolat S, Rudka T, Hartmann D, Costa V, Serneels L, Craessaerts K, et al. Mitochondrial rhomboid PARL regulates cytochrome c release during apoptosis via OPA1-dependent cristae remodeling. Cell. 2006; 126: 163-75.

51

Arnoult D, Grodet A, Lee YJ, Estaquier J, Blackstone C. Release of OPA1 during apoptosis participates in the rapid and complete release of cytochrome c and subsequent mitochondrial fragmentation. J Biol Chem. 2005; 280: 35742-50.

Cancer Biology & Medicine
Pages 1315-1333
Cite this article:
Xu H, Chen J, Cao Z, et al. Glycolytic potential enhanced by blockade of pyruvate influx into mitochondria sensitizes prostate cancer to detection and radiotherapy. Cancer Biology & Medicine, 2022, 19(9): 1315-1333. https://doi.org/10.20892/j.issn.2095-3941.2021.0638

147

Views

1

Downloads

1

Crossref

1

Web of Science

1

Scopus

Altmetrics

Received: 29 November 2021
Accepted: 02 March 2022
Published: 22 September 2022
©2022 Cancer Biology & Medicine.

Creative Commons Attribution-NonCommercial 4.0 International License

Return