Cyclin-dependent kinase 12 deficiency reprogrammes cellular metabolism to alleviate ferroptosis potential and promote the progression of castration-resistant prostate cancer

Haozhe Zhang , Yi Zhou , Yating Feng , Wenli Hou , Yafei Chen , Zengzhen Xing , Yifan Zhang , Qiang Wei , Yu Yin , Ju Guo , Hailiang Hu

Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (5) : e1678

PDF
Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (5) : e1678 DOI: 10.1002/ctm2.1678
RESEARCH ARTICLE

Cyclin-dependent kinase 12 deficiency reprogrammes cellular metabolism to alleviate ferroptosis potential and promote the progression of castration-resistant prostate cancer

Author information +
History +
PDF

Abstract

• CDK12 deficiency promotes castration-resistant prostate cancer (CRPC) progression by reprogramming cellular metabolism.

• CDK12 deficiency in CRPC leads to a more active mitochondrial electron transport chain (ETC), ensuring efficient cell energy supply.

• CDK12 phosphorylates RNA Pol II to ensure the transcription of ACSL4 to regulate ferroptosis.

• Mitochondrial ETC inhibitors exhibit better selectivity for CDK12-deficient CRPC cells, offering a promising new therapeutic approach for this subtype of CRPC patients.

Keywords

CDK12 / CRPC / ferroptosis / metabolism

Cite this article

Download citation ▾
Haozhe Zhang, Yi Zhou, Yating Feng, Wenli Hou, Yafei Chen, Zengzhen Xing, Yifan Zhang, Qiang Wei, Yu Yin, Ju Guo, Hailiang Hu. Cyclin-dependent kinase 12 deficiency reprogrammes cellular metabolism to alleviate ferroptosis potential and promote the progression of castration-resistant prostate cancer. Clinical and Translational Medicine, 2024, 14(5): e1678 DOI:10.1002/ctm2.1678

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

FinleyLWS. What is cancer metabolism?Cell. 2023;186:1670-1688.

[2]

Martínez-ReyesI, Chandel NS. Cancer metabolism: looking forward. Nat Rev Cancer. 2021;21:669-680.

[3]

PavlovaNN, ZhuJ, ThompsonCB. The hallmarks of cancer metabolism: still emerging. Cell Metab. 2022;34:355-377.

[4]

Vander HeidenMG, Cantley LC, ThompsonCB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324:1029-1033.

[5]

ZongW-X, Rabinowitz JD, WhiteE. Mitochondria and cancer. Mol Cell. 2016;61:667-676.

[6]

PicardM, Shirihai OS. Mitochondrial signal transduction. Cell Metab. 2022;34:1620-1653.

[7]

PorporatoPE, Filigheddu N, PedroJMB-S, KroemerG, Galluzzi L. Mitochondrial metabolism and cancer. Cell Res. 2018;28:265-280.

[8]

ZorovDB, Juhaszova M, SollottSJ. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol Rev. 2014;94:909-950.

[9]

BasitF, van Oppen LM, SchöckelL, et al. Mitochondrial complex I inhibition triggers a mitophagy-dependent ROS increase, leading to necroptosis and ferroptosis in melanoma cells. Cell Death Dis. 2017;8:e271610.

[10]

BockFJ, TaitSWG. Mitochondria as multifaceted regulators of cell death. Nat Rev Mol Cell Biol. 2020;21:85-100.

[11]

JiangX, Stockwell BR, ConradM. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 2021;22:266-282.

[12]

BartkowiakB, LiuP, PhatnaniHP, et al. CDK12 is a transcription elongation associated with CTD kinase, the metazoan ortholog of yeast Ctk1. Gene Dev. 2010;24:2303-2316.

[13]

KoTK, KellyE, PinesJ. CrkRS: a novel conserved Cdc2-related protein kinase that colocalizes with SC35 speckles. J Cell Sci. 2001;114:2591-2603.

[14]

ChenH-H, WangY-C, FannM-J. Identification and characterization of the CDK12/cyclin L1 complex involved in alternative splicing regulation. Mol Cell Biol. 2006;26:2736-2745.

[15]

BlazekD, Kohoutek J, BartholomeeusenK, et al. The Cyclin K/Cdk12 complex maintains genomic stability by regulating the expression of DNA damage response genes. Gene Dev. 2011;25:2158-2172.

[16]

DubburySJ, BoutzPL, SharpPA. CDK12 regulates DNA repair genes by suppressing intronic polyadenylation. Nature. 2018;564:141.

[17]

PanL, XieW, LiK-L, et al. Heterochromatin remodeling by CDK12 contributes to learning. Proc Natl Acad Sci USA. 2015;112:13988-13993.

[18]

DaiQ, LeiT, ZhaoC, et al. Cyclin K-containing kinase complexes maintain self-renewal in murine embryonic stem cells. J Biol Chem. 2012;287:25344-25352.

[19]

WuY-M, Cieślik M, LonigroRJ, et al. Inactivation of CDK12 delineates a distinct immunogenic class of advanced prostate cancer. Cell. 2018;173:1770-1782. e1714

[20]

QueredaV, BayleS, VenaF, et al. Therapeutic targeting of CDK12/CDK13 in triple-negative breast cancer. Cancer Cell. 2019;36:545.

[21]

DieterSM, SieglC, CodóPL, et al. Degradation of CCNK/CDK12 is a druggable vulnerability of colorectal cancer. Cell Rep. 2021;36:10939.

[22]

ChengL, ZhouS, ZhouS, et al. Dual inhibition of CDK12/CDK13 targets both tumor and immune cells in ovarian cancer. Cancer Res. 2022;82:3588-3602.

[23]

HoulesT, LavoieG, NourreddineS, et al. CDK12 is hyperactivated and a synthetic-lethal target in BRAF-mutated melanoma. Nat Commun. 2022;13:6457.

[24]

MateoJ, PortaN, BianchiniD, et al. Olaparib in patients with metastatic castration-resistant prostate cancer with DNA repair gene aberrations (TOPARP-B): a multicentre, open-label, randomized, phase 2 trial. Lancet Oncol. 2020;21:162-174.

[25]

AbidaW, Campbell D, PatnaikA, et al. Non-BRCA DNA damage repair gene alterations and response to the PARP inhibitor rucaparib in metastatic castration-resistant prostate cancer: analysis from the phase II TRITON2 study. Clin Cancer Res. 2020;26:2487-2496.

[26]

DongB, FanL, YangB, et al. Use of circulating tumor DNA for the clinical management of metastatic castration-resistant prostate cancer: a multicenter, real-world study. J Natl Compr Canc Netw. 2021;19:905.

[27]

KwonDH, ChouJ, YipSM, et al. Differential treatment outcomes in BRCA1/2-, ATM-, and CDK12-mutated metastatic castration-resistant prostate cancer. Cancer. 2021;127:1965-1973.

[28]

RescignoP, GurelB, PereiraR, et al. Characterizing CDK12-mutated prostate cancers. Clin Cancer Res. 2021;27:566-574.

[29]

NguyenB, MotaJM, NandakumarS, et al. Pan-cancer analysis of alterations identifies a subset of prostate cancers with distinct genomic and clinical characteristics. Eur Urol. 2020;78:671-679.

[30]

GaoM, YiJ, ZhuJ, et al. Role of mitochondria in ferroptosis. Mol Cell. 2019;73:354.

[31]

XuL, YinYu, LiY, et al. A glutaminase isoform switch drives therapeutic resistance and disease progression of prostate cancer. Proc Natl Acad Sci USA. 2021;118:e2012748118.

[32]

CulpMB, Soerjomataram I, EfstathiouJA, BrayF, JemalA. Recent global patterns in prostate cancer incidence and mortality rates. Eur Urol. 2020;77:38-52.

[33]

FanL, FeiX, ZhuY, et al. Comparative analysis of genomic alterations across castration sensitive and castration resistant prostate cancer via circulating tumor DNA sequencing. J Urol. 2021;205:461-469.

[34]

CaiM, SongX-Lu, LiX-An, et al. Current therapy and drug resistance in metastatic castration-resistant prostate cancer. Drug Resist Updat. 2023;68:10096.

[35]

DuY, GuoZ. Recent progress in ferroptosis: inducers and inhibitors. Cell Death Discov. 2022;8:50.

[36]

WuJ, Minikes AM, GaoM, et al. Intercellular interaction dictates cancer cell ferroptosis via NF2-YAP signaling. Nature. 2019;572:402.

[37]

WangM-E, ChenJ, LuY, et al. RB1-deficient prostate tumor growth and metastasis are vulnerable to ferroptosis induction via the E2F/ACSL4 axis. J Clin Invest. 2023;133:e166647.

[38]

VercellinoI, Sazanov LA. The assembly, regulation, and function of the mitochondrial respiratory chain. Nat Rev Mol Cell Biol. 2022;23:141-161.

[39]

StineZE, SchugZT, SalvinoJM, Dang CV. Targeting cancer metabolism in the era of precision oncology. Nat Rev Drug Discov. 2022;21:141-162.

[40]

MolinaJR, SunY, ProtopopovaM, et al. An inhibitor of oxidative phosphorylation exploits cancer vulnerability. Nat Med. 2018;24:1036.

[41]

YapTA, DaverN, MahendraM, et al. Complex I inhibitor of oxidative phosphorylation in advanced solid tumors and acute myeloid leukemia: phase I trials. Nat Med. 2023;29(1):115-126.

[42]

FreiK, Schecher S, DaherT, et al. Inhibition of the Cyclin K-CDK12 complex induces DNA damage and increases the effect of androgen deprivation therapy in prostate cancer. Int J Cancer. 2024;154:1082-1096.

[43]

LeiH, WangZ, JiangD, et al. CRISPR screening identifies CDK12 as a conservative vulnerability of prostate cancer. Cell Death Dis. 2021;12:740.

[44]

LiuH, ShinSH, ChenH, et al. CDK12 and PAK2 as novel therapeutic targets for human gastric cancer. Theranostics. 2020;10:6201-6215.

[45]

IniguezAB, StolteB, WangEJ, et al. EWS/FLI confers tumor cell synthetic lethality to CDK12 inhibition in Ewing sarcoma. Cancer Cell. 2018;33:202-216. e206

[46]

SunR, WeiT, DingD, et al. CYCLIN K down-regulation induces androgen receptor gene intronic polyadenylation, variant expression, and PARP inhibitor vulnerability in castration-resistant prostate cancer. Proc Natl Acad Sci USA. 2022;119:e2205509119.

[47]

BajramiI, Frankum JR, KondeA, et al. Genome-wide profiling of genetic synthetic lethality identifies CDK12 as a novel determinant of PARP1/2 inhibitor sensitivity. Cancer Res. 2014;74:287-297.

[48]

LordCJ, Ashworth A. PARP inhibitors: synthetic lethality in the clinic. Science. 2017;355:1152-1158.

[49]

CarreiraS, PortaN, Arce-GallegoS, et al. Biomarkers associating with PARP inhibitor benefit in prostate cancer in the TOPARP-B trial. Cancer Discov. 2021;11:2812-2827.

[50]

HussainM, MateoJ, FizaziK, et al. Survival with olaparib in metastatic castration-resistant prostate cancer. N Engl J Med. 2020;383:2345-2357.

[51]

AntonarakisES. Cyclin-dependent kinase 12, immunity, and prostate cancer. N Engl J Med. 2018;379:1087-1089.

[52]

RebelloRJ, OingC, KnudsenKE, et al. Prostate cancer. Nat Rev Dis Primers. 2021;7:9.

[53]

FilipponeMG, GaglioD, BonfantiR, et al. CDK12 promotes tumorigenesis but induces vulnerability to therapies inhibiting folate one-carbon metabolism in breast cancer. Nat Commun. 2022;13:264210.

[54]

FarberS, Diamond LK, MercerRD, SylvesterRF, WolffJA. Temporary remissions in acute leukemia in children produced by folic acid antagonist, 4-aminopteroyl-glutamic acid (aminopterin). N Engl J Med. 1948;238:787-793.

[55]

MonteDAD. The environment and Parkinson's disease: is the nigrostriatal system preferentially targeted by neurotoxins?Lancet Neurol. 2003;2:531-538.

[56]

RohlenaJ, DongL-F, RalphSJ, Neuzil J. Anticancer drugs targeting the mitochondrial electron transport chain. Antioxid Redox Signal. 2011;15:2951-2974.

[57]

Sainero-AlcoladoL, Liaño-Pons J, Ruiz-PérezMV, Arsenian-HenrikssonM. Targeting mitochondrial metabolism for precision medicine in cancer. Cell Death Differ. 2022;29:1304-1317.

RIGHTS & PERMISSIONS

2024 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

144

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/