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 (1.6 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

Akt isoforms differentially provide for chemoresistance in prostate cancer

Bo Ma1,2,3 ( )Hanshuang Shao2,3Xia Jiang4Zhou Wang2,5,6,7Chuanyue (Cary) Wu2Diana Whaley2,3Alan Wells2,3,7 ( )
Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA
Pittsburgh VA Healthcare System, Pittsburgh, PA 15213, USA
Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15261, USA
Department of Urology, University of Pittsburgh, Pittsburgh, PA 15261, USA
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
Show Author Information

Abstract

Objective

Early prostate cancer micrometastatic foci undergo a mesenchymal to epithelial reverting transition, not only aiding seeding and colonization, but also rendering the tumor cells generally chemoresistant. We previously found that upregulated E-cadherin in the epithelial micrometastases activated canonical survival pathways, including PI3K-Akt, that protected the tumor cells from death; however, the extent of protection from blocking the pathway in its entirety was modest, because different isoforms may have alternately affected cell functioning. Here, we characterized Akt isoform expressions in primary and metastatic prostate cancers, as well as their individual contributions to chemoresistance.

Methods

Akt isoforms and E-cadherin were manipulated with drugs, knocked down, and over expressed. Tumor cell killing was determined in vitro and in vivo. Overall survival was calculated from patient records and specimens.

Results

Pan-Akt inhibition sensitized tumor cells to chemotherapy, and specific blockade of Akt1 or/and Akt2 caused cells to be more chemoresponsive. Overexpression of Akt3 induced apoptosis. A low dose of Akt1 or Akt2 inhibitor enabled standard chemotherapies to significantly eradicate metastatic prostate tumors in a mouse model, acting as chemosensitizers. In human specimens, we found Akt1 and Akt2 positively correlated, whereas Akt3 inversely correlated, with the overall survival of prostate cancer patients. Akt1high/Akt2high/Akt3low tumors had the worst outcomes.

Conclusions

E-cadherin-induced activation of Akt1/2 isoforms was the essential mechanism of chemoresistance, whereas Akt3 made cells more fragile. These findings emphasized the need to target Akt1/2, rather than pan-Akt, as a rational therapeutic approach.

Electronic Supplementary Material

Download File(s)
cbm-19-5-635-ESM.pdf (736.5 KB)

References

1

Seyfried TN, Huysentruyt LC. On the origin of cancer metastasis. Crit Rev Oncog. 2013; 18: 43-73.

2

Wells A, Grahovac J, Wheeler S, Ma B, Lauffenburger D. Targeting tumor cell motility as a strategy against invasion and metastasis. Trends Pharmacol Sci. 2013; 34: 283-9.

3

Meads MB, Gatenby RA, Dalton WS. Environment-mediated drug resistance: a major contributor to minimal residual disease. Nat Rev Cancer. 2009; 9: 665-74.

4

Singh A, Settleman J. Emt, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene. 2010; 29: 4741-51.

5

Wilson TR, Fridlyand J, Yan Y, Penuel E, Burton L, Chan E, et al. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature. 2012; 487: 505-9.

6

Ma B, Wells A, Clark AM. The pan-therapeutic resistance of disseminated tumor cells: role of phenotypic plasticity and the metastatic microenvironment. Semin Cancer Biol. 2020; 60: 138-47.

7

Mahon KL, Henshall SM, Sutherland RL, Horvath LG. Pathways of chemotherapy resistance in castration-resistant prostate cancer. Endocr Relat Cancer. 2011; 18: R103-23.

8

Shah NL, Sanda M. Health-related quality of life in treatment for prostate cancer: looking beyond survival. Support Cancer Ther. 2004; 1: 230-6.

9

Di Lorenzo G, Buonerba C, Autorino R, De Placido S, Sternberg CN. Castration-resistant prostate cancer: current and emerging treatment strategies. Drugs. 2010; 70: 983-1000.

10

Hotte SJ, Saad F. Current management of castrate-resistant prostate cancer. Curr Oncol. 2010; 17(Suppl 2): S72-9.

11

Gunasinghe NPAD, Wells A, Thompson EW, Hugo HJ. Mesenchymal-epithelial transition (met) as a mechanism for metastatic colonisation in breast cancer. Cancer Metast Rev. 2012; 31: 469-78.

12

Hugo H, Ackland ML, Blick T, Lawrence MG, Clements JA, Williams ED, et al. Epithelial--mesenchymal and mesenchymal--epithelial transitions in carcinoma progression. J Cell Physiol. 2007; 213: 374-83.

13

Tam WL, Weinberg RA. The epigenetics of epithelial-mesenchymal plasticity in cancer. Nat Med. 2013; 19: 1438-49.

14

Wells A, Chao YL, Grahovac J, Wu Q, Lauffenburger DA. Epithelial and mesenchymal phenotypic switchings modulate cell motility in metastasis. Front Biosci (Landmark Ed). 2011; 16: 815-37.

15

Chao Y, Wu Q, Shepard C, Wells A. Hepatocyte induced re-expression of e-cadherin in breast and prostate cancer cells increases chemoresistance. Clin Exp Metastas. 2012; 29: 39-50.

16

Ma B, Wells A. The mitogen-activated protein (MAP) kinases p38 and extracellular signal-regulated kinase (ERK) are involved in hepatocyte-mediated phenotypic switching in prostate cancer cells. J Biol Chem. 2014; 289: 11153-61.

17

Ma B, Khazali A, Shao H, Jiang Y, Wells A. Expression of E-cadherin and specific CXCR3 isoforms impact each other in prostate cancer. Cell Commun Signal. 2019; 17: 164.

18

Ma B, Wells A, Wei L, Zheng J. Prostate cancer liver metastasis: dormancy and resistance to therapy. Semin Cancer Biol. 2021; 71: 2-9.

19

Ma B, Wheeler SE, Clark AM, Whaley DL, Yang M, Wells A. Liver protects metastatic prostate cancer from induced death by activating e-cadherin signaling. Hepatology. 2016; 64: 1725-42.

20

LoPiccolo J, Granville CA, Gills JJ, Dennis PA. Targeting akt in cancer therapy. Anticancer Drugs. 2007; 18: 861-74.

21

Martelli AM, Tabellini G, Bressanin D, Ognibene A, Goto K, Cocco L, et al. The emerging multiple roles of nuclear Akt. Biochim Biophys Acta. 2012; 1823: 2168-78.

22

Al-Qatati A, Akrong C, Stevic I, Pantel K, Awe J, Saranchuk J, et al. Plasma microrna signature is associated with risk stratification in prostate cancer patients. Int J Cancer. 2017; 141: 1231-9.

23

Larsson P, Syed Khaja AS, Semenas J, Wang T, Sarwar M, Dizeyi N, et al. The functional interlink between ar and mmp9/vegf signaling axis is mediated through pip5k1alpha/pakt in prostate cancer. Int J Cancer. 2020; 146: 1686-99.

24

Nitulescu GM, Margina D, Juzenas P, Peng Q, Olaru OT, Saloustros E, et al. Akt inhibitors in cancer treatment: the long journey from drug discovery to clinical use (review). Int J Oncol. 2016; 48: 869-85.

25

Brodbeck D, Hill MM, Hemmings BA. Two splice variants of protein kinase b gamma have different regulatory capacity depending on the presence or absence of the regulatory phosphorylation site serine 472 in the carboxyl-terminal hydrophobic domain. J Biol Chem. 2001; 276: 29550-8.

26

Zinda MJ, Johnson MA, Paul JD, Horn C, Konicek BW, Lu ZH, et al. Akt-1, -2, and -3 are expressed in both normal and tumor tissues of the lung, breast, prostate, and colon. Clin Cancer Res. 2001; 7: 2475-9.

27

Kim D, Dan HC, Park S, Yang L, Liu Q, Kaneko S, et al. AKT/PKB signaling mechanisms in cancer and chemoresistance. Front. Biosci. 2005; 10: 975-87.

28

Sheng S, Qiao M, Pardee AB. Metastasis and AKT activation. J cell Physiol. 2009; 218: 451-4.

29

Toker A, Yoeli-Lerner M. Akt signaling and cancer: surviving but not moving on. Cancer Res. 2006; 66: 3963-6.

30

Luo Y, Shoemaker AR, Liu X, Woods KW, Thomas SA, de Jong R, et al. Potent and selective inhibitors of Akt kinases slow the progress of tumors in vivo. Mol Cancer Ther. 2005; 4: 977-86.

31

Yap TA, Walton MI, Hunter LJ, Valenti M, de Haven Brandon A, Eve PD, et al. Preclinical pharmacology, antitumor activity, and development of pharmacodynamic markers for the novel, potent AKT inhibitor CCT128930. Mol Cancer Ther. 2011; 10: 360-71.

32

Hirai H, Sootome H, Nakatsuru Y, Miyama K, Taguchi S, Tsujioka K, et al. Mk-2206, an allosteric Akt inhibitor, enhances antitumor efficacy by standard chemotherapeutic agents or molecular targeted drugs in vitro and in vivo. Mol Cancer Ther. 2010; 9: 1956-67.

33

Chorner PM, Moorehead RA. A-674563, a putative AKT1 inhibitor that also suppresses CDK2 activity, inhibits human NSCLC cell growth more effectively than the pan-AKT inhibitor, MK-2206. PLoS One. 2018; 13: e0193344.

34

Zheng HC. The molecular mechanisms of chemoresistance in cancers. Oncotarget. 2017; 8: 59950-64.

35

Yates CC, Shepard CR, Stolz DB, Wells A. Co-culturing human prostate carcinoma cells with hepatocytes leads to increased expression of e-cadherin. Br J Cancer. 2007; 96: 1246-52.

36

Rodriguez FJ, Lewis-Tuffin LJ, Anastasiadis PZ. E-cadherin’s dark side: possible role in tumor progression. Biochim Biophys Acta. 2012; 1826: 23-31.

37

De Santis G, Miotti S, Mazzi M, Canevari S, Tomassetti A. E-cadherin directly contributes to PI3K/AKT activation by engaging the Pi3K-P85 regulatory subunit to adherens junctions of ovarian carcinoma cells. Oncogene. 2009; 28: 1206-17.

38

Chao Y, Wu Q, Acquafondata M, Dhir R, Wells A. Partial mesenchymal to epithelial reverting transition in breast and prostate cancer metastases. Cancer Microenviron. 2012; 5: 19-28.

39

Mulholland DJ, Tran LM, Li Y, Cai H, Morim A, Wang S, et al. Cell autonomous role of pten in regulating castration-resistant prostate cancer growth. Cancer Cell. 2011; 19: 792-804.

40

Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, et al. Integrative genomic profiling of human prostate cancer. Cancer Cell. 2010; 18: 11-22.

41

Yoshimoto M, Cunha IW, Coudry RA, Fonseca FP, Torres CH, Soares FA, et al. Fish analysis of 107 prostate cancers shows that pten genomic deletion is associated with poor clinical outcome. Br J Cancer. 2007; 97: 678-85.

42

Rychahou PG, Kang J, Gulhati P, Doan HQ, Chen LA, Xiao SY, et al. Akt2 overexpression plays a critical role in the establishment of colorectal cancer metastasis. Proc Natl Acad Sci USA. 2008; 105: 20315-20.

43

Attoub S, Arafat K, Hammadi NK, Mester J, Gaben AM. Akt2 knock-down reveals its contribution to human lung cancer cell proliferation, growth, motility, invasion and endothelial cell tube formation. Sci Rep. 2015; 5: 12759.

44

Iliopoulos D, Polytarchou C, Hatziapostolou M, Kottakis F, Maroulakou IG, Struhl K, et al. Micrornas differentially regulated by akt isoforms control emt and stem cell renewal in cancer cells. Sci Signal. 2009; 2: ra62.

45

Grottke A, Ewald F, Lange T, Norz D, Herzberger C, Bach J, et al. Downregulation of AKT3 increases migration and metastasis in triple negative breast cancer cells by upregulating S100A4. PloS One. 2016; 11: e0146370.

46

Politz O, Siegel F, Barfacker L, Bomer U, Hagebarth A, Scott WJ, et al. BAY 1125976, a selective allosteric AKT1/2 inhibitor, exhibits high efficacy on AKT signaling-dependent tumor growth in mouse models. Int J Cancer. 2017; 140: 449-59.

47

West KA, Castillo SS, Dennis PA. Activation of the PI3K/Akt pathway and chemotherapeutic resistance. Drug Resist Updat. 2002; 5: 234-48.

48

Thamilselvan V, Menon M, Thamilselvan S. Combination of carmustine and selenite effectively inhibits tumor growth by targeting androgen receptor, androgen receptor-variants, and Akt in preclinical models: new hope for patients with castration resistant prostate cancer. Int J Cancer. 2016; 139: 1632-47.

49

Giantonio BJ, Derry C, McAleer C, McPhillips JJ, O’Dwyer PJ. Phase Ⅰ and pharmacokinetic study of the cytotoxic ether lipid ilmofosine administered by weekly two-hour infusion in patients with advanced solid tumors. Clin Cancer Res. 2004; 10: 1282-8.

50

Sampath D, Malik A, Plunkett W, Nowak B, Williams B, Burton M, et al. Phase i clinical, pharmacokinetic, and pharmacodynamic study of the akt-inhibitor triciribine phosphate monohydrate in patients with advanced hematologic malignancies. Leuk Res. 2013; 37: 1461-7.

51

Hinz N, Baranowsky A, Horn M, Kriegs M, Sibbertsen F, Smit DJ, et al. Knockdown of AKT3 activates HER2 and DDR kinases in bone-seeking breast cancer cells, promotes metastasis in vivo and attenuates the TGFbeta/CTGF axis. Cells. 2021; 10: 430.

52

Phung TL, Du W, Xue Q, Ayyaswamy S, Gerald D, Antonello Z, et al. Akt1 and akt3 exert opposing roles in the regulation of vascular tumor growth. Cancer Res. 2015; 75: 40-50.

53

Wiesehofer M, Czyrnik ED, Spahn M, Ting S, Reis H, Dankert JT, et al. Increased expression of AKT3 in neuroendocrine differentiated prostate cancer cells alters the response towards anti-androgen treatment. Cancers (Basel) 2021; 13: 578.

54

Buikhuisen JY, Gomez Barila PM, Torang A, Dekker D, de Jong JH, Cameron K, et al. AKT3 expression in mesenchymal colorectal cancer cells drives growth and is associated with epithelial-mesenchymal transition. Cancers (Basel). 2021; 13: 801.

55

Shu G, Su H, Wang Z, Lai S, Wang Y, Liu X, et al. LINC00680 enhances hepatocellular carcinoma stemness behavior and chemoresistance by sponging miR-568 to upregulate AKT3. J Exp Clin Cancer Res. 2021; 40: 45.

56

Sui GQ, Fei D, Guo F, Zhen X, Luo Q, Yin S, et al. MicroRNA-338-3p inhibits thyroid cancer progression through targeting AKT3. Am J Cancer Res. 2017; 7: 1177-87.

57

Polytarchou C, Hatziapostolou M, Yau TO, Christodoulou N, Hinds PW, Kottakis F, et al. Akt3 induces oxidative stress and DNA damage by activating the NADPH oxidase via phosphorylation of p47(phox). Proc Natl Acad Sci USA. 2020; 117: 28806-15.

Cancer Biology & Medicine
Pages 635-650
Cite this article:
Ma B, Shao H, Jiang X, et al. Akt isoforms differentially provide for chemoresistance in prostate cancer. Cancer Biology & Medicine, 2022, 19(5): 635-650. https://doi.org/10.20892/j.issn.2095-3941.2020.0747

200

Views

2

Downloads

7

Crossref

9

Web of Science

8

Scopus

Altmetrics

Received: 07 December 2020
Accepted: 01 April 2021
Published: 24 June 2022
©2022 Cancer Biology & Medicine.

Creative Commons Attribution-NonCommercial 4.0 International License

Return