51
|
Su Q, Luo S, Tan Q, Deng J, Zhou S, Peng M, Tao T, Yang X. The role of pyruvate kinase M2 in anticancer therapeutic treatments. Oncol Lett 2019; 18:5663-5672. [PMID: 31788038 PMCID: PMC6865080 DOI: 10.3892/ol.2019.10948] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 09/06/2019] [Indexed: 12/13/2022] Open
Abstract
Cancer cells are characterized by a high glycolytic rate, which leads to energy regeneration and anabolic metabolism; a consequence of this is the abnormal expression of pyruvate kinase isoenzyme M2 (PKM2). Multiple studies have demonstrated that the expression levels of PKM2 are upregulated in numerous cancer types. Consequently, the mechanism of action of certain anticancer drugs is to downregulate PKM2 expression, indicating the significance of PKM2 in a chemotherapeutic setting. Furthermore, it has previously been highlighted that the downregulation of PKM2 expression, using either inhibitors or short interfering RNA, enhances the anticancer effect exerted by THP treatment on bladder cancer cells, both in vitro and in vivo. The present review summarizes the detailed mechanisms and therapeutic relevance of anticancer drugs that inhibit PKM2 expression. In addition, the relationship between PKM2 expression levels and drug resistance were explored. Finally, future directions, such as the targeting of PKM2 as a strategy to explore novel anticancer agents, were suggested. The current review explored and highlighted the important role of PKM2 in anticancer treatments.
Collapse
Affiliation(s)
- Qiongli Su
- Department of Pharmacy, Zhuzhou Central Hospital, Zhuzhou, Hunan 412000, P.R. China
| | - Shengping Luo
- Department of Pharmacy, Zhuzhou Central Hospital, Zhuzhou, Hunan 412000, P.R. China
| | - Qiuhong Tan
- Department of Pharmacy, Zhuzhou Central Hospital, Zhuzhou, Hunan 412000, P.R. China
| | - Jun Deng
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - Sichun Zhou
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - Mei Peng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Ting Tao
- Department of Pharmacy, Yueyang Maternal-Child Medicine Health Hospital, Yueyang, Hunan 414000, P.R. China
| | - Xiaoping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
52
|
Han CY, Patten DA, Lee SG, Parks RJ, Chan DW, Harper M, Tsang BK. p53 Promotes chemoresponsiveness by regulating hexokinase II gene transcription and metabolic reprogramming in epithelial ovarian cancer. Mol Carcinog 2019; 58:2161-2174. [DOI: 10.1002/mc.23106] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 08/16/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Chae Young Han
- Department of Obstetrics & Gynecology and Cellular & Molecular MedicineUniversity of Ottawa; Chronic Disease Program, Ottawa Hospital Research Institute Ottawa Ontario Canada
| | - David A. Patten
- Department of Biochemistry, Microbiology and ImmunologyUniversity of Ottawa, Institute of Systems Biology Ottawa Ontario Canada
| | - Seung Gee Lee
- Department of Obstetrics & Gynecology and Cellular & Molecular MedicineUniversity of Ottawa; Chronic Disease Program, Ottawa Hospital Research Institute Ottawa Ontario Canada
| | - Robin J. Parks
- Department of Biochemistry, Microbiology & ImmunologyUniversity of Ottawa and Regenerative Medicine Program, Ottawa Hospital Research Institute Ottawa Ontario Canada
| | - David W. Chan
- Department of Obstetrics and GynecologyThe University of Hong Kong Hong Kong SAR P.R. China
| | - Mary‐Ellen Harper
- Department of Biochemistry, Microbiology and ImmunologyUniversity of Ottawa, Institute of Systems Biology Ottawa Ontario Canada
| | - Benjamin K. Tsang
- Department of Obstetrics & Gynecology and Cellular & Molecular MedicineUniversity of Ottawa; Chronic Disease Program, Ottawa Hospital Research Institute Ottawa Ontario Canada
| |
Collapse
|
53
|
Liu W, Woolbright BL, Pirani K, Didde R, Abbott E, Kaushik G, Martin P, Hamilton-Reeves J, Taylor JA, Holzbeierlein JM, Anant S, Lee EK. Tumor M2-PK: A novel urine marker of bladder cancer. PLoS One 2019; 14:e0218737. [PMID: 31246990 PMCID: PMC6597081 DOI: 10.1371/journal.pone.0218737] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 06/07/2019] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Bladder cancer is a "Warburg-like" tumor characterized by a reliance on aerobic glycolysis and expression of pyruvate kinase M2 (PKM2). PKM2 oscillates between an active tetramer and an inactive dimer. We aim to further characterize PKM2, in particular PKM2 dimer, as a urinary biomarker of bladder cancer and a potential target for treatment. METHODS HTB-9, HTB-5, and UM-UC3 bladder cancer cells were assessed for proliferation under differential glucose levels using the hexosaminidase assay. Western blot and Blue-native analysis was performed for protein expression of PKM2. Shikonin, an herb that is known to bind and inhibit PKM2, was utilized to determine if PKM2 has a role in glucose usage and cellular proliferation in bladder cancer cells by caspase activity assay. Institutional review board approval was obtained to collect healthy control and bladder cancer patient urine samples. The ScheBo M2-PK EDTA Plasma Test was performed on urine samples to assess urine Tumor M2-PK values. RESULTS The three bladder cancer cell lines tested all demonstrate statistically significant increases in proliferation when exposed to higher level of glucose (200mg/dL). Similarly, low doses of glucose (25mg/dL) result in reduced proliferation. Increased cell growth in higher glucose concentration correlated with up-regulation of PKM2 protein expression. Shikonin, a PKM2 inhibitor, reduced cell proliferation and switched PKM2 isoforms from the dimer to tetramer. Lastly, dimer PKM2 (Tumor-M2PK) levels were assessed in the urine samples from bladder cancer (Bca) patients and healthy controls. Tumor M2-PK significantly correlated with the presence of BCa in our subjects. CONCLUSIONS Our studies demonstrate the potential of PKM2, specifically the dimer (Tumor-M2PK) as a target of drug therapy and as a urinary marker for bladder cancer.
Collapse
Affiliation(s)
- Weiya Liu
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Benjamin L. Woolbright
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Karim Pirani
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Ryan Didde
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Erika Abbott
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Gaurav Kaushik
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Paige Martin
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Jill Hamilton-Reeves
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - John A. Taylor
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Jeffrey M. Holzbeierlein
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Shrikant Anant
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Eugene K. Lee
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| |
Collapse
|
54
|
Shikonin derivatives for cancer prevention and therapy. Cancer Lett 2019; 459:248-267. [PMID: 31132429 DOI: 10.1016/j.canlet.2019.04.033] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/15/2019] [Accepted: 04/26/2019] [Indexed: 12/25/2022]
Abstract
Phytochemicals gained considerable interest during the past years as source to develop new treatment options for chemoprevention and cancer therapy. Motivated by the fact that a majority of established anticancer drugs are derived in one way or another from natural resources, we focused on shikonin, a naphthoquinone with high potentials to be further developed as preventive or therapeutic drug to fight cancer. Shikonin is the major chemical component of Lithospermum erythrorhizon (Purple Cromwell) roots. Traditionally, the root extract has been applied to cure dermatitis, burns, and wounds. Over the past three decades, the anti-inflammatory and anticancer effects of root extracts, isolated shikonin as well as semi-synthetic and synthetic derivatives and nanoformulations have been described. In vitro and in vivo experiments were conducted to understand the effect of shikonin at cellular and molecular levels. Preliminary clinical trials indicate the potential of shikonin for translation into clinical oncology. Shikonin exerts additive and synergistic interactions in combination with established chemotherapeutics, immunotherapeutic approaches, radiotherapy and other treatment modalities, which further underscores the potential of this phytochemical to be integrated into standard treatment regimens.
Collapse
|
55
|
Wang F, Yao X, Zhang Y, Tang J. Synthesis, biological function and evaluation of Shikonin in cancer therapy. Fitoterapia 2019; 134:329-339. [DOI: 10.1016/j.fitote.2019.03.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/04/2019] [Accepted: 03/07/2019] [Indexed: 12/16/2022]
|
56
|
Wang Y, Hao F, Nan Y, Qu L, Na W, Jia C, Chen X. PKM2 Inhibitor Shikonin Overcomes the Cisplatin Resistance in Bladder Cancer by Inducing Necroptosis. Int J Biol Sci 2018; 14:1883-1891. [PMID: 30443191 PMCID: PMC6231221 DOI: 10.7150/ijbs.27854] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 09/08/2018] [Indexed: 02/06/2023] Open
Abstract
Cisplatin-based chemotherapy often results in the development of chemo-resistance when used to treat bladder cancer (BC), which is difficult to overcome. Recent data indicate that pyruvate kinase M2 (PKM2), a glycolytic enzyme for Warburg effect, is strongly upregulated in BC, and contributes to the cisplatin resistance in BC. However, the underlying mechanisms remain unclear. In this study, we also found that the expression level of PKM2 is also higher in cisplatin resistant BC cells and tumors. Down-regulation of PKM2 by siRNA or inhibition of PKM2 by shikonin re-sensitized the cisplatin resistant T24 cells. Shikonin and cisplatin together exhibit significantly greater killing effects than when used alone. Interestingly, we found shikonin kills the T24 cisplatin resistant cells by inducing necroptosis, as the cell death could not inhibited by apoptosis inhibitor, z-VAD, but compromised by RIP3 inhibitor, GSK872, or RIP3 siRNA. In contrast, shikonin induced apoptosis in T24 parental cells. We further investigate the underlying mechanism, and found that the dysregulation of Bcl-2 family proteins, including Bcl-2, PUMA, Bax, play an important role in deciding that shikonin kills the BC cells by necroptosis or apoptosis. Collectively, our results suggested that inducing necroptosis is an alternative way to overcome the apoptosis resistant in BC therapy, and orchestrating the regulation of Bcl-2, PUMA, and Bax in BC cisplatin resistant cells may improve the therapy effect of cisplatin in BC tumor.
Collapse
Affiliation(s)
- Yonggang Wang
- Department of Urology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Fangshi Hao
- Department of Urology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yonghao Nan
- Department of Urology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Licheng Qu
- Center for Reproductive Medicine and Center for Prenatal Diagnosis, First Hospital, Jilin University, Changchun, China
| | - Wanli Na
- Department of Urology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Chunshu Jia
- Center for Reproductive Medicine and Center for Prenatal Diagnosis, First Hospital, Jilin University, Changchun, China
| | - Xiaoliang Chen
- Department of Urology, China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
57
|
Gupta A, Ajith A, Singh S, Panday RK, Samaiya A, Shukla S. PAK2-c-Myc-PKM2 axis plays an essential role in head and neck oncogenesis via regulating Warburg effect. Cell Death Dis 2018; 9:825. [PMID: 30068946 PMCID: PMC6070504 DOI: 10.1038/s41419-018-0887-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/07/2018] [Accepted: 07/18/2018] [Indexed: 12/21/2022]
Abstract
The histone modifiers (HMs) are crucial for chromatin dynamics and gene expression; however, their dysregulated expression has been observed in various abnormalities including cancer. In this study, we have analyzed the expression of HMs in microarray profiles of head and neck cancer (HNC), wherein a highly significant overexpression of p21-activated kinase 2 (PAK2) was identified which was further validated in HNC patients. The elevated expression of PAK2 positively correlated with enhanced cell proliferation, aerobic glycolysis and chemoresistance and was associated with the poor clinical outcome of HNC patients. Further, dissection of molecular mechanism revealed an association of PAK2 with c-Myc and c-Myc-dependent PKM2 overexpression, wherein we showed that PAK2 upregulates c-Myc expression and c-Myc thereby binds to PKM promoter and induces PKM2 expression. We observed that PAK2-c-Myc-PKM2 axis is critical for oncogenic cellular proliferation. Depletion of PAK2 disturbs the axis and leads to downregulation of c-Myc and thereby PKM2 expression, which resulted in reduced aerobic glycolysis, proliferation and chemotherapeutic resistance of HNC cells. Moreover, the c-Myc complementation rescued PAK2 depletion effects and restored aerobic glycolysis, proliferation, migration and invasion in PAK2-depleted cells. The global transcriptome analysis of PAK2-depleted HNC cells revealed the downregulation of various genes involved in active cell proliferation, which indicates that PAK2 overexpression is critical for HNC progression. Together, these results suggest that the axis of PAK2-c-Myc-PKM2 is critical for HNC progression and could be a therapeutic target to reduce the cell proliferation and acquired chemoresistance and might enhance the efficacy of standard chemotherapy which will help in better management of HNC patients.
Collapse
Affiliation(s)
- Amit Gupta
- Epigenetics and RNA Processing Lab, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh, 462066, India
| | - Athira Ajith
- Epigenetics and RNA Processing Lab, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh, 462066, India
- Lab No. 315, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology, Madras, Tamil Nadu, 600036, India
| | - Smriti Singh
- Epigenetics and RNA Processing Lab, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh, 462066, India
| | | | - Atul Samaiya
- Department of Surgical Oncology, Bansal Hospital, Bhopal, Madhya Pradesh, 462016, India
| | - Sanjeev Shukla
- Epigenetics and RNA Processing Lab, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh, 462066, India.
| |
Collapse
|
58
|
Ni F, Yan CY, Zhou S, Hui PY, Du YH, Zheng L, Yu J, Hu XJ, Zhang ZG. Repression of GRIM19 expression potentiates cisplatin chemoresistance in advanced bladder cancer cells via disrupting ubiquitination-mediated Bcl-xL degradation. Cancer Chemother Pharmacol 2018; 82:593-605. [PMID: 30032449 DOI: 10.1007/s00280-018-3651-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/18/2018] [Indexed: 01/24/2023]
Abstract
OBJECTIVE The mainstay of treatment for advanced bladder cancer (BC) is cisplatin (CDDP)-based systematic chemotherapy. However, acquired chemoresistance induced by as yet unidentified mechanisms is encountered frequently and often results in treatment failure and disease progression. The present study was designed to elucidate the expression and potential role of the gene associated with retinoid-interferon-induced mortality-19 (GRIM19) in the pathogenesis of CDDP resistance in BC. METHODS RT-qPCR and immunoblotting were employed to evaluate the expression profile of GRIM19 in clinical BC samples and in different BC cells. Using cell viability assay, apoptotic ELISA, xenografts mouse model, and Transwell assay, the effects of GRIM19 inhibition or GRIM19 overexpression on CDDP resistance were determined in different BC cells. Lastly, using co-immunoprecipitation, we provided the molecular evidence for the interaction between GRIM19 and Bcl-xL. RESULTS Expression levels of GRIM19 were significantly down-regulated in recurrent BC specimens, and in experimentally induced CDDP-resistant BC cells. Functionally, overexpression of the exogenous GRIM19 potentiated CDDP sensitivity and suppressed the survival and invasion of BC cells in the presence of CDDP challenge. Mechanistically, the compromised CDDP chemosensitization induced by GRIM19 loss was at least partially attributed to the attenuation of Bcl-xL polyubiquitination and subsequent degradation, because (1) GRIM19 colocalized with Bcl-xL in the mitochondria of BC cells and (2) GRIM19 overexpression promoted the ubiquitination of Bcl-xL, and this event could be effectively reversed by pretreatment with inhibitors of p38-MAPK and JNK pathways, indicating that GRIM19 overexpression-induced Bcl-xL ubiquitination may achieve in a p38/JNK-dependent manner. Using the UMUC-3 cells stably depleted of endogenous GRIM19, we further show that inhibition of Bcl-xL rectified GRIM19 deficiency-caused CDDP resistance in BC cells. In addition, BCL2L1 mRNA levels were negatively correlated with GRIM19 mRNA levels in CDDP-associated clinical BC tissues. CONCLUSIONS Disruption of GRIM19/Bcl-xL is a key mechanism of CDDP resistance in advanced BC. Therapeutically, enhancement of GRIM19 expression or employment of p38/JNK inhibitors may serve as resensitizing therapies for subgroups of CDDP-resistant or refractory BC patients.
Collapse
Affiliation(s)
- Feng Ni
- Department of Urology, The 2nd Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Chang-You Yan
- Family Planning Service Stations of Health and Family Planning Commission of Chengcheng County, Chengcheng County, Weinan City, 714000, Shaanxi Province, China
| | - Sheng Zhou
- Department of Anorectal Surgery, The 2nd Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Baqiao District, Xi'an, 710038, Shaanxi Province, China.
| | - Peng-Yu Hui
- Department of Urology, The 2nd Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Yong-Hui Du
- Department of Urology, The 2nd Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Liang Zheng
- Department of Urology, The 2nd Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Jin Yu
- Department of Urology, The 2nd Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Xiao-Jian Hu
- Department of Urology, The 2nd Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Zhi-Gang Zhang
- Department of Urology, The 2nd Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi Province, China
| |
Collapse
|
59
|
Woolbright BL, Ayres M, Taylor JA. Metabolic changes in bladder cancer. Urol Oncol 2018; 36:327-337. [DOI: 10.1016/j.urolonc.2018.04.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/05/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022]
|
60
|
Woolbright BL, Choudhary D, Mikhalyuk A, Trammel C, Shanmugam S, Abbott E, Pilbeam CC, Taylor JA. The Role of Pyruvate Dehydrogenase Kinase-4 (PDK4) in Bladder Cancer and Chemoresistance. Mol Cancer Ther 2018; 17:2004-2012. [PMID: 29907593 DOI: 10.1158/1535-7163.mct-18-0063] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/18/2018] [Accepted: 06/11/2018] [Indexed: 12/12/2022]
Abstract
Advanced bladder cancer remains a major source of mortality, with poor treatment options. Cisplatin-based chemotherapy is the standard treatment, however many patients are or become resistant. One potential cause of chemoresistance is the Warburg effect, a metabolic switch to aerobic glycolysis that occurs in many cancers. Upregulation of the pyruvate dehydrogenase kinase family (PDK1-PDK4) is associated with aerobic glycolysis and chemoresistance through inhibition of the pyruvate dehydrogenase complex (PDH). We have previously observed upregulation of PDK4 in high-grade compared with low-grade bladder cancers. We initiated this study to determine if inhibition of PDK4 could reduce tumor growth rates or sensitize bladder cancer cells to cisplatin. Upregulation of PDK4 in malignant bladder cancer cell lines as compared with benign transformed urothelial cells was confirmed using qPCR. Inhibition of PDK4 with dichloroacetate (DCA) resulted in increased PDH activity, reduced cell growth, and G0-G1 phase arrest in bladder cancer cells. Similarly, siRNA knockdown of PDK4 inhibited bladder cancer cell proliferation. Cotreatment of bladder cancer cells with cisplatin and DCA did not increase caspase-3 activity but did enhance overall cell death in vitro Although daily treatment with 200 mg/kg DCA alone did not reduce tumor volumes in a xenograft model, combination treatment with cisplatin resulted in dramatically reduced tumor volumes as compared with either DCA or cisplatin alone. This was attributed to substantial intratumoral necrosis. These findings indicate inhibition of PDK4 may potentiate cisplatin-induced cell death and warrant further studies investigating the mechanism through which this occurs. Mol Cancer Ther; 17(9); 2004-12. ©2018 AACR.
Collapse
Affiliation(s)
| | | | - Andrew Mikhalyuk
- University of Connecticut School of Medicine, Farmington, Connecticut
| | - Cassandra Trammel
- University of Connecticut School of Medicine, Farmington, Connecticut
| | | | - Erika Abbott
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas
| | - Carol C Pilbeam
- Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut
| | - John A Taylor
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas.
| |
Collapse
|
61
|
Shimada N, Takasawa R, Tanuma SI. Interdependence of GLO I and PKM2 in the Metabolic shift to escape apoptosis in GLO I-dependent cancer cells. Arch Biochem Biophys 2018; 638:1-7. [PMID: 29225125 DOI: 10.1016/j.abb.2017.12.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/06/2017] [Accepted: 12/07/2017] [Indexed: 12/27/2022]
Abstract
Many cancer cells undergo metabolic reprogramming known as the Warburg effect, which is characterized by a greater dependence on glycolysis for ATP generation, even under normoxic conditions. Glyoxalase I (GLO I) is a rate-limiting enzyme involved in the detoxification of cytotoxic methylglyoxal formed in glycolysis and which is known to be highly expressed in many cancer cells. Thus, specific inhibitors of GLO I are expected to be effective anticancer drugs. We previously discovered a novel GLO I inhibitor named TLSC702. Although the strong inhibitory activity of TLSC702 was observed in the in vitro enzyme assay, higher concentrations were required to induce apoptosis at the cellular level. One of the proposed reasons for this difference is that cancer cells alter the energy metabolism leading them to become more dependent on mitochondrial respiration than glycolysis (Metabolic shift) to avoid apoptosis induction. Thus, we assumed that combination of TLSC702 with shikonin-a specific inhibitor of pyruvate kinase M2 (PKM2) that acts as a driver of TCA cycle by supplying pyruvate and which is known to be specifically expressed in cancer cells-would have anticancer effects. We herein show the anticancer effects of combination treatment with TLSC702 and shikonin, and a possible anticancer mechanism.
Collapse
Affiliation(s)
- Nami Shimada
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Ryoko Takasawa
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Sei-Ichi Tanuma
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan; Genome & Drug Research Center, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan.
| |
Collapse
|
62
|
Buss JH, Begnini KR, Bender CB, Pohlmann AR, Guterres SS, Collares T, Seixas FK. Nano-BCG: A Promising Delivery System for Treatment of Human Bladder Cancer. Front Pharmacol 2018; 8:977. [PMID: 29379438 PMCID: PMC5770893 DOI: 10.3389/fphar.2017.00977] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 12/21/2017] [Indexed: 01/06/2023] Open
Abstract
Mycobacterium bovis bacillus Calmette–Guerin (BCG) remains at the forefront of immunotherapy for treating bladder cancer patients. However, the incidence of recurrence and progression to invasive cancer is commonly observed. There are no established effective intravesical therapies available for patients, whose tumors recur following BCG treatment, representing an important unmet clinical need. In addition, there are very limited options for patients who do not respond to or tolerate chemotherapy due to toxicities, resulting in poor overall treatment outcomes. Within this context, nanotechnology is an emergent and promising tool for: (1) controlling drug release for extended time frames, (2) combination therapies due to the ability to encapsulate multiple drugs simultaneously, (3) reducing systemic side effects, (4) increasing bioavailability, (5) and increasing the viability of various routes of administration. Moreover, bladder cancer is often characterized by high mutation rates and over expression of tumor antigens on the tumor cell surface. Therapeutic targeting of these biomolecules may be improved by nanotechnology strategies. In this mini-review, we discuss how nanotechnology can help overcome current obstacles in bladder cancer treatment, and how nanotechnology can facilitate combination chemotherapeutic and BCG immunotherapies for the treatment of non-muscle invasive urothelial bladder cancer.
Collapse
Affiliation(s)
- Julieti Huch Buss
- Laboratory of Cancer Biotechnology, Biotechnology Graduate Program, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Karine Rech Begnini
- Laboratory of Cancer Biotechnology, Biotechnology Graduate Program, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Camila Bonemann Bender
- Laboratory of Cancer Biotechnology, Biotechnology Graduate Program, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Adriana R Pohlmann
- Pharmaceutical Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Institute of Chemistry, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Silvia S Guterres
- Pharmaceutical Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Tiago Collares
- Laboratory of Cancer Biotechnology, Biotechnology Graduate Program, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Fabiana Kömmling Seixas
- Laboratory of Cancer Biotechnology, Biotechnology Graduate Program, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| |
Collapse
|
63
|
Yang P, Ding GB, Liu W, Fu R, Sajid A, Li Z. Tannic acid directly targets pyruvate kinase isoenzyme M2 to attenuate colon cancer cell proliferation. Food Funct 2018; 9:5547-5559. [DOI: 10.1039/c8fo01161c] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Tannic acid, which ubiquitously exists in grapes and green tea, binds to K433 to trigger dissociation of PKM2 tetramers and further block the metabolic activity of PKM2 to suppress colorectal cancer cell proliferation.
Collapse
Affiliation(s)
- Peng Yang
- Institute of Biotechnology
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education
- Shanxi University
- Taiyuan 030006
- China
| | - Guo-Bin Ding
- Institute of Biotechnology
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education
- Shanxi University
- Taiyuan 030006
- China
| | - Wen Liu
- Institute of Biotechnology
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education
- Shanxi University
- Taiyuan 030006
- China
| | - Rong Fu
- Institute of Biotechnology
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education
- Shanxi University
- Taiyuan 030006
- China
| | - Amin Sajid
- Institute of Biotechnology
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education
- Shanxi University
- Taiyuan 030006
- China
| | - Zhuoyu Li
- Institute of Biotechnology
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education
- Shanxi University
- Taiyuan 030006
- China
| |
Collapse
|
64
|
He Y, Gao M, Cao Y, Tang H, Liu S, Tao Y. Nuclear localization of metabolic enzymes in immunity and metastasis. Biochim Biophys Acta Rev Cancer 2017; 1868:359-371. [PMID: 28757126 DOI: 10.1016/j.bbcan.2017.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/19/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023]
Abstract
Metabolism is essential to all living organisms that provide cells with energy, regulators, building blocks, enzyme cofactors and signaling molecules, and is in tune with nutritional conditions and the function of cells to make the appropriate developmental decisions or maintain homeostasis. As a fundamental biological process, metabolism state affects the production of multiple metabolites and the activation of various enzymes that participate in regulating gene expression, cell apoptosis, cancer progression and immunoreactions. Previous studies generally focus on the function played by the metabolic enzymes in the cytoplasm and mitochondrion. In this review, we conclude the role of them in the nucleus and their implications for cancer progression, immunity and metastasis.
Collapse
Affiliation(s)
- Yuchen He
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Cancer Research Institute, School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410078, China; Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Menghui Gao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Cancer Research Institute, School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410078, China; Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yiqu Cao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Cancer Research Institute, School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410078, China; Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Haosheng Tang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Cancer Research Institute, School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410078, China; Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Shuang Liu
- Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Cancer Research Institute, School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410078, China; Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
65
|
Friend or foe? Mitochondria as a pharmacological target in cancer treatment. Future Med Chem 2017; 9:2197-2210. [PMID: 29182013 DOI: 10.4155/fmc-2017-0110] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Mitochondria have acquired numerous functions over the course of evolution, such as those involved in controlling energy production, cellular metabolism, cell survival, apoptosis and autophagy within host cells. Tumor cells can develop defects in mitochondrial function, presenting a potential strategy for designing selective anticancer therapies. Therefore, cancer has been the main focus of recent research to uncover possible mitochondrial targets for therapeutic benefit. This comprehensive review covers not only the recent discoveries of the roles of mitochondria in cancer development, progression and therapeutic implications but also the findings regarding emerging mitochondrial therapeutic targets and mitochondria-targeted agents. Current challenges and future directions for developments and applications of mitochondrial-targeted therapeutics are also discussed.
Collapse
|
66
|
Abate M, Laezza C, Pisanti S, Torelli G, Seneca V, Catapano G, Montella F, Ranieri R, Notarnicola M, Gazzerro P, Bifulco M, Ciaglia E. Deregulated expression and activity of Farnesyl Diphosphate Synthase (FDPS) in Glioblastoma. Sci Rep 2017; 7:14123. [PMID: 29075041 PMCID: PMC5658376 DOI: 10.1038/s41598-017-14495-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/11/2017] [Indexed: 01/09/2023] Open
Abstract
Glioblastoma (GBM), the most aggressive brain cancer, is highly dependent on the mevalonate (MVA) pathway for the synthesis of lipid moieties critical for cell proliferation but the function and regulation of key intermediate enzymes like farnesyl-diphosphate synthase (FDPS), up to now, remained unknown. A deregulated expression and activity of FDPS was the central research idea of the present study. FDPS mRNA, protein and enzyme activity were analyzed in a cohort of stage III-IV glioma patients (N = 49) and primary derived cells. FDPS silencing helped to clarify its function in the maintenance of malignant phenotype. Interestingly, compared to tumor-free peripheral (TFB) brain and normal human astrocytes (NHA), FDPS protein expression and enzyme activity were detected at high degree in tumor mass where a correlation with canonical oncogenic signaling pathways such as STAT3, ERK and AKT was also documented. Further, FDPS knockdown in U87 and GBM primary cells but not in NHA, enhanced apoptosis. With the effort to develop a more refined map of the connectivity between signal transduction pathways and metabolic networks in cancer FDPS as a new candidate metabolic oncogene in glioblastoma, might suggest to further target MVA pathway as valid therapeutic tool.
Collapse
Affiliation(s)
- Mario Abate
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Salvatore Allende, 84081, Baronissi Salerno, Italy
| | - Chiara Laezza
- Institute of Endocrinology and Experimental Oncology, IEOS CNR, Via Pansini 5, 80131, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini, 80131, Naples, Italy
| | - Simona Pisanti
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Salvatore Allende, 84081, Baronissi Salerno, Italy
| | - Giovanni Torelli
- Neurosurgery Unit A.O. San Giovanni di Dio e Ruggi d' Aragona - Salerno's School of Medicine Largo Città di Ippocrate, 84131, Salerno, Italy
| | - Vincenzo Seneca
- "G.Rummo" Medical Hospital, Department of Neurosurgery, Benevento, Italy
| | - Giuseppe Catapano
- "G.Rummo" Medical Hospital, Department of Neurosurgery, Benevento, Italy
| | - Francesco Montella
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Salvatore Allende, 84081, Baronissi Salerno, Italy
| | - Roberta Ranieri
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Salvatore Allende, 84081, Baronissi Salerno, Italy
| | - Maria Notarnicola
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Bari, 70013, Italy
| | - Patrizia Gazzerro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084, Fisciano Salerno, Italy
| | - Maurizio Bifulco
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Salvatore Allende, 84081, Baronissi Salerno, Italy. .,Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini, 80131, Naples, Italy.
| | - Elena Ciaglia
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Salvatore Allende, 84081, Baronissi Salerno, Italy.
| |
Collapse
|