1
|
Biswal S, Sahoo SK, Biswal BK. Shikonin a potent phytotherapeutic: a comprehensive review on metabolic reprogramming to overcome drug resistance in cancer. Mol Biol Rep 2025; 52:347. [PMID: 40156720 DOI: 10.1007/s11033-025-10459-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/20/2025] [Indexed: 04/01/2025]
Abstract
Drug resistance remains a major challenge in cancer therapy, often leading to treatment failure. Metabolic reprogramming, a hallmark of cancer, plays a pivotal role in drug resistance. Phytocompounds, particularly shikonin, a naphthoquinone derived from Lithospermum erythrorhizon, have garnered significant interest as potential alternatives for cancer prevention and treatment. This review focuses on the anticancer properties of shikonin, particularly its ability to modulate metabolic reprogramming and overcome drug resistance. This review, based on extensive searches in databases like PubMed, Web of Science, Google Scholar, and Scopus, highlights shikonin's potential as a therapeutic agent. Shikonin exhibits a wide range of anticancer activities, including induction of apoptosis, autophagy, necroptosis, inhibition of angiogenesis, invasion, and migration, as well as disruption of the cell cycle and promotion of DNA damage. It targets altered cancer cell metabolism to inhibit proliferation and reverse drug resistance, making it a promising candidate for therapeutic development. Preliminary clinical trials suggest that shikonin can enhance the efficacy of established chemotherapeutic agents, immunotherapies, and radiation through additive and synergistic interactions. Despite its promise, further research is needed to elucidate the precise mechanisms underlying shikonin's metabolic reprogramming effects in cancer. A comprehensive understanding could pave the way for its integration into standard oncological treatments. With its capacity to act on multiple cancer pathways and enhance conventional treatments, shikonin stands out as a viable candidate for combating drug-resistant cancers and advancing clinical oncology.
Collapse
Affiliation(s)
- Stuti Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India
| | | | - Bijesh K Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India.
| |
Collapse
|
2
|
Zou H, Deng W, Xu L, Shi M, Liu L, Gong L, Cui D, Zhang F. CircRNA-0013747 Promotes Mesangial Cell Proliferation in Immunoglobulin A Nephropathy through Modulation of the Warburg Effect. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2025; 68:43-56. [PMID: 39904903 DOI: 10.4103/ejpi.ejpi-d-24-00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/20/2024] [Indexed: 02/06/2025]
Abstract
ABSTRACT Immunoglobulin A nephropathy (IgAN) is characterized by aberrant mesangial cell (MC) proliferation, which is a critical determinant of glomerular sclerosis and renal dysfunction. Previous studies have highlighted the role of pyruvate kinase M2 (PKM2)- mediated aerobic glycolysis in promoting MC growth and the progression of kidney diseases. However, the precise mechanisms underlying PKM2 dysregulation in IgAN remain unclear. Circular RNAs (circRNAs), a class of noncoding RNAs, have emerged as pivotal regulators in various diseases, yet their role in IgAN has not been fully elucidated. In this study, we investigated the expression and functional significance of circRNA_0013747 in IgAN, focusing on its interaction with microRNA-330-3p (miR-330-3p) and its downstream effects on PKM2-mediated aerobic glycolysis. Our results demonstrated a significant upregulation of circRNA_0013747 in kidney biopsy samples from IgAN patients. Functional analyses revealed that circRNA_0013747 promoted MC proliferation and activated PKM2-mediated aerobic glycolysis. Importantly, these effects were attenuated by the upregulation of miR-330-3p, which was found to physically interact with circRNA_0013747, thereby inhibiting its function. Mechanistically, circRNA_0013747 acted as a sponge for miR-330-3p, relieving its suppressive effects on PKM2 expression. These findings suggest that circRNA_0013747 enhances glycolysis and proliferation in MCs through modulation of the miR-330-3p/PKM2 signaling axis. These results offer novel insights into the pathogenesis of IgAN and could contribute to new therapeutic approaches for this disease. Specifically, targeting circRNA_0013747 or modulating its interaction with miR-330-3p may provide a means to inhibit MC proliferation and aerobic glycolysis, thereby slowing the progression of IgAN and preserving renal function. Such therapeutic strategies hold the promise of substantial benefits for patients with IgAN and could pave the path toward developing more potent treatments for a wider range of renal diseases.
Collapse
Affiliation(s)
- Huimei Zou
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, China
- Department of Fundamental Nursing, School of Nursing, Guizhou Medical University, Guiyang, China
| | - Wenli Deng
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, China
| | - Lifen Xu
- Department of Pathology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Mingjun Shi
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, China
| | - Lingling Liu
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, China
| | - Lei Gong
- School of Basic Medicine, Qujing Medical College, Qujing, China
| | - Daolin Cui
- School of Basic Medicine, Qujing Medical College, Qujing, China
| | - Fan Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, China
| |
Collapse
|
3
|
Li Y, Wang W, Xu D, Liang H, Yu H, Zhou Y, Liang J, Sun H, Liu X, Xue M, Ling B, Feng D. PIWIL2/PDK1 Axis Promotes the Progression of Cervical Epithelial Lesions via Metabolic Reprogramming to Maintain Tumor-Initiating Cell Stemness. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2410756. [PMID: 39499767 DOI: 10.1002/advs.202410756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/26/2024] [Indexed: 11/07/2024]
Abstract
When PIWIL2 expression is restored via heterogeneous integration of human papillomavirus, cellular reprogramming is initiated to form tumor-initiating cells (TICs), which triggers cervical squamous intraepithelial lesions (SIL). TIC stemness is critical for the prognosis of SIL. However, the mechanisms underlying TIC stemness maintenance and tumorigenicity remain unclear. Here, it is revealed that aberrant pyruvate dehydrogenase kinase 1 (PDK1) expression is closely related to aerobic glycolysis in SIL and poor survival in patients with cervical cancer. Mechanistically, that PIWIL2, which induced by stable transfection of either PIWIL2 or HPV16 oncogene E6 in human primary cervical basal epithelial cells and keratinocyte cell line HaCaT, upregulates PDK1 expression via the LIN28/let-7 axis, hence reprogramming metabolism to activate glycolysis and synchronize with TIC formation. It is further demonstrate that PDK1 is critical for TIC stemness maintenance and tumorigenicity via the PI3K/AKT/mTOR pathway both in vitro and in vivo, revealing a previously unclear mechanism for SIL progression, regression or relapse. Therefore, this findings suggest a potential rationale for prognostic predictions and selecting targeted therapy for cervical lesions.
Collapse
Affiliation(s)
- Yuebo Li
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, 100029, China
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Wenhui Wang
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Dongkui Xu
- VIP Department, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Haiyan Liang
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Huan Yu
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Ying Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, 230001, China
| | - Jing Liang
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Heming Sun
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Xiaodie Liu
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, 100029, China
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Ming Xue
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, 100029, China
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Bin Ling
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Dingqing Feng
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, 100029, China
| |
Collapse
|
4
|
Angulo-Elizari E, Henriquez-Figuereo A, Morán-Serradilla C, Plano D, Sanmartín C. Unlocking the potential of 1,4-naphthoquinones: A comprehensive review of their anticancer properties. Eur J Med Chem 2024; 268:116249. [PMID: 38458106 DOI: 10.1016/j.ejmech.2024.116249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 03/10/2024]
Abstract
Cancer encompasses a group of pathologies with common characteristics, high incidence, and prevalence in all countries. Although there are treatments available for this disease, they are not always effective or safe, often failing to achieve the desired results. This is why it is necessary to continue the search for new therapies. One of the strategies for obtaining new antitumor drugs is the use of 1,4-naphthoquinone as a scaffold in synthetic or natural products with antitumor activity. This review focuses on compiling studies related to the antitumor activity of 1,4-naphthoquinone and its natural and synthetic derivatives over the last 10 years. The work describes the main natural naphthoquinones with antitumor activity and classifies the synthetic naphthoquinones based on the structural modifications made to the scaffold. Additionally, the formation of metal complexes using naphthoquinones as a ligand is considered. After a thorough review, 197 synthetic compounds with potent biological activity against cancer have been classified according to their chemical structures and their mechanisms of action have been described.
Collapse
Affiliation(s)
- Eduardo Angulo-Elizari
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain
| | - Andreina Henriquez-Figuereo
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain
| | - Cristina Morán-Serradilla
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain
| | - Daniel Plano
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain; Navarra Institute for Health Research (IdisNA), 31008, Pamplona, Spain.
| | - Carmen Sanmartín
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain; Navarra Institute for Health Research (IdisNA), 31008, Pamplona, Spain.
| |
Collapse
|
5
|
Zuo Y, Long Z, Li R, Le Y, Zhang S, He H, Yan L. Design, synthesis and antitumor activity of 4-arylamine substituted pyrimidine derivatives as noncovalent EGFR inhibitors overcoming C797S mutation. Eur J Med Chem 2024; 265:116106. [PMID: 38169271 DOI: 10.1016/j.ejmech.2023.116106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/26/2023] [Accepted: 12/26/2023] [Indexed: 01/05/2024]
Abstract
Clinical researches have shown that epidermal growth factor receptor (EGFR) is a key target for treatment of non-small cell lung cancer (NSCLC). Many EGFR inhibitors were successfully developed as ani-tumor drugs to treat NSCLC patients. Unfortunately, drug resistances were found in clinic. To overcome C797S mutation in EGFR, a novel series of 4-arylamine substituted pyrimidine derivatives were designed and synthesized under the principle of structure-based drug design. Interestingly, compounds 6e and 9i demonstrated the best anti-proliferative activity against A549, NCI-H1975, and HCC827 cells. In particular, the IC50 values against HCC827 cells reached to 24.6 nM and 31.6 nM, which were much lower than human normal cells 2BS and LO2. Furthermore, compounds 6e and 9i showed extraordinary activity against EGFR19del/T790M/C797S (IC50 = 16.06 nM and 37.95 nM) and EGFRL858R/T790M/C797S (IC50 = 11.81 nM and 26.68 nM), which were potent than Osimertinib (IC50 = 52.28 nM and 157.60 nM). Further studies have shown that compounds 6e and 9i could pertain inhibition of HCC827 colony formation, and arrest HCC827 cells at G2/M phase. Moreover, the most promising compound 6e could inhibit the migration of HCC827 cells, induce HCC827 cells apoptosis, and significantly inhibit the phosphorylation of EGFR, AKT and Erk1/2. In vivo xenograft mouse model with HCC827 cells, compound 6e resulted in remarkable tumor regression without obvious toxicity. In addition, molecular docking studies suggested that compound 6e could firmly combine with T790M-mutant, T790 M/C797S-mutant, and L858R/T790 M/C797S-mutant EGFR kinases as ATP-competitive inhibitor.
Collapse
Affiliation(s)
- Yaqing Zuo
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, 550025, China
| | - Zhiwu Long
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, 550025, China
| | - Rongrong Li
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, 550025, China
| | - Yi Le
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, 550025, China; State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
| | - Silong Zhang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, 550025, China
| | - Huan He
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, 550025, China
| | - Longjia Yan
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, 550025, China; State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China.
| |
Collapse
|
6
|
Lin H, Ai D, Liu Q, Wang X, Chen Q, Hong Z, Tao Y, Gao J, Wang L. Natural isoflavone glabridin targets PI3Kγ as an adjuvant to increase the sensitivity of MDA-MB-231 to tamoxifen and DU145 to paclitaxel. J Steroid Biochem Mol Biol 2024; 236:106426. [PMID: 37984749 DOI: 10.1016/j.jsbmb.2023.106426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/21/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023]
Abstract
Glabridin is a natural isoflavone with estrogen receptor agonism and significant anti-tumor activity. Additionally, glabridin has a regulation effect on PI3K/AKT/mTOR pathway, but its exact target remains unclear. In this study, we evaluated the antitumor activity of glabridin against breast cancer and prostate cancer cells, and further clarified its targeting to PI3K. We found that glabridin could significantly inhibit the cell viability of human breast cancer and prostate cancer cell lines. It induced caspase activation cascade and cell apoptosis through decreasing the mitochondrial transmembrane potential and increasing the intracellular reactive oxygen species (ROS). Moreover, glabridin could attenuate epithelial-mesenchymal transition (EMT) progression by inhibiting cell migration. PharmMapper calculation showed that PI3Kγ might be the most potential target protein because of the highest Normal Fit score (0.9735) and z'-score (0.9797). Molecular docking and bio-layer interferometry (BLI) analysis further demonstrated the PI3Kγ targeting of glabridin. In vivo experiments showed that glabridin can effectively inhibit the tumor growth of breast cancer xenograft model, and does not show obvious hepatorenal toxicity. Moreover, glabridin could effectively promote the anti-proliferation and pro-apoptotic effects of tamoxifen on MDA-MB-231 cell and taxol on DU145 cell. Elucidating the targeting of glabridin to PI3K may lay a theoretical foundation for the structural derivatization of glabridin, which is expected to greatly promote the application and development of glabridin in the field of cancer therapy.
Collapse
Affiliation(s)
- Hongyan Lin
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Dongxuan Ai
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Qingqing Liu
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Xinling Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Qingqing Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Zhongbin Hong
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Yuheng Tao
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Jian Gao
- School of Medicine, Anhui University of Science and Technology, Huainan 232002, Anhui, China.
| | - Liqun Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| |
Collapse
|
7
|
Gan L, Yang Y, Liang Z, Zhang M, He Y, Zhang SL. Targeting the pyruvate dehydrogenase complex/pyruvate dehydrogenase kinase (PDC/PDK) axis to discover potent PDK inhibitors through structure-based virtual screening and pharmacological evaluation. Eur J Med Chem 2024; 264:116008. [PMID: 38056298 DOI: 10.1016/j.ejmech.2023.116008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/17/2023] [Accepted: 11/24/2023] [Indexed: 12/08/2023]
Abstract
Proliferating cancer cells are characterized by the Warburg effect, a metabolic alteration in which ATP is generated from cytoplasmic glycolysis instead of oxidative phosphorylation. The pyruvate dehydrogenase complex/pyruvate dehydrogenase kinase (PDC/PDK) axis plays a crucial role in this effect and has been identified as a potential target for anticancer drug development. Herein, we present the discovery and pharmacological evaluation of potent PDK inhibitors targeting the PDK/PDC axis. We successfully identified 6 compounds from a small molecule library through a structure-based virtual screening campaign and evaluated their enzymatic inhibitory potencies for PDK1-4. Our results indicated that compound 1 exhibited submicromolar inhibitory activities against PDK1-3 (IC50 = 109.3, 135.8, and 458.7 nM, respectively), but is insensitive to PDK4 (IC50 = 8.67 μM). Furthermore, compound 1 inhibited the proliferation of A549 cells with an EC50 value of 10.7 μM. In addition, compound 1 induced cell apoptosis, arrested the cell cycle at the S phase, and reduced cell invasion and migration, while showing low in vivo toxicity at a high dose. Based on these observations, it can be concluded that compound 1 is a promising anti-PDK1-3 lead that merits further investigation.
Collapse
Affiliation(s)
- Linling Gan
- Chongqing Engineering Research Center of Pharmaceutical Sciences, School of Pharmacy, Chongqing Medical and Pharmaceutical College, Chongqing, 401331, PR China
| | - Ying Yang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing, 401331, PR China
| | - Zizhen Liang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing, 401331, PR China
| | - Maojie Zhang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing, 401331, PR China
| | - Yun He
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing, 401331, PR China.
| | - Shao-Lin Zhang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing, 401331, PR China.
| |
Collapse
|
8
|
Chen P, Lou L, Sharma B, Li M, Xie C, Yang F, Wu Y, Xiao Q, Gao L. Recent Advances on PKM2 Inhibitors and Activators in Cancer Applications. Curr Med Chem 2024; 31:2955-2973. [PMID: 37455458 DOI: 10.2174/0929867331666230714144851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 05/28/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023]
Abstract
Metabolic reprogramming of cells, from the normal mode of glucose metabolism named glycolysis, is a pivotal characteristic of impending cancerous cells. Pyruvate kinase M2 (PKM2), an important enzyme that catalyzes the final rate-limiting stage during glycolysis, is highly expressed in numerous types of tumors and aids in development of favorable conditions for the survival of tumor cells. Increasing evidence has suggested that PKM2 is one of promising targets for innovative drug discovery, especially for the developments of antitumor therapeutics. Herein, we systematically summarize the recent advancement on PKM2 modulators including inhibitors and activators in cancer applications. We also discussed the classifications of pyruvate kinases in mammals and the biological functions of PKM2 in this review. We do hope that this review would provide a comprehensive understanding of the current research on PKM2 modulators, which may benefit the development of more potent PKM2-related drug candidates to treat PKM2-associated diseases including cancers in future.
Collapse
Affiliation(s)
- Peng Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Liang Lou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Bigyan Sharma
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Mengchu Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Chengliang Xie
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Fen Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Yihang Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Qicai Xiao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Liqian Gao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| |
Collapse
|
9
|
Zhao H, Jiang R, Feng Z, Wang X, Zhang C. Transcription factor LHX9 (LIM Homeobox 9) enhances pyruvate kinase PKM2 activity to induce glycolytic metabolic reprogramming in cancer stem cells, promoting gastric cancer progression. J Transl Med 2023; 21:833. [PMID: 37980488 PMCID: PMC10657563 DOI: 10.1186/s12967-023-04658-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 10/25/2023] [Indexed: 11/20/2023] Open
Abstract
BACKGROUND Glycolytic metabolic reprogramming is a phenomenon in which cells undergo altered metabolic patterns during malignant transformation, mainly involving various aspects of glycolysis, electron transport chain, oxidative phosphorylation, and pentose phosphate pathway. This reprogramming phenomenon can be used as one of the markers of tumorigenesis and development. Pyruvate kinase is the third rate-limiting enzyme in the sugar metabolism process by specifically catalyzing the irreversible conversion of PEP to pyruvate. PURPOSE This study aimed to reveal the critical mediator(s) that regulate glycolytic metabolism reprogramming in gastric cancer and their underlying molecular mechanism and then explore the molecular mechanisms by which LHX9 may be involved in regulating gastric cancer (GC) progression. METHODS Firstly, we downloaded the GC and glycolysis-related microarray datasets from TCGA and MSigDB databases and took the intersection to screen out the transcription factor LHX9 that regulates GC glycolytic metabolic reprogramming. Software packages were used for differential analysis, single gene predictive analysis, and Venn diagram. In addition, an enrichment analysis of the glycolytic pathway was performed. Immunohistochemical staining was performed for LHX9 and PKM2 protein expression in 90 GC patients, and the association between their expressions was evaluated by Spearman's correlation coefficient method. Three human GC cell lines (AGS, NCI-N87, HGC-27) were selected for in vitro experimental validation. Flow cytometry was utilized to determine the stem cell marker CD44 expression status in GCSCs. A sphere formation assay was performed to evaluate the sphere-forming capabilities of GCSCs. In addition, RT-qPCR and Western blot experiments were employed to investigate the tumor stem cell markers OCT4 and SOX2 expression levels in GCSCs. Furthermore, a lentiviral expression vector was constructed to assess the impact of downregulating LHX9 or PKM2 on the glycolytic metabolic reprogramming of GCSCs. The proliferation, migration, and invasion of GCSCs were then detected by CCK-8, EdU, and Transwell assays. Subsequently, the mutual binding of LHX9 and PKM2 was verified using chromatin immunoprecipitation and dual luciferase reporter genes. In vivo experiments were verified by establishing a subcutaneous transplantation tumor model in nude mice, observing the size and volume of tumors in vivo in nude mice, and obtaining fresh tissues for subsequent experiments. RESULTS Bioinformatics analysis revealed that LHX9 might be involved in the occurrence and development of GC through regulating glycolytic metabolism. High LHX9 expression could be used as a reference marker for prognosis prediction of GC patients. Clinical tissue assays revealed that LHX9 and PKM2 were highly expressed in GC tissues. Meanwhile, GC tissues also highly expressed glycolysis-associated protein GLUT1 and tumor cell stemness marker CD44. In vitro cellular assays showed that LHX9 could enhance its activity and induce glycolytic metabolic reprogramming in GCSCs through direct binding to PKM2. In addition, the knockdown of LHX9 inhibited PKM2 activity and glycolytic metabolic reprogramming and suppressed the proliferation, migration, and invasive ability of GCSCs. In vivo animal experiments further confirmed that the knockdown of LHX9 could reduce the tumorigenic ability of GCSCs in nude mice by inhibiting PKM2 activity and glycolytic metabolic reprogramming. CONCLUSION The findings suggest that both LHX9 and PKM2 are highly expressed in GCs, and LHX9 may induce the reprogramming of glycolytic metabolism through transcriptional activation of PKM2, enhancing the malignant biological properties of GCSCs and ultimately promoting GC progression.
Collapse
Affiliation(s)
- Hongying Zhao
- Department of Oncology, Xuzhou City Cancer Hospital, Xuzhou Third People's Hospital, Jiangsu Province, Xuzhou Hospital Affiliated to Jiangsu University, No. 131, Huancheng Road, Gulou District, Xuzhou, 221000, People's Republic of China.
| | - Rongke Jiang
- Department of Oncology, Xuzhou City Cancer Hospital, Xuzhou Third People's Hospital, Jiangsu Province, Xuzhou Hospital Affiliated to Jiangsu University, No. 131, Huancheng Road, Gulou District, Xuzhou, 221000, People's Republic of China
| | - Zhijing Feng
- Jiangsu University, Zhenjiang, 212013, People's Republic of China
| | - Xue Wang
- Department of Oncology, Xuzhou City Cancer Hospital, Xuzhou Third People's Hospital, Jiangsu Province, Xuzhou Hospital Affiliated to Jiangsu University, No. 131, Huancheng Road, Gulou District, Xuzhou, 221000, People's Republic of China
| | - Chunmei Zhang
- Department of Oncology, Xuzhou City Cancer Hospital, Xuzhou Third People's Hospital, Jiangsu Province, Xuzhou Hospital Affiliated to Jiangsu University, No. 131, Huancheng Road, Gulou District, Xuzhou, 221000, People's Republic of China
| |
Collapse
|
10
|
She W, Liu T, Li H, Wang Z, Guo Z, Liu Y, Liu Y. Reprogramming Energy Metabolism with Synthesized PDK Inhibitors Based on Dichloroacetate Derivatives and Targeted Delivery Systems for Enhanced Cancer Therapy. J Med Chem 2023; 66:14683-14699. [PMID: 37688544 DOI: 10.1021/acs.jmedchem.3c01197] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2023]
Abstract
In many types of cancers, pyruvate dehydrogenase kinase (PDK) is abnormally overexpressed and has become a promising target for cancer therapy. However, few highly effective inhibitors of PDK have been reported to date. Herein, we designed and synthesized a series of PDK inhibitors based on dichloroacetate (DCA) and arsenicals. Of the 27 compounds, 1f demonstrated PDK inhibition with high efficiency at a cellular level (IC50 = 2.0 μM) and an enzyme level (EC50 = 68 nM), far more effective than that of DCA. In silico, in vitro, and in vivo studies demonstrated that 1f inhibited PDK, shifted the energy metabolism from aerobic glycolysis to oxidative phosphorylation, and induced cell apoptosis. Moreover, new 1f-loaded nanoparticles were developed, and the administration of high-drug-loading nanoparticles (0.15 mg/kg) caused up to 90% tumor shrinkage without any apparent toxicity. Hence, this study provided a novel metabolic therapy for cancer treatment.
Collapse
Affiliation(s)
- Wenyan She
- College of Chemistry and Molecular Science, Wuhan University, Wuhan 430072, P. R. China
| | - Tingting Liu
- State Key Laboratory of Separation Membranes and Membrane Processes, School of Chemistry & School of Material Science and Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Haimei Li
- College of Chemistry and Molecular Science, Wuhan University, Wuhan 430072, P. R. China
| | - Zichen Wang
- College of Chemistry and Molecular Science, Wuhan University, Wuhan 430072, P. R. China
| | - Zhibin Guo
- State Key Laboratory of Separation Membranes and Membrane Processes, School of Chemistry & School of Material Science and Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Yi Liu
- College of Chemistry and Molecular Science, Wuhan University, Wuhan 430072, P. R. China
- State Key Laboratory of Separation Membranes and Membrane Processes, School of Chemistry & School of Material Science and Engineering, Tiangong University, Tianjin 300387, P. R. China
- School of Chemical and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, P. R. China
- Hubei Key Laboratory of Radiation Chemistry and Functional Materials, Hubei University of Science and Technology, Xianning 437100, P. R. China
| | - Yujiao Liu
- State Key Laboratory of Separation Membranes and Membrane Processes, School of Chemistry & School of Material Science and Engineering, Tiangong University, Tianjin 300387, P. R. China
| |
Collapse
|
11
|
Ma Y, Lai X, Wen Z, Zhou Z, Yang M, Chen Q, Wang X, Mei F, Yang L, Yin T, Sun S, Lu G, Qi J, Lin H, Han H, Yang Y. Design, synthesis and biological evaluation of novel modified dual-target shikonin derivatives for colorectal cancer treatment. Bioorg Chem 2023; 139:106703. [PMID: 37399615 DOI: 10.1016/j.bioorg.2023.106703] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/15/2023] [Accepted: 06/25/2023] [Indexed: 07/05/2023]
Abstract
Warburg effect provides energy and material essential for tumor proliferation, the reverse of Warburg effect provides insights into the development of a novel anti-cancer strategy. Pyruvate kinase 2 (PKM2) and pyruvate dehydrogenase kinase 1 (PDK1) are two key enzymes in tumor glucose metabolism pathway that not only contribute to the Warburg effect through accelerating aerobic glycolysis, but also serve as druggable target for colorectal cancer (CRC). Considering that targeting PKM2 or PDK1 alone does not seem to be sufficient to remodel abnormal glucose metabolism and achieve significant antitumor activity, a series of novel benzenesulfonyl shikonin derivatives were designed to regulate PKM2 and PDK1 simultaneously. By means of molecular docking and antiproliferative screen, we found that compound Z10 could act as the combination of PKM2 activator and PDK1 inhibitor, thereby significantly inhibited glycolysis that reshaping tumor metabolism. Moreover, Z10 could inhibit proliferation, migration and induce apoptosis in CRC cell HCT-8. Finally, the in vivo anti-tumor activity of Z10 was evaluated in a colorectal cancer cell xenograft model in nude mice and the results demonstrated that Z10 induced tumor cell apoptosis and inhibited tumor cell proliferation with lower toxicity than shikonin. Our findings indicated that it is feasible to alter tumor energy metabolism through multi-target synergies, and the dual-target benzenesulfonyl shikonin derivative Z10 could be a potential anti-CRC agent.
Collapse
Affiliation(s)
- Yudi Ma
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Xiaohui Lai
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; School of Biology and Geography Science, Yili Normal University, Yining 835000, China
| | - Zhongling Wen
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Ziling Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Minkai Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Qingqing Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Xuan Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Feng Mei
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Liu Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Tongming Yin
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Shucun Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Guihua Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Jinliang Qi
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Hongyan Lin
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China; School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Hongwei Han
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China.
| | - Yonghua Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China.
| |
Collapse
|