1
|
Lin TP, Chen PC, Lin CY, Wang BJ, Kuo YY, Yeh CC, Tseng JC, Huo C, Kao CL, Shih LJ, Chen JK, Li CY, Hour TC, Chuu CP. Prostate cancer cells elevate glycolysis and G6PD in response to caffeic acid phenethyl ester-induced growth inhibition. BMC Cancer 2025; 25:95. [PMID: 39819475 PMCID: PMC11737093 DOI: 10.1186/s12885-025-13477-6] [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: 05/03/2024] [Accepted: 01/08/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Caffeic acid phenethyl ester (CAPE) is the main bioactive component of poplar type propolis. We previously reported that treatment with caffeic acid phenethyl ester (CAPE) suppressed the cell proliferation, tumor growth, as well as migration and invasion of prostate cancer (PCa) cells via inhibition of signaling pathways of AKT, c-Myc, Wnt and EGFR. We also demonstrated that combined treatment of CAPE and docetaxel altered the genes involved in glycolysis and tricarboxylic acid (TCA) cycle. We therefore suspect that CAPE treatment may interfere glucose metabolism in PCa cells. METHODS Seahorse Bioenergetics platform was applied to analyzed the extra cellular acidification rate (ECAR) and oxygen consumption rate (OCR) of PCa cells under CAPE treatment. UPLC-MSMS with Multiple Reaction Monitoring (MRM), PCR, and western blot were used to analyze the effects of CAPE on metabolites, genes, and proteins involved in glycolysis, TCA cycle and pentose phosphate pathway in PCa cells. Flow cytometry and ELISA were used to determine the level of reactive oxygen species in PCa cells being treated with CAPE. RESULTS Seahorse Bioenergetics analysis revealed that ECAR, glycolysis, OCR, and ATP production were elevated in C4-2B cells under CAPE treatment. Protein levels of glucose-6-phosphate dehydrogenase (G6PD), phosphogluconate dehydrogenase (PGD), glutaminase (GLS), phospho-AMPK Thr172 as well as abundance of pyruvate, lactate, ribulose-5-phosphate, and sedoheptulose-7-phosphate were increased in CAPE-treated C4-2B cells. ROS level decreased 48 h after treatment with CAPE. Co-treatment of AMPK inhibitor with CAPE exhibited additive growth inhibition on PCa cells. CONCLUSIONS Our study indicated that PCa cells attempted to overcome the CAPE-induced stress by upregulation of glycolysis and G6PD but failed to impede the growth inhibition caused by CAPE. Concurrent treatment of CAPE and inhibitors targeting glycolysis may be effective therapy for advanced PCa.
Collapse
Affiliation(s)
- Tzu-Ping Lin
- Faculty of Medicine, National Yang Ming Chiao Tung University, Hsinchu City, 30010, Taiwan
- Department of Urology, Taipei Veterans General Hospital, Taipei City, 11217, Taiwan
| | - Pei-Chun Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Ching-Yu Lin
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, Taipei Medical University, Taipei City, 11031, Taiwan
| | - Bi-Juan Wang
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Ying-Yu Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Chien-Chih Yeh
- Department of Education and Medical Research, Taoyuan Armed Forces General Hospital, Taoyuan City, 325208, Taiwan
- Division of Colon and Rectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114202, Taiwan
| | - Jen-Chih Tseng
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan
- Immunology Research Center, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Chieh Huo
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Cheng-Li Kao
- Division of Urology, Departments of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114202, Taiwan
- Division of Urology, Department of Surgery, Taoyuan Armed Forces General Hospital, Taoyuan City, 325208, Taiwan
| | - Li-Jane Shih
- Division of Colon and Rectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114202, Taiwan
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei City, 114202, Taiwan
| | - Jen-Kun Chen
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City, 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University, Kaohsiung City, 80708, Taiwan
| | - Tzyh-Chyuan Hour
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung City, 80708, Taiwan
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung City, 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung City, 80708, Taiwan
| | - Chih-Pin Chuu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan.
- PhD Program for Aging, China Medical University, Taichung City, 40402, Taiwan.
- Biotechnology Center, National Chung Hsing University, Taichung City, 40227, Taiwan.
- Department of Life Sciences, National Central University, Taoyuan City, 32031, Taiwan.
| |
Collapse
|
2
|
Gao F, Huang Y, Yang M, He L, Yu Q, Cai Y, Shen J, Lu B. Machine learning-based cell death marker for predicting prognosis and identifying tumor immune microenvironment in prostate cancer. Heliyon 2024; 10:e37554. [PMID: 39309810 PMCID: PMC11414577 DOI: 10.1016/j.heliyon.2024.e37554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024] Open
Abstract
Background Prostate cancer (PCa) incidence and mortality rates are rising, necessitating precise prognostic tools to guide personalized treatment. Dysregulation of programmed cell death pathways in tumor suppression and cancer development has garnered increasing attention, providing a new research direction for identifying biomarkers and potential therapeutic targets. Methods Integrating multiple database resources, we constructed and optimized a prognostic signature based on the expression of programmed cell death-related genes (PCDRG) using ten machine learning algorithms. Model performance and prognostic effects were further evaluated. We analyzed the relationships between signature and clinicopathological features, somatic mutations, drug sensitivity, and the tumor immune microenvironment, and constructed a nomogram. The expression level of PCDRGs were evaluated and compared. Results Of 1560 PCDRGs, 149 were differentially expressed in PCa, with 34 associated with biochemical recurrence. The PCDRG-derived index (PCDI), constructed using the random forest algorithm, exhibited optimal prognostic performance, successfully stratifying PCa patients into two groups with significant prognostic differences. Patients with high PCDI scores exhibited poorer survival and lower immunotherapy benefit. PCDI was closely associated with the infiltration of specific immune cells, particularly positive correlations with macrophages and T helper cells, and negative correlations with neutrophils, suggesting that PCDI may influence the tumor immune microenvironment, thereby affecting patient prognosis and treatment response. PCDI was associated with age, pathological stage, somatic mutations, and drug sensitivity. The PCDI-based nomogram demonstrated excellent performance in predicting biochemical recurrence in PCa patients. Finally, the differential expression of these PCDRGs was verified based on cell lines and PCa patient expression profile data. Conclusion This study developed an effective prognostic indicator for prostate cancer, PCDI, using machine learning approaches. PCDI reflects the link between aberrant programmed cell death pathways and disease progression and treatment response.
Collapse
Affiliation(s)
- Feng Gao
- Department of Urology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310007, China
| | - Yasheng Huang
- Department of Urology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310007, China
| | - Mei Yang
- Department of Urology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310007, China
| | - Liping He
- Department of Urology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310007, China
| | - Qiqi Yu
- Department of Urology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310007, China
| | - Yueshu Cai
- Department of Urology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310007, China
| | - Jie Shen
- Department of Urology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310007, China
| | - Bingjun Lu
- Department of Urology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310007, China
| |
Collapse
|
3
|
Gonçalves J, Feijó M, Socorro S, Luís Â, Gallardo E, Duarte AP. The Role of Ayahuasca in Colorectal Adenocarcinoma Cell Survival, Proliferation and Oxidative Stress. Pharmaceuticals (Basel) 2024; 17:719. [PMID: 38931386 PMCID: PMC11207024 DOI: 10.3390/ph17060719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
The psychedelic beverage ayahuasca is originally obtained by Banisteriopsis caapi (B. caapi) (BC) and Psychotria viridis (P. viridis) (PV). However, sometimes these plant species are replaced by others that mimic the original effects, such as Mimosa hostilis (M. hostilis) (MH) and Peganum harmala (P. harmala) (PH). Its worldwide consumption and the number of studies on its potential therapeutic effects has increased. This study aimed to evaluate the anticancer properties of ayahuasca in human colorectal adenocarcinoma cells. Thus, the maximum inhibitory concentration (IC50) of decoctions of MH, PH, and a mixture of these (MHPH) was determined. The activities of caspases 3 and 9 were evaluated, and the cell proliferation index was determined through immunocytochemical analysis (Ki-67). Two fluorescent probes were used to evaluate the production of oxidative stress and the activity of the antioxidant enzymes superoxide dismutase (SOD) and glutathione peroxidase (GPx) was also evaluated. It was demonstrated that exposure to the extracts significantly induced apoptosis in Caco-2 cells, while decreasing cell proliferation. MH and MHPH samples significantly reduced oxidative stress and significantly increased glutathione peroxidase activity. No significant differences were found in SOD activity. Overall, it was demonstrated that the decoctions have a potential anticancer activity in Caco-2 cells.
Collapse
Affiliation(s)
- Joana Gonçalves
- Centro de Investigação em Ciências da Saúde (CICS-UBI), University of Beira Interior, 6200-506 Covilhã, Portugal; (J.G.); (M.F.); (S.S.); (E.G.); (A.P.D.)
- Laboratório de Fármaco-Toxicologia, UBIMedical, University of Beira Interior, 6200-284 Covilhã, Portugal
| | - Mariana Feijó
- Centro de Investigação em Ciências da Saúde (CICS-UBI), University of Beira Interior, 6200-506 Covilhã, Portugal; (J.G.); (M.F.); (S.S.); (E.G.); (A.P.D.)
| | - Sílvia Socorro
- Centro de Investigação em Ciências da Saúde (CICS-UBI), University of Beira Interior, 6200-506 Covilhã, Portugal; (J.G.); (M.F.); (S.S.); (E.G.); (A.P.D.)
| | - Ângelo Luís
- Centro de Investigação em Ciências da Saúde (CICS-UBI), University of Beira Interior, 6200-506 Covilhã, Portugal; (J.G.); (M.F.); (S.S.); (E.G.); (A.P.D.)
- Laboratório de Fármaco-Toxicologia, UBIMedical, University of Beira Interior, 6200-284 Covilhã, Portugal
| | - Eugenia Gallardo
- Centro de Investigação em Ciências da Saúde (CICS-UBI), University of Beira Interior, 6200-506 Covilhã, Portugal; (J.G.); (M.F.); (S.S.); (E.G.); (A.P.D.)
- Laboratório de Fármaco-Toxicologia, UBIMedical, University of Beira Interior, 6200-284 Covilhã, Portugal
| | - Ana Paula Duarte
- Centro de Investigação em Ciências da Saúde (CICS-UBI), University of Beira Interior, 6200-506 Covilhã, Portugal; (J.G.); (M.F.); (S.S.); (E.G.); (A.P.D.)
- Laboratório de Fármaco-Toxicologia, UBIMedical, University of Beira Interior, 6200-284 Covilhã, Portugal
| |
Collapse
|
4
|
Nayeem MJ, Yamamura A, Hayashi H, Muramatsu H, Nakamura K, Sassa N, Sato M. Imatinib mesylate inhibits androgen-independent PC-3 cell viability, proliferation, migration, and tumor growth by targeting platelet-derived growth factor receptor-α. Life Sci 2022; 288:120171. [PMID: 34822800 DOI: 10.1016/j.lfs.2021.120171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 11/08/2021] [Accepted: 11/18/2021] [Indexed: 12/18/2022]
Abstract
AIM The abnormal expression of oncogenic tyrosine kinase receptors such as platelet-derived growth factor receptors (PDGFRs) has been reported in cancer progression. However, the role of PDGFRs in the human androgen-independent prostate cancer PC-3 cell line is not well understood. Thus, this study examined the role of PDGFRs in androgen-independent PC-3 cells. MAIN METHODS PDGFR mRNA and protein expression was determined by quantitative real-time PCR and western blotting, respectively. The effects of the tyrosine kinase inhibitor imatinib (imatinib mesylate) and small interfering RNAs (siRNAs) were determined by a Cell Counting Kit-8 assay, bromodeoxyuridine assay, and Transwell migration assay. The in vivo effect of imatinib was analyzed using a tumor formation assay in nude mice. KEY FINDINGS PDGFRα was upregulated in androgen-independent PC-3 cells compared with normal prostate epithelial cells. PDGF-BB induced the phosphorylation of PDGFRα and downstream signaling molecules, including Akt, in a dose-dependent manner. Imatinib reduced the phosphorylation of the PDGFRα/Akt axis. Imatinib also suppressed the viability, proliferation, migration, and tumor growth of PC-3 cells. PDGFRα knockdown by siRNA decreased the viability and migration of PC-3 cells. SIGNIFICANCE These results demonstrated the distinct contribution of PDGFRα signaling to the proliferation and migration of PC-3 cells and suggested the potential for PDGFRα as a therapeutic target for metastatic and androgen-independent prostate cancer.
Collapse
Affiliation(s)
| | - Aya Yamamura
- Department of Physiology, Aichi Medical University, Japan
| | - Hisaki Hayashi
- Department of Physiology, Aichi Medical University, Japan
| | | | | | - Naoto Sassa
- Department of Urology, Aichi Medical University, Japan
| | - Motohiko Sato
- Department of Physiology, Aichi Medical University, Japan.
| |
Collapse
|
5
|
Cardoso HJ, Figueira MI, Vaz CV, Carvalho TMA, Brás LA, Madureira PA, Oliveira PJ, Sardão VA, Socorro S. Glutaminolysis is a metabolic route essential for survival and growth of prostate cancer cells and a target of 5α-dihydrotestosterone regulation. Cell Oncol (Dordr) 2021; 44:385-403. [PMID: 33464483 DOI: 10.1007/s13402-020-00575-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Resistance to androgen-deprivation therapies and progression to so-called castrate-resistant prostate cancer (CRPC) remain challenges in prostate cancer (PCa) management and treatment. Among other alterations, CRPC has been associated with metabolic reprogramming driven by androgens. Here, we investigated the role of androgens in regulating glutaminolysis in PCa cells and determined the relevance of this metabolic route in controlling the survival and growth of androgen-sensitive (LNCaP) and CRPC (DU145 and PC3) cells. METHODS PCa cells (LNCaP, DU145 and PC3) and 3-month old rats were treated with 5α-dihydrotestosterone (DHT). Alternatively, LNCaP cells were exposed to the glutaminase inhibitor BPTES, alone or in combination with the anti-androgen bicalutamide. Biochemical, Western blot and extracellular flux assays were used to evaluate the viability, proliferation, migration and metabolism of PCa cells in response to DHT treatment or glutaminase inhibition. RESULTS We found that DHT up-regulated the expression of the glutamine transporter ASCT2 and glutaminase, both in vitro in LNCaP cells and in vivo in rat prostate cells. BPTES diminished the viability and migration of PCa cells, while increasing caspase-3 activity. CRPC cells were found to be more dependent on glutamine and more sensitive to glutaminase inhibition. BPTES and bicalutamide co-treatment had an additive effect on suppressing LNCaP cell viability. Finally, we found that inhibition of glutaminolysis differentially affected glycolysis and lipid metabolism in both androgen-sensitive and CRPC cells. CONCLUSION Our data reveal glutaminolysis as a central metabolic route controlling PCa cell fate and highlight the relevance of targeting glutaminase for CRPC treatment.
Collapse
Affiliation(s)
- Henrique J Cardoso
- CICS-UBI, Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal
- Centre for Biomedical Research (CBMR), Campus of Gambelas, University of Algarve, Faro, Portugal
| | - Marília I Figueira
- CICS-UBI, Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal
| | - Cátia V Vaz
- CICS-UBI, Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal
| | - Tiago M A Carvalho
- CICS-UBI, Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal
| | - Luís A Brás
- CICS-UBI, Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal
| | - Patrícia A Madureira
- Centre for Biomedical Research (CBMR), Campus of Gambelas, University of Algarve, Faro, Portugal
- Brain Tumour Research Centre of Excellence, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK
| | - Paulo J Oliveira
- CNC - Center for Neuroscience and Cell Biology, UC-Biotech, University of Coimbra, Biocant Park, Cantanhede, Portugal
| | - Vilma A Sardão
- CNC - Center for Neuroscience and Cell Biology, UC-Biotech, University of Coimbra, Biocant Park, Cantanhede, Portugal
| | - Sílvia Socorro
- CICS-UBI, Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal.
| |
Collapse
|
6
|
Vaghari-Tabari M, Ferns GA, Qujeq D, Andevari AN, Sabahi Z, Moein S. Signaling, metabolism, and cancer: An important relationship for therapeutic intervention. J Cell Physiol 2021; 236:5512-5532. [PMID: 33580511 DOI: 10.1002/jcp.30276] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 11/05/2022]
Abstract
In cancerous cells, significant changes occur in the activity of signaling pathways affecting a wide range of cellular activities ranging from growth and proliferation to apoptosis, invasiveness, and metastasis. Extensive changes also happen with respect to the metabolism of a cancerous cell encompassing a wide range of functions that include: nutrient acquisition, biosynthesis of macromolecules, and energy generation. These changes are important and some therapeutic approaches for treating cancers have focused on targeting the metabolism of cancerous cells. Oncogenes and tumor suppressor genes have a significant effect on the metabolism of cells. There appears to be a close interaction between metabolism and the signaling pathways in a cancerous cell, in which the interaction provides the metabolic needs of a cancerous cell for uncontrolled proliferation, resistance to apoptosis, and metastasis. In this review, we have reviewed the latest findings in this regard and briefly review the most recent research findings regarding targeting the metabolism of cancer cells as a therapeutic approach for treatment of cancer.
Collapse
Affiliation(s)
- Mostafa Vaghari-Tabari
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gordon A Ferns
- Department of Medical Education, Brighton & Sussex Medical School, Falmer, Brighton, Sussex, UK
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | - Ali Nosrati Andevari
- Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Zahra Sabahi
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soheila Moein
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
7
|
Marcucci F, Rumio C. Glycolysis-induced drug resistance in tumors-A response to danger signals? Neoplasia 2021; 23:234-245. [PMID: 33418276 PMCID: PMC7804361 DOI: 10.1016/j.neo.2020.12.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/25/2020] [Accepted: 12/29/2020] [Indexed: 12/15/2022] Open
Abstract
Tumor cells often switch from mitochondrial oxidative metabolism to glycolytic metabolism even under aerobic conditions. Tumor cell glycolysis is accompanied by several nonenzymatic activities among which induction of drug resistance has important therapeutic implications. In this article, we review the main aspects of glycolysis-induced drug resistance. We discuss the classes of antitumor drugs that are affected and the components of the glycolytic pathway (transporters, enzymes, metabolites) that are involved in the induction of drug resistance. Glycolysis-associated drug resistance occurs in response to stimuli, either cell-autonomous (e.g., oncoproteins) or deriving from the tumor microenvironment (e.g., hypoxia or pseudohypoxia, mechanical cues, etc.). Several mechanisms mediate the induction of drug resistance in response to glycolytic metabolism: inhibition of apoptosis, induction of epithelial-mesenchymal transition, induction of autophagy, inhibition of drug influx and increase of drug efflux. We suggest that drug resistance in response to glycolysis comes into play in presence of qualitative (e.g., expression of embryonic enzyme isoforms, post-translational enzyme modifications) or quantitative (e.g., overexpression of enzymes or overproduction of metabolites) alterations of glycolytic metabolism. We also discern similarities between changes occurring in tumor cells in response to stimuli inducing glycolysis-associated drug resistance and those occurring in cells of the innate immune system in response to danger signals and that have been referred to as danger-associated metabolic modifications. Eventually, we briefly address that also mitochondrial oxidative metabolism may induce drug resistance and discuss the therapeutic implications deriving from the fact that the main energy-generating metabolic pathways may be both at the origin of antitumor drug resistance.
Collapse
Affiliation(s)
- Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.
| | - Cristiano Rumio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| |
Collapse
|
8
|
Cardoso HJ, Carvalho TMA, Fonseca LRS, Figueira MI, Vaz CV, Socorro S. Revisiting prostate cancer metabolism: From metabolites to disease and therapy. Med Res Rev 2020; 41:1499-1538. [PMID: 33274768 DOI: 10.1002/med.21766] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/24/2020] [Accepted: 11/22/2020] [Indexed: 12/24/2022]
Abstract
Prostate cancer (PCa), one of the most commonly diagnosed cancers worldwide, still presents important unmet clinical needs concerning treatment. In the last years, the metabolic reprogramming and the specificities of tumor cells emerged as an exciting field for cancer therapy. The unique features of PCa cells metabolism, and the activation of specific metabolic pathways, propelled the use of metabolic inhibitors for treatment. The present work revises the knowledge of PCa metabolism and the metabolic alterations that underlie the development and progression of the disease. A focus is given to the role of bioenergetic sources, namely, glucose, lipids, and glutamine sustaining PCa cell survival and growth. Moreover, it is described as the action of oncogenes/tumor suppressors and sex steroid hormones in the metabolic reprogramming of PCa. Finally, the status of PCa treatment based on the inhibition of metabolic pathways is presented. Globally, this review updates the landscape of PCa metabolism, highlighting the critical metabolic alterations that could have a clinical and therapeutic interest.
Collapse
Affiliation(s)
- Henrique J Cardoso
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Tiago M A Carvalho
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Lara R S Fonseca
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Marília I Figueira
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Cátia V Vaz
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Sílvia Socorro
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
9
|
Li X, Chen Z, Li Z, Huang G, Lin J, Wei Q, Liang J, Li W. The metabolic role of PFKFB4 in androgen-independent growth in vitro and PFKFB4 expression in human prostate cancer tissue. BMC Urol 2020; 20:61. [PMID: 32487245 PMCID: PMC7268689 DOI: 10.1186/s12894-020-00635-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/22/2020] [Indexed: 01/13/2023] Open
Abstract
Background It is well known that androgen-deprivation therapy (ADT) can inevitably drive prostate cancer (PCa) cells into a castration-resistant state. According to the “Warburg effect”, the metabolism of aggressive tumor cells increases significantly. The growth of cancer cells depends on glycolysis, which may be a potential target for cancer control. 6-Phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 (PFKFB4) plays key roles in the proliferation and metastasis of PCa cells. However, there is very limited knowledge on the role of PFKFB4 in the conversion to castration resistance. The present study aimed to determine the changes in glucose consumption and PFKFB4 expression in LNCaP cells and androgen-independent LNCaP (LNCaP-AI) cells during the whole process of androgen-independent growth. Additionally, PFKFB4 expression in human PCa tissues was evaluated. Methods We established an androgen-independent LNCaP-AI cell line derived from LNCaP cells to mimic the traits of castration resistance in vitro. LNCaP-AI and LNCaP cells were cultured in the corresponding medium containing the same amount of glucose. At the end of experiments, the medium supernatant and blank medium were collected, and absorbance was measured. LNCaP-AI and LNCaP cells were harvested to detect PFKFB4 expression by Western blotting. Prostate tissue samples including PCa tissue, carcinoma-adjacent tissue and benign prostatic hyperplasia (BPH) tissue specimens were evaluated for PFKFB4 expression using immunohistochemistry. Results In 18 h supernatant samples, the glucose consumption and lactate secretion of LNCaP-AI cells were higher than those of LNCaP cells. The Western blot results indicated that PFKFB4 expression was increased in LNCaP-AI cells compared with LNCaP cells. Immunohistochemistry revealed that the expression of PFKFB4 in PCa tissue specimens was higher than that in BPH and adjacent tissue specimens. However, the differences in PCa tissue before and after ADT were not statistically significant. Conclusion PFKFB4 may be associated with enhanced glycolysis during the androgen-independent growth of PCa cells in vitro. PFKFB4 may be a marker of PCa progression. Our results provide a rationale for further clinical investigation of PCa treatment focused on controlling PFKFB4 expression.
Collapse
Affiliation(s)
- Ximing Li
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China.,Graduate School, Guangxi Medical University, Nanning, China
| | - Zhi Chen
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zuan Li
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Guihai Huang
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Junhao Lin
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Qiang Wei
- Department of Pathology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jianbo Liang
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Wei Li
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China.
| |
Collapse
|
10
|
Çoban Ö, Değim Z, Yılmaz Ş, Altıntaş L, Arsoy T, Sözmen M. Efficacy of targeted liposomes and nanocochleates containing imatinib plus dexketoprofen against fibrosarcoma. Drug Dev Res 2019; 80:556-565. [PMID: 30901500 DOI: 10.1002/ddr.21530] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/27/2019] [Accepted: 03/03/2019] [Indexed: 11/06/2022]
Abstract
The main challenges in treating cancer using chemotherapeutics are insufficient dose at the target site and the development of drug resistance, while higher doses can induce side effects by damaging nontarget tissues. Combinatorial drug therapy may overcome these limitations by permitting lower doses and more specific targeting, thereby mitigating drug resistance and nontarget side effects. Recent reports indicate that nonsteroidal anti-inflammatory drugs (NSAIDs) have anticancer potential and can be used together with conventional chemotherapeutics to improve efficacy and safety. In the present study, imatinib mesylate and dexketoprofen trometamol were selected as model drugs to develop targeted surface-modified liposome and nanocochleate formulations for fibrosarcoma treatment. The physicochemical properties and in vitro efficacy of various formulations were evaluated by measurement of particle size distribution, polydispersity index, zeta potential, encapsulation efficiency, diffusion through Caco-2 cells, and toxicity in culture. Selected formulations were then evaluated in fibrosarcoma-bearing model mice by histopathological observations and tyrosine kinase receptor inhibition assays. The most effective formulation on the fibrosarcoma model was a PEGylated nanocochleate formulation. These findings provide a foundation for developing more effective formulations and chemotherapeutic strategies for the treatment of fibrosarcoma and other types of cancer.
Collapse
Affiliation(s)
- Özlem Çoban
- Department of Pharmaceutical Technology, Karadeniz Technical University Faculty of Pharmacy, Trabzon, Turkey
| | - Zelihagül Değim
- Department of Pharmaceutical Technology, Biruni University Faculty of Pharmacy, İstanbul, Turkey
| | - Şükran Yılmaz
- Department of Cell Bank, Food and Mouth Diseases Institute, Ankara, Turkey
| | - Levent Altıntaş
- Department of Pharmacology and Toxicology, Ankara University Faculty of Veterinary Medicine, Ankara, Turkey
| | - Taibe Arsoy
- Department of Cell Bank, Food and Mouth Diseases Institute, Ankara, Turkey
| | - Mahmut Sözmen
- Department of Preclinical Sciences, OndokuzMayıs University, Faculty of Veterinary Medicine, Samsun, Turkey
| |
Collapse
|