1
|
Anjaly K, Tiku AB. Exogenous Treatment of Caffeic Acid and Methylglyoxal Synergistically Enhances Anticancer Effect in Prostate Cancer via Inhibition of Glyoxalase-1. Prostate 2025. [PMID: 39748483 DOI: 10.1002/pros.24849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 11/08/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Caffeic acid (CA), a dietary compound, has been studied for its potential impact on inhibiting prostate cancer (PCa) growth. PCa is often associated with heightened expression of glyoxalase-1 (Glo-1), making it a target for potential therapeutic interventions. CA's mechanisms in suppressing Glo-1 expression and its effects on PCa cell proliferation are areas of interest for understanding its potential as an anticancer agent. METHODS Cellular viability and proliferation were evaluated through MTT and clonogenic assays. The expression levels of particular proteins were assessed using western blot analysis and immunocytochemistry. RESULTS Results indicated significant reduction in PCa cell proliferation by CA, accompanied by induction of DNA double-strand breaks, leading to apoptotic cell death through decreased pro-caspases expression. Additionally, CA was found to inhibit Glo-1 expression. To enhance CA's anticancer effect, a novel approach was taken by combining it with methylglyoxal (MG). Exogenous MG treatment, a glycolysis by-product and glyoxalase enzyme substrate, exhibited dose and time-dependent toxicity in PCa cells when combined with CA. This combination treatment showed heightened toxicity against PCa cells, attributed to CA's inhibition of Glo-1 expression and the nontoxic doses of exogenous MG. Consequently, increased levels of endogenous MG were observed, leading to apoptosis and suggesting a promising strategy for targeting glyoxalase oncogenic signaling pathways in PCa with minimal adverse effects. CONCLUSION The study highlights the potential of CA as a therapeutic agent for inhibiting PCa growth through multiple mechanisms, including the induction of apoptotic cell death and inhibition of Glo-1 expression. Combining CA with MG enhances its anticancer effects, offering a promising strategy for targeting glyoxalase oncogenic pathways in PCa.
Collapse
Affiliation(s)
- Km Anjaly
- Radiation and Cancer Therapeutics Lab, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Ashu Bhan Tiku
- Radiation and Cancer Therapeutics Lab, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
2
|
Matsuoka I, Kasai T, Onaga C, Ozaki A, Motomura H, Maemura Y, Tada Y, Mori H, Hara Y, Xiong Y, Sato K, Tamori S, Sasaki K, Ohno S, Akimoto K. Co‑expression of SLC20A1 and ALDH1A3 is associated with poor prognosis, and SLC20A1 is required for the survival of ALDH1‑positive pancreatic cancer stem cells. Oncol Lett 2024; 28:426. [PMID: 39021737 PMCID: PMC11253103 DOI: 10.3892/ol.2024.14558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 02/23/2024] [Indexed: 07/20/2024] Open
Abstract
Solute carrier family 20 member 1 (SLC20A1) is a sodium/inorganic phosphate symporter, which has been identified as a prognostic marker in several types of cancer, including pancreatic cancer. However, to the best of our knowledge, the association between SLC20A1 expression and cancer stem cell (CSC) markers, such as aldehyde dehydrogenase 1 (ALDH1), in pancreatic ductal adenocarcinoma (PDAC), and the role of SLC20A1 in PDAC CSCs remains unclear. In the present study, a genomic dataset of primary pancreatic cancer (The Cancer Genome Atlas, Pan-Cancer Atlas) was downloaded and analyzed. Kaplan-Meier analysis and multivariate Cox regression analysis were performed to evaluate the overall survival, disease-specific survival (DSS), disease-free interval (DFI) and progression-free interval (PFI). Subsequently, SLC20A1 small interfering RNA (siRNA) knockdown (KD) was induced in the PANC-1 and MIA-PaCa-2 PDAC cell lines, and in sorted high ALDH1 activity (ALDH1high) cells, after which, cell viability, in vitro tumor sphere formation, cell death and caspase-3 activity were examined. The results revealed that patients with high expression of SLC20A1 (SLC20A1 high) at tumor stage I had a poor prognosis compared with patients with low expression of SLC20A1 (SLC20A1 low) in terms of DSS, DFI and PFI. In addition, patients with high expression of SLC20A1 and ALDH1A3 (SLC20A1 high ALDH1A3 high) exhibited poorer clinical outcomes than patients with high expression of SLC20A1 and low expression of ALDH1A3 (SLC20A1 high ALDH1A3 low), low expression of SLC20A1 and high expression of ALDH1A3 (SLC20A1 low ALDH1A3 high) and SLC20A1 low ALDH1A3 low. SLC20A1 siRNA KD in ALDH1high cells isolated from PANC-1 and MIA-PaCa-2 cell lines resulted in suppression of in vitro tumorsphere formation, and enhancement of cell death and caspase-3 activity. These results suggested that SLC20A1 was involved in cell survival via the suppression of caspase-3-dependent apoptosis, and contributed to cancer progression and poor clinical outcomes in PDAC. In conclusion, SLC20A1 may be used as a prognostic marker and novel therapeutic target of ALDH1-positive pancreatic CSCs.
Collapse
Affiliation(s)
- Izumi Matsuoka
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Takahiro Kasai
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Chotaro Onaga
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Ayaka Ozaki
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Hitomi Motomura
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Yuki Maemura
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Yuna Tada
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Haruka Mori
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Yasushi Hara
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Yuyun Xiong
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Keiko Sato
- Research Division of Medical Data Science, Research Institute for Science and Technology, Tokyo University of Science, Noda, Chiba 278-8510, Japan
- Department of Information Sciences, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Shoma Tamori
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
- Research Division of Medical Data Science, Research Institute for Science and Technology, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Kazunori Sasaki
- Laboratory of Cancer Biology, Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Shigeo Ohno
- Laboratory of Cancer Biology, Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Kazunori Akimoto
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
- Research Division of Medical Data Science, Research Institute for Science and Technology, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| |
Collapse
|
3
|
Kim JY, Jung JH, Jung S, Lee S, Lee HA, Ouh YT, Hong SH. Glyoxalase 1: Emerging biomarker and therapeutic target in cervical cancer progression. PLoS One 2024; 19:e0299345. [PMID: 38870176 PMCID: PMC11175447 DOI: 10.1371/journal.pone.0299345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/26/2024] [Indexed: 06/15/2024] Open
Abstract
INTRODUCTION Cervical cancer presents a significant global health challenge, disproportionately impacting underserved populations with limited access to healthcare. Early detection and effective management are vital in addressing this public health concern. This study focuses on Glyoxalase-1 (GLO1), an enzyme crucial for methylglyoxal detoxification, in the context of cervical cancer. METHODS We assessed GLO1 expression in cervical cancer patient samples using immunohistochemistry. In vitro experiments using HeLa cells were conducted to evaluate the impact of GLO1 inhibition on cell viability and migration. Single-cell RNA sequencing (scRNA-seq) and gene set variation analysis were utilized to investigate the role of GLO1 in the metabolism of cervical cancer. Additionally, public microarray data were analyzed to determine GLO1 expression across various stages of cervical cancer. RESULTS Our analysis included 58 cervical cancer patients, and showed that GLO1 is significantly upregulated in cervical cancer tissues compared to normal cervical tissues, independent of pathological findings and disease stage. In vitro experiments indicated that GLO1 inhibition by S-p-bromobenzylglutathione cyclopentyl diester decreased cell viability and migration in cervical cancer cell lines. Analyses of scRNA-seq data and public gene expression datasets corroborated the overexpression of GLO1 and its involvement in cancer metabolism, particularly glycolysis. An examination of expression data from precancerous lesions revealed a progressive increase in GLO1 expression from normal tissue to invasive cervical cancer. CONCLUSIONS This study highlights the critical role of GLO1 in the progression of cervical cancer, presenting it as a potential biomarker and therapeutic target. These findings contribute valuable insights towards personalized treatment approaches and augment the ongoing efforts to combat cervical cancer. Further research is necessary to comprehensively explore GLO1's potential in clinical applications.
Collapse
Affiliation(s)
- Ji-Young Kim
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Ji-Hye Jung
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Soryung Jung
- Department of Life Science, Ewha Womans University, Seoul, Republic of Korea
| | - Sanghyuk Lee
- Department of Life Science, Ewha Womans University, Seoul, Republic of Korea
| | - Hyang Ah Lee
- Department of Obstetrics and Gynecology, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Yung-Taek Ouh
- Department of Obstetrics and Gynecology, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
- Department of Obstetrics and Gynecology, Ansan Hospital, Korea University College of Medicine, Gyeonggi, Republic of Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
- KW-Bio Co., Ltd, Chuncheon, Republic of Korea
| |
Collapse
|
4
|
Alhujaily M. Glyoxalase System in Breast and Ovarian Cancers: Role of MEK/ERK/SMAD1 Pathway. Biomolecules 2024; 14:584. [PMID: 38785990 PMCID: PMC11117840 DOI: 10.3390/biom14050584] [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: 03/26/2024] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024] Open
Abstract
The glyoxalase system, comprising GLO1 and GLO2 enzymes, is integral in detoxifying methylglyoxal (MGO) generated during glycolysis, with dysregulation implicated in various cancer types. The MEK/ERK/SMAD1 signaling pathway, crucial in cellular processes, influences tumorigenesis, metastasis, and angiogenesis. Altered GLO1 expression in cancer showcases its complex role in cellular adaptation and cancer aggressiveness. GLO2 exhibits context-dependent functions, contributing to both proapoptotic and antiapoptotic effects in different cancer scenarios. Research highlights the interconnected nature of these systems, particularly in ovarian cancer and breast cancer. The glyoxalase system's involvement in drug resistance and its impact on the MEK/ERK/SMAD1 signaling cascade underscore their clinical significance. Furthermore, this review delves into the urgent need for effective biomarkers, exemplified in ovarian cancer, where the RAGE-ligand pathway emerges as a potential diagnostic tool. While therapeutic strategies targeting these pathways hold promise, this review emphasizes the challenges posed by context-dependent effects and intricate crosstalk within the cellular milieu. Insights into the molecular intricacies of these pathways offer a foundation for developing innovative therapeutic approaches, providing hope for enhanced cancer diagnostics and tailored treatment strategies.
Collapse
Affiliation(s)
- Muhanad Alhujaily
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| |
Collapse
|
5
|
Alhujaily M. Molecular Assessment of Methylglyoxal-Induced Toxicity and Therapeutic Approaches in Various Diseases: Exploring the Interplay with the Glyoxalase System. Life (Basel) 2024; 14:263. [PMID: 38398772 PMCID: PMC10890012 DOI: 10.3390/life14020263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
This comprehensive exploration delves into the intricate interplay of methylglyoxal (MG) and glyoxalase 1 (GLO I) in various physiological and pathological contexts. The linchpin of the narrative revolves around the role of these small molecules in age-related issues, diabetes, obesity, cardiovascular diseases, and neurodegenerative disorders. Methylglyoxal, a reactive dicarbonyl metabolite, takes center stage, becoming a principal player in the development of AGEs and contributing to cell and tissue dysfunction. The dual facets of GLO I-activation and inhibition-unfold as potential therapeutic avenues. Activators, spanning synthetic drugs like candesartan to natural compounds like polyphenols and isothiocyanates, aim to restore GLO I function. These molecular enhancers showcase promising outcomes in conditions such as diabetic retinopathy, kidney disease, and beyond. On the contrary, GLO I inhibitors emerge as crucial players in cancer treatment, offering new possibilities in diseases associated with inflammation and multidrug resistance. The symphony of small molecules, from GLO I activators to inhibitors, presents a nuanced understanding of MG regulation. From natural compounds to synthetic drugs, each element contributes to a molecular orchestra, promising novel interventions and personalized approaches in the pursuit of health and wellbeing. The abstract concludes with an emphasis on the necessity of rigorous clinical trials to validate these findings and acknowledges the importance of individual variability in the complex landscape of health.
Collapse
Affiliation(s)
- Muhanad Alhujaily
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| |
Collapse
|
6
|
Inoue M, Nakagawa Y, Azuma M, Akahane H, Chimori R, Mano Y, Takasawa R. The PKM2 inhibitor shikonin enhances piceatannol-induced apoptosis of glyoxalase I-dependent cancer cells. Genes Cells 2024; 29:52-62. [PMID: 37963646 PMCID: PMC11448369 DOI: 10.1111/gtc.13084] [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: 06/20/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 11/16/2023]
Abstract
Glyoxalase I (GLO I), a major enzyme involved in the detoxification of the anaerobic glycolytic byproduct methylglyoxal, is highly expressed in various tumors, and is regarded as a promising target for cancer therapy. We recently reported that piceatannol potently inhibits human GLO I and induces the death of GLO I-dependent cancer cells. Pyruvate kinase M2 (PKM2) is also a potential therapeutic target for cancer treatment, so we evaluated the combined anticancer efficacy of piceatannol plus low-dose shikonin, a potent and specific plant-derived PKM2 inhibitor, in two GLO I-dependent cancer cell lines, HL-60 human myeloid leukemia cells and NCI-H522 human non-small-cell lung cancer cells. Combined treatment with piceatannol and low-dose shikonin for 48 h synergistically reduced cell viability, enhanced apoptosis rate, and increased extracellular methylglyoxal accumulation compared to single-agent treatment, but did not alter PKM1, PKM2, or GLO I protein expression. Taken together, these results indicate that concomitant use of low-dose shikonin potentiates piceatannol-induced apoptosis of GLO I-dependent cancer cells by augmenting methylglyoxal accumulation.
Collapse
Affiliation(s)
- Manami Inoue
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Yuki Nakagawa
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Miku Azuma
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Haruka Akahane
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Ryusei Chimori
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Yasunari Mano
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Ryoko Takasawa
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| |
Collapse
|
7
|
Lu KJ, Yang CH, Sheu JR, Chung CL, Jayakumar T, Chen CM, Hsieh CY. Overexpressing glyoxalase 1 attenuates acute hyperglycemia-exacerbated neurological deficits of ischemic stroke in mice. Transl Res 2023; 261:57-68. [PMID: 37419278 DOI: 10.1016/j.trsl.2023.07.002] [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: 02/11/2023] [Revised: 06/22/2023] [Accepted: 07/02/2023] [Indexed: 07/09/2023]
Abstract
Stress-induced hyperglycemia (SIH) is associated with poor functional recovery and high mortality in patients with acute ischemic stroke (AIS). However, intensive controlling of blood glucose by using insulin was not beneficial in patients with AIS and acute hyperglycemia. This study investigated the therapeutic effects of the overexpression of glyoxalase I (GLO1), a detoxifying enzyme of glycotoxins, on acute hyperglycemia-aggravated ischemic brain injury. In the present study, adeno-associated viral (AAV)-mediated GLO1 overexpression reduced infarct volume and edema level but did not improve neurofunctional recovery in the mice with middle cerebral artery occlusion (MCAO). AAV-GLO1 infection significantly enhanced neurofunctional recovery in the MCAO mice with acute hyperglycemia but not in the mice with normoglycemia. Methylglyoxal (MG)-modified proteins expression significantly increased in the ipsilateral cortex of the MCAO mice with acute hyperglycemia. AAV-GLO1 infection attenuated the induction of MG-modified proteins, ER stress formation, and caspase 3/7 activation in MG-treated Neuro-2A cells, and reductions in synaptic plasticity and microglial activation were mitigated in the injured cortex of the MCAO mice with acute hyperglycemia. Treatment with ketotifen, a potent GLO1 stimulator, after surgery, alleviated neurofunctional deficits and ischemic brain damage in the MCAO mice with acute hyperglycemia. Altogether, our data substantiate that, in ischemic brain injury, GLO1 overexpression can alleviate pathologic alterations caused by acute hyperglycemia. Upregulation of GLO1 may be a therapeutic strategy for alleviating SIH-aggravated poor functional outcomes in patients with AIS.
Collapse
Affiliation(s)
- Kuan-Jung Lu
- College of Medicine, Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | - Chih-Hao Yang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Joen-Rong Sheu
- College of Medicine, Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan; Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chi-Li Chung
- Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tanasekar Jayakumar
- Department of Ecology & Environmental Sciences, School of Life Science, Pondicherry University, Kalapet, Puducherry, India
| | - Chieh-Min Chen
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Ying Hsieh
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
8
|
Franco I, Alshalalfa M, Hernandez A, Mahal BA, Nguyen T, Wang L, Punglia R, Swami N, Goel N. Genomic Characterization of Aggressive Breast Cancer in Younger Women. Ann Surg Oncol 2023; 30:7569-7578. [PMID: 37550448 DOI: 10.1245/s10434-023-14080-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 07/20/2023] [Indexed: 08/09/2023]
Abstract
PURPOSE Although breast cancer (BC) risk increases with age, BC in younger women is more aggressive with higher mortality compared with older women. We characterize the genomic landscape of BCs in younger women. METHODS Clinicopathologic, molecular, and genomic differences across age groups (< 40 years, 40-60 years, > 60 years) in female BC patients were investigated in two large cohorts [AACR-GENIE8.1 (n = 11,594) and METABRIC (n = 2509)]. Cox-proportional regression analyzed the prognostic impact of age groups for disease-specific survival (DSS) and recurrence-free survival (RFS) in METABRIC and progression-free survival (PFS) in GENIE cohorts. Chi-squared test was used to assess statistical associations between genomic alterations and age groups. RESULTS Survival analysis showed that women < 40 years had shorter DSS [hazard ratio (HR): 1.52, p = 0.005], RFS (HR: 1.4, p = 0.006), and PFS (HR: 1.82, p = 0.0003) compared with women 40-60 years, and shorter RFS (HR: 1.5, p = 0.001) and PFS (HR: 2.95, p < 0.0001) compared with women > 60 years. Molecular subtypes in the METABRIC cohort showed women < 40 years were enriched with basal, and HER2+ subtypes, and less enriched with luminal A and B subtype (p < 0.0001). Characterization of genomic alterations in both cohorts demonstrated that BCs in women < 40 years were more enriched with TP53 mutations (FDR < 0.0001), BRCA1 mutations (FDR = 0.01), ERBB2 amplifications (FDR < 0.001), CDK12 amplifications (FDR < 0.001), and PPM1D amplifications (FDR < 0.001). In contrast, BCs in older women (> 60 years) were more enriched with PIK3CA, KMT2C, and CDH1 mutations (FDR < 0.0001). CONCLUSIONS BCs in young women are associated with shorter survival and more aggressive genomic features, including mutations in TP53 and BRCA1, and amplifications in ERBB2 and CDK12. These findings have the potential to impact clinical trial design and treatment.
Collapse
Affiliation(s)
- Idalid Franco
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mohammed Alshalalfa
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alexandra Hernandez
- Department of Surgical Oncology, Department of Surgery, University of Miami, Miami, FL, USA
| | - Brandon A Mahal
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Tiffany Nguyen
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lora Wang
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rinaa Punglia
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nishwant Swami
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Neha Goel
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.
- Department of Surgical Oncology, Department of Surgery, University of Miami, Miami, FL, USA.
| |
Collapse
|
9
|
Kasai T, Tamori S, Takasaki Y, Matsuoka I, Ozaki A, Matsuda C, Harada Y, Sasaki K, Ohno S, Akimoto K. High expression of PKCλ and ALDH1A3 indicates a poor prognosis, and PKCλ is required for the asymmetric cell division of ALDH1A3-positive cancer stem cells in PDAC. Biochem Biophys Res Commun 2023; 669:85-94. [PMID: 37267864 DOI: 10.1016/j.bbrc.2023.05.086] [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: 05/09/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the cancer with the poorest prognosis. One of the major properties reflecting its poor prognosis is high-grade heterogeneity, which leads to insensitivity to anticancer treatments. Cancer stem cells (CSCs) acquire phenotypic heterogeneity, generating abnormally differentiated cells by asymmetric cell division. However, the detailed mechanism leading to phenotypic heterogeneity is largely unknown. Here, we showed that PDAC patients with co-upregulation of PKCλ and ALDH1A3 had the poorest clinical outcome. PKCλ knockdown by DsiRNA in the ALDH1high population of PDAC MIA-PaCa-2 cells attenuated the asymmetric distribution of the ALDH1A3 protein. To monitor asymmetric cell division of ALDH1A3-positive PDAC CSCs, we established stable Panc-1 PDAC clones expressing ALDH1A3-turboGFP (Panc-1-ALDH1A3-turboGFP cells). In addition to MIA-PaCa-2-ALDH1high cells, turboGFPhigh cells sorted from Panc-1-ALDH1A3-turboGFP cells showed asymmetric cell propagation of ALDH1A3 protein. PKCλ DsiRNA in Panc-1-ALDH1A3-turboGFP cells also attenuated the asymmetric distribution of ALDH1A3 protein. These results suggest that PKCλ regulates the asymmetric cell division of ALDH1A3-positive PDAC CSCs. Furthermore, Panc-1-ALDH1A3-turboGFP cells can be useful for the visualization and monitoring of CSC properties such as asymmetric cell division of ALDH1A3-positive PDAC CSCs in time-lapse imaging.
Collapse
Affiliation(s)
- Takahiro Kasai
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Shoma Tamori
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan; Research Division of Medical Data Science, Research Institute for Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Yuta Takasaki
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Izumi Matsuoka
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Ayaka Ozaki
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Chika Matsuda
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Yohsuke Harada
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Kazunori Sasaki
- Laboratory of Cancer Biology, Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo, Japan
| | - Shigeo Ohno
- Laboratory of Cancer Biology, Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo, Japan
| | - Kazunori Akimoto
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan; Research Division of Medical Data Science, Research Institute for Science and Technology, Tokyo University of Science, Chiba, Japan.
| |
Collapse
|
10
|
Kim JY, Jung JH, Lee SJ, Han SS, Hong SH. Glyoxalase 1 as a Therapeutic Target in Cancer and Cancer Stem Cells. Mol Cells 2022; 45:869-876. [PMID: 36172978 PMCID: PMC9794553 DOI: 10.14348/molcells.2022.0109] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 01/11/2023] Open
Abstract
Methylglyoxal (MG) is a dicarbonyl compound formed in cells mainly by the spontaneous degradation of the triose phosphate intermediates of glycolysis. MG is a powerful precursor of advanced glycation end products, which lead to strong dicarbonyl and oxidative stress. Although divergent functions of MG have been observed depending on its concentration, MG is considered to be a potential anti-tumor factor due to its cytotoxic effects within the oncologic domain. MG detoxification is carried out by the glyoxalase system. Glyoxalase 1 (Glo1), the ubiquitous glutathione-dependent enzyme responsible for MG degradation, is considered to be a tumor promoting factor due to it catalyzing the removal of cytotoxic MG. Indeed, various cancer types exhibit increased expression and activity of Glo1 that closely correlate with tumor cell growth and metastasis. Furthermore, mounting evidence suggests that Glo1 contributes to cancer stem cell survival. In this review, we discuss the role of Glo1 in the malignant progression of cancer and its possible use as a promising therapeutic target for tumor therapy. We also summarize therapeutic outcomes of Glo1 inhibitors as prospective treatments for the prevention of cancer.
Collapse
Affiliation(s)
- Ji-Young Kim
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Ji-Hye Jung
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Seung-Joon Lee
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Seon-Sook Han
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
- Institute of Medical Science, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
- KW-Bio Co., Ltd., Wonju 26487, Korea
| |
Collapse
|
11
|
Lai SWT, Lopez Gonzalez EDJ, Zoukari T, Ki P, Shuck SC. Methylglyoxal and Its Adducts: Induction, Repair, and Association with Disease. Chem Res Toxicol 2022; 35:1720-1746. [PMID: 36197742 PMCID: PMC9580021 DOI: 10.1021/acs.chemrestox.2c00160] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Metabolism is an essential part of life that provides energy for cell growth. During metabolic flux, reactive electrophiles are produced that covalently modify macromolecules, leading to detrimental cellular effects. Methylglyoxal (MG) is an abundant electrophile formed from lipid, protein, and glucose metabolism at intracellular levels of 1-4 μM. MG covalently modifies DNA, RNA, and protein, forming advanced glycation end products (MG-AGEs). MG and MG-AGEs are associated with the onset and progression of many pathologies including diabetes, cancer, and liver and kidney disease. Regulating MG and MG-AGEs is a potential strategy to prevent disease, and they may also have utility as biomarkers to predict disease risk, onset, and progression. Here, we review recent advances and knowledge surrounding MG, including its production and elimination, mechanisms of MG-AGEs formation, the physiological impact of MG and MG-AGEs in disease onset and progression, and the latter in the context of its receptor RAGE. We also discuss methods for measuring MG and MG-AGEs and their clinical application as prognostic biomarkers to allow for early detection and intervention prior to disease onset. Finally, we consider relevant clinical applications and current therapeutic strategies aimed at targeting MG, MG-AGEs, and RAGE to ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Seigmund Wai Tsuen Lai
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California 91010, United States
| | - Edwin De Jesus Lopez Gonzalez
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California 91010, United States
| | - Tala Zoukari
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California 91010, United States
| | - Priscilla Ki
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California 91010, United States
| | - Sarah C Shuck
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California 91010, United States
| |
Collapse
|
12
|
Opławski M, Średnicka A, Niewiadomska E, Boroń D, Januszyk P, Grabarek BO. Clinical and molecular evaluation of patients with ovarian cancer in the context of drug resistance to chemotherapy. Front Oncol 2022; 12:954008. [PMID: 35992817 PMCID: PMC9389532 DOI: 10.3389/fonc.2022.954008] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
The present study aimed to evaluate changes in the expression patterns at the gene and protein levels associated with drug resistance. The study group included 48 women who had a histopathologically confirmed diagnosis of stage I-IV ovarian cancer, they were divided into two subgroups (groups A and B). In group A, there were 36 patients in whom surgical treatment was supplemented with first-line chemotherapy according to current standards. Within this patient group, 5 had stage I (14%), 5 had stage II (14%), 25 had stage III (69%), and 1 had stage IV ovarian cancer (3%). Drug resistance was found after the third cycle of chemotherapy in 17 patients (71%) and after the sixth cycle in 7 patients (29%). Group B included 12 women with type I ovarian cancer, including 11 with stage I and 1 patient with stage IV ovarian cancer. The oncological treatment required only surgery. The control group (C) included 50 women in whom the uterus and adnexa were surgically removed for non-oncological reasons. Significantly higher levels of carcinoma antigen 125 CA-125 and human epididymis protein 4 HE4 were observed in group A and in menopausal women. Moreover, drug resistance was associated with significantly higher levels of CA-125 (p < 0.05). The genes UBA2, GLO1, STATH, and TUFT1 were differentiated in test samples from control samples. Moreover, drug resistance was associated with significantly higher expression of GLO1. The results of these assessments indicated the strong link between UBA2 and hsa-miR-133a-3p and hsa-miR-133b; GLO1 and hsa-miR-561-5p; STATH and hsa-miR-137-3p and hsa-miR-580-3p; and TUFT1 and hsa-miR-1233-3p and hsa-miR-2052. Correlation analysis showed a significant correlation between CA-125 and HE4 levels. Moreover, a significant correlation between TUFT1 mRNA and UBA2, GLO1, STATH (negative correlation), and TUFT1 in relation to CA-125 and HE4 (p < 0.05) was noted in all patients. In view of the lack of screening tests for ovarian cancer, the occurrence of the described correlation may be inscribed as an attempt to establish an assay that meets the criteria of a screening test and thus increase the early diagnosis of ovarian cancer.
Collapse
Affiliation(s)
- Marcin Opławski
- Department of Gynecology and Obstetrics with Gynecologic Oncology, Ludwik Rydygier Memorial Specialized Hospital, Kraków, Poland
- Department of Gynecology and Obstetrics, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski University in Kraków, Kraków, Poland
| | - Agata Średnicka
- Department of Gynecology and Obstetrics with Gynecologic Oncology, Ludwik Rydygier Memorial Specialized Hospital, Kraków, Poland
| | - Ewa Niewiadomska
- Department of Epidemiology and Biostatistics, School of Health Sciences in Bytom, Medical University of Silesia, Katowice, Poland
| | - Dariusz Boroń
- Department of Gynecology and Obstetrics with Gynecologic Oncology, Ludwik Rydygier Memorial Specialized Hospital, Kraków, Poland
- Department of Histology, Cytophysiology and Embryology, Faculty of Medicine, University of Technology, Academia of Silesia in Katowice, Zabrze, Poland
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Technology, Academia of Silesia in Katowice, Zabrze, Poland
| | - Piotr Januszyk
- Department of Biochemistry, Faculty of Medicine, University of Technology, Academia of Silesia in Katowice, Zabrze, Poland
| | - Beniamin Oskar Grabarek
- Department of Gynecology and Obstetrics with Gynecologic Oncology, Ludwik Rydygier Memorial Specialized Hospital, Kraków, Poland
- Department of Histology, Cytophysiology and Embryology, Faculty of Medicine, University of Technology, Academia of Silesia in Katowice, Zabrze, Poland
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Technology, Academia of Silesia in Katowice, Zabrze, Poland
- GynCentrum, Laboratory of Molecular Biology and Virology, Katowice, Poland
| |
Collapse
|
13
|
Wang J, Yang X, Wang Z, Wang J. Role of the Glyoxalase System in Breast Cancer and Gynecological Cancer-Implications for Therapeutic Intervention: a Review. Front Oncol 2022; 12:857746. [PMID: 35898868 PMCID: PMC9309216 DOI: 10.3389/fonc.2022.857746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 06/17/2022] [Indexed: 12/24/2022] Open
Abstract
Methyglyoxal (MGO), an essential endogenous dicarbonyl metabolite, can lead to multiple physiological problems including hyperglycemia, kidney diseases, malignant tumors, beyond its normal concentration range. The glyoxalase system, making MGO maintained at a low level, links glycation to carcinogenesis, growth, metastasis, and cancer chemotherapy. The glyoxalase system comprises glyoxalase 1 (Glo1) and glyoxalase 2 (Glo2), which is often overexpressed in various tumor tissues. However, very little is known about the glyoxalase system in breast cancer and gynecological cancer. In this review, we introduce the role of the glyoxalase system in breast cancer, endometrial cancer, ovarian cancer and cervical cancer, and highlight the potential of the glyoxalase system to be both as a marker for diagnosis and a novel target for antitumor therapy. However, the intrinsic molecular biology and mechanisms of the glyoxalase system in breast cancer and gynecological cancer need further exploration.
Collapse
|
14
|
Onaga C, Tamori S, Matsuoka I, Ozaki A, Motomura H, Nagashima Y, Sato T, Sato K, Tahata K, Xiong Y, Nakano Y, Mano Y, Miyazaki S, Sasaki K, Ohno S, Akimoto K. High SLC20A1 Expression Is Associated With Poor Prognosis for Radiotherapy of Estrogen Receptor-positive Breast Cancer. CANCER DIAGNOSIS & PROGNOSIS 2022; 2:429-442. [PMID: 35813014 PMCID: PMC9254097 DOI: 10.21873/cdp.10126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 05/25/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND/AIM Radiotherapy is one of the main treatments for estrogen receptor-positive (ER+) breast cancer. However, in some ER+ breast cancer cases, radiotherapy is insufficient to inhibit progression and there is a lack of markers to predict radiotherapy insensitivity. Solute carrier family 20 member 1 (SLC20A1) is a sodium/inorganic phosphate symporter, which has been proposed to be a viable prognostic marker for luminal A and B types of ER+ breast cancer. The present study examined the possibility of SLC20A1 as a novel biomarker for the prediction of radiotherapy efficiency. PATIENTS AND METHODS The Molecular Taxonomy of Breast Cancer International Consortium dataset was downloaded from cBioportal and the prognosis of patients with high SLC20A1 expression (SLC20A1 high ) was compared with that of patients with low SLC20A1 expression, without or with radiotherapy and tumor stages I, II, and III, using the Kaplan-Meier method and multivariate Cox regression analyses of disease-specific and relapse-free survival. RESULTS Patients in the SLC20A1 high group with radiotherapy showed poor clinical outcomes in both luminal A and luminal B breast cancers. Furthermore, in luminal A breast cancer at tumor stage I, patients in the SLC20A1 high group with radiotherapy also showed poor clinical outcomes. Therefore, these results suggest that radiotherapy is insufficient for patients in the SLC20A1 high group for both luminal A and B types, and especially for the luminal A type at tumor stage I. CONCLUSION SLC20A1 can be used as a prognostic marker for the prediction of the efficacy of radiotherapy for luminal A and luminal B breast cancers.
Collapse
Affiliation(s)
- Chotaro Onaga
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Shoma Tamori
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Izumi Matsuoka
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Ayaka Ozaki
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Hitomi Motomura
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Yuka Nagashima
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Tsugumichi Sato
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Keiko Sato
- Department of Information Sciences, Faculty of Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Kouji Tahata
- Department of Information Sciences, Faculty of Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Yuyun Xiong
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Yoshio Nakano
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Yasunari Mano
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Satoru Miyazaki
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Kazunori Sasaki
- Laboratory of Cancer Biology, Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo, Japan
| | - Shigeo Ohno
- Laboratory of Cancer Biology, Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo, Japan
| | - Kazunori Akimoto
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| |
Collapse
|
15
|
Onaga C, Tamori S, Matsuoka I, Ozaki A, Motomura H, Nagashima Y, Sato T, Sato K, Xiong Y, Sasaki K, Ohno S, Akimoto K. High expression of SLC20A1 is less effective for endocrine therapy and predicts late recurrence in ER-positive breast cancer. PLoS One 2022; 17:e0268799. [PMID: 35605014 PMCID: PMC9126382 DOI: 10.1371/journal.pone.0268799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/08/2022] [Indexed: 12/02/2022] Open
Abstract
Estrogen receptor-positive (ER+) breast cancer intrinsically confers satisfactory clinical outcomes in response to endocrine therapy. However, a significant proportion of patients with ER+ breast cancer do not respond well to this treatment. Therefore, to evaluate the effects of endocrine therapy, there is a need for identification of novel markers that can be used at the time of diagnosis for predicting clinical outcomes, especially for early-stage and late recurrence. Solute carrier family 20 member 1 (SLC20A1) is a sodium/inorganic phosphate symporter that has been proposed to be a viable prognostic marker for the luminal A and luminal B types of ER+ breast cancer. In the present study, we examined the possible association of SLC20A1 expression with tumor staging, endocrine therapy and chemotherapy in the luminal A and luminal B subtypes of breast cancer. In addition, we analyzed the relationship between SLC20A1 expression and late recurrence in patients with luminal A and luminal B breast cancer following endocrine therapy. We showed that patients with higher levels of SLC20A1 expression (SLC20A1high) exhibited poorer clinical outcomes in those with tumor stage I luminal A breast cancer. In addition, this SLC20A1high subgroup of patients exhibited less responses to endocrine therapy, specifically in those with the luminal A and luminal B subtypes of breast cancer. However, patients with SLC20A1high showed good clinical outcomes following chemotherapy. Patients tested to be in the SLC20A1high group at the time of diagnosis also showed a higher incidence of recurrence compared with those with lower expression levels of SLC20A1, at >15 years for luminal A breast cancer and at 10–15 years for luminal B breast cancer. Therefore, we conclude that SLC20A1high can be used as a prognostic biomarker for predicting the efficacy of endocrine therapy and late recurrence for ER+ breast cancer.
Collapse
Affiliation(s)
- Chotaro Onaga
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Shoma Tamori
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Izumi Matsuoka
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Ayaka Ozaki
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Hitomi Motomura
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Yuka Nagashima
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Tsugumichi Sato
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Keiko Sato
- Department of Information Sciences, Faculty of Sciences and Technology, Tokyo University of Science, Chiba, Japan
| | - Yuyun Xiong
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Kazunori Sasaki
- Laboratory of Cancer Biology, Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo, Japan
| | - Shigeo Ohno
- Laboratory of Cancer Biology, Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo, Japan
| | - Kazunori Akimoto
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
- * E-mail:
| |
Collapse
|
16
|
Usami M, Ando K, Shibuya A, Takasawa R, Yokoyama H. Crystal structures of human glyoxalase I and its complex with TLSC702 reveal inhibitor binding mode and substrate preference. FEBS Lett 2022; 596:1458-1467. [PMID: 35363883 DOI: 10.1002/1873-3468.14344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 11/11/2022]
Abstract
Human glyoxalase I (hGLO I) is an enzyme for detoxification of methylglyoxal (MG), and has been considered an attractive target for the development of new anti-cancer drugs. In our previous report, the GLO I inhibitor TLSC702 induced apoptosis in tumor cells. Here, we determined the crystal structures of hGLO I and its complex with TLSC702. In the complex, the carboxy O atom of TLSC702 is coordinated to Zn2+ , and TLSC702 mainly shows van der Waals interaction with hydrophobic residues. In the inhibitor-unbound structure, glycerol, which has similar functional groups to MG, was bound to Zn2+ , indicating that GLO I can easily bind to MG. This study provides a structural basis to develop better anticancer drugs.
Collapse
Affiliation(s)
- Midori Usami
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Koki Ando
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Asuka Shibuya
- 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
| | - Hideshi Yokoyama
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| |
Collapse
|
17
|
Abdul Kader S, Dib S, Achkar IW, Thareja G, Suhre K, Rafii A, Halama A. Defining the landscape of metabolic dysregulations in cancer metastasis. Clin Exp Metastasis 2021; 39:345-362. [PMID: 34921655 PMCID: PMC8971193 DOI: 10.1007/s10585-021-10140-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/10/2021] [Indexed: 02/07/2023]
Abstract
Metastasis is the primary cause of cancer related deaths due to the limited number of efficient druggable targets. Signatures of dysregulated cancer metabolism could serve as a roadmap for the determination of new treatment strategies. However, the metabolic signatures of metastatic cells remain vastly elusive. Our aim was to determine metabolic dysregulations associated with high metastatic potential in breast cancer cell lines. We have selected 5 triple negative breast cancer (TNBC) cell lines including three with high metastatic potential (HMP) (MDA-MB-231, MDA-MB-436, MDA-MB-468) and two with low metastatic potential (LMP) (BT549, HCC1143). The normal epithelial breast cell line (hTERT-HME1) was also investigated. The untargeted metabolic profiling of cells and growth media was conducted and total of 479 metabolites were quantified. First we characterized metabolic features differentiating TNBC cell lines from normal cells as well as identified cell line specific metabolic fingerprints. Next, we determined 92 metabolites in cells and 22 in growth medium that display significant differences between LMP and HMP. The HMP cell lines had elevated level of molecules involved in glycolysis, TCA cycle and lipid metabolism. We identified metabolic advantages of cell lines with HMP beyond enhanced glycolysis by pinpointing the role of branched chain amino acids (BCAA) catabolism as well as molecules supporting coagulation and platelet activation as important contributors to the metastatic cascade. The landscape of metabolic dysregulations, characterized in our study, could serve as a roadmap for the identification of treatment strategies targeting cancer cells with enhanced metastatic potential.
Collapse
Affiliation(s)
- Sara Abdul Kader
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, 24144, Doha, Qatar
- University of Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Shaima Dib
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, 24144, Doha, Qatar
| | - Iman W Achkar
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, 24144, Doha, Qatar
| | - Gaurav Thareja
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, 24144, Doha, Qatar
- University of Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, 24144, Doha, Qatar
- Department of Biophysics and Physiology, Weill Cornell Medicine, New York, USA
| | - Arash Rafii
- Department of Genetic Medicine, Weill Cornell Medicine, New York, USA
- Genetic Intelligence Laboratory, Weill Cornell Medicine in Qatar, Qatar Foundation, Doha, Qatar
| | - Anna Halama
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, 24144, Doha, Qatar.
- Department of Biophysics and Physiology, Weill Cornell Medicine, New York, USA.
| |
Collapse
|
18
|
Motomura H, Ozaki A, Tamori S, Onaga C, Nozaki Y, Waki Y, Takasawa R, Yoshizawa K, Mano Y, Sato T, Sasaki K, Ishiguro H, Miyagi Y, Nagashima Y, Yamamoto K, Sato K, Hanawa T, Tanuma SI, Ohno S, Akimoto K. Glyoxalase 1 and protein kinase Cλ as potential therapeutic targets for late-stage breast cancer. Oncol Lett 2021; 22:547. [PMID: 34093768 PMCID: PMC8170180 DOI: 10.3892/ol.2021.12808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 03/23/2021] [Indexed: 01/20/2023] Open
Abstract
Cancer cells upregulate the expression levels of glycolytic enzymes in order to reach the increased glycolysis required. One such upregulated glycolytic enzyme is glyoxalase 1 (GLO 1), which catalyzes the conversion of toxic methylglyoxal to nontoxic S-D-lactoylglutathione. Protein kinase Cλ (PKCλ) is also upregulated in various types of cancer and is involved in cancer progression. In the present study, the association between enhanced glycolysis and PKCλ in breast cancer was investigated. In human breast cancer, high GLO 1 expression was associated with high PKCλ expression at the protein (P<0.01) and mRNA levels (P<0.01). Furthermore, Wilcoxon and Cox regression model analysis revealed that patients with stage III-IV tumors with high GLO 1 and PKCλ expression had poor overall survival compared with patients expressing lower levels of these genes [P=0.040 (Gehan-Breslow generalized Wilcoxon test) and P=0.031 (hazard ratio, 2.36; 95% confidence interval, 1.08-5.16), respectively]. Treatment of MDA-MB-157 and MDA-MB-468 human basal-like breast cancer cells with TLSC702 (a GLO 1 inhibitor) and/or aurothiomalate (a PKCλ inhibitor) reduced both cell viability and tumor-sphere formation. These results suggested that GLO 1 and PKCλ were cooperatively involved in cancer progression and contributed to a poor prognosis in breast cancer. In conclusion, GLO 1 and PKCλ serve as potentially effective therapeutic targets for treatment of late-stage human breast cancer.
Collapse
Affiliation(s)
- Hitomi Motomura
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan
| | - Ayaka Ozaki
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan
| | - Shoma Tamori
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan
| | - Chotaro Onaga
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan
| | - Yuka Nozaki
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan
| | - Yuko Waki
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan
| | - Ryoko Takasawa
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan
| | - Kazumi Yoshizawa
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan
| | - Yasunari Mano
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan
| | - Tsugumichi Sato
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan
| | - Kazunori Sasaki
- Department of Molecular Biology, Yokohama City University, School of Medicine, Kanagawa 236-0004, Japan
| | - Hitoshi Ishiguro
- Department of Urology, Yokohama City University Graduate School of Medicine, Kanagawa 236-0004, Japan
- Photocatalyst Group, Research and Development Department, Kanagawa Institute of Industrial Science and Technology, Kanagawa 210-0821, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Kanagawa 241-8515, Japan
| | - Yoji Nagashima
- Department of Surgical Pathology, Tokyo Women's Medical University Hospital, Tokyo 162-8666, Japan
| | - Kouji Yamamoto
- Department of Biostatistics, Yokohama City University, School of Medicine, Kanagawa 236-0004, Japan
| | - Keiko Sato
- Department of Information Sciences, Faculty of Science and Technology, Tokyo University of Science, Chiba 278-8510, Japan
| | - Takehisa Hanawa
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan
| | - Sei-Ichi Tanuma
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan
- Department of Genomic Medicinal Science, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, Chiba 278-8510, Japan
| | - Shigeo Ohno
- Department of Molecular Biology, Yokohama City University, School of Medicine, Kanagawa 236-0004, Japan
- Department of Cancer Biology, Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Kazunori Akimoto
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan
| |
Collapse
|
19
|
Addition of hydrophobic side chains improve the apoptosis inducibility of the human glyoxalase I inhibitor, TLSC702. Bioorg Med Chem Lett 2021; 40:127918. [PMID: 33711442 DOI: 10.1016/j.bmcl.2021.127918] [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: 10/12/2020] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 11/22/2022]
Abstract
Glyoxalase I (GLO I) is a known therapeutic target in cancer. Even though TLSC702, a GLO I inhibitor that we discovered, induces apoptosis in tumor cells, exceptionally higher doses are required compared with those needed to inhibit GLO I activity in vitro. In this work, structure-activity optimization studies were conducted on four sections of the TLSC702 molecule to determine the partial structural features necessary for the inhibition of GLO I. Herein, we found that the carboxy group in TLSC702 was critical for binding with the divalent zinc at the active site of GLO I. In contrast, the side chain substituents in the meta- and para- positions of the benzene ring had little influence on the in vitro inhibition of GLO I. The CLogP values of the TLSC702 derivatives showed a positive correlation with the antiproliferative effects on NCI-H522 cells. Thus, two derivatives of TLSC702, which displayed either high or low lipophilicity due to the types of substituents at the phenyl position, were selected. Even though both derivatives showed comparable inhibitory effects as that of their parent compound, the derivative with the high CLogP value was distinctly more antiproliferative than TLSC702. In contrast, the derivative with the low CLogP value did not decrease cell viability in NCI-H522 and HL-60 cells. These findings suggested that structural improvements, such as the addition of hydrophobic moieties to the phenyl group, enhanced the ability of TLSC702 to induce apoptosis by increasing cell membrane permeability.
Collapse
|
20
|
Leone A, Nigro C, Nicolò A, Prevenzano I, Formisano P, Beguinot F, Miele C. The Dual-Role of Methylglyoxal in Tumor Progression - Novel Therapeutic Approaches. Front Oncol 2021; 11:645686. [PMID: 33869040 PMCID: PMC8044862 DOI: 10.3389/fonc.2021.645686] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
One of the hallmarks of cancer cells is their metabolic reprogramming, which includes the preference for the use of anaerobic glycolysis to produce energy, even in presence of normal oxygen levels. This phenomenon, known as “Warburg effect”, leads to the increased production of reactive intermediates. Among these Methylglyoxal (MGO), a reactive dicarbonyl known as the major precursor of the advanced glycated end products (AGEs), is attracting great attention. It has been well established that endogenous MGO levels are increased in several types of cancer, however the MGO contribution in tumor progression is still debated. Although an anti-cancer role was initially attributed to MGO due to its cytotoxicity, emerging evidence has highlighted its pro-tumorigenic role in several types of cancer. These apparently conflicting results are explained by the hormetic potential of MGO, in which lower doses of MGO are able to establish an adaptive response in cancer cells while higher doses cause cellular apoptosis. Therefore, the extent of MGO accumulation and the tumor context are crucial to establish MGO contribution to cancer progression. Several therapeutic approaches have been proposed and are currently under investigation to inhibit the pro-tumorigenic action of MGO. In this review, we provide an overview of the early and latest evidence regarding the role of MGO in cancer, in order to define its contribution in tumor progression, and the therapeutic strategies aimed to counteract the tumor growth.
Collapse
Affiliation(s)
- Alessia Leone
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Cecilia Nigro
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Antonella Nicolò
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Immacolata Prevenzano
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Pietro Formisano
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Francesco Beguinot
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Claudia Miele
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| |
Collapse
|
21
|
Jandova J, Wondrak GT. Genomic GLO1 deletion modulates TXNIP expression, glucose metabolism, and redox homeostasis while accelerating human A375 malignant melanoma tumor growth. Redox Biol 2021; 39:101838. [PMID: 33360689 PMCID: PMC7772567 DOI: 10.1016/j.redox.2020.101838] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/29/2020] [Accepted: 12/02/2020] [Indexed: 12/26/2022] Open
Abstract
Glyoxalase 1 (encoded by GLO1) is a glutathione-dependent enzyme detoxifying the glycolytic byproduct methylglyoxal (MG), an oncometabolite involved in metabolic reprogramming. Recently, we have demonstrated that GLO1 is overexpressed in human malignant melanoma cells and patient tumors and substantiated a novel role of GLO1 as a molecular determinant of invasion and metastasis in melanoma. Here, employing NanoString™ gene expression profiling (nCounter™ 'PanCancer Progression Panel'), we report that CRISPR/Cas 9-based GLO1 deletion from human A375 malignant melanoma cells alters glucose metabolism and redox homeostasis, observable together with acceleration of tumorigenesis. Nanostring™ analysis identified TXNIP (encoding thioredoxin-interacting protein), a master regulator of cellular energy metabolism and redox homeostasis, displaying the most pronounced expression change in response to GLO1 elimination, confirmed by RT-qPCR and immunoblot analysis. TXNIP was also upregulated in CRISPR/Cas9-engineered DU145 prostate carcinoma cells lacking GLO1, and treatment with MG or a pharmacological GLO1 inhibitor (TLSC702) mimicked GLO1_KO status, suggesting that GLO1 controls TXNIP expression through regulation of MG. GLO1_KO status was characterized by (i) altered oxidative stress response gene expression, (ii) attenuation of glucose uptake and metabolism with downregulation of gene expression (GLUT1, GFAT1, GFAT2, LDHA) and depletion of related key metabolites (glucose-6-phosphate, UDP-N-acetylglucosamine), and (iii) immune checkpoint modulation (PDL1). While confirming our earlier finding that GLO1 deletion limits invasion and metastasis with modulation of EMT-related genes (e.g. TGFBI, MMP9, ANGPTL4, TLR4, SERPINF1), we observed that GLO1_KO melanoma cells displayed a shortened population doubling time, cell cycle alteration with increased M-phase population, and enhanced anchorage-independent growth, a phenotype supported by expression analysis (CXCL8, CD24, IL1A, CDKN1A). Concordantly, an accelerated growth rate of GLO1_KO tumors, accompanied by TXNIP overexpression and metabolic reprogramming, was observable in a SCID mouse melanoma xenograft model, demonstrating that A375 melanoma tumor growth and metastasis can be dysregulated in opposing ways as a consequence of GLO1 elimination.
Collapse
Affiliation(s)
- Jana Jandova
- Department of Pharmacology and Toxicology, College of Pharmacy and UA Cancer Center, University of Arizona, Tucson, AZ, USA
| | - Georg T Wondrak
- Department of Pharmacology and Toxicology, College of Pharmacy and UA Cancer Center, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
22
|
Shan NL, Shin Y, Yang G, Furmanski P, Suh N. Breast cancer stem cells: A review of their characteristics and the agents that affect them. Mol Carcinog 2021; 60:73-100. [PMID: 33428807 DOI: 10.1002/mc.23277] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
The evolving concept that cancer stem cells (CSCs) are the driving element in cancer development, evolution and heterogeneity, has overridden the previous model of a tumor consisting of cells all with similar sequentially acquired mutations and a similar potential for renewal, invasion and metastasis. This paradigm shift has focused attention on therapeutically targeting CSCs directly as a means of eradicating the disease. In breast cancers, CSCs can be identified by cell surface markers and are characterized by their ability to self-renew and differentiate, resist chemotherapy and radiation, and initiate new tumors upon serial transplantation in xenografted mice. These functional properties of CSCs are regulated by both intracellular and extracellular factors including pluripotency-related transcription factors, intracellular signaling pathways and external stimuli. Several classes of natural products and synthesized compounds have been studied to target these regulatory elements and force CSCs to lose stemness and/or terminally differentiate and thereby achieve a therapeutic effect. However, realization of an effective treatment for breast cancers, focused on the biological effects of these agents on breast CSCs, their functions and signaling, has not yet been achieved. In this review, we delineate the intrinsic and extrinsic factors identified to date that control or promote stemness in breast CSCs and provide a comprehensive compilation of potential agents that have been studied to target breast CSCs, transcription factors and stemness-related signaling. Our aim is to stimulate further study of these agents that could become the basis for their use as stand-alone treatments or components of combination therapies effective against breast cancers.
Collapse
Affiliation(s)
- Naing L Shan
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Yoosub Shin
- Yonsei University, College of Medicine, Seoul, Republic of Korea
| | - Ge Yang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Philip Furmanski
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Nanjoo Suh
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| |
Collapse
|
23
|
High PKCλ expression is required for ALDH1-positive cancer stem cell function and indicates a poor clinical outcome in late-stage breast cancer patients. PLoS One 2020; 15:e0235747. [PMID: 32658903 PMCID: PMC7357771 DOI: 10.1371/journal.pone.0235747] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 06/22/2020] [Indexed: 12/19/2022] Open
Abstract
Despite development of markers for identification of cancer stem cells, the mechanism underlying the survival and division of cancer stem cells in breast cancer remains unclear. Here we report that PKCλ expression was enriched in basal-like breast cancer, among breast cancer subtypes, and was correlated with ALDH1A3 expression (p = 0.016, χ2-test). Late stage breast cancer patients expressing PKCλhigh and ALDH1A3high had poorer disease-specific survival than those expressing PKCλlow and ALDH1A3low (p = 0.018, log rank test for Kaplan-Meier survival curves: hazard ratio 2.58, 95% CI 1.24–5.37, p = 0.011, multivariate Cox regression analysis). Functional inhibition of PKCλ through siRNA-mediated knockdown or CRISPR-Cas9-mediated knockout in ALDH1high MDA-MB 157 and MDA-MB 468 basal-like breast cancer cells led to increases in the numbers of trypan blue-positive and active-caspase 3-positive cells, as well as suppression of tumor-sphere formation and cell migration. Furthermore, the amount of CASP3 and PARP mRNA and the level of cleaved caspase-3 protein were enhanced in PKCλ-deficient ALDH1high cells. An Apoptosis inhibitor (z-VAD-FMK) suppressed the enhancement of cell death as well as the levels of cleaved caspase-3 protein in PKCλ deficient ALDH1high cells. It also altered the asymmetric/symmetric distribution ratio of ALDH1A3 protein. In addition, PKCλ knockdown led to increases in cellular ROS levels in ALDH1high cells. These results suggest that PKCλ is essential for cancer cell survival and migration, tumorigenesis, the asymmetric distribution of ALDH1A3 protein among cancer cells, and the maintenance of low ROS levels in ALDH1-positive breast cancer stem cells. This makes it a key contributor to the poorer prognosis seen in late-stage breast cancer patients.
Collapse
|
24
|
Jandova J, Perer J, Hua A, Snell JA, Wondrak GT. Genetic Target Modulation Employing CRISPR/Cas9 Identifies Glyoxalase 1 as a Novel Molecular Determinant of Invasion and Metastasis in A375 Human Malignant Melanoma Cells In Vitro and In Vivo. Cancers (Basel) 2020; 12:E1369. [PMID: 32466621 PMCID: PMC7352620 DOI: 10.3390/cancers12061369] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/15/2020] [Accepted: 05/22/2020] [Indexed: 12/22/2022] Open
Abstract
Metabolic reprogramming is a molecular hallmark of cancer. Recently, we have reported the overexpression of glyoxalase 1 (encoded by GLO1), a glutathione-dependent enzyme involved in detoxification of the reactive glycolytic byproduct methylglyoxal, in human malignant melanoma cell culture models and clinical samples. However, the specific role of GLO1 in melanomagenesis remains largely unexplored. Here, using genetic target modulation, we report the identification of GLO1 as a novel molecular determinant of invasion and metastasis in malignant melanoma. First, A375 human malignant melanoma cells with GLO1 deletion (A375-GLO1_KO) were engineered using CRISPR/Cas9, and genetic rescue clones were generated by stable transfection of KO clones employing a CMV-driven GLO1 construct (A375-GLO1_R). After confirming GLO1 target modulation at the mRNA and protein levels (RT-qPCR, immunodetection, enzymatic activity), phenotypic characterization indicated that deletion of GLO1 does not impact proliferative capacity while causing significant sensitization to methylglyoxal-, chemotherapy-, and starvation-induced cytotoxic stress. Employing differential gene expression array analysis (A375-GLO1_KO versus A375-GLO1_WT), pronounced modulation of epithelial--mesenchymal transition (EMT)-related genes [upregulated: CDH1, OCLN, IL1RN, PDGFRB, SNAI3; (downregulated): BMP1, CDH2, CTNNB1, FN1, FTH1, FZD7, MELTF, MMP2, MMP9, MYC, PTGS2, SNAI2, TFRC, TWIST1, VIM, WNT5A, ZEB1, and ZEB2 (up to tenfold; p < 0.05)] was observed-all of which are consistent with EMT suppression as a result of GLO1 deletion. Importantly, these expression changes were largely reversed upon genetic rescue employing A375-GLO1_R cells. Differential expression of MMP9 as a function of GLO1 status was further substantiated by enzymatic activity and ELISA analysis; phenotypic assessment revealed the pronounced attenuation of morphological potential, transwell migration, and matrigel 3D-invasion capacity displayed by A375-GLO1_KO cells, reversed again in genetic rescue clones. Strikingly, in a SCID mouse metastasis model, lung tumor burden imposed by A375-GLO1_KO cells was strongly attenuated as compared to A375-GLO1_WT cells. Taken together, these prototype data provide evidence in support of a novel function of GLO1 in melanoma cell invasiveness and metastasis, and ongoing investigations explore the function and therapeutic potential of GLO1 as a novel melanoma target.
Collapse
Affiliation(s)
| | | | | | | | - Georg T. Wondrak
- Department of Pharmacology and Toxicology, College of Pharmacy & UA Cancer Center, University of Arizona, Tucson, AZ 85724, USA; (J.J.); (J.P.); (A.H.); (J.A.S.)
| |
Collapse
|
25
|
Brown JM, Wasson MCD, Marcato P. The Missing Lnc: The Potential of Targeting Triple-Negative Breast Cancer and Cancer Stem Cells by Inhibiting Long Non-Coding RNAs. Cells 2020; 9:E763. [PMID: 32244924 PMCID: PMC7140662 DOI: 10.3390/cells9030763] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/10/2020] [Accepted: 03/18/2020] [Indexed: 12/14/2022] Open
Abstract
Treatment decisions for breast cancer are based on staging and hormone receptor expression and include chemotherapies and endocrine therapy. While effective in many cases, some breast cancers are resistant to therapy, metastasize and recur, leading to eventual death. Higher percentages of tumor-initiating cancer stem cells (CSCs) may contribute to the increased aggressiveness, chemoresistance, and worse outcomes among breast cancer. This may be particularly true in triple-negative breast cancers (TNBCs) which have higher percentages of CSCs and are associated with worse outcomes. In recent years, increasing numbers of long non-coding RNAs (lncRNAs) have been identified as playing an important role in breast cancer progression and some of these have been specifically associated within the CSC populations of breast cancers. LncRNAs are non-protein-coding transcripts greater than 200 nucleotides which can have critical functions in gene expression regulation. The preclinical evidence regarding lncRNA antagonists for the treatment of cancer is promising and therefore, presents a potential novel approach for treating breast cancer and targeting therapy-resistant CSCs within these tumors. Herein, we summarize the lncRNAs that have been identified as functionally relevant in breast CSCs. Furthermore, our review of the literature and analysis of patient datasets has revealed that many of these breast CSC-associated lncRNAs are also enriched in TNBC. Together, this suggests that these lncRNAs may be playing a particularly important role in TNBC. Thus, certain breast cancer-promoting/CSC-associated lncRNAs could be targeted in the treatment of TNBCs and the CSCs within these tumors should be susceptible to anti-lncRNA therapy.
Collapse
Affiliation(s)
- Justin M Brown
- Departments of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (J.M.B.); (M.-C.D.W.)
| | - Marie-Claire D Wasson
- Departments of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (J.M.B.); (M.-C.D.W.)
| | - Paola Marcato
- Departments of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (J.M.B.); (M.-C.D.W.)
- Departments of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
26
|
Legoff L, Dali O, D'Cruz SC, Suglia A, Gely-Pernot A, Hémery C, Kernanec PY, Demmouche A, Kervarrec C, Tevosian S, Multigner L, Smagulova F. Ovarian dysfunction following prenatal exposure to an insecticide, chlordecone, associates with altered epigenetic features. Epigenetics Chromatin 2019; 12:29. [PMID: 31084621 PMCID: PMC6515617 DOI: 10.1186/s13072-019-0276-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 05/02/2019] [Indexed: 02/07/2023] Open
Abstract
Chlordecone (CD) is an insecticide that was used in the French West Indies for several years to control the banana root borer pest. Given its nonsignificant degradation, it persists in the environment. CD is a carcinogenic compound with reproductive and developmental toxicity and is a recognized endocrine-disrupting chemical. In this study, we examined the effects of CD on female reproductive system of mice with the focus on epigenetic features in ovary. Our data show that gestational exposure to low dose of CD affects meiotic double-strand breaks repair in female embryos. In adult mice derived from CD-treated pregnant females, we observed delayed puberty, decreased number of primordial and increased number of atretic follicles. Gene expression analysis revealed that Rcbtb2 and Rbpms genes were not expressed in embryonic gonads. Estrogen signaling- and oocyte maturation-associated genes were downregulated in adult ovaries. The morphological changes were associated with altered epigenetic features: increased H2Aub and increased H3K27me3 and decreased H4ac and H3K4me3 in embryonic oocytes. The DNA damage-associated, γH2AX marks were detected in the follicles of treated but not control adult ovaries. We also found reduced H3K4me3 and H4ac in fully grown oocytes of the treated ovaries. The ChIP-seq analysis of H3K4me3 in adult ovaries showed that target genes of ZFP57 and TRIM28, which regulate pluripotency and imprinting, were significantly enriched in altered regions. Our study clearly demonstrates that gestational exposure to a low dose of CD impairs the function of female reproductive system and the changes are associated with altered epigenetic features.
Collapse
Affiliation(s)
- Louis Legoff
- EHESP, Inserm, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Univ Rennes, 35000, Rennes, France
| | - Ouzna Dali
- EHESP, Inserm, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Univ Rennes, 35000, Rennes, France.,Biotoxicology Laboratory, Department of Biology, Faculty of Natural Sciences and Life, Djillali Liabes University, 22000, Sidi Bel Abbès, Algeria
| | - Shereen Cynthia D'Cruz
- EHESP, Inserm, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Univ Rennes, 35000, Rennes, France
| | - Antonio Suglia
- EHESP, Inserm, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Univ Rennes, 35000, Rennes, France
| | - Aurore Gely-Pernot
- EHESP, Inserm, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Univ Rennes, 35000, Rennes, France
| | - Chloé Hémery
- EHESP, Inserm, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Univ Rennes, 35000, Rennes, France
| | - Pierre-Yves Kernanec
- EHESP, Inserm, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Univ Rennes, 35000, Rennes, France
| | - Abbassia Demmouche
- Biotoxicology Laboratory, Department of Biology, Faculty of Natural Sciences and Life, Djillali Liabes University, 22000, Sidi Bel Abbès, Algeria
| | - Christine Kervarrec
- EHESP, Inserm, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Univ Rennes, 35000, Rennes, France
| | - Sergei Tevosian
- Department of Physiological Sciences, University of Florida, Box 100144, 1333 Center Drive, Gainesville, FL, 32610, USA
| | - Luc Multigner
- EHESP, Inserm, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Univ Rennes, 35000, Rennes, France
| | - Fatima Smagulova
- EHESP, Inserm, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Univ Rennes, 35000, Rennes, France.
| |
Collapse
|