1
|
Li W, Zhang Y, Zhuang Y, Chen R, Xiong Z, Li K, Liu F, Xu H, Li D, Peng J. Effects of Simvastatin on Inflammatory Response and Biological Behaviour of Adamantinomatous Craniopharyngioma. Neuroendocrinology 2024; 114:934-949. [PMID: 38964285 DOI: 10.1159/000539821] [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: 12/02/2023] [Accepted: 06/03/2024] [Indexed: 07/06/2024]
Abstract
INTRODUCTION The aim of this study was to investigate the autoinflammatory effect and biological behaviour of simvastatin (SIM) on adamantinomatous craniopharyngioma (ACP) cells. METHODS Craniopharyngiomas imaging, intraoperative observations, and tumour histopathology were employed to investigate the correlation between esters and craniopharyngiomas. Filipin III fluorescent probe verified the validity of SIM on the alternations of synthesized cholesterol in craniopharyngioma cells. The cell counting kit-8 (CCK8) assay detected the impacts of SIM on cell proliferation and determined the IC50 value of tumour cells. Reverse transcription polymerase chain reaction (RT-PCR) measured the expression of inflammatory factors. Flow cytometry technique detected the cell cycle and apoptosis, and cell scratch assay judged the cell migration. Meanwhile, Western blot was adopted to determine the expression of proteins related to inflammation, proliferation, and apoptosis signalling pathways. RESULTS In the ACP tumour parenchyma, many cholesterol crystalline clefts were observed, and the deposition of esters was closely associated with craniopharyngioma inflammation. After SIM intervention, a reduction in cholesterol synthesis within ACP was noted. RT-PCR analysis revealed SIM inhibited the transcription of inflammatory factors in ACP cells. Western blot analysis demonstrated SIM inhibited nuclear factor-kappa B p65 activation expression while promoted the expressions of Cl-caspase-3 and P38 MAPK. CCK8 assay indicated a decrease in ACP cell activity upon SIM treatment. Scratch assay signified that SIM hindered ACP cell migration. Flow cytometry results suggested that the drug promoted ACP cell apoptosis. CONCLUSION SIM suppressed the inflammatory response to craniopharyngiomas by inhibiting craniopharyngioma cholesterol synthesis, inhibited proliferation of ACP cells, and promoted their apoptosis.
Collapse
Affiliation(s)
- Weizhao Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China,
| | - Yunxiao Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yishan Zhuang
- Department of Biostatistics, School of Public Health, Southern Medical University, Guangzhou, China
| | - Rongjun Chen
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiwei Xiong
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kai Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fang Liu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haiyan Xu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Danling Li
- Department of Biostatistics, School of Public Health, Southern Medical University, Guangzhou, China
| | - Junxiang Peng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Lu J, Chen S, Bai X, Liao M, Qiu Y, Zheng LL, Yu H. Targeting cholesterol metabolism in Cancer: From molecular mechanisms to therapeutic implications. Biochem Pharmacol 2023; 218:115907. [PMID: 37931664 DOI: 10.1016/j.bcp.2023.115907] [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: 07/18/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/08/2023]
Abstract
Cholesterol is an essential component of cell membranes and helps to maintain their structure and function. Abnormal cholesterol metabolism has been linked to the development and progression of tumors. Changes in cholesterol metabolism triggered by internal or external stimuli can promote tumor growth. During metastasis, tumor cells require large amounts of cholesterol to support their growth and colonization of new organs. Recent research has shown that cholesterol metabolism is reprogrammed during tumor development, and this can also affect the anti-tumor activity of immune cells in the surrounding environment. However, identifying the specific targets in cholesterol metabolism that regulate cancer progression and the tumor microenvironment is still a challenge. Additionally, exploring the potential of combining statin drugs with other therapies for different types of cancer could be a worthwhile avenue for future drug development. In this review, we focus on the molecular mechanisms of cholesterol and its derivatives in cell metabolism and the tumor microenvironment, and discuss specific targets and relevant therapeutic agents that inhibit aspects of cholesterol homeostasis.
Collapse
Affiliation(s)
- Jia Lu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Siwei Chen
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xuejiao Bai
- Department of Anesthesiology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Minru Liao
- Department of Anesthesiology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| | - Ling-Li Zheng
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China.
| | - Haiyang Yu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
3
|
Lee H, Ha S, Choi S, Do S, Yoon S, Kim YK, Kim WY. Oncogenic Impact of TONSL, a Homologous Recombination Repair Protein at the Replication Fork, in Cancer Stem Cells. Int J Mol Sci 2023; 24:ijms24119530. [PMID: 37298484 DOI: 10.3390/ijms24119530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/15/2023] [Accepted: 05/18/2023] [Indexed: 06/12/2023] Open
Abstract
We investigated the role of TONSL, a mediator of homologous recombination repair (HRR), in stalled replication fork double-strand breaks (DSBs) in cancer. Publicly available clinical data (tumors from the ovary, breast, stomach and lung) were analyzed through KM Plotter, cBioPortal and Qomics. Cancer stem cell (CSC)-enriched cultures and bulk/general mixed cell cultures (BCCs) with RNAi were employed to determine the effect of TONSL loss in cancer cell lines from the ovary, breast, stomach, lung, colon and brain. Limited dilution assays and ALDH assays were used to quantify the loss of CSCs. Western blotting and cell-based homologous recombination assays were used to identify DNA damage derived from TONSL loss. TONSL was expressed at higher levels in cancer tissues than in normal tissues, and higher expression was an unfavorable prognostic marker for lung, stomach, breast and ovarian cancers. Higher expression of TONSL is partly associated with the coamplification of TONSL and MYC, suggesting its oncogenic role. The suppression of TONSL using RNAi revealed that it is required in the survival of CSCs in cancer cells, while BCCs could frequently survive without TONSL. TONSL dependency occurs through accumulated DNA damage-induced senescence and apoptosis in TONSL-suppressed CSCs. The expression of several other major mediators of HRR was also associated with worse prognosis, whereas the expression of error-prone nonhomologous end joining molecules was associated with better survival in lung adenocarcinoma. Collectively, these results suggest that TONSL-mediated HRR at the replication fork is critical for CSC survival; targeting TONSL may lead to the effective eradication of CSCs.
Collapse
Affiliation(s)
- Hani Lee
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Sojung Ha
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - SeokGyeong Choi
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Soomin Do
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Sukjoon Yoon
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Yong Kee Kim
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, Republic of Korea
- Research Institute of Pharmacal Research, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Woo-Young Kim
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
- Research Institute of Pharmacal Research, Sookmyung Women's University, Seoul 04310, Republic of Korea
| |
Collapse
|
4
|
Min JY, Kim DH. Stearoyl-CoA Desaturase 1 as a Therapeutic Biomarker: Focusing on Cancer Stem Cells. Int J Mol Sci 2023; 24:ijms24108951. [PMID: 37240297 DOI: 10.3390/ijms24108951] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
The dysregulation of lipid metabolism and alterations in the ratio of monounsaturated fatty acids (MUFAs) to saturated fatty acids (SFAs) have been implicated in cancer progression and stemness. Stearoyl-CoA desaturase 1 (SCD1), an enzyme involved in lipid desaturation, is crucial in regulating this ratio and has been identified as an important regulator of cancer cell survival and progression. SCD1 converts SFAs into MUFAs and is important for maintaining membrane fluidity, cellular signaling, and gene expression. Many malignancies, including cancer stem cells, have been reported to exhibit high expression of SCD1. Therefore, targeting SCD1 may provide a novel therapeutic strategy for cancer treatment. In addition, the involvement of SCD1 in cancer stem cells has been observed in various types of cancer. Some natural products have the potential to inhibit SCD1 expression/activity, thereby suppressing cancer cell survival and self-renewal activity.
Collapse
Affiliation(s)
- Jin-Young Min
- Department of Chemistry, College of Convergence and Integrated Science, Kyonggi University, Suwon 16227, Gyeonggi-do, Republic of Korea
| | - Do-Hee Kim
- Department of Chemistry, College of Convergence and Integrated Science, Kyonggi University, Suwon 16227, Gyeonggi-do, Republic of Korea
| |
Collapse
|
5
|
Zhang Y, Wang Y, Zhao G, Orsulic S, Matei D. Metabolic dependencies and targets in ovarian cancer. Pharmacol Ther 2023; 245:108413. [PMID: 37059310 DOI: 10.1016/j.pharmthera.2023.108413] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/31/2023] [Accepted: 04/10/2023] [Indexed: 04/16/2023]
Abstract
Reprogramming of cellular metabolism is a hallmark of cancer. Cancer cells undergo metabolic adaptations to maintain tumorigenicity and survive under the attack of immune cells and chemotherapy in the tumor microenvironment. Metabolic alterations in ovarian cancer in part overlap with findings from other solid tumors and in part reflect unique traits. Altered metabolic pathways not only facilitate ovarian cancer cells' survival and proliferation but also endow them to metastasize, acquire resistance to chemotherapy, maintain cancer stem cell phenotype and escape the effects of anti-tumor immune defense. In this review, we comprehensively review the metabolic signatures of ovarian cancer and their impact on cancer initiation, progression, and resistance to treatment. We highlight novel therapeutic strategies targeting metabolic pathways under development.
Collapse
Affiliation(s)
- Yaqi Zhang
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Driskill Graduate Training Program in Life Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yinu Wang
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Guangyuan Zhao
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Driskill Graduate Training Program in Life Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Sandra Orsulic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Daniela Matei
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Jesse Brown VA Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
6
|
Liu Z, Lei J, Wu T, Hu W, Zheng M, Wang Y, Song J, Ruan H, Xu L, Ren T, Xu W, Wen Z. Lipogenesis promotes mitochondrial fusion and maintains cancer stemness in human NSCLC. JCI Insight 2023; 8:158429. [PMID: 36809297 PMCID: PMC10070109 DOI: 10.1172/jci.insight.158429] [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: 01/12/2022] [Accepted: 02/15/2023] [Indexed: 02/23/2023] Open
Abstract
Cancer stem-like cells (CSCs) are critically involved in cancer metastasis and chemoresistance, acting as one major obstacle in clinical practice. While accumulating studies have implicated the metabolic reprogramming of CSCs, mitochondrial dynamics in such cells remain poorly understood. Here we pinpointed OPA1hi with mitochondrial fusion as a metabolic feature of human lung CSCs, licensing their stem-like properties. Specifically, human lung CSCs exerted enhanced lipogenesis, inducing OPA1 expression via transcription factor SAM Pointed Domain containing ETS transcription Factor (SPDEF). In consequence, OPA1hi promoted mitochondrial fusion and stemness of CSCs. Such lipogenesishi, SPDEFhi, and OPA1hi metabolic adaptions were verified with primary CSCs from lung cancer patients. Accordingly, blocking lipogenesis and mitochondrial fusion efficiently impeded CSC expansion and growth of organoids derived from patients with lung cancer. Together, lipogenesis regulates mitochondrial dynamics via OPA1 for controlling CSCs in human lung cancer.
Collapse
Affiliation(s)
- Zhen Liu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Jiaxin Lei
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Tong Wu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Weijie Hu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Ming Zheng
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Ying Wang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Jingdong Song
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hang Ruan
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Lin Xu
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou, China
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi, Guizhou, China
| | - Tao Ren
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Xu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Zhenke Wen
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
7
|
Papadaki S, Magklara A. Regulation of Metabolic Plasticity in Cancer Stem Cells and Implications in Cancer Therapy. Cancers (Basel) 2022; 14:5912. [PMID: 36497394 PMCID: PMC9741285 DOI: 10.3390/cancers14235912] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
Cancer stem cells (CSCs), a subpopulation of tumor cells with self-renewal capacity, have been associated with tumor initiation, progression, and therapy resistance. While the bulk of tumor cells mainly use glycolysis for energy production, CSCs have gained attention for their ability to switch between glycolysis and oxidative phosphorylation, depending on their energy needs and stimuli from their microenvironment. This metabolic plasticity is mediated by signaling pathways that are also implicated in the regulation of CSC properties, such as the Wnt/β-catenin, Notch, and Hippo networks. Two other stemness-associated processes, autophagy and hypoxia, seem to play a role in the metabolic switching of CSCs as well. Importantly, accumulating evidence has linked the metabolic plasticity of CSCs to their increased resistance to treatment. In this review, we summarize the metabolic signatures of CSCs and the pathways that regulate them; we especially highlight research data that demonstrate the metabolic adaptability of these cells and their role in stemness and therapy resistance. As the development of drug resistance is a major challenge for successful cancer treatment, the potential of specific elimination of CSCs through targeting their metabolism is of great interest and it is particularly examined.
Collapse
Affiliation(s)
- Styliani Papadaki
- Department of Clinical Chemistry, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece
| | - Angeliki Magklara
- Department of Clinical Chemistry, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece
- Biomedical Research Institute–Foundation for Research and Technology, 45110 Ioannina, Greece
- Institute of Biosciences, University Research Center of Ioannina (URCI), 45110 Ioannina, Greece
| |
Collapse
|
8
|
Screening of the siGPCR library in combination with cisplatin against lung cancers. Sci Rep 2022; 12:17358. [PMID: 36253428 PMCID: PMC9576725 DOI: 10.1038/s41598-022-21063-0] [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: 06/08/2022] [Accepted: 09/22/2022] [Indexed: 01/10/2023] Open
Abstract
The screening of siRNAs targeting 390 human G protein-coupled receptors (GPCRs) was multiplexed in combination with cisplatin against lung cancer cells. While the cell viability measure hardly captured the anticancer effect of siGPCRs, the direct cell count revealed the anticancer potential of diverse GPCRs (46 hits with > twofold growth inhibition, p-value < 0.01). In combined treatment with cisplatin, siRNAs against five genes (ADRA2A, F2RL3, NPSR1, NPY and TACR3) enhanced the anti-proliferation efficacy on cancer cells and reduced the self-recovery ability of surviving cells after the removal of the combined treatment. Further on-target validation confirmed that the knockdown of TACR3 expression exhibited anticancer efficacy under both single and combined treatment with cisplatin. Q-omics ( http://qomics.io ) analysis showed that high expression of TACR3 was unfavorable for patient survival, particularly with mutations in GPCR signaling pathways. The present screening data provide a useful resource for GPCR targets and biomarkers for improving the efficacy of cisplatin treatment.
Collapse
|
9
|
Cholesterol Synthesis Is Important for Breast Cancer Cell Tumor Sphere Formation and Invasion. Biomedicines 2022; 10:biomedicines10081908. [PMID: 36009455 PMCID: PMC9405659 DOI: 10.3390/biomedicines10081908] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/25/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
Breast cancer has a high risk of recurrence and distant metastasis after remission. Controlling distant metastasis is important for reducing breast cancer mortality, but accomplishing this goal remains elusive. In this study, we investigated the molecular pathways underlying metastasis using cells that mimic the breast cancer distant metastasis process. HCC1143 breast cancer cells were cultured under two-dimensional (2D)-adherent, tumor sphere (TS), and reattached (ReA) culture conditions to mimic primary tumors, circulating tumor cells, and metastasized tumors, respectively. ReA cells demonstrated increased TS formation and enhanced invasion capacity compared to the original 2D-cultured parental cells. In addition, ReA cells had a higher frequency of ESA+CD44+CD24− population, which represents a stem-cell-like cell population. RNA sequencing identified the cholesterol synthesis pathway as one of the most significantly increased pathways in TS and ReA cells compared to parental cells, which was verified by measuring intracellular cholesterol levels. Furthermore, the pharmacological inhibition of the cholesterol synthesis pathway decreased the ability of cancer cells to form TSs and invade. Our results suggest that the cholesterol synthesis pathway plays an important role in the distant metastasis of breast cancer cells by augmenting TS formation and invasion capacity.
Collapse
|
10
|
Lee H, Choi S, Ha S, Yoon S, Kim WY. ARL2 is required for homologous recombination repair and colon cancer stem cell survival. FEBS Open Bio 2022; 12:1523-1533. [PMID: 35567502 PMCID: PMC9340879 DOI: 10.1002/2211-5463.13438] [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: 02/05/2022] [Revised: 04/16/2022] [Accepted: 05/12/2022] [Indexed: 11/10/2022] Open
Abstract
ARL2 regulates the dynamics of cytological components and is highly expressed in colon cancer tissues. Here, we report novel roles of ARL2 in the cell nucleus and colon cancer stem cells (CSCs). ARL2 is expressed at relatively low levels in K‐RAS active colon cancer cells, but its expression is induced in CSCs. Depletion of ARL2 results in M phase arrest exclusively in non‐CSC cultured cells; in addition, DNA break stress accumulates in CSCs leading to apoptosis. ARL2 expression is positively associated with the expression of all six RAD51 family genes, which are essential for homologous recombination repair (HRR). Furthermore, ARL2 is required for HRR and detected within chromatin compartments. These results demonstrate the requirement of ARL2 in colon CSC maintenance, which possibly occurs through mediating double‐strand break DNA repair in the nucleus.
Collapse
Affiliation(s)
- Hani Lee
- College of Pharmacy, Sookmyung Women's University, Cheongparo 47 gil, Yongsangu, Seoul, 04312, Korea
| | - SeokGyeong Choi
- College of Pharmacy, Sookmyung Women's University, Cheongparo 47 gil, Yongsangu, Seoul, 04312, Korea
| | - Sojung Ha
- College of Pharmacy, Sookmyung Women's University, Cheongparo 47 gil, Yongsangu, Seoul, 04312, Korea
| | - Sukjoon Yoon
- Department of Biological Sciences, Sookmyung Women's University, Cheongparo 47 gil, Yongsangu, Seoul, 04312, Korea
| | - Woo-Young Kim
- College of Pharmacy, Sookmyung Women's University, Cheongparo 47 gil, Yongsangu, Seoul, 04312, Korea.,Research Institute of Pharmacal Research, Sookmyung Women's University, Cheongparo 47 gil, Yongsangu, Seoul, 04312, Korea
| |
Collapse
|
11
|
Wang SY, Hu QC, Wu T, Xia J, Tao XA, Cheng B. Abnormal lipid synthesis as a therapeutic target for cancer stem cells. World J Stem Cells 2022; 14:146-162. [PMID: 35432735 PMCID: PMC8963380 DOI: 10.4252/wjsc.v14.i2.146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/19/2021] [Accepted: 02/20/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) comprise a subpopulation of cancer cells with stem cell properties, which exhibit the characteristics of high tumorigenicity, self-renewal, and tumor initiation and are associated with the occurrence, metastasis, therapy resistance, and relapse of cancer. Compared with differentiated cells, CSCs have unique metabolic characteristics, and metabolic reprogramming contributes to the self-renewal and maintenance of stem cells. It has been reported that CSCs are highly dependent on lipid metabolism to maintain stemness and satisfy the requirements of biosynthesis and energy metabolism. In this review, we demonstrate that lipid anabolism alterations promote the survival of CSCs, including de novo lipogenesis, lipid desaturation, and cholesterol synthesis. In addition, we also emphasize the molecular mechanism underlying the relationship between lipid synthesis and stem cell survival, the signal trans-duction pathways involved, and the application prospect of lipid synthesis reprogramming in CSC therapy. It is demonstrated that the dependence on lipid synthesis makes targeting of lipid synthesis metabolism a promising therapeutic strategy for eliminating CSCs. Targeting key molecules in lipid synthesis will play an important role in anti-CSC therapy.
Collapse
Affiliation(s)
- Si-Yu Wang
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Qin-Chao Hu
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Tong Wu
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Juan Xia
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Xiao-An Tao
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Bin Cheng
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| |
Collapse
|
12
|
Addeo M, Di Paola G, Verma HK, Laurino S, Russi S, Zoppoli P, Falco G, Mazzone P. Gastric Cancer Stem Cells: A Glimpse on Metabolic Reprogramming. Front Oncol 2021; 11:698394. [PMID: 34249759 PMCID: PMC8262334 DOI: 10.3389/fonc.2021.698394] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 05/31/2021] [Indexed: 12/11/2022] Open
Abstract
Gastric cancer (GC) is one of the most widespread causes of cancer-related death worldwide. Recently, emerging implied that gastric cancer stem cells (GCSCs) play an important role in the initiation and progression of GC. This subpopulation comprises cells with several features, such as self-renewal capability, high proliferating rate, and ability to modify their metabolic program, which allow them to resist current anticancer therapies. Metabolic pathway intermediates play a pivotal role in regulating cell differentiation both in tumorigenesis and during normal development. Thus, the dysregulation of both anabolic and catabolic pathways constitutes a significant opportunity to target GCSCs in order to eradicate the tumor progression. In this review, we discuss the current knowledge about metabolic phenotype that supports GCSC proliferation and we overview the compounds that selectively target metabolic intermediates of CSCs that can be used as a strategy in cancer therapy.
Collapse
Affiliation(s)
- Martina Addeo
- Istituto di Ricerche Genetiche Gaetano Salvatore Biogem Scarl, Ariano Irpino, Italy.,Department of Biology, University of Naples Federico II, Naples, Italy
| | - Giuseppina Di Paola
- Istituto di Ricerche Genetiche Gaetano Salvatore Biogem Scarl, Ariano Irpino, Italy
| | - Henu Kumar Verma
- Istituto di Ricerche Genetiche Gaetano Salvatore Biogem Scarl, Ariano Irpino, Italy.,IEOS-CNR, Institute of Experimental Endocrinology and Oncology "G. Salvatore" - National Research Council, Naples, Italy
| | - Simona Laurino
- Laboratory of Pre-Clinical and Translational Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)-centro di riferimento oncologico della basilicata (CROB), Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Sabino Russi
- Laboratory of Pre-Clinical and Translational Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)-centro di riferimento oncologico della basilicata (CROB), Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Pietro Zoppoli
- Laboratory of Pre-Clinical and Translational Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)-centro di riferimento oncologico della basilicata (CROB), Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Geppino Falco
- Istituto di Ricerche Genetiche Gaetano Salvatore Biogem Scarl, Ariano Irpino, Italy.,Department of Biology, University of Naples Federico II, Naples, Italy.,IEOS-CNR, Institute of Experimental Endocrinology and Oncology "G. Salvatore" - National Research Council, Naples, Italy.,Laboratory of Pre-Clinical and Translational Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)-centro di riferimento oncologico della basilicata (CROB), Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Pellegrino Mazzone
- Istituto di Ricerche Genetiche Gaetano Salvatore Biogem Scarl, Ariano Irpino, Italy
| |
Collapse
|
13
|
Yu Y, Kim H, Choi S, Yu J, Lee JY, Lee H, Yoon S, Kim WY. Targeting a Lipid Desaturation Enzyme, SCD1, Selectively Eliminates Colon Cancer Stem Cells through the Suppression of Wnt and NOTCH Signaling. Cells 2021; 10:cells10010106. [PMID: 33430034 PMCID: PMC7826607 DOI: 10.3390/cells10010106] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 12/13/2022] Open
Abstract
The elimination of the cancer stem cell (CSC) population may be required to achieve better outcomes of cancer therapy. We evaluated stearoyl-CoA desaturase 1 (SCD1) as a novel target for CSC-selective elimination in colon cancer. CSCs expressed more SCD1 than bulk cultured cells (BCCs), and blocking SCD1 expression or function revealed an essential role for SCD1 in the survival of CSCs, but not BCCs. The CSC potential selectively decreased after treatment with the SCD1 inhibitor in vitro and in vivo. The CSC-selective suppression was mediated through the induction of apoptosis. The mechanism leading to selective CSC death was investigated by performing a quantitative RT-PCR analysis of 14 CSC-specific signaling and marker genes after 24 and 48 h of treatment with two concentrations of an inhibitor. The decrease in the expression of Notch1 and AXIN2 preceded changes in the expression of all other genes, at 24 h of treatment in a dose-dependent manner, followed by the downregulation of most Wnt- and NOTCH-signaling genes. Collectively, we showed that not only Wnt but also NOTCH signaling is a primary target of suppression by SCD1 inhibition in CSCs, suggesting the possibility of targeting SCD1 against colon cancer in clinical settings.
Collapse
Affiliation(s)
- Yeongji Yu
- College of Pharmacy, Sookmyung Women’s University, Seoul 04312, Korea; (Y.Y.); (H.K.); (S.C.); (J.Y.); (J.Y.L.); (H.L.)
| | - Hyejin Kim
- College of Pharmacy, Sookmyung Women’s University, Seoul 04312, Korea; (Y.Y.); (H.K.); (S.C.); (J.Y.); (J.Y.L.); (H.L.)
| | - SeokGyeong Choi
- College of Pharmacy, Sookmyung Women’s University, Seoul 04312, Korea; (Y.Y.); (H.K.); (S.C.); (J.Y.); (J.Y.L.); (H.L.)
| | - JinSuh Yu
- College of Pharmacy, Sookmyung Women’s University, Seoul 04312, Korea; (Y.Y.); (H.K.); (S.C.); (J.Y.); (J.Y.L.); (H.L.)
| | - Joo Yeon Lee
- College of Pharmacy, Sookmyung Women’s University, Seoul 04312, Korea; (Y.Y.); (H.K.); (S.C.); (J.Y.); (J.Y.L.); (H.L.)
| | - Hani Lee
- College of Pharmacy, Sookmyung Women’s University, Seoul 04312, Korea; (Y.Y.); (H.K.); (S.C.); (J.Y.); (J.Y.L.); (H.L.)
| | - Sukjoon Yoon
- Research Institute of Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea;
| | - Woo-Young Kim
- Research Institute of Pharmaceutical Sciences, Sookmyung Women’s University, Seoul 04312, Korea
- Correspondence: ; Tel.: +82-2-2077-7587
| |
Collapse
|
14
|
Ehmsen S, Ditzel HJ. Signaling pathways essential for triple-negative breast cancer stem-like cells. Stem Cells 2020; 39:133-143. [PMID: 33211379 DOI: 10.1002/stem.3301] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/31/2020] [Indexed: 12/24/2022]
Abstract
Since the discovery of breast cancer stem cells (CSCs), a significant effort has been made to identify and characterize these cells. It is a generally believe that CSCs play an important role in cancer initiation, therapy resistance, and progression of triple-negative breast cancer (TNBC), an aggressive breast cancer subtype with poor prognosis. Thus, therapies targeting these cells would be a valuable addition to standard treatments that primarily target more differentiated, rapidly dividing TNBC cells. Although several cell surface and intracellular proteins have been described as biomarkers for CSCs, none of these are specific to this population of cells. Recent research is moving toward cellular signaling pathways as targets and biomarkers for CSCs. The WNT pathway, the nuclear factor-kappa B (NF-κB) pathway, and the cholesterol biosynthesis pathway have recently been identified to play a key role in proliferation, survival, and differentiation of CSCs, including those of breast cancer. In this review, we assess recent findings related to these three pathways in breast CSC, with particular focus on TNBC CSCs, and discuss how targeting these pathways, in combination with current standard of care, might prove effective and improve the prognosis of TNBC patients.
Collapse
Affiliation(s)
- Sidse Ehmsen
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Oncology, Odense University Hospital, Odense, Denmark.,Research Unit of Oncology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
| | - Henrik J Ditzel
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Oncology, Odense University Hospital, Odense, Denmark.,Research Unit of Oncology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
| |
Collapse
|
15
|
Qiu T, Cao J, Chen W, Wang J, Wang Y, Zhao L, Liu M, He L, Wu G, Li H, Gu H. 24-Dehydrocholesterol reductase promotes the growth of breast cancer stem-like cells through the Hedgehog pathway. Cancer Sci 2020; 111:3653-3664. [PMID: 32713162 PMCID: PMC7540995 DOI: 10.1111/cas.14587] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/17/2020] [Accepted: 07/20/2020] [Indexed: 12/19/2022] Open
Abstract
Cholesterol is a risk factor for breast cancer. However, it is still unclear whether the cholesterol biosynthesis pathway plays any significant role in breast carcinogenesis. 24-Dehydrocholesterol reductase (DHCR24) is a key enzyme in the cholesterol synthesis pathway. Although DHCR24 is reported to have different functions in different cancers, it is not clear whether DHCR24 is involved in breast cancer. In this study, we found that DHCR24 expression was higher in breast cancer especially in luminal and HER2 positive breast cancer tissues compared with normal breast. Changes in DHCR24 expression altered cellular cholesterol content without affecting the adherent growth of breast cancer cells. However, DHCR24 knockdown reduced whereas DHCR24 overexpression enhanced breast cancer stem-like cell populations such as mammosphere and aldehyde dehydrogenase positive cell numbers. In addition, DHCR24 overexpression increased the expression of the Hedgehog pathway-regulated genes. Treating DHCR24 overexpressing breast cancer cell lines with the Hedgehog pathway inhibitor GANT61 blocked DHCR24-induced mammosphere growth and increased mRNA levels of the Hedgehog regulated genes. Furthermore, expression of a constitutively activated mutant of Smoothened, a key hedgehog signal transducer, rescued the decreases in mammosphere growth and Hedgehog regulated gene expression induced by knockdown of DHCR24. These results indicate that DHCR24 promotes the growth of breast cancer stem-like cells in part through enhancing the Hedgehog signaling pathway. Our data suggest that cholesterol contribute to breast carcinogenesis by enhancing Hedgehog signaling and cancer stem-like cell populations. Enzymes including DHCR24 involved in cholesterol biosynthesis should be considered as potential treatment targets for breast cancer.
Collapse
Affiliation(s)
- Ting Qiu
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jiawei Cao
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wanzhou Chen
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jieyi Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yaqi Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Lingjie Zhao
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Min Liu
- Department of Orthopedics, Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Licai He
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Guang Wu
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hongzhi Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Haihua Gu
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
16
|
Ehmsen S, Pedersen MH, Wang G, Terp MG, Arslanagic A, Hood BL, Conrads TP, Leth-Larsen R, Ditzel HJ. Increased Cholesterol Biosynthesis Is a Key Characteristic of Breast Cancer Stem Cells Influencing Patient Outcome. Cell Rep 2020; 27:3927-3938.e6. [PMID: 31242424 DOI: 10.1016/j.celrep.2019.05.104] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 04/01/2019] [Accepted: 05/29/2019] [Indexed: 12/12/2022] Open
Abstract
Tumor eradication may be greatly improved by targeting cancer stem cells (CSCs), as they exhibit resistance to conventional therapy. To gain insight into the unique biology of CSCs, we developed patient-derived xenograft tumors (PDXs) from ER- breast cancers from which we isolated mammospheres that are enriched for CSCs. Comparative global proteomic analysis was performed on patient tumor tissues and corresponding PDXs and mammospheres. Mammospheres exhibited increased expression of proteins associated with de novo cholesterol synthesis. The clinical relevance of increased cholesterol biosynthesis was verified in a large breast cancer cohort showing correlation with shorter relapse-free survival. RNAi and chemical inhibition of the cholesterol biosynthesis pathway reduced mammosphere formation, which could be rescued by a downstream metabolite. Our findings identify the cholesterol biosynthesis pathway as central for CSC propagation and a potential therapeutic target, as well as providing a mechanistic explanation for the therapeutic benefit of statins in breast cancer.
Collapse
Affiliation(s)
- Sidse Ehmsen
- Department of Molecular Medicine, Cancer, and Inflammation Research Unit, University of Southern Denmark, 5000 Odense, Denmark; Department of Oncology, Odense University Hospital, 5000 Odense, Denmark; Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, 5000 Odense, Denmark
| | - Martin H Pedersen
- Department of Molecular Medicine, Cancer, and Inflammation Research Unit, University of Southern Denmark, 5000 Odense, Denmark
| | - Guisong Wang
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Annandale, VA 22003, USA
| | - Mikkel G Terp
- Department of Molecular Medicine, Cancer, and Inflammation Research Unit, University of Southern Denmark, 5000 Odense, Denmark
| | - Amina Arslanagic
- Department of Molecular Medicine, Cancer, and Inflammation Research Unit, University of Southern Denmark, 5000 Odense, Denmark
| | - Brian L Hood
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Annandale, VA 22003, USA
| | - Thomas P Conrads
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Annandale, VA 22003, USA; Inova Schar Cancer Institute, Inova Center for Personalized Health, Fairfax, VA 22031, USA
| | - Rikke Leth-Larsen
- Department of Molecular Medicine, Cancer, and Inflammation Research Unit, University of Southern Denmark, 5000 Odense, Denmark; Department of Regional Health Research, University of Southern Denmark, 5000 Odense, Denmark.
| | - Henrik J Ditzel
- Department of Molecular Medicine, Cancer, and Inflammation Research Unit, University of Southern Denmark, 5000 Odense, Denmark; Department of Oncology, Odense University Hospital, 5000 Odense, Denmark; Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark; Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, 5000 Odense, Denmark.
| |
Collapse
|
17
|
Li H, Feng Z, He ML. Lipid metabolism alteration contributes to and maintains the properties of cancer stem cells. Theranostics 2020; 10:7053-7069. [PMID: 32641978 PMCID: PMC7330842 DOI: 10.7150/thno.41388] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 04/28/2020] [Indexed: 12/11/2022] Open
Abstract
Lipids, the basic components of the cell membrane, execute fundamental roles in almost all the cell activities including cell-cell recognition, signalling transduction and energy supplies. Lipid metabolism is elementary for life sustentation that balances activity between synthesis and degradation. An accumulating amount of data has indicated abnormal lipid metabolism in cancer stem cells (CSCs), and that the alteration of lipid metabolism exerts a great impact on CSCs' properties such as the capability of self-renewal, differentiation, invasion, metastasis, and drug sensitivity and resistance. CSCs' formation and maintenance cannot do without the regulation of fatty acids and cholesterol. In normal cells and embryonic development, fatty acids and cholesterol metabolism are regulated by some important signalling pathways (such as Hedgehog, Notch, Wnt signalling pathways); these signalling pathways also play crucial roles in initiating and/or maintaining CSCs' properties, and such signalling is shown to be commonly modulated by the abnormal lipid metabolism in CSCs; on the other hand, the altered lipid metabolism in turn modifies the cell signalling and generates additional impacts on CSCs. Metabolic rewiring is considered as an ideal hallmark of CSCs, and metabolic alterations would be promising therapeutic targets of CSCs for aggressive tumors. In this review, we summarize the most updated findings of lipid metabolic abnormalities in CSCs and prospect the potential applications of targeting lipid metabolism for anticancer treatment.
Collapse
|
18
|
Kim H, Yu Y, Choi S, Lee H, Yu J, Lee JH, Kim WY. Evodiamine Eliminates Colon Cancer Stem Cells via Suppressing Notch and Wnt Signaling. Molecules 2019; 24:molecules24244520. [PMID: 31835579 PMCID: PMC6943729 DOI: 10.3390/molecules24244520] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 12/09/2019] [Accepted: 12/09/2019] [Indexed: 02/06/2023] Open
Abstract
Evodiamine, an alkaloid contained in traditional Asian herbal medicines that have been used for hundreds years, is interesting due to its cytotoxic effects against many cancers. We examined the effect of evodiamine on the cancer stem cell (CSC) population and the bulk cultured cancer cells (BCC) of colon cancers to examine the double targeting effect. We found that three colon cancer cell lines’ BCC and CSC are effectively targeted by evodiamine. Evodiamine was able to suppress BCC proliferation and induce apoptosis of the cells captured in G2/M phase, as previously reported. However, evodiamine did not cause the accumulation of CSCs at a certain stage of the cell cycle, resulting in the elimination of stemness through an unknown mechanism. By analyzing the expression of 84 genes related to CSCs in two colon cancer cell lines’ CSC, as well as performing further informatics analyses, and quantitative RT-PCR analyses of 24 CSC genes, we found that evodiamine suppressed the expression of the genes that control key signaling pathways of CSC, namely, WNT and NOTCH signaling, to lead CSC elimination. These results suggest that evodiamine should be further developed for targeting both BCCs and CSCs in colon cancers.
Collapse
Affiliation(s)
- Hyejin Kim
- College of Pharmacy, Sookmyung Women’s University, Cheongparo-47 Gil, Yongsan Gu, Seoul 04312, Korea; (H.K.); (Y.Y.); (S.C.); (H.L.); (J.Y.)
| | - Yeongji Yu
- College of Pharmacy, Sookmyung Women’s University, Cheongparo-47 Gil, Yongsan Gu, Seoul 04312, Korea; (H.K.); (Y.Y.); (S.C.); (H.L.); (J.Y.)
| | - SeokGyeong Choi
- College of Pharmacy, Sookmyung Women’s University, Cheongparo-47 Gil, Yongsan Gu, Seoul 04312, Korea; (H.K.); (Y.Y.); (S.C.); (H.L.); (J.Y.)
| | - Hani Lee
- College of Pharmacy, Sookmyung Women’s University, Cheongparo-47 Gil, Yongsan Gu, Seoul 04312, Korea; (H.K.); (Y.Y.); (S.C.); (H.L.); (J.Y.)
| | - Jinsuh Yu
- College of Pharmacy, Sookmyung Women’s University, Cheongparo-47 Gil, Yongsan Gu, Seoul 04312, Korea; (H.K.); (Y.Y.); (S.C.); (H.L.); (J.Y.)
| | - Jeong-Ho Lee
- Inland Aquaculture Research Center, National Institute of Fisheries Science, Changwon 51688, Korea;
| | - Woo-Young Kim
- College of Pharmacy, Sookmyung Women’s University, Cheongparo-47 Gil, Yongsan Gu, Seoul 04312, Korea; (H.K.); (Y.Y.); (S.C.); (H.L.); (J.Y.)
- Research Institute of Pharmaceutical Sciences, Sookmyung Women’s University, Cheongparo-47 Gil, Yongsan Gu, Seoul 04312, Korea
- Correspondence: ; Tel.: +82-2-2077-7587
| |
Collapse
|
19
|
Park C, Lee Y, Je S, Chang S, Kim N, Jeong E, Yoon S. Overexpression and Selective Anticancer Efficacy of ENO3 in STK11 Mutant Lung Cancers. Mol Cells 2019; 42:804-809. [PMID: 31697874 PMCID: PMC6883975 DOI: 10.14348/molcells.2019.0099] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 10/02/2019] [Accepted: 10/13/2019] [Indexed: 02/06/2023] Open
Abstract
Oncogenic gain-of-function mutations are clinical biomarkers for most targeted therapies, as well as represent direct targets for drug treatment. Although loss-of-function mutations involving the tumor suppressor gene, STK11 (LKB1) are important in lung cancer progression, STK11 is not the direct target for anticancer agents. We attempted to identify cancer transcriptome signatures associated with STK11 loss-offunction mutations. Several new sensitive and specific gene expression markers (ENO3, TTC39C, LGALS3, and MAML2) were identified using two orthogonal measures, i.e., fold change and odds ratio analyses of transcriptome data from cell lines and tissue samples. Among the markers identified, the ENO3 gene over-expression was found to be the direct consequence of STK11 loss-of-function. Furthermore, the knockdown of ENO3 expression exhibited selective anticancer effect in STK11 mutant cells compared with STK11 wild type (or recovered) cells. These findings suggest that ENO3 -based targeted therapy might be promising for patients with lung cancer harboring STK11 mutations.
Collapse
Affiliation(s)
- Choa Park
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310,
Korea
| | - Yejin Lee
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310,
Korea
| | - Soyeon Je
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310,
Korea
| | - Shengzhi Chang
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310,
Korea
| | - Nayoung Kim
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310,
Korea
| | - Euna Jeong
- Research Institute of Women’s Health, Sookmyung Women’s University, Seoul 04310,
Korea
| | - Sukjoon Yoon
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310,
Korea
- Research Institute of Women’s Health, Sookmyung Women’s University, Seoul 04310,
Korea
| |
Collapse
|
20
|
Ovarian Cancer-Why Lipids Matter. Cancers (Basel) 2019; 11:cancers11121870. [PMID: 31769430 PMCID: PMC6966536 DOI: 10.3390/cancers11121870] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/23/2019] [Accepted: 11/25/2019] [Indexed: 12/25/2022] Open
Abstract
This review highlights recent advances in the understanding of the relevance of altered lipid metabolic pathways contributing to the poor prognosis of high grade serous ovarian cancer, as they relate to cancer metastasis and cancer stemness. Increased lipid uptake regulated by the receptor CD36 and the transport protein FABP4 has been implicated in ovarian cancer metastasis. The symbiotic relationship between ovarian cancer cells and adipocytes was shown to be important for sustaining widespread peritoneal and omental metastasis. Increased lipogenesis dependent on the fatty acid desaturase SCD1 was detected in ovarian cancer stem cells. Furthermore, response to therapy, specifically to platinum, was linked to increased fatty acid biogenesis, while the survival of drug tolerant cells was shown to depend on lipid peroxidation. These recent findings suggest that lipids are necessary elements supporting oncogenic signaling and the energetic needs of rapidly proliferating cancer cells. New strategies targeting key enzymes involved in lipid uptake or utilization in cancer cells have been shown to exert anti-tumor effects and are being developed as cancer interventions in combination with chemotherapy or immunotherapy.
Collapse
|
21
|
Begicevic RR, Arfuso F, Falasca M. Bioactive lipids in cancer stem cells. World J Stem Cells 2019; 11:693-704. [PMID: 31616544 PMCID: PMC6789187 DOI: 10.4252/wjsc.v11.i9.693] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/08/2019] [Accepted: 08/20/2019] [Indexed: 02/06/2023] Open
Abstract
Tumours are known to be a heterogeneous group of cells, which is why they are difficult to eradicate. One possible cause for this is the existence of slow-cycling cancer stem cells (CSCs) endowed with stem cell-like properties of self-renewal, which are responsible for resistance to chemotherapy and radiotherapy. In recent years, the role of lipid metabolism has garnered increasing attention in cancer. Specifically, the key roles of enzymes such as stearoyl-CoA desaturase-1 and 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase in CSCs, have gained particular interest. However, despite accumulating evidence on the role of proteins in controlling lipid metabolism, very little is known about the specific role played by lipid products in CSCs. This review highlights recent findings on the role of lipid metabolism in CSCs, focusing on the specific mechanism by which bioactive lipids regulate the fate of CSCs and their involvement in signal transduction pathways.
Collapse
Affiliation(s)
- Romana-Rea Begicevic
- Metabolic Signalling Group, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
| | - Marco Falasca
- Metabolic Signalling Group, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
| |
Collapse
|
22
|
Choi S, Yoo YJ, Kim H, Lee H, Chung H, Nam MH, Moon JY, Lee HS, Yoon S, Kim WY. Clinical and biochemical relevance of monounsaturated fatty acid metabolism targeting strategy for cancer stem cell elimination in colon cancer. Biochem Biophys Res Commun 2019; 519:100-105. [PMID: 31481234 DOI: 10.1016/j.bbrc.2019.08.137] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 08/24/2019] [Indexed: 12/12/2022]
Abstract
Lipid metabolism is associated with colon cancer prognosis and incidence. Stearoyl-CoA desaturase 1 (SCD1), which converts fully saturated fatty acids (SFAs) to monounsaturated fatty acids (MUFAs), has been suggested as a vulnerable target for selective elimination of cancer stem cells (CSCs). However, the clinical significance and physiological role of SCD1 in CSCs has not been well demonstrated. Here, we showed the clinical and biochemical relevance of blocking SCD1 to target CSCs by analyzing human colon cancer data from TCGA and through lipidomic profiling of CSCs with or without SCD1 inhibition using mass spectrometry. Positive associations between SCD1 expression and colorectal cancer patient clinical status and the expression of CSC-related genes (WNT and NOTCH signaling) were found based on TCGA data analysis. Lipidomic profiling of CSCs and bulk cancer cells (BCCs) using mass spectrometry revealed that colon CSCs contained a distinctive lipid profile, with higher free MUFA and lower free SFA levels than in BCCs, suggesting that enhanced SCD1 activity generates MUFAs that may support WNT signaling in CSCs. In addition, all identified phosphatidyl-ethanolamine-containing MUFAs were found at higher levels in CSCs. Interestingly, we observed lower phosphatidyl-serine (18:1/18:0), phosphatidyl-choline (PC; p-18:0/18:1)), and sphingomyelin (SM; d18:1/20:0 or d16:1/22:0) levels in CSCs than in BCCs. Of those, SCD1 inhibition, which efficiently diminished free MUFA levels, increased those specific PC and SM and MUFAs in CSCs promptly. These results suggest that these specific lipid composition is critical for CSC stem cell maintenance. In addition, not only free MUFAs, which are known to be required for WNT signaling, but also other phospholipids, such as SM, which are important for lipid raft formation, may mediate other cell signaling pathways that support CSC maintenance. Comparison of the lipidomic profiles of colon cancer cells with those of previously reported for glioma cells further demonstrated the tissue specific characteristics of lipid metabolism in CSCs.
Collapse
Affiliation(s)
- SeokGyeong Choi
- College of Pharmacy, Sookmyung Women's University, Seoul, 04312, South Korea
| | - Young Ji Yoo
- College of Pharmacy, Sookmyung Women's University, Seoul, 04312, South Korea
| | - Hyejin Kim
- College of Pharmacy, Sookmyung Women's University, Seoul, 04312, South Korea
| | - Hani Lee
- College of Pharmacy, Sookmyung Women's University, Seoul, 04312, South Korea
| | - Hayung Chung
- Environmental Risk and Welfare Research Team, Korea Basic Science Institute, Seoul, 02841, South Korea
| | - Myung-Hee Nam
- Environmental Risk and Welfare Research Team, Korea Basic Science Institute, Seoul, 02841, South Korea
| | - Ju-Yeon Moon
- Drug Metabolism & Bioanalysis Laboratory, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, South Korea
| | - Hye Suk Lee
- Drug Metabolism & Bioanalysis Laboratory, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, South Korea
| | - Sukjoon Yoon
- Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, South Korea
| | - Woo-Young Kim
- College of Pharmacy, Sookmyung Women's University, Seoul, 04312, South Korea; Research Institute of Pharmaceutical Sciences, Sookmyung Women's University, Seoul, 04312, South Korea.
| |
Collapse
|
23
|
Dissecting phenotypic responses of the druggable targetome in cancers. Sci Rep 2019; 9:12513. [PMID: 31467349 PMCID: PMC6715751 DOI: 10.1038/s41598-019-48989-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/16/2019] [Indexed: 02/02/2023] Open
Abstract
Although a large amount of screening data comprising target genes and/or drugs tested against cancer cell line panels are available, different assay conditions and readouts limit the integrated analysis and batch-to-batch comparison of these data. Here, we systematically produced and analyzed the anticancer effect of the druggable targetome to understand the varied phenotypic outcomes of diverse functional classes of target genes. A library of siRNAs targeting ~4,800 druggable genes was screened against cancer cell lines under 2D and/or 3D assay conditions. The anticancer effect was simultaneously measured by quantifying cell proliferation and/or viability. Hit rates varied significantly depending on assay conditions and/or phenotypic readouts. Functional classes of hit genes were correlated with the microenvironment difference between the 2D monolayer cell proliferation and 3D sphere formation assays. Furthermore, multiplexing of cell proliferation and viability measures enabled us to compare the sensitivity and resistance responses to the gene knockdown. Many target genes that inhibited cell proliferation increased the single-cell-level viability of surviving cells, leading to an increase in self-renewal potential. In this study, combinations of parallel 2D/3D assays and multiplexing of cell proliferation and viability measures provided functional insights into the varied phenotypic outcomes of the cancer targetome.
Collapse
|
24
|
Therapeutic targeting of lipid synthesis metabolism for selective elimination of cancer stem cells. Arch Pharm Res 2018; 42:25-39. [PMID: 30536027 DOI: 10.1007/s12272-018-1098-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/01/2018] [Indexed: 02/06/2023]
Abstract
Cancer stem cells (CSCs) are believed to have an essential role in tumor resistance and metastasis; however, no therapeutic strategy for the selective elimination of CSCs has been established. Recently, several studies have shown that the metabolic regulation for ATP synthesis and biological building block generation in CSCs are different from that in bulk cancer cells and rather similar to that in normal tissue stem cells. To take advantage of this difference for CSC elimination therapy, many studies have tested the effect of blocking these metabolism. Two specific processes for lipid biosynthesis, i.e., fatty acid unsaturation and cholesterol biosynthesis, have been shown to be very effective and selective for CSC targets. In this review, lipid metabolism specific to CSCs are summarized. In addition, how monounsaturated fatty acid and cholesterol synthesis may contribute to CSC maintenance are discussed. Specifically, the molecular mechanism required for lipid synthesis and essential for stem cell biology is highlighted. The limit and preview of the lipid metabolism targeting for CSCs are also discussed.
Collapse
|
25
|
Gao F, Yu X, Meng R, Wang J, Jia L. STARD13 is positively correlated with good prognosis and enhances 5-FU sensitivity via suppressing cancer stemness in hepatocellular carcinoma cells. Onco Targets Ther 2018; 11:5371-5381. [PMID: 30214243 PMCID: PMC6126513 DOI: 10.2147/ott.s170775] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background STARD13 has been revealed to suppress tumor progression. However, the roles in regulating the stemness of hepatocellular carcinoma (HCC) cells are unclear. Methods Quantitative real-time PCR (qRT-PCR) was used to detect STARD13 expression in HCC tissues and normal adjacent tissues. Kaplan Meier (KM)-plotter analysis was performed to analyze the correlation between STARD13 expression and overall survival of HCC patients. Cell spheroid formation and ALDH1 activity analysis were carried out to detect the effects of STARD13 on the stemness of HCC cells. Furthermore, immunofluorescent, luciferase reporter, RhoA GTPase and F-actin visualization assays were performed to explore the mechanisms contributing to STARD13-mediated effects. Results STARD13 expression was significantly downregulated in HCC tissues compared with normal adjacent tissues, and was positively correlated with the overall survival of HCC patients. Functionally, overexpression of STARD13 inhibited cells stemness and enhanced 5-FU sensitivity in HCC cells. Mechanistically, STRAD13 overexpression suppressed RhoGTPase signaling and thus inhibited transcriptional factor YAP translocation from nuclear to cytoplasm, leading to the downregulation of transcriptional activity of YAP. Notably, the inhibitory effects of STARD13 on HCC cells stemness and 5-FU sensitivity were rescued by RhoA or YAP-5SA overexpression. Conclusion Our results indicate that STARD13 could enhances 5-FU sensitivity by suppressing cancer stemness in hepatocellular carcinoma cells via attenuating YAP transcriptional activity.
Collapse
Affiliation(s)
- Fei Gao
- Department of Oncology, Sichuan Mental Health Center, The Third Hospital of Mianyang, Mianyang, People's Republic of China,
| | - Xiaolin Yu
- Department of Oncology, AVIC 363 Hospital, Chengdu, People's Republic of China
| | - Rongqin Meng
- Department of Oncology, AVIC 363 Hospital, Chengdu, People's Republic of China
| | - Jisheng Wang
- Department of Oncology, Sichuan Mental Health Center, The Third Hospital of Mianyang, Mianyang, People's Republic of China,
| | - Lin Jia
- Department of Oncology, Sichuan Mental Health Center, The Third Hospital of Mianyang, Mianyang, People's Republic of China,
| |
Collapse
|
26
|
Six2 is negatively correlated with good prognosis and decreases 5-FU sensitivity via suppressing E-cadherin expression in hepatocellular carcinoma cells. Biomed Pharmacother 2018; 104:204-210. [PMID: 29772441 DOI: 10.1016/j.biopha.2018.05.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/08/2018] [Accepted: 05/08/2018] [Indexed: 11/20/2022] Open
Abstract
This work aims to study the roles and related mechanisms of six2 in 5-FU sensitivity of hepatocellular carcinoma (HCC) cells. KM-Plotter analysis showed that HCC patients with higher six2 expression levels had shorter overall survival. Six2 expression was higher in clinical HCC tissues than in normal tissues, and was negatively correlated with E-cadherin expression. Additionally, six2 overexpression decreased the sensitivity of HCC cells to 5-Fu, characterized as attenuating 5-FU-induced cell apoptosis and downregulation of cell viability, and promoted HCC cells stemness. Mechanistically, six2 overexpression repressed E-cadherin expression via stimulating promoter methylation of the E-cadherin. And E-cadherin overexpression rescued six2-induced decrease of 5-FU sensitivity and promotion on HCC cells stemness. Therefore, our results suggest that Six2 is negatively correlated with good prognosis and decreases 5-FU sensitivity via suppressing E-cadherin expression in HCC cells.
Collapse
|
27
|
Carnosol controls the human glioblastoma stemness features through the epithelial-mesenchymal transition modulation and the induction of cancer stem cell apoptosis. Sci Rep 2017; 7:15174. [PMID: 29123181 PMCID: PMC5680298 DOI: 10.1038/s41598-017-15360-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 10/25/2017] [Indexed: 12/18/2022] Open
Abstract
A high cell proliferation rate, invasiveness and resistance to chemotherapy are the main features of glioblastoma (GBM). GBM aggressiveness has been widely associated both with a minor population of cells presenting stem-like properties (cancer stem-like cells, CSCs) and with the ability of tumor cells to acquire a mesenchymal phenotype (epithelial-mesenchymal transition, EMT). Carnosol (CAR), a natural inhibitor of MDM2/p53 complex, has been attracted attention for its anti-cancer effects on several tumor types, including GBM. Herein, the effects of CAR on U87MG-derived CSC viability and stemness features were evaluated. CAR decreased the rate of CSC formation and promoted the CSC apoptotic cell death through p53 functional reactivation. Moreover, CAR was able to control the TNF-α/TGF-β-induced EMT, counteracting the effects of the cytokine on EMT master regulator genes (Slug, Snail, Twist and ZEB1) and modulating the activation of miR-200c, a key player in the EMT process. Finally, CAR was able to increase the temozolomide (TMZ) anti-proliferative effects. These findings demonstrate that CAR affected the different intracellular mechanism of the complex machinery that regulates GBM stemness. For the first time, the diterpene was highlighted as a promising lead for the development of agents able to decrease the stemness features, thus controlling GBM aggressiveness.
Collapse
|
28
|
Transcriptome modeling and phenotypic assays for cancer precision medicine. Arch Pharm Res 2017; 40:906-914. [PMID: 28766239 DOI: 10.1007/s12272-017-0940-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 07/27/2017] [Indexed: 01/02/2023]
Abstract
Cancer precision medicine requires clinically actionable biomarkers for patient stratification and a better prediction of clinical outcome. Although thousands of cancer-enriched mutated genes have been reported by global sequencing projects, to date, only a few oncogenic mutations have been confirmed as effective biomarkers in cancer therapies. The low frequency and varied profile (i.e., allele frequency, mutation position) of mutant genes among cancer types limit the utility of predictive biomarkers. The recent explosion of cancer transcriptome and phenotypic screening data provides another opportunity for finding transcript-level biomarkers and targets, thus overcoming the limitation of cancer mutation analyses. Technological developments enable the rapid and extensive discovery of potential target-biomarker combinations from large-scale transcriptome-level screening combined with physiologically relevant phenotypic assays. Here, we summarized recent progress as well as discussed the outlook of transcriptome-oriented data mining strategies and phenotypic assays for the identification of non-genetic biomarkers and targets in cancer drug discovery.
Collapse
|
29
|
Lipid Storage and Autophagy in Melanoma Cancer Cells. Int J Mol Sci 2017; 18:ijms18061271. [PMID: 28617309 PMCID: PMC5486093 DOI: 10.3390/ijms18061271] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 06/12/2017] [Accepted: 06/13/2017] [Indexed: 01/06/2023] Open
Abstract
Cancer stem cells (CSC) represent a key cellular subpopulation controlling biological features such as cancer progression in all cancer types. By using melanospheres established from human melanoma patients, we compared less differentiated melanosphere-derived CSC to differentiating melanosphere-derived cells. Increased lipid uptake was found in melanosphere-derived CSC vs. differentiating melanosphere-derived cells, paralleled by strong expression of lipogenic factors Sterol Regulatory Element-Binding Protein-1 (SREBP-1) and Peroxisome Proliferator-Activated Receptor-γ (PPAR-γ). An inverse relation between lipid-storing phenotype and autophagy was also found, since microtubule-associated protein 1A/1B-Light Chain 3 (LC3) lipidation is reduced in melanosphere-derived CSC. To investigate upstream autophagy regulators, Phospho-AMP activated Protein Kinase (P-AMPK) and Phospho-mammalian Target of Rapamycin (P-mTOR) were analyzed; lower P-AMPK and higher P-mTOR expression in melanosphere-derived CSC were found, thus explaining, at least in part, their lower autophagic activity. In addition, co-localization of LC3-stained autophagosome spots and perilipin-stained lipid droplets was demonstrated mainly in differentiating melanosphere-derived cells, further supporting the role of autophagy in lipid droplets clearance. The present manuscript demonstrates an inverse relationship between lipid-storing phenotype and melanoma stem cells differentiation, providing novel indications involving autophagy in melanoma stem cells biology.
Collapse
|
30
|
Wong TL, Che N, Ma S. Reprogramming of central carbon metabolism in cancer stem cells. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1728-1738. [PMID: 28502706 DOI: 10.1016/j.bbadis.2017.05.012] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/23/2017] [Accepted: 05/10/2017] [Indexed: 12/19/2022]
Abstract
Cancer metabolism has been studied for years and adopted in the clinic for monitoring disease progression and therapy response. Despite our growing knowledge of a distinctly altered metabolic behavior in cancer, drugs targeting cancer metabolism have remained less than promising. Recent efforts in cancer stem cell (CSC) biology suggest that a subpopulation of tumor-initiating cells within the tumor bulk represents the root of tumor recurrence and therapy resistance. In recent years, metabolic phenotype of CSCs of various tumor types has been identified. This breakthrough has shed light on the underlying mechanism by which CSCs maintain stemness, confer resistance to therapies and initiate tumor relapse. The distinct metabolic characteristics of CSCs compared to non-CSCs provide an opportunity to target CSCs more specifically and have become a major focus in cancer research in recent years with substantial efforts conducted towards discovering clinical targets. This perspective article summarizes the current knowledge of CSC metabolism in carcinogenesis and highlights the potential of targeting CSC metabolism for therapy.
Collapse
Affiliation(s)
- Tin Lok Wong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Noélia Che
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Stephanie Ma
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; State Key Laboratory for Liver Research, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.
| |
Collapse
|