1
|
Hou Y, Fu Z, Wang C, Kucharzewska P, Guo Y, Zhang S. 27-Hydroxycholesterol in cancer development and drug resistance. J Enzyme Inhib Med Chem 2025; 40:2507670. [PMID: 40401382 PMCID: PMC12100970 DOI: 10.1080/14756366.2025.2507670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 04/25/2025] [Accepted: 05/13/2025] [Indexed: 05/23/2025] Open
Abstract
27-Hydroxycholesterol (27HC), a cholesterol metabolite, functions both as a selective oestrogen receptor (ER) modulator and a ligand for liver X receptors (LXRs). The discovery of 27HC involvement in carcinogenesis has unveiled new research avenues, yet its precise role remains controversial and context-dependent. In this review, we provide an overview of the biosynthesis and metabolism of 27HC and explore its cancer-associated signalling, with a particular focus on ER- and LXR-mediated pathways. Given the tissue-specific dual role of 27HC, we discuss its differential impact across various cancer types. Furthermore, we sort out 27HC-contributed drug resistance mechanisms from the perspectives of drug efflux, cellular proliferation, apoptosis, epithelial-mesenchymal transition (EMT), antioxidant defence, epigenetic modification, and metabolic reprogramming. Finally, we highlight the chemical inhibitors to mitigate 27HC-driven cancer progression and drug resistance. This review offers an updated role of 27HC in cancer biology, setting the stage for future research and the development of targeted therapeutics.
Collapse
Affiliation(s)
- Yaxin Hou
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, China
| | - Zhiguang Fu
- Department of Tumor Radiotherapy, Air Force Medical Center, People’s Liberation Army of China (PLA), Beijing, China
| | - Chenhui Wang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, China
| | - Paulina Kucharzewska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, Warsaw, Poland
| | - Yuan Guo
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, China
| | - Sihe Zhang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
2
|
Ewendt F, Janjetovic Z, Kim TK, Mobley AA, Brożyna AA, Ravichandran S, Fabisiak A, Brzeminski P, Sicinski RR, Stangl GI, Tuckey RC, Slominski AT. The vitamin D 3 hormone, 1,25(OH) 2D 3, regulates fibroblast growth factor 23 (FGF23) production in human skin cells. Am J Physiol Cell Physiol 2025; 328:C1177-C1192. [PMID: 40055144 DOI: 10.1152/ajpcell.00827.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/11/2024] [Accepted: 02/12/2025] [Indexed: 03/25/2025]
Abstract
The bone hormone fibroblast growth factor 23 (FGF23) regulates renal phosphate reabsorption and the enzymatic production of active vitamin D3 [1,25(OH)2D3]. Therefore, FGF23 production in bone cells is closely regulated by 1,25(OH)2D3 acting via the vitamin D receptor (VDR). Skin cells can produce hydroxyvitamin D3 metabolites from its precursor D3 made through ultraviolet B light exposure. Interestingly, the expression of Fgf23 has been found in rodent skin, but its expression, regulation, and role in human skin are unclear. Therefore, we investigated whether hydroxyvitamin D3 metabolites regulate FGF23 in human skin cells. Primary adult and neonatal epidermal keratinocytes (HEKn), melanocytes (HEMn), dermal fibroblasts (HDFn), as well as human melanoma cells, HaCaT, HaCaT VDR KO, and A431 epidermoid cells, were used to assess FGF23 gene expression (quantitative reverse-transcription real-time PCR), cellular FGF23 protein (Western blot), or secreted FGF23 protein (ELISA) after treatment with hydroxyvitamin D3 metabolites. HaCaT cells treated with recombinant FGF23 were used to explore its function in skin. Human skin cells can synthesize FGF23. Treatment with 1,25(OH)2D3 significantly increased FGF23 mRNA levels in HaCaT and HDFn cells, and moderately in HEKn cells, mediated in part by the VDR. It also moderately enhanced mRNA levels of the FGF23-processing enzyme GALNT3 and stimulated secretion of hormonally active FGF23 from HaCaT cells. Treatment of HaCaT cells with FGF23 increased mRNA levels of the cholesterol- and vitamin D-metabolizing enzymes, CYP11A1 and CYP27A1. In conclusion, human skin cells express and secrete FGF23, which is regulated by 1,25(OH)2D3 acting in part by the VDR. FGF23 affects the expression of cutaneous sterol-metabolizing enzymes.NEW & NOTEWORTHY This study shows for the first time the expression and secretion of the FGF23 hormone by human skin cells. In addition, we identified the active vitamin D3 hormone, 1,25(OH)2D3, to be a potent regulator of dermal FGF23 expression and protein secretion, partly involving the vitamin D receptor. Furthermore, we provide initial evidence demonstrating that FGF23 upregulates the gene expression of CYP11A1 and CYP27A1 in keratinocytes.
Collapse
Affiliation(s)
- Franz Ewendt
- Department of Dermatology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Zorica Janjetovic
- Department of Dermatology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Tae-Kang Kim
- Department of Dermatology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Alisa A Mobley
- Department of Dermatology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Anna A Brożyna
- Department of Human Biology, Institute of Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Toruń, Poland
| | - Senthilkumar Ravichandran
- Department of Dermatology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Adrian Fabisiak
- Department of Chemistry, University of Warsaw, Warsaw, Poland
| | | | | | - Gabriele I Stangl
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Robert C Tuckey
- School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Andrzej T Slominski
- Department of Dermatology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
- Cancer Chemoprevention Program, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
- VA Medical Center, Birmingham, Alabama, United States
| |
Collapse
|
3
|
Liu Y, Qin J, Li X, Wu G. Oxysterols in tumor immune microenvironment (TIME). J Steroid Biochem Mol Biol 2025; 245:106634. [PMID: 39551164 DOI: 10.1016/j.jsbmb.2024.106634] [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: 07/29/2024] [Revised: 10/22/2024] [Accepted: 11/09/2024] [Indexed: 11/19/2024]
Abstract
Oxysterols are compounds generated through oxidative reactions involving cholesterol and other steroid molecules. They play a crucial role in the tumor immune microenvironment by interacting with molecules such as the cell membrane receptor EBI2 and nuclear receptors like LXR and PXR. This interaction regulates immune cell signaling pathways, affecting proliferation, apoptosis, migration, and invasion in tumor-related processes. Activating these receptors alters the function and behavior of immune cells-such as macrophages, T cells, and dendritic cells-within the tumor microenvironment, thus promoting or inhibiting tumor development. Certain oxidized steroids can increase both the number and activation of infiltrating T cells, synergizing with anti-PD-1 to enhance anti-tumor efficacy. An in-depth study of the biological mechanisms of oxidized sterols will not only enhance our understanding of the complexity of the tumor immune microenvironment but may also reveal new therapeutic targets, providing innovative strategies for tumor immunotherapy.
Collapse
Affiliation(s)
- Yuanxin Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Jie Qin
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Xiaorui Li
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China.
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
4
|
Woo SY, Shim WS, Lee H, Baryawno N, Song P, Kim BS, Yoon S, Oh SO, Lee D. 27-Hydroxycholesterol Negatively Affects the Function of Bone Marrow Endothelial Cells in the Bone Marrow. Int J Mol Sci 2024; 25:10517. [PMID: 39408846 PMCID: PMC11477443 DOI: 10.3390/ijms251910517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Hematopoietic stem cells (HSCs) reside in specific microenvironments that facilitate their regulation through both internal mechanisms and external cues. Bone marrow endothelial cells (BMECs), which are found in one of these microenvironments, play a vital role in controlling the self-renewal and differentiation of HSCs during hematological stress. We previously showed that 27-hydroxycholesterol (27HC) administration of exogenous 27HC negatively affected the population of HSCs and progenitor cells by increasing the reactive oxygen species levels in the bone marrow. However, the effect of 27HC on BMECs is unclear. To determine the function of 27HC in BMECs, we employed magnetic-activated cell sorting to isolate CD31+ BMECs and CD31- cells. We demonstrated the effect of 27HC on CD31+ BMECs and HSCs. Treatment with exogenous 27HC led to a decrease in the number of BMECs and reduced the expression of adhesion molecules that are crucial for maintaining HSCs. Our results demonstrate that BMECs are sensitively affected by 27HC and are crucial for HSC survival.
Collapse
Affiliation(s)
- Soo-Yeon Woo
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.-Y.W.); (W.-S.S.); (H.L.); (P.S.)
| | - Wan-Seog Shim
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.-Y.W.); (W.-S.S.); (H.L.); (P.S.)
| | - Hyejin Lee
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.-Y.W.); (W.-S.S.); (H.L.); (P.S.)
| | - Ninib Baryawno
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden;
| | - Parkyong Song
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.-Y.W.); (W.-S.S.); (H.L.); (P.S.)
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Sik Yoon
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.Y.); (S.-O.O.)
| | - Sae-Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.Y.); (S.-O.O.)
| | - Dongjun Lee
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.-Y.W.); (W.-S.S.); (H.L.); (P.S.)
- Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| |
Collapse
|
5
|
Wang X, Zhong F, Chen T, Wang H, Wang W, Jin H, Li C, Guo X, Liu Y, Zhang Y, Li B. Cholesterol neutralized vemurafenib treatment by promoting melanoma stem-like cells via its metabolite 27-hydroxycholesterol. Cell Mol Life Sci 2024; 81:226. [PMID: 38775844 PMCID: PMC11111659 DOI: 10.1007/s00018-024-05267-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/14/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024]
Abstract
Vemurafenib has been used as first-line therapy for unresectable or metastatic melanoma with BRAFV600E mutation. However, overall survival is still limited due to treatment resistance after about one year. Therefore, identifying new therapeutic targets for melanoma is crucial for improving clinical outcomes. In the present study, we found that lowering intracellular cholesterol by knocking down DHCR24, the limiting synthetase, impaired tumor cell proliferation and migration and abrogated the ability to xenotransplant tumors. More importantly, administration of DHCR24 or cholesterol mediated resistance to vemurafenib and promoted the growth of melanoma spheroids. Mechanistically, we identified that 27-hydroxycholesterol (27HC), a primary metabolite of cholesterol synthesized by the enzyme cytochrome P450 27A1 (CYP27A1), reproduces the phenotypes induced by DHCR24 or cholesterol administration and activates Rap1-PI3K/AKT signaling. Accordingly, CYP27A1 is highly expressed in melanoma patients and upregulated by DHCR24 induction. Dafadine-A, a CYP27A1 inhibitor, attenuates cholesterol-induced growth of melanoma spheroids and abrogates the resistance property of vemurafenib-resistant melanoma cells. Finally, we confirmed that the effects of cholesterol on melanoma resistance require its metabolite 27HC through CYP27A1 catalysis, and that 27HC further upregulates Rap1A/Rap1B expression and increases AKT phosphorylation. Thus, our results suggest that targeting 27HC may be a useful strategy to overcome treatment resistance in metastatic melanoma.
Collapse
Affiliation(s)
- Xiaohong Wang
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-Related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Feiliang Zhong
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, People's Republic of China
| | - Tingting Chen
- School of Basic Medicine, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Hongbo Wang
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-Related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Weifang Wang
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-Related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Hongkai Jin
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-Related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Chouyang Li
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Xuan Guo
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-Related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Ying Liu
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-Related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Yu Zhang
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-Related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China.
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China.
| | - Bo Li
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-Related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China.
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China.
| |
Collapse
|
6
|
Dias IHK, Shokr H. Oxysterols as Biomarkers of Aging and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:307-336. [PMID: 38036887 DOI: 10.1007/978-3-031-43883-7_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Oxysterols derive from either enzymatic or non-enzymatic oxidation of cholesterol. Even though they are produced as intermediates of bile acid synthesis pathway, they are recognised as bioactive compounds in cellular processes. Therefore, their absence or accumulation have been shown to be associated with disease phenotypes. This chapter discusses the contribution of oxysterol to ageing, age-related diseases such as neurodegeneration and various disorders such as cancer, cardiovascular disease, diabetes, metabolic and ocular disorders. It is clear that oxysterols play a significant role in development and progression of these diseases. As a result, oxysterols are being investigated as suitable markers for disease diagnosis purposes and some drug targets are in development targeting oxysterol pathways. However, further research will be needed to confirm the suitability of these potentials.
Collapse
Affiliation(s)
- Irundika H K Dias
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, UK.
| | - Hala Shokr
- Manchester Pharmacy School, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| |
Collapse
|
7
|
Dai L, Wang J, Zhang X, Yan M, Zhou L, Zhang G, Meng L, Chen L, Cao X, Zhang Z, Wang G, Zhang Z. 27-Hydroxycholesterol Drives the Spread of α-Synuclein Pathology in Parkinson's Disease. Mov Disord 2023; 38:2005-2018. [PMID: 37593929 DOI: 10.1002/mds.29577] [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: 03/18/2023] [Accepted: 07/28/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND The accumulation and aggregation of α-synuclein (α-Syn) are characteristic of Parkinson's disease (PD). Epidemiological evidence indicates that hyperlipidemia is associated with an increased risk of PD. The levels of 27-hydroxycholesterol (27-OHC), a cholesterol oxidation derivative, are increased in the brain and cerebrospinal fluid of patients with PD. However, whether 27-OHC plays a role in α-Syn aggregation and propagation remains elusive. OBJECTIVE The aim of this study was to determine whether 27-OHC regulates α-Syn aggregation and propagation. METHODS Purified recombinant α-Syn, neuronal cultures, and α-Syn fibril-injected mouse model of PD were treated with 27-OHC. In addition, CYP27A1 knockout mice were used to investigate the effect of lowering 27-OHC on α-Syn pathology in vivo. RESULTS 27-OHC accelerates the aggregation of α-Syn and enhances the seeding activity of α-Syn fibrils. Furthermore, the 27-OHC-modified α-Syn fibrils localize to the mitochondria and induce mitochondrial dysfunction and neurotoxicity. Injection of 27-OHC-modified α-Syn fibrils induces enhanced spread of α-Syn pathology and dopaminergic neurodegeneration compared with pure α-Syn fibrils. Similarly, subcutaneous administration of 27-OHC facilitates the seeding of α-Syn pathology. Genetic deletion of cytochrome P450 27A1 (CYP27A1), the enzyme that converts cholesterol to 27-OHC, ameliorates the spread of pathologic α-Syn, degeneration of the nigrostriatal dopaminergic pathway, and motor impairments. These results indicate that the cholesterol metabolite 27-OHC plays an important role in the pathogenesis of PD. CONCLUSIONS 27-OHC promotes the aggregation and spread of α-Syn. Strategies aimed at inhibiting the CYP27A1-27-OHC axis may hold promise as a disease-modifying therapy to halt the progression of α-Syn pathology in PD. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Lijun Dai
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiannan Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xingyu Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mingmin Yan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lingyan Zhou
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guoxin Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Liam Chen
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Xuebing Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
8
|
Roumain M, Guillemot-Legris O, Ameraoui H, Alhouayek M, Muccioli GG. Identification and in vivo detection of side-chain hydroxylated metabolites of 4β-hydroxycholesterol. J Steroid Biochem Mol Biol 2023; 234:106376. [PMID: 37604319 DOI: 10.1016/j.jsbmb.2023.106376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 08/13/2023] [Accepted: 08/18/2023] [Indexed: 08/23/2023]
Abstract
Oxysterols are oxidized derivatives of cholesterol that are formed by enzymatic processes or through the action of reactive oxygen species. Several of these bioactive lipids have been shown to be affected and/or play a role in inflammatory processes. 4β-hydroxycholesterol is one of the major oxysterols in mice and humans and its levels are affected by inflammatory diseases. However, apart from its long half-life, little is known about its catabolism. By incubating 4β-hydroxycholesterol with mouse mitochondria-enriched liver fractions, as well as 25-hydroxycholesterol and 27-hydroxycholesterol with recombinant CYP3A4, we identified 4β,25-dihydroxycholesterol and 4β,27-dihydroxycholesterol as 4β-hydroxycholesterol metabolites. Supporting the biological relevance of this metabolism, we detected both metabolites after incubation of J774, primary mouse peritoneal macrophages and PMA-differentiated THP-1 cells with 4β-hydroxycholesterol. Across our experiments, the incubation of cells with lipopolysaccharides differentially affected the levels of the 25- and 27-hydroxylated metabolites of 4β-hydroxycholesterol. Finally, 4β,27-dihydroxycholesterol was also detected in mice liver and plasma after intraperitoneal administration of 4β-hydroxycholesterol. To our knowledge, this is the first report of the in vitro and in vivo detection and quantification of 4β-hydroxycholesterol metabolites.
Collapse
Affiliation(s)
- Martin Roumain
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Belgium
| | - Owein Guillemot-Legris
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Belgium
| | - Hafsa Ameraoui
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Belgium
| | - Mireille Alhouayek
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Belgium.
| |
Collapse
|
9
|
DeRouen MC, Yang J, Li Y, Franke AA, Tome AN, White KK, Hernandez BY, Shvetsov Y, Setiawan V, Wu AH, Wilkens LR, Le Marchand L, Loo LWM, Cheng I. Circulating 27-hydroxycholesterol, lipids, and steroid hormones in breast cancer risk: a nested case-control study of the Multiethnic Cohort Study. Breast Cancer Res 2023; 25:95. [PMID: 37580793 PMCID: PMC10424359 DOI: 10.1186/s13058-023-01693-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 08/01/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Laboratory studies have indicated that a cholesterol metabolite and selective estrogen receptor modulator, 27-hydroxycholesterol (27HC), may be important in breast cancer etiology and explain associations between obesity and postmenopausal breast cancer risk. Epidemiologic evidence for 27HC in breast cancer risk is limited, particularly in multiethnic populations. METHODS In a nested case-control study of 1470 breast cancer cases and 1470 matched controls within the Multiethnic Cohort Study, we examined associations of pre-diagnostic circulating 27HC with breast cancer risk among African American, Japanese American, Native Hawaiian, Latino, and non-Latino White postmenopausal females. We used multivariable logistic regression adjusted for age, education, parity, body mass index, and smoking status. Stratified analyses were conducted across racial and ethnic groups, hormone receptor (HR) status, and use of lipid-lowering drugs. We assessed interactions of 27HC with steroid hormones. RESULTS 27HC levels were inversely related to breast cancer risk (odds ratio [OR] 0.80; 95% confidence interval [CI] 0.58, 1.12), but the association was not statistically significant in the full model. Directions of associations differed by racial and ethnic group. Results suggested an inverse association with HR-negative breast cancer (OR 0.46; 95% CI 0.20, 1.06). 27HC interacted with testosterone, but not estrone, on risk of breast cancer; 27HC was only inversely associated with risk among those with the highest levels of testosterone (OR 0.46; 95% CI 0.24, 0.86). CONCLUSION This is the first US study to examine circulating 27HC and breast cancer risk and reports a weak inverse association that varies across racial and ethnic groups and testosterone level.
Collapse
Affiliation(s)
- Mindy C DeRouen
- Department of Epidemiology and Biostatistics, School of Medicine, University of California San Francisco, 550 16th Street, San Francisco, CA, 94538, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, 550 16th Street, San Francisco, CA, 94538, USA.
| | - Juan Yang
- Department of Epidemiology and Biostatistics, School of Medicine, University of California San Francisco, 550 16th Street, San Francisco, CA, 94538, USA
| | - Yuqing Li
- Department of Epidemiology and Biostatistics, School of Medicine, University of California San Francisco, 550 16th Street, San Francisco, CA, 94538, USA
| | - Adrian A Franke
- Cancer Biology Program, University of Hawai'i Cancer Center, Honolulu, HI, USA
| | - Anne N Tome
- Population Sciences of the Pacific Program, University of Hawai'i Cancer Center, Honolulu, HI, USA
| | - Kami K White
- Population Sciences of the Pacific Program, University of Hawai'i Cancer Center, Honolulu, HI, USA
| | - Brenda Y Hernandez
- Population Sciences of the Pacific Program, University of Hawai'i Cancer Center, Honolulu, HI, USA
| | - Yurii Shvetsov
- Population Sciences of the Pacific Program, University of Hawai'i Cancer Center, Honolulu, HI, USA
| | - Veronica Setiawan
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Anna H Wu
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lynne R Wilkens
- Population Sciences of the Pacific Program, University of Hawai'i Cancer Center, Honolulu, HI, USA
| | - Loïc Le Marchand
- Population Sciences of the Pacific Program, University of Hawai'i Cancer Center, Honolulu, HI, USA
| | - Lenora W M Loo
- Cancer Biology Program, University of Hawai'i Cancer Center, Honolulu, HI, USA
| | - Iona Cheng
- Department of Epidemiology and Biostatistics, School of Medicine, University of California San Francisco, 550 16th Street, San Francisco, CA, 94538, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, 550 16th Street, San Francisco, CA, 94538, USA.
| |
Collapse
|
10
|
Lee-Rueckert M, Canyelles M, Tondo M, Rotllan N, Kovanen PT, Llorente-Cortes V, Escolà-Gil JC. Obesity-induced changes in cancer cells and their microenvironment: Mechanisms and therapeutic perspectives to manage dysregulated lipid metabolism. Semin Cancer Biol 2023; 93:36-51. [PMID: 37156344 DOI: 10.1016/j.semcancer.2023.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/05/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
Obesity has been closely related to cancer progression, recurrence, metastasis, and treatment resistance. We aim to review recent progress in the knowledge on the obese macroenvironment and the generated adipose tumor microenvironment (TME) inducing lipid metabolic dysregulation and their influence on carcinogenic processes. Visceral white adipose tissue expansion during obesity exerts systemic or macroenvironmental effects on tumor initiation, growth, and invasion by promoting inflammation, hyperinsulinemia, growth-factor release, and dyslipidemia. The dynamic relationship between cancer and stromal cells of the obese adipose TME is critical for cancer cell survival and proliferation as well. Experimental evidence shows that secreted paracrine signals from cancer cells can induce lipolysis in cancer-associated adipocytes, causing them to release free fatty acids and acquire a fibroblast-like phenotype. Such adipocyte delipidation and phenotypic change is accompanied by an increased secretion of cytokines by cancer-associated adipocytes and tumor-associated macrophages in the TME. Mechanistically, the availability of adipose TME free fatty acids and tumorigenic cytokines concomitant with the activation of angiogenic processes creates an environment that favors a shift in the cancer cells toward an aggressive phenotype associated with increased invasiveness. We conclude that restoring the aberrant metabolic alterations in the host macroenvironment and in adipose TME of obese subjects would be a therapeutic option to prevent cancer development. Several dietary, lipid-based, and oral antidiabetic pharmacological therapies could potentially prevent tumorigenic processes associated with the dysregulated lipid metabolism closely linked to obesity.
Collapse
Affiliation(s)
| | - Marina Canyelles
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Mireia Tondo
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Noemi Rotllan
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | | | - Vicenta Llorente-Cortes
- Wihuri Research Institute, Helsinki, Finland; Institute of Biomedical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain; CIBERCV, Institute of Health Carlos III, 28029 Madrid, Spain.
| | - Joan Carles Escolà-Gil
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.
| |
Collapse
|
11
|
Role of reactive oxygen species in regulating 27-hydroxycholesterol-induced apoptosis of hematopoietic progenitor cells and myeloid cell lines. Cell Death Dis 2022; 13:916. [PMID: 36316327 PMCID: PMC9622808 DOI: 10.1038/s41419-022-05360-0] [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: 07/17/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/07/2022]
Abstract
Oxysterols are oxygenated derivatives of cholesterol that contain an additional hydroxy, epoxide, or ketone group in the sterol nucleus and/or a hydroxyl group in the side chain of the cholesterol molecule. 27-Hydroxycholesterol (27HC) is a side-chain oxysterol that is oxygenated at the 27th carbon atom of cholesterol. The oxysterol (27HC) is produced via oxidation by sterol 27-hydroxylase (CYP27A1) and metabolized via oxysterol 7a-hydroxylase (CYP7B1) for bile acid synthesis in the liver. A previous study has demonstrated that treatment with the alternative Estrogen receptor alpha (ERα) ligand 27HC induces ERα-dependent hematopoietic stem cell (HSC) mobilization. In addition, Cyp27a1-deficient mice demonstrate significantly reduced 27HC levels and HSC mobilization. Here, we report that exogenous 27HC treatment leads to a substantial reduction in the hematopoietic stem and progenitor cell (HSPC) population owing to significantly increased reactive oxygen species (ROS) levels and apoptosis in the bone marrow (BM). However, 27HC does not influence the population of mature hematopoietic cells in the BM. Furthermore, exogenous 27HC treatment suppresses cell growth and promotes ROS production and apoptosis in leukemic cells. Moreover, acute myeloid leukemia (AML) patients with high CYP7B1 expression (expected to have inhibition of 27HC) had significantly shorter survival than those with low CYP7B1 expression (expected to have an elevation of 27HC). Single-cell RNA-sequencing (scRNA seq) analysis revealed that the expression of CYP7B1 was significantly increased in AML patients. Thus, our study suggests that 27HC may serve as a potent agent for regulating pools of HSPCs and may have an application as a novel therapeutic target for hematological malignancies. Collectively, pharmacological inhibition of CYP7B1 (expected to have an elevation of 27HC) would potentially have fewer long-term hematological side effects, particularly when used in combination with chemotherapy or radiation for the treatment of leukemia patients.
Collapse
|
12
|
Ma L, Cho W, Nelson ER. Our evolving understanding of how 27-hydroxycholesterol influences cancer. Biochem Pharmacol 2022; 196:114621. [PMID: 34043965 PMCID: PMC8611110 DOI: 10.1016/j.bcp.2021.114621] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 02/09/2023]
Abstract
Cholesterol has been implicated in the pathophysiology and progression of several cancers now, although the mechanisms by which it influences cancer biology are just emerging. Two likely contributing mechanisms are the ability for cholesterol to directly regulate signaling molecules within the membrane, and certain metabolites acting as signaling molecules. One such metabolite is the oxysterol 27-hydroxycholesterol (27HC), which is a primary metabolite of cholesterol synthesized by the enzyme Cytochrome P450 27A1 (CYP27A1). Physiologically, 27HC is involved in the regulation of cholesterol homeostasis and contributes to cholesterol efflux through liver X receptor (LXR) and inhibition of de novo cholesterol synthesis through the insulin-induced proteins (INSIGs). 27HC is also a selective modulator of the estrogen receptors. An increasing number of studies have identified its importance in cancer progression of various origins, especially in breast cancer. In this review, we discuss the physiological roles of 27HC targeting these two nuclear receptors and the subsequent contribution to cancer progression. We describe how 27HC promotes tumor growth directly through cancer-intrinsic factors, and indirectly through its immunomodulatory roles which lead to decreased immune surveillance and increased tumor invasion. This review underscores the importance of the cholesterol metabolic pathway in cancer progression and the potential therapeutic utility of targeting this metabolic pathway.
Collapse
Affiliation(s)
- Liqian Ma
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL
| | - Wonhwa Cho
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL
| | - Erik R. Nelson
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL,Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL,Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL,University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL,Carl R. Woese Institute for Genomic Biology, Anticancer Discovery from Pets to People Theme, University of Illinois Urbana-Champaign, Urbana, IL,To whom correspondence and reprint requests should be addressed: Erik R. Nelson. University of Illinois at Urbana-Champaign. 407 S Goodwin Ave (MC-114), Urbana, IL, 61801. Phone: 217-244-5477. Fax: 217-333-1133.
| |
Collapse
|
13
|
Garcia-Ruiz C, Conde de la Rosa L, Ribas V, Fernandez-Checa JC. MITOCHONDRIAL CHOLESTEROL AND CANCER. Semin Cancer Biol 2021; 73:76-85. [PMID: 32805396 PMCID: PMC7882000 DOI: 10.1016/j.semcancer.2020.07.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/22/2020] [Accepted: 07/29/2020] [Indexed: 12/11/2022]
Abstract
Cholesterol is a crucial component of membrane bilayers that determines their physical and functional properties. Cells largely satisfy their need for cholesterol through the novo synthesis from acetyl-CoA and this demand is particularly critical for cancer cells to sustain dysregulated cell proliferation. However, the association between serum or tissue cholesterol levels and cancer development is not well established as epidemiologic data do not consistently support this link. While most preclinical studies focused on the role of total celular cholesterol, the specific contribution of the mitochondrial cholesterol pool to alterations in cancer cell biology has been less explored. Although low compared to other bilayers, the mitochondrial cholesterol content plays an important physiological function in the synthesis of steroid hormones in steroidogenic tissues or bile acids in the liver and controls mitochondrial function. In addition, mitochondrial cholesterol metabolism generates oxysterols, which in turn, regulate multiple pathways, including cholesterol and lipid metabolism as well as cell proliferation. In the present review, we summarize the regulation of mitochondrial cholesterol, including its role in mitochondrial routine performance, cell death and chemotherapy resistance, highlighting its potential contribution to cancer. Of particular relevance is hepatocellular carcinoma, whose incidence in Western countries had tripled in the past decades due to the obesity and type II diabetes epidemic. A better understanding of the role of mitochondrial cholesterol in cancer development may open up novel opportunities for cancer therapy.
Collapse
Affiliation(s)
- Carmen Garcia-Ruiz
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain; Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Laura Conde de la Rosa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Vicent Ribas
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Jose C Fernandez-Checa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain; Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
14
|
Cardoso HJ, Carvalho TMA, Fonseca LRS, Figueira MI, Vaz CV, Socorro S. Revisiting prostate cancer metabolism: From metabolites to disease and therapy. Med Res Rev 2020; 41:1499-1538. [PMID: 33274768 DOI: 10.1002/med.21766] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/24/2020] [Accepted: 11/22/2020] [Indexed: 12/24/2022]
Abstract
Prostate cancer (PCa), one of the most commonly diagnosed cancers worldwide, still presents important unmet clinical needs concerning treatment. In the last years, the metabolic reprogramming and the specificities of tumor cells emerged as an exciting field for cancer therapy. The unique features of PCa cells metabolism, and the activation of specific metabolic pathways, propelled the use of metabolic inhibitors for treatment. The present work revises the knowledge of PCa metabolism and the metabolic alterations that underlie the development and progression of the disease. A focus is given to the role of bioenergetic sources, namely, glucose, lipids, and glutamine sustaining PCa cell survival and growth. Moreover, it is described as the action of oncogenes/tumor suppressors and sex steroid hormones in the metabolic reprogramming of PCa. Finally, the status of PCa treatment based on the inhibition of metabolic pathways is presented. Globally, this review updates the landscape of PCa metabolism, highlighting the critical metabolic alterations that could have a clinical and therapeutic interest.
Collapse
Affiliation(s)
- Henrique J Cardoso
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Tiago M A Carvalho
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Lara R S Fonseca
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Marília I Figueira
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Cátia V Vaz
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Sílvia Socorro
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
15
|
Revilla G, Cedó L, Tondo M, Moral A, Pérez JI, Corcoy R, Lerma E, Fuste V, Reddy ST, Blanco-Vaca F, Mato E, Escolà-Gil JC. LDL, HDL and endocrine-related cancer: From pathogenic mechanisms to therapies. Semin Cancer Biol 2020; 73:134-157. [PMID: 33249202 DOI: 10.1016/j.semcancer.2020.11.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/19/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023]
Abstract
Cholesterol is essential for a variety of functions in endocrine-related cells, including hormone and steroid production. We have reviewed the progress to date in research on the role of the main cholesterol-containing lipoproteins; low-density lipoprotein (LDL) and high-density lipoprotein (HDL), and their impact on intracellular cholesterol homeostasis and carcinogenic pathways in endocrine-related cancers. Neither LDL-cholesterol (LDL-C) nor HDL-cholesterol (HDL-C) was consistently associated with endocrine-related cancer risk. However, preclinical studies showed that LDL receptor plays a critical role in endocrine-related tumor cells, mainly by enhancing circulating LDL-C uptake and modulating tumorigenic signaling pathways. Although scavenger receptor type BI-mediated uptake of HDL could enhance cell proliferation in breast, prostate, and ovarian cancer, these effects may be counteracted by the antioxidant and anti-inflammatory properties of HDL. Moreover, 27-hydroxycholesterol a metabolite of cholesterol promotes tumorigenic processes in breast and epithelial thyroid cancer. Furthermore, statins have been reported to reduce the incidence of breast, prostate, pancreatic, and ovarian cancer in large clinical trials, in part because of their ability to lower cholesterol synthesis. Overall, cholesterol homeostasis deregulation in endocrine-related cancers offers new therapeutic opportunities, but more mechanistic studies are needed to translate the preclinical findings into clinical therapies.
Collapse
Affiliation(s)
- Giovanna Revilla
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, C/ Sant Quintí 77, 08041 Barcelona Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, C/ Antoni M. Claret 167, 08025 Barcelona, Spain
| | - Lídia Cedó
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, C/ Sant Quintí 77, 08041 Barcelona Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), C/ Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Mireia Tondo
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, C/ Sant Quintí 77, 08041 Barcelona Spain; Servei de Bioquímica, Hospital de la Santa Creu i Sant Pau, C/ Sant Quintí 89, 08041 Barcelona, Spain
| | - Antonio Moral
- Department of General Surgery, Hospital de la Santa Creu i Sant Pau, C/ Sant Quintí 89, 08041 Barcelona, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, C/ Antoni M. Claret 167, 08025 Barcelona, Spain
| | - José Ignacio Pérez
- Department of General Surgery, Hospital de la Santa Creu i Sant Pau, C/ Sant Quintí 89, 08041 Barcelona, Spain
| | - Rosa Corcoy
- Departament de Medicina, Universitat Autònoma de Barcelona, C/ Antoni M. Claret 167, 08025 Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/ Monforte de Lemos 3-5, 28029 Madrid, Spain; Department of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau, C/ Sant Quintí 89, 08041 Barcelona, Spain
| | - Enrique Lerma
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, C/ Sant Quintí 77, 08041 Barcelona Spain; Department of Anatomic Pathology, Hospital de la Santa Creu i Sant Pau, C/ Sant Quintí 89, 08041 Barcelona, Spain
| | - Victoria Fuste
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, C/ Sant Quintí 77, 08041 Barcelona Spain; Department of Anatomic Pathology, Hospital de la Santa Creu i Sant Pau, C/ Sant Quintí 89, 08041 Barcelona, Spain
| | - Srivinasa T Reddy
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA
| | - Francisco Blanco-Vaca
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, C/ Sant Quintí 77, 08041 Barcelona Spain; Servei de Bioquímica, Hospital de la Santa Creu i Sant Pau, C/ Sant Quintí 89, 08041 Barcelona, Spain.
| | - Eugènia Mato
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, C/ Sant Quintí 77, 08041 Barcelona Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/ Monforte de Lemos 3-5, 28029 Madrid, Spain.
| | - Joan Carles Escolà-Gil
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, C/ Sant Quintí 77, 08041 Barcelona Spain.
| |
Collapse
|
16
|
Kloudova-Spalenkova A, Holy P, Soucek P. Oxysterols in cancer management: From therapy to biomarkers. Br J Pharmacol 2020; 178:3235-3247. [PMID: 32986851 DOI: 10.1111/bph.15273] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/03/2020] [Accepted: 09/11/2020] [Indexed: 12/20/2022] Open
Abstract
Oxysterols are oxidized derivatives of cholesterol, both endogenous and exogenous. They have been implicated in numerous pathologies, including cancer. In addition to their roles in carcinogenesis, proliferation, migration, apoptosis, and multiple signalling pathways, they have been shown to modulate cancer therapy. They are known to affect therapy of hormonally positive breast cancer through modulating oestrogen receptor activity. Oxysterols have also been shown in various in vitro models to influence efficacy of chemotherapeutics, such as doxorubicin, vincristine, cisplatin, 5-fluorouracil, and others. Their effects on the immune system should also be considered in immunotherapy. Selective anti-cancer cytotoxic properties of some oxysterols make them candidates for new therapeutic molecules. Finally, differences in oxysterol levels in blood of cancer patients in different stages or versus healthy controls, and in tumour versus non-tumour tissues, show potential of oxysterols as biomarkers for cancer management and patient stratification for optimization of therapy. LINKED ARTICLES: This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc.
Collapse
Affiliation(s)
- Alzbeta Kloudova-Spalenkova
- Department of Toxicogenomics, National Institute of Public Health, Prague, Czech Republic.,Third Faculty of Medicine, Charles University, Prague, Czech Republic.,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Petr Holy
- Department of Toxicogenomics, National Institute of Public Health, Prague, Czech Republic.,Third Faculty of Medicine, Charles University, Prague, Czech Republic.,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pavel Soucek
- Department of Toxicogenomics, National Institute of Public Health, Prague, Czech Republic.,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| |
Collapse
|
17
|
Cholesterol metabolism: New functions and therapeutic approaches in cancer. Biochim Biophys Acta Rev Cancer 2020; 1874:188394. [PMID: 32698040 DOI: 10.1016/j.bbcan.2020.188394] [Citation(s) in RCA: 207] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/08/2020] [Accepted: 07/12/2020] [Indexed: 02/05/2023]
Abstract
Cholesterol and its metabolites (precursors and derivatives) play an important role in cancer. In recent years, numerous studies have reported the functions of cholesterol metabolism in the regulation of tumor biological processes, especially oncogenic signaling pathways, ferroptosis, and tumor microenvironment. Preclinical studies have over the years indicated the inhibitory effects of blocking cholesterol synthesis and uptake on tumor formation and growth. Besides, some new cholesterol metabolic molecules such as SOAT1, SQLE, and NPC1 have recently emerged as promising drug targets for cancer treatment. Here, we systematically review the roles of cholesterol and its metabolites, and the latest advances in cancer therapy targeting cholesterol metabolism.
Collapse
|
18
|
Asghari A, Umetani M. Obesity and Cancer: 27-Hydroxycholesterol, the Missing Link. Int J Mol Sci 2020; 21:E4822. [PMID: 32650428 PMCID: PMC7404106 DOI: 10.3390/ijms21144822] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023] Open
Abstract
Obesity is currently affecting more than 40% of the Americans, and if it progresses with this rate, soon one out of two Americans will be obese. Obesity is an important risk factor for several disorders including cardiovascular disease, the first cause of death in the United States. Cancer follows as the second deadliest disease, and a link between obesity and cancer has been suggested. However, it is very hard to establish an exact connection between obesity and cancers due to the multifactorial nature of obesity. Hypercholesterolemia is a comorbidity of obesity and also linked to several cancers. Recently a cholesterol metabolite 27-hydroxycholesterol (27HC) was found to be an endogenous selective estrogen receptor modulator (SERM), which opened new doors toward several interesting studies on the role of this molecule in biological disorders. It is speculated that 27HC might be the missing link in the obesity and cancer chain. Here, we explored the effects of 27-hydroxycholesterol on obesity and cancers with a focus on the SERM capacity of 27HC.
Collapse
Affiliation(s)
- Arvand Asghari
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204-5056, USA;
| | - Michihisa Umetani
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204-5056, USA;
- HEALTH Research Institute, University of Houston, Houston, TX 77204-5056, USA
| |
Collapse
|
19
|
Riscal R, Skuli N, Simon MC. Even Cancer Cells Watch Their Cholesterol! Mol Cell 2019; 76:220-231. [PMID: 31586545 DOI: 10.1016/j.molcel.2019.09.008] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/30/2019] [Accepted: 09/05/2019] [Indexed: 02/07/2023]
Abstract
Deregulated cell proliferation is an established feature of cancer, and altered tumor metabolism has witnessed renewed interest over the past decade, including the study of how cancer cells rewire metabolic pathways to renew energy sources and "building blocks" that sustain cell division. Microenvironmental oxygen, glucose, and glutamine are regarded as principal nutrients fueling tumor growth. However, hostile tumor microenvironments render O2/nutrient supplies chronically insufficient for increased proliferation rates, forcing cancer cells to develop strategies for opportunistic modes of nutrient acquisition. Recent work shows that cancer cells overcome this nutrient scarcity by scavenging other substrates, such as proteins and lipids, or utilizing adaptive metabolic pathways. As such, reprogramming lipid metabolism plays important roles in providing energy, macromolecules for membrane synthesis, and lipid-mediated signaling during cancer progression. In this review, we highlight more recently appreciated roles for lipids, particularly cholesterol and its derivatives, in cancer cell metabolism within intrinsically harsh tumor microenvironments.
Collapse
Affiliation(s)
- Romain Riscal
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicolas Skuli
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
20
|
Liang Z, Chen Y, Wang L, Li D, Yang X, Ma G, Wang Y, Li Y, Zhao H, Liang Y, Niu H. CYP27A1 inhibits bladder cancer cells proliferation by regulating cholesterol homeostasis. Cell Cycle 2018; 18:34-45. [PMID: 30563407 DOI: 10.1080/15384101.2018.1558868] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
CYP27A1, an enzyme involved in regulating cellular cholesterol homeostasis, converts cholesterol into 27-hydroxycholesterol (27-HC). The relationship between CYP27A1 and cell proliferation was studied to determine the role of CYP27A1 in bladder cancer. The expression of CYP27A1 in three bladder cancer cell lines (T24, UM-UC-3 and 5637) were assessed by qRT-PCR and Western blotting, and cells with stable CYP27A1 expression were generated by lentiviral infection. Cell proliferation was detected by MTT assays, colony formation assays and a tumor xenograft model in vitro and in vivo, and the intracellular 27-HC and cholesterol secretion levels were detected by enzyme-linked immunosorbent assays (ELISA). The results revealed that CYP27A1 expression was downregulated in androgen receptor (AR)-positive T24/UM-UC-3 cells compared with AR-negative 5637 cell. After CYP27A1 expression was restored, cell proliferation was inhibited in vitro and in vivo because much more intracellular 27-HC was produced in the CYP27A1-overexpressing cells than in the control cells. Both T24 and UM-UC-3 cells treated with 27-HC showed similar results. In addition, CYP27A1/27HC could reduce the cellular cholesterol level in both T24 and UM-UC-3 cells by upregulating ATP-binding cassette transporters G1 and A1 (ABCG1 and ABCA1) through Liver X receptors (LXRs) pathway and downregulating low-density lipoprotein receptor (LDLR) expression. These findings all suggest that CYP27A1 is a critical cholesterol sensor in bladder cancer cells that may contribute significantly to bladder cancer proliferation.
Collapse
Affiliation(s)
- Zhijuan Liang
- a Key Laboratory, Department of Urology and Andrology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Yuanbin Chen
- a Key Laboratory, Department of Urology and Andrology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Liping Wang
- a Key Laboratory, Department of Urology and Andrology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Dan Li
- a Key Laboratory, Department of Urology and Andrology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Xuecheng Yang
- b Department of Urology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Guofeng Ma
- b Department of Urology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Yonghua Wang
- b Department of Urology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Yongxin Li
- c Department of Vascular Surgery , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Han Zhao
- d Department of Pathology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Ye Liang
- a Key Laboratory, Department of Urology and Andrology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Haitao Niu
- a Key Laboratory, Department of Urology and Andrology , Affiliated Hospital of Qingdao University , Qingdao , China.,b Department of Urology , Affiliated Hospital of Qingdao University , Qingdao , China
| |
Collapse
|
21
|
Maldonado-Pereira L, Schweiss M, Barnaba C, Medina-Meza IG. The role of cholesterol oxidation products in food toxicity. Food Chem Toxicol 2018; 118:908-939. [DOI: 10.1016/j.fct.2018.05.059] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/17/2018] [Accepted: 05/25/2018] [Indexed: 01/10/2023]
|
22
|
27-hydroxycholesterol decreases cell proliferation in colon cancer cell lines. Biochimie 2018; 153:171-180. [PMID: 30009860 DOI: 10.1016/j.biochi.2018.07.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/11/2018] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is the third most diagnosed cancer in the western world, affecting 1 out of approximately 22 people in their lifetime. Several epidemiological studies suggest a positive association between high plasma cholesterol levels and colorectal cancer. However, the molecular mechanisms by which cholesterol may alter the risk of colorectal cancer (CRC) are ill-defined as the cholesterol lowering drugs statins do not appear to decrease a patient's risk of developing colorectal cancer. Cholesterol is metabolized to active derivatives including cholesterol oxidization products (COP), known as oxysterols, which have been shown to alter cellular proliferation. These metabolites and not cholesterol per se, may therefore affect the risk of developing colorectal cancer. The cholesterol metabolite or the oxysterol 27-hydroxycholesterol (27-OHC) is the most abundant oxysterol in the plasma and has been shown to be involved in the pathogenesis of several cancers including breast and prostate cancer. However, the role of 27-OHC in colorectal cancer has not been investigated. We treated Caco2 and SW620, two well characterized colon cancer cells with low, physiological and high concentrations of 27-OHC, and found that 27-OHC reduces cellular proliferation in these cells. We also found that the effects of 27-OHC on cell proliferation are not due to cellular cytotoxicity or apoptotic cellular death. Additionally, 27-OHC-induced reduction in cell proliferation is independent of actions on its target nuclear receptors, liver-X-receptors (LXR) and estrogen receptors (ER) activation. Instead, our study demonstrates that 27-OHC significantly decreases AKT activation, a major protein kinase involved in the pathogenesis of cancer as it regulates cell cycle progression, protein synthesis, and cellular survival. Our data shows that treatment with 27-OHC substantially decreases the activation of AKT by reducing levels of its active form, p-AKT, in Caco2 cells but not SW620 cells. All-together, our results show for the first time that the cholesterol metabolite 27-OHC reduces cell proliferation in colorectal cancer cells.
Collapse
|
23
|
Sottero B, Leonarduzzi G, Testa G, Gargiulo S, Poli G, Biasi F. Lipid Oxidation Derived Aldehydes and Oxysterols Between Health and Disease. EUR J LIPID SCI TECH 2018. [DOI: 10.1002/ejlt.201700047] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Barbara Sottero
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Simona Gargiulo
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Fiorella Biasi
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| |
Collapse
|
24
|
He S, Nelson ER. 27-Hydroxycholesterol, an endogenous selective estrogen receptor modulator. Maturitas 2017; 104:29-35. [PMID: 28923174 DOI: 10.1016/j.maturitas.2017.07.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 07/26/2017] [Accepted: 07/28/2017] [Indexed: 12/11/2022]
Abstract
Estrogen receptors (ERs) mediate the actions of the steroidal estrogens, and are important for the regulation of several physiological and pathophysiological processes, including reproduction, bone physiology, cardiovascular physiology and breast cancer. The unique pharmacology of the ERs allows for certain ligands, such as tamoxifen, to elicit tissue- and context-specific responses, ligands now referred to as selective estrogen receptor modulators (SERMs). Recently, the cholesterol metabolite 27-hydroxychoelsterol (27HC) has been defined as an endogenous SERM, with activities in atherosclerosis, osteoporosis, breast and prostate cancers, and neural degenerative diseases. Since 27HC concentrations closely mirror those of cholesterol, it is possible that 27HC mediates many of the biological effects of cholesterol. This paper provides an overview of ER pharmacology and summarizes the work to date implicating 27HC in various diseases. Wherever possible, we highlight clinical data in support of a role for 27HC in the diseases discussed.
Collapse
Affiliation(s)
- Sisi He
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; University of Illinois Cancer Center, Chicago, IL, USA; Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
25
|
Kloudova A, Guengerich FP, Soucek P. The Role of Oxysterols in Human Cancer. Trends Endocrinol Metab 2017; 28:485-496. [PMID: 28410994 PMCID: PMC5474130 DOI: 10.1016/j.tem.2017.03.002] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/20/2017] [Indexed: 12/12/2022]
Abstract
Oxysterols are oxygenated derivatives of cholesterol formed in the human body or ingested in the diet. By modulating the activity of many proteins [e.g., liver X receptors (LXRs), oxysterol-binding proteins (OSBPs), some ATP-binding cassette (ABC) transporters], oxysterols can affect many cellular functions and influence various physiological processes (e.g., cholesterol metabolism, membrane fluidity regulation, intracellular signaling pathways). Therefore, the role of oxysterols is also important in pathological conditions (e.g., atherosclerosis, diabetes mellitus type 2, neurodegenerative disorders). Finally, current evidence suggests that oxysterols play a role in malignancies such as breast, prostate, colon, and bile duct cancer. This review summarizes the physiological importance of oxysterols in the human body with a special emphasis on their roles in various tumors.
Collapse
Affiliation(s)
- Alzbeta Kloudova
- Department of Toxicogenomics, National Institute of Public Health, Prague 100 42, Czech Republic; Third Faculty of Medicine, Charles University, Prague 100 00, Czech Republic
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Pavel Soucek
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen 323 00, Czech Republic.
| |
Collapse
|
26
|
Marwarha G, Raza S, Hammer K, Ghribi O. 27-hydroxycholesterol: A novel player in molecular carcinogenesis of breast and prostate cancer. Chem Phys Lipids 2017; 207:108-126. [PMID: 28583434 DOI: 10.1016/j.chemphyslip.2017.05.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/31/2017] [Accepted: 05/31/2017] [Indexed: 12/13/2022]
Abstract
Several studies have suggested an etiological role for hypercholesterolemia in the pathogenesis of breast cancer and prostate cancer (PCa). However, the molecular mechanisms that underlie and mediate the hypercholesterolemia-fostered increased risk for breast cancer and PCa are yet to be determined. The discovery that the most abundant cholesterol oxidized metabolite in the plasma, 27 hydroxycholesterol (27-OHC), is a selective estrogen receptor modulator (SERM) and an agonist of Liver X receptors (LXR) partially fills the void in our understanding and knowledge of the mechanisms that may link hypercholesterolemia to development and progression of breast cancer and PCa. The wide spectrum and repertoire of SERM and LXR-dependent effects of 27-OHC in the context of all facets and aspects of breast cancer and prostate cancer biology are reviewed in this manuscript in a very comprehensive manner. This review highlights recent findings pertaining to the role of 27-OHC in breast cancer and PCa and delineates the signaling mechanisms involved in the governing of different facets of tumor biology, that include tumor cell proliferation, epithelial-mesenchymal transition (EMT), as well as tumor cell invasion, migration, and metastasis. We also discuss the limitations of contemporary studies and lack of our comprehension of the entire gamut of effects exerted by 27-OHC that may be relevant to the pathogenesis of breast cancer and PCa. We unveil and propose potential future directions of research that may further our understanding of the role of 27-OHC in breast cancer and PCa and help design therapeutic interventions against endocrine therapy-resistant breast cancer and PCa.
Collapse
Affiliation(s)
- Gurdeep Marwarha
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, 58202, USA
| | - Shaneabbas Raza
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, 58202, USA
| | - Kimberly Hammer
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, 58202, USA; Department of Veteran Affairs, Fargo VA Health Care System, Fargo, North Dakota 58102, USA
| | - Othman Ghribi
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, 58202, USA.
| |
Collapse
|
27
|
Raza S, Meyer M, Goodyear C, Hammer KDP, Guo B, Ghribi O. The cholesterol metabolite 27-hydroxycholesterol stimulates cell proliferation via ERβ in prostate cancer cells. Cancer Cell Int 2017; 17:52. [PMID: 28503095 PMCID: PMC5425984 DOI: 10.1186/s12935-017-0422-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 05/02/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND For every six men, one will be diagnosed with prostate cancer (PCa) in their lifetime. Estrogen receptors (ERs) are known to play a role in prostate carcinogenesis. However, it is unclear whether the estrogenic effects are mediated by estrogen receptor α (ERα) or estrogen receptor β (ERβ). Although it is speculated that ERα is associated with harmful effects on PCa, the role of ERβ in PCa is still ill-defined. The cholesterol oxidized metabolite 27-hydroxycholesterol (27-OHC) has been found to bind to ERs and act as a selective ER modulator (SERM). Increased 27-OHC levels are found in individuals with hypercholesterolemia, a condition that is suggested to be a risk factor for PCa. METHODS In the present study, we determined the extent to which 27-OHC causes deleterious effects in the non-tumorigenic RWPE-1, the low tumorigenic LNCaP, and the highly tumorigenic PC3 prostate cancer cells. We conducted cell metabolic activity and proliferation assays using MTS and CyQUANT dyes, protein expression analyses via immunoblots and gene expression analyses via RT-PCR. Additionally, immunocytochemistry and invasion assays were performed to analyze intracellular protein distribution and quantify transepithelial cell motility. RESULTS We found that incubation of LNCaP and PC3 cells with 27-OHC significantly increased cell proliferation. We also demonstrate that the ER inhibitor ICI 182,780 (fulvestrant) significantly reduced 27-OH-induced cell proliferation, indicating the involvement of ERs in proliferation. Interestingly, ERβ levels, and to a lesser extent ERα, were significantly increased following incubation of PCa cells with 27-OHC. Furthermore, in the presence of the ERβ specific inhibitor, PHTPP, 27-OHC-induced proliferation is attenuated. CONCLUSIONS Altogether, our results show for the first time that 27-OHC, through ER activation, triggers deleterious effect in prostate cancer cell lines. We propose that dysregulated levels of 27-OHC may trigger or exacerbate prostate cancer via acting on ERβ.
Collapse
Affiliation(s)
- Shaneabbas Raza
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 501 North Columbia Road, Grand Forks, ND 58202 USA
| | - Megan Meyer
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 501 North Columbia Road, Grand Forks, ND 58202 USA
| | - Casey Goodyear
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 501 North Columbia Road, Grand Forks, ND 58202 USA
| | - Kimberly D P Hammer
- Department of Veteran Affairs, Fargo VA Health Care System, Fargo, ND 58102 USA
| | - Bin Guo
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58108 USA
| | - Othman Ghribi
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 501 North Columbia Road, Grand Forks, ND 58202 USA
| |
Collapse
|
28
|
Shen Z, Zhu D, Liu J, Chen J, Liu Y, Hu C, Li Z, Li Y. 27-Hydroxycholesterol induces invasion and migration of breast cancer cells by increasing MMP9 and generating EMT through activation of STAT-3. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 51:1-8. [PMID: 28257824 DOI: 10.1016/j.etap.2017.02.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/25/2017] [Accepted: 02/02/2017] [Indexed: 06/06/2023]
Abstract
Breast carcinoma plays a vital role in the reasons of global women's death. ER-related invasion and migration play an important part in the development and prognosis of breast cancer. Here, we found that 27-Hydroxycholesterol (27HC) could induce epithelial-mesenchymal transition (EMT) and increase the expression of the matrix metalloproteinase 9 (MMP9) at mRNA level and the active form. Meanwhile, interestingly, we found 27HC activated signal transducer and activator of transcription 3 (STAT-3) in ER positive cells except activation of ER signaling. Furthermore, inhibition of STAT-3 by siRNA attenuated the 27HC-induced improvement of MMP9 and decreased the invasion and migration ability in MCF7 and T47D cells. In addition, 27HC could also promote MMP9, vimentin and active STAT-3 in the ER negative cells MDA-MB-231. All these results not only raise a mechanism whereby 27HC enhances the invasion and metastasis, but also is helpful to realize 27HC as a potential endogenous detrimental factor in breast tumor patients.
Collapse
Affiliation(s)
- Zhaoxia Shen
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Dongmei Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Jiao Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Juan Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yun Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Chunyan Hu
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Zhong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| | - Yuan Li
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
29
|
Alfaqih MA, Nelson ER, Liu W, Safi R, Jasper JS, Macias E, Geradts J, Thompson JW, Dubois LG, Freeman MR, Chang CY, Chi JT, McDonnell DP, Freedland SJ. CYP27A1 Loss Dysregulates Cholesterol Homeostasis in Prostate Cancer. Cancer Res 2017; 77:1662-1673. [PMID: 28130224 PMCID: PMC5687884 DOI: 10.1158/0008-5472.can-16-2738] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 01/10/2017] [Accepted: 01/10/2017] [Indexed: 11/16/2022]
Abstract
In this study, we used a bioinformatic approach to identify genes whose expression is dysregulated in human prostate cancers. One of the most dramatically downregulated genes identified encodes CYP27A1, an enzyme involved in regulating cellular cholesterol homeostasis. Importantly, lower CYP27A1 transcript levels were associated with shorter disease-free survival and higher tumor grade. Loss of CYP27A1 in prostate cancer was confirmed at the protein level by immunostaining for CYP27A1 in annotated tissue microarrays. Restoration of CYP27A1 expression in cells where its gene was silenced attenuated their growth in vitro and in tumor xenografts. Studies performed in vitro revealed that treatment of prostate cancer cells with 27-hydroxycholesterol (27HC), an enzymatic product of CYP27A1, reduced cellular cholesterol content in prostate cancer cell lines by inhibiting the activation of sterol regulatory-element binding protein 2 and downregulating low-density lipoprotein receptor expression. Our findings suggest that CYP27A1 is a critical cellular cholesterol sensor in prostate cells and that dysregulation of the CYP27A1/27HC axis contributes significantly to prostate cancer pathogenesis. Cancer Res; 77(7); 1662-73. ©2017 AACR.
Collapse
Affiliation(s)
- Mahmoud A Alfaqih
- Department of Surgery, Duke University, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Erik R Nelson
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign; and University of Illinois Cancer Center, Chicago, Illinois
| | - Wen Liu
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Rachid Safi
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Jeffery S Jasper
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Everardo Macias
- Department of Surgery, Duke University, Durham, North Carolina
- Department of Surgery and Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Joseph Geradts
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - J Will Thompson
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
- Department of Proteomics and Metabolomics Shared Resource, Duke University, Durham, North Carolina
| | - Laura G Dubois
- Department of Proteomics and Metabolomics Shared Resource, Duke University, Durham, North Carolina
| | - Michael R Freeman
- Department of Surgery and Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Ching-Yi Chang
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Jen-Tsan Chi
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina.
| | - Stephen J Freedland
- Department of Surgery and Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California.
- Surgery Section, Durham VA Medical Center, Durham, North Carolina
| |
Collapse
|
30
|
Wang H, Yuan L, Ma W, Han J, Lu Y, Feng L, Xiao R. The cytotoxicity of 27-hydroxycholesterol in co-cultured SH-SY5Y cells and C6 cells. Neurosci Lett 2016; 632:209-17. [DOI: 10.1016/j.neulet.2016.08.056] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 08/26/2016] [Accepted: 08/30/2016] [Indexed: 12/16/2022]
|