51
|
Cedó L, Reddy ST, Mato E, Blanco-Vaca F, Escolà-Gil JC. HDL and LDL: Potential New Players in Breast Cancer Development. J Clin Med 2019; 8:jcm8060853. [PMID: 31208017 PMCID: PMC6616617 DOI: 10.3390/jcm8060853] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/11/2019] [Accepted: 06/12/2019] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most prevalent cancer and primary cause of cancer-related mortality in women. The identification of risk factors can improve prevention of cancer, and obesity and hypercholesterolemia represent potentially modifiable breast cancer risk factors. In the present work, we review the progress to date in research on the potential role of the main cholesterol transporters, low-density and high-density lipoproteins (LDL and HDL), on breast cancer development. Although some studies have failed to find associations between lipoproteins and breast cancer, some large clinical studies have demonstrated a direct association between LDL cholesterol levels and breast cancer risk and an inverse association between HDL cholesterol and breast cancer risk. Research in breast cancer cells and experimental mouse models of breast cancer have demonstrated an important role for cholesterol and its transporters in breast cancer development. Instead of cholesterol, the cholesterol metabolite 27-hydroxycholesterol induces the proliferation of estrogen receptor-positive breast cancer cells and facilitates metastasis. Oxidative modification of the lipoproteins and HDL glycation activate different inflammation-related pathways, thereby enhancing cell proliferation and migration and inhibiting apoptosis. Cholesterol-lowering drugs and apolipoprotein A-I mimetics have emerged as potential therapeutic agents to prevent the deleterious effects of high cholesterol in breast cancer.
Collapse
Affiliation(s)
- Lídia Cedó
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Sant Quintí 77, 08041 Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Monforte de Lemos 3-5, 28029 Madrid, Spain.
| | - Srinivasa T Reddy
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA.
| | - Eugènia Mato
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Sant Quintí 77, 08041 Barcelona, Spain.
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Monforte de Lemos 3-5, 28029 Madrid, Spain.
| | - Francisco Blanco-Vaca
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Sant Quintí 77, 08041 Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Monforte de Lemos 3-5, 28029 Madrid, Spain.
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Av. de Can Domènech 737, 08193 Cerdanyola del Vallès, Spain.
| | - Joan Carles Escolà-Gil
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Sant Quintí 77, 08041 Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Monforte de Lemos 3-5, 28029 Madrid, Spain.
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Av. de Can Domènech 737, 08193 Cerdanyola del Vallès, Spain.
| |
Collapse
|
52
|
Mikó E, Kovács T, Sebő É, Tóth J, Csonka T, Ujlaki G, Sipos A, Szabó J, Méhes G, Bai P. Microbiome-Microbial Metabolome-Cancer Cell Interactions in Breast Cancer-Familiar, but Unexplored. Cells 2019; 8:E293. [PMID: 30934972 PMCID: PMC6523810 DOI: 10.3390/cells8040293] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/18/2022] Open
Abstract
Breast cancer is a leading cause of death among women worldwide. Dysbiosis, an aberrant composition of the microbiome, characterizes breast cancer. In this review we discuss the changes to the metabolism of breast cancer cells, as well as the composition of the breast and gut microbiome in breast cancer. The role of the breast microbiome in breast cancer is unresolved, nevertheless it seems that the gut microbiome does have a role in the pathology of the disease. The gut microbiome secretes bioactive metabolites (reactivated estrogens, short chain fatty acids, amino acid metabolites, or secondary bile acids) that modulate breast cancer. We highlight the bacterial species or taxonomical units that generate these metabolites, we show their mode of action, and discuss how the metabolites affect mitochondrial metabolism and other molecular events in breast cancer. These metabolites resemble human hormones, as they are produced in a "gland" (in this case, the microbiome) and they are subsequently transferred to distant sites of action through the circulation. These metabolites appear to be important constituents of the tumor microenvironment. Finally, we discuss how bacterial dysbiosis interferes with breast cancer treatment through interfering with chemotherapeutic drug metabolism and availability.
Collapse
Affiliation(s)
- Edit Mikó
- Department of Medical Chemistry, University of Debrecen, 4032 Debrecen, Hungary.
- Department of Microbiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - Tünde Kovács
- Department of Medical Chemistry, University of Debrecen, 4032 Debrecen, Hungary.
| | - Éva Sebő
- Kenézy Breast Center, Kenézy Gyula County Hospital, 4032 Debrecen, Hungary.
| | - Judit Tóth
- Kenézy Breast Center, Kenézy Gyula County Hospital, 4032 Debrecen, Hungary.
| | - Tamás Csonka
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - Gyula Ujlaki
- Department of Medical Chemistry, University of Debrecen, 4032 Debrecen, Hungary.
| | - Adrienn Sipos
- Department of Medical Chemistry, University of Debrecen, 4032 Debrecen, Hungary.
| | - Judit Szabó
- Department of Microbiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - Péter Bai
- Department of Medical Chemistry, University of Debrecen, 4032 Debrecen, Hungary.
- MTA-DE Lendület Laboratory of Cellular Metabolism, 4032 Debrecen, Hungary.
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| |
Collapse
|
53
|
Włodarczyk M, Ciebiera M, Nowicka G. TNF-α G-308A genetic variants, serum CRP-hs concentration and DNA damage in obese women. Mol Biol Rep 2019; 47:855-866. [PMID: 30900134 PMCID: PMC7340642 DOI: 10.1007/s11033-019-04764-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/14/2019] [Indexed: 12/21/2022]
Abstract
Obesity is associated with inflammation, which can disturb genome stability. Tumor necrosis factor (TNF-α) polymorphism was found to affect TNF-α protein production and inflammation. Therefore, the present study illustrates the relationship between TNF-α polymorphism, the degree of inflammation assessed by serum high sensitivity C-reactive protein concentration (CRP-hs) and basal DNA damage in patients with obesity (BMI 30–34.9 kg/m2) and control subjects with proper body mass (BMI < 25 kg/m2). A total of 115 participants (75 obese premenopausal women; and 40 age-, and gender-matched controls) were included. Biochemical parameters (serum concentrations of total-cholesterol, HDL-cholesterol, LDL- cholesterol, triglycerides, glucose, apolipoprotein AI, CRP-hs) and endogenous DNA damage (determined by comet assay) were measured. TNF-α G-308A polymorphism (rs1800629) was analyzed by PCR-RFLP (PCR-restriction fragments length polymorphism). An effect of TNF-α genotype on serum CRP-hs concentration was noted (p = 0.031). In general, carriers of the rare A allele of the TNF-α G-308A polymorphism had significantly lower endogenous DNA damage and serum CRP-hs concentrations than GG homozygotes, however, the protective effect of the A allele was especially visible in non-obese women. Serum CRP-hs concentrations and levels of DNA damage (% DNA in tail) were significantly higher in obese than in controls (p = 0.001 and p < 0.0001, respectively). The adjusted multiple linear regression analyses revealed a significant, independent impact of obesity on DNA damage (p = 0.00000) and no effect of other covariates i.e. age, TNF-α genotype and serum CRP-hs concentration. Our study showed that obesity has a significant impact on the levels of endogenous DNA damage. Obesity abolished the protective effect of A allele of the TNF-α G-308A polymorphism on DNA damage and on inflammation development observed in non-obese A allele carriers.
Collapse
Affiliation(s)
- Marta Włodarczyk
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy with Division of Laboratory Medicine, Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland. .,Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland.
| | - Michał Ciebiera
- II Department of Obstetrics and Gynecology, The Centre of Postgraduate Medical Education, Cegłowska 80, 01-809, Warsaw, Poland
| | - Grażyna Nowicka
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy with Division of Laboratory Medicine, Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland.,Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| |
Collapse
|
54
|
Luis C, Duarte F, Faria I, Jarak I, Oliveira PF, Alves MG, Soares R, Fernandes R. Warburg Effect Inversion: Adiposity shifts central primary metabolism in MCF-7 breast cancer cells. Life Sci 2019; 223:38-46. [PMID: 30862570 DOI: 10.1016/j.lfs.2019.03.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 12/22/2022]
Abstract
AIMS Obesity is a complex health disorder and a trigger to many diseases like Diabetes mellitus (DM) and breast cancer (BrCa), both leading causes of morbidity and mortality worldwide. Also evidence demonstrates that abnormal glucose metabolism termed 'the Warburg effect' in cancer cell is closely associated with malignant phenotypes and promote the aggressiveness of several types of cancer, including BrCa. In this study, we evaluated the breast cancer cell metabolism in normoglycemia, hyperglycemia and in an obesity condition in order to clarify the potential underlined mechanisms that link these disorders. MATERIALS AND METHODS MCF-7 cells were exposed to low and high glucose levels, the latter either in the presence of 3T3-L1 adipocyte conditioned medium (CM), thus mimicking the adiposity observed in obese patients. Cell viability, migration, proliferation, cytotoxicity and cell death assays were performed under the different culture conditions. Hormonal and lipid profile were also characterized by biochemical assays and primary metabolism was determined by Nuclear Magnetic Resonance (NMR)-based metabolomics. RESULTS Our results show an increased aggressiveness in the condition mimicking diabetogenic obesity with an altered energy/lipid metabolism. Interestingly in the experimental obesity-mimicking status, lipids and amino acids were expended while glucose was produced by tumor cells from lactate. These findings reveal a shift on tumor cells metabolism that is opposite to 'the Warburg effect'. CONCLUSIONS Overall, this experimentally obesity-mimicking condition not only revealed an increased tumor proliferation and aggressiveness but also disclosed a new mechanism of cancer metabolism, the 'Warburg Effect Inversion'.
Collapse
Affiliation(s)
- Carla Luis
- School of Health, Polytechnic of Porto (ESS/P.PORTO), Porto, Portugal; Biochemistry Unit, Department of Biomedicine, Faculty of Medicine, University of Porto (FMUP), Porto, Portugal; Instituto de Inovação e Investigação em Saúde (I3S), University of Porto, Portugal
| | - Fernanda Duarte
- School of Health, Polytechnic of Porto (ESS/P.PORTO), Porto, Portugal; CoreLab, Hospital Centre of Porto University (CHUP), Porto, Portugal
| | - Isabel Faria
- School of Health, Polytechnic of Porto (ESS/P.PORTO), Porto, Portugal
| | - Ivana Jarak
- Department of Life Sciences, Faculty of Sciences and Technology, Centre for Functional Ecology (CFE), University of Coimbra, Coimbra; Laboratory of Cell Biology, Unit for Multidisciplinary Research in Biomedicine (UMIB), Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Pedro F Oliveira
- Instituto de Inovação e Investigação em Saúde (I3S), University of Porto, Portugal; Laboratory of Cell Biology, Unit for Multidisciplinary Research in Biomedicine (UMIB), Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal; Department of Genetics, Faculty of Medicine, University of Porto, Portugal
| | - Marco G Alves
- Laboratory of Cell Biology, Unit for Multidisciplinary Research in Biomedicine (UMIB), Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Raquel Soares
- Biochemistry Unit, Department of Biomedicine, Faculty of Medicine, University of Porto (FMUP), Porto, Portugal; Instituto de Inovação e Investigação em Saúde (I3S), University of Porto, Portugal
| | - Rúben Fernandes
- School of Health, Polytechnic of Porto (ESS/P.PORTO), Porto, Portugal; Instituto de Inovação e Investigação em Saúde (I3S), University of Porto, Portugal; Faculty of Medicine, University of Santiago de Compostela, Galiza, Spain.
| |
Collapse
|
55
|
Borgquist S, Broberg P, Tojjar J, Olsson H. Statin use and breast cancer survival - a Swedish nationwide study. BMC Cancer 2019; 19:54. [PMID: 30634941 PMCID: PMC6330431 DOI: 10.1186/s12885-018-5263-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 12/28/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND A sizeable body of evidence suggests that statins can cease breast cancer progression and prevent breast cancer recurrence. The latest studies have, however, not been supportive of such clinically beneficial effects. These discrepancies may be explained by insufficient power. This considerably sized study investigates the association between both pre- and post-diagnostic statin use and breast cancer outcome. METHODS A Swedish nation-wide retrospective cohort study of 20,559 Swedish women diagnosed with breast cancer (July 1st, 2005 through 2008). Dispensed statin medication was identified through the Swedish Prescription Registry. Breast cancer related death information was obtained from the national cause-of-death registry until December 31st, 2012. Cox regression models yielded hazard ratios (HR) and 95% confidence intervals (CI) regarding associations between statin use and breast cancer-specific and overall mortality. RESULTS During a median follow-up time of 61.6 months, a total of 4678 patients died, of which 2669 were considered breast cancer related deaths. Compared to non- or irregular use, regular pre-diagnostic statin use was associated with lower risk of breast cancer related deaths (HR = 0.77; 95% CI 0.63-0.95, P = 0.014). Similarly, post-diagnostic statin use compared to non-use was associated with lower risk of breast cancer related deaths (HR = 0.83; 95% CI 0.75-0.93, P = 0.001). CONCLUSION This study supports the notion that statin use is protective regarding breast cancer related mortality in agreement with previous Scandinavian studies, although less so with studies in other populations. These disparities should be further investigated to pave the way for future randomized clinical trials investigating the role of statins in breast cancer.
Collapse
Affiliation(s)
- Signe Borgquist
- Division of Oncology and Pathology, Department of Clinical Sciences, Barngatan 4, SE-221 85, Lund, Sweden. .,Department of Oncology, Aarhus University Hospital, Aarhus, Denmark.
| | - Per Broberg
- Division of Cancer Epidemiology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jasaman Tojjar
- Division of Cancer Epidemiology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Håkan Olsson
- Division of Oncology and Pathology, Department of Clinical Sciences, Barngatan 4, SE-221 85, Lund, Sweden.,Division of Cancer Epidemiology, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
56
|
Bleach R, McIlroy M. The Divergent Function of Androgen Receptor in Breast Cancer; Analysis of Steroid Mediators and Tumor Intracrinology. Front Endocrinol (Lausanne) 2018; 9:594. [PMID: 30416486 PMCID: PMC6213369 DOI: 10.3389/fendo.2018.00594] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/19/2018] [Indexed: 12/16/2022] Open
Abstract
Androgen receptor (AR) is the most widely expressed steroid receptor protein in normal breast tissue and is detectable in approximately 90% of primary breast cancers and 75% of metastatic lesions. However, the role of AR in breast cancer development and progression is mired in controversy with evidence suggesting it can either inhibit or promote breast tumorigenesis. Studies have shown it to antagonize estrogen receptor alpha (ERα) DNA binding, thereby preventing pro-proliferative gene transcription; whilst others have demonstrated AR to take on the mantle of a pseudo ERα particularly in the setting of triple negative breast cancer. Evidence for a potentiating role of AR in the development of endocrine resistant breast cancer has also been mounting with reports associating high AR expression with poor response to endocrine treatment. The resurgence of interest into the function of AR in breast cancer has resulted in various emergent clinical trials evaluating anti-AR therapy and selective androgen receptor modulators in the treatment of advanced breast cancer. Trials have reported varied response rates dependent upon subtype with overall clinical benefit rates of ~19-29% for anti-androgen monotherapy, suggesting that with enhanced patient stratification AR could prove efficacious as a breast cancer therapy. Androgens and AR have been reported to facilitate tumor stemness in some cancers; a process which may be mediated through genomic or non-genomic actions of the AR, with the latter mechanism being relatively unexplored in breast cancer. Steroidogenic ligands of the AR are produced in females by the gonads and as sex-steroid precursors secreted from the adrenal glands. These androgens provide an abundant reservoir from which all estrogens are subsequently synthesized and their levels are undiminished in the event of standard hormonal therapeutic intervention in breast cancer. Steroid levels are known to be altered by lifestyle factors such as diet and exercise; understanding their potential role in dictating the function of AR in breast cancer development could therefore have wide-ranging effects in prevention and treatment of this disease. This review will outline the endogenous biochemical drivers of both genomic and non-genomic AR activation and how these may be modulated by current hormonal therapies.
Collapse
Affiliation(s)
| | - Marie McIlroy
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
57
|
Munir MT, Ponce C, Powell CA, Tarafdar K, Yanagita T, Choudhury M, Gollahon LS, Rahman SM. The contribution of cholesterol and epigenetic changes to the pathophysiology of breast cancer. J Steroid Biochem Mol Biol 2018; 183:1-9. [PMID: 29733910 DOI: 10.1016/j.jsbmb.2018.05.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 04/12/2018] [Accepted: 05/03/2018] [Indexed: 12/30/2022]
Abstract
Breast cancer is one of the most commonly diagnosed cancers in women. Accumulating evidence suggests that cholesterol plays an important role in the development of breast cancer. Even though the mechanistic link between these two factors is not well understood, one possibility is that dysregulated cholesterol metabolism may affect lipid raft and membrane fluidity and can promote tumor development. Current studies have shown oxysterol 27-hydroxycholesterol (27-HC) as a critical regulator of cholesterol and breast cancer pathogenesis. This is supported by the significantly higher expression of CYP27A1 (cytochrome P450, family 27, subfamily A, polypeptide 1) in breast cancers. This enzyme is responsible for 27-HC synthesis from cholesterol. It has been shown that 27-HC can not only increase the proliferation of estrogen receptor (ER)-positive breast cancer cells but also stimulate tumor growth and metastasis in several breast cancer models. This phenomenon is surprising since 27-HC and other oxysterols generally reduce intracellular cholesterol levels by activating the liver X receptors (LXRs). Resolving this paradox will elucidate molecular pathways by which cholesterol, ER, and LXR are connected to breast cancer. These findings will also provide the rationale for evaluating pharmaceutical approaches that manipulate cholesterol or 27-HC synthesis in order to mitigate the impact of cholesterol on breast cancer pathophysiology. In addition to cholesterol, epigenetic changes including non-coding RNAs, and microRNAs, DNA methylation, and histone modifications, have all been shown to control tumorigenesis. The purpose of this review is to discuss the link between altered cholesterol metabolism and epigenetic modification during breast cancer progression.
Collapse
Affiliation(s)
- Maliha T Munir
- Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| | | | - Catherine A Powell
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Sciences Center, College Station, Texas, USA
| | | | | | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Sciences Center, College Station, Texas, USA
| | - Lauren S Gollahon
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Shaikh M Rahman
- Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA.
| |
Collapse
|
58
|
Challenges and perspectives in the treatment of diabetes associated breast cancer. Cancer Treat Rev 2018; 70:98-111. [PMID: 30130687 DOI: 10.1016/j.ctrv.2018.08.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/02/2018] [Accepted: 08/09/2018] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes mellitus is one of the most common chronic disease worldwide and affects all cross-sections of the society including children, women, youth and adults. Scientific evidence has linked diabetes to higher incidence, accelerated progression and increased aggressiveness of different cancers. Among the different forms of cancer, research has reinforced a link between diabetes and the risk of breast cancer. Some studies have specifically linked diabetes to the highly aggressive, triple negative breast cancers (TNBCs) which do not respond to conventional hormonal/HER2 targeted interventions, have chances of early recurrence, metastasize, tend to be more invasive in nature and develop drug resistance. Commonly used anti-diabetic drugs, such as metformin, have recently gained importance in the treatment of breast cancer due to their proposed anti-cancer properties. Here we discuss the link between diabetes and breast cancer, the metabolic disturbances in diabetes that support the development of breast cancer, the challenges involved and future perspective and directions. We link the three main metabolic disturbances (dyslipidemia, hyperinsulinemia and hyperglycemia) that occur in diabetes to potential aberrant molecular pathways that may lead to the development of an oncogenic phenotype of the breast tissue, thereby leading to acceleration of cell growth, proliferation, migration, inflammation, angiogenesis, EMT and metastasis and inhibition of apoptosis in breast cancer cells. Furthermore, managing diabetes and treating cancer using a combination of anti-diabetic and classical anti-cancer drugs should prove to be more efficient in the treatment diabetes associated cancers.
Collapse
|
59
|
Huang D, Yuan W, Li H, Li S, Chen Z, Yang H. Identification of key pathways and biomarkers in sorafenib-resistant hepatocellular carcinoma using bioinformatics analysis. Exp Ther Med 2018; 16:1850-1858. [PMID: 30186410 PMCID: PMC6122189 DOI: 10.3892/etm.2018.6427] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 04/26/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant types of cancer, with a high mortality rate. Sorafenib is the sole approved oral clinical therapy against advanced HCC. However, individual patients exhibit varying responses to sorafenib and the development of sorafenib resistance has been a new challenge for its clinical efficacy. The current study identified gene biomarkers and key pathways in sorafenib-resistant HCC using bioinformatics analysis. Gene dataset GSE73571 was obtained from the Gene Expression Omnibus (GEO) database, including four sorafenib-acquired resistant and three sorafenib-sensitive HCC phenotypes. Differentially expressed genes (DEGs) were identified using the web tool GEO2R. Functional and pathway enrichment of DEGs were analyzed using the Database for Annotation, Visualization and Integrated Discovery and the protein-protein interaction (PPI) network was constructed using the Search Tool for the Retrieval of Interacting Genes/Proteins and Cytoscape. A total of 1,319 DEGs were selected, which included 593 upregulated and 726 downregulated genes. Functional and pathway enrichment analysis revealed DEGs enriched in negative regulation of endopeptidase activity, cholesterol homeostasis, DNA replication and repair, coagulation cascades, insulin resistance, RNA transport, cell cycle and others. Eight hub genes, including kininogen 1, vascular cell adhesion molecule 1, apolipoprotein C3, alpha 2-HS glycoprotein, erb-b2 receptor tyrosine kinase 2, secreted protein acidic and cysteine rich, vitronectin and vimentin were identified from the PPI network. In conclusion, the present study identified DEGs and key genes in sorafenib-resistant HCC, which further the knowledge of potential mechanisms in the development of sorafenib resistance and may provide potential targets for early diagnosis and new treatments for sorafenib-resistant HCC.
Collapse
Affiliation(s)
- Danping Huang
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Weiqu Yuan
- Acupuncture Department, The Fourth Clinical Medical College of Guangzhou University Chinese Medicine, Shenzhen, Guangdong 518000, P.R. China
| | - Hanmin Li
- Hepatopathy Institution, Affiliated Hospital Hubei University Chinese Medicine, Wuhan, Hubei 430061, P.R. China
| | - Shaodong Li
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Zuanguang Chen
- Pharmaceutical Analysis Department, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Hongzhi Yang
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|
60
|
Haque MW, Bose P, Siddique MUM, Sunita P, Lapenna A, Pattanayak SP. Taxifolin binds with LXR (α & β) to attenuate DMBA-induced mammary carcinogenesis through mTOR/Maf-1/PTEN pathway. Biomed Pharmacother 2018; 105:27-36. [PMID: 29843042 DOI: 10.1016/j.biopha.2018.05.114] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/23/2018] [Accepted: 05/23/2018] [Indexed: 01/27/2023] Open
Abstract
AIM 7,12-dimethylbenz(a)anthracene(DMBA), a PAH derivative initializes cascades of signaling events that alters a variety of enzymes responsible for lipid and glucose homeostasis resulting in enhanced availability and consumption of energy producing molecules for the development of carcinogenesis. 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMG-CoAR) is a key enzyme regulating the pathway of synthesis of cholesterol whereas liver-X-receptor (LXR) regulates lipid, carbohydrate metabolism in various malignancies including mammary carcinogenesis (MC). In this study Taxifolin (TAX), a potential flavanoid has been subjected to evaluate its anti-cancer potential on (MC). METHODS We designed to screen the molecular docking analysis of TAX on LXRα, LXRβ, HMG-CoAR, mTOR and PTEN using MAESTRO tool comparing with their reference ligands. MC was developed by the administration of DMBA in the air pouch (under the mammary fat pad) of the female Sprague-Dawley rats (55 days old). After 90 days of cancer induction, the chemotherapeutic potential of TAX was evaluated by administering TAX at different doses (10, 20 and 40 mg/kg b.w./day). Then western blot and RT-qPCR analysis were performed for determination of the protein and mRNA expressions respectively. RESULTS The docking analysis revealed significant interaction with LXR (α&β), HMG-CoAR, mTOR and PTEN. The docking results were validated with the enzyme inhibition assay using HMG-CoAR (EC 1.1.1.34). TAX inhibited the HMG-CoAR activity with an IC50 value of 97.54 ± 2.5 nM whereas the reference molecule pavastatin revealed an IC50 value of 84.35 ± 1.2 nM. Moreover, TAX modulated the energy regulation on DMBA-induced MC in SD-rats by significantly restoring the cancer-induced alterations in body weight, tumor growth and lipid, lipoproteins, lipid metabolizing enzymes and glycolytic enzymes. TAX interacted with LXRs, HMG-CoAR, metabolic enzymes and restored the altered metabolism that accelerates uncontrolled cell proliferation in MC. Moreover, TAX also altered the mRNA and protein expressions of HMG-CoAR, LXR (α,β), Maf1, PTEN, phosphoinositide 3-kinase (PI3K), Akt, mTOR, fatty acid synthase (FASN) and acetyl-CoA carboxylase 1 (ACC1) in a dose dependent manner. CONCLUSION These results validate the anti-cancer potential of TAX in DMBA-induced MC through LXR-mTOR/Maf1/PTEN axis.
Collapse
Affiliation(s)
- Md Wasimul Haque
- Division of Advanced Pharmacology, Department of Pharm. Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, India
| | - Pritha Bose
- Division of Advanced Pharmacology, Department of Pharm. Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, India
| | - Mohd Usman Mohd Siddique
- Division of Pharmaceutical Chemistry, Department of Pharm. Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India
| | - Priyashree Sunita
- Government Pharmacy Institute, Department of Health, Education & Family Welfare, Govt. of Jharkhand, Bariatu, Ranchi, 834009, India
| | - Antonio Lapenna
- Academic Unit of Inflammation & Tumor Targeting, Dept. of Oncology and Metabolism, University of Sheffield Medical School, Beech Hill Road, Sheffield, S10 2RX, United Kingdom
| | - Shakti P Pattanayak
- Division of Advanced Pharmacology, Department of Pharm. Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, India.
| |
Collapse
|
61
|
Zhang RX, Li J, Zhang T, Amini MA, He C, Lu B, Ahmed T, Lip H, Rauth AM, Wu XY. Importance of integrating nanotechnology with pharmacology and physiology for innovative drug delivery and therapy - an illustration with firsthand examples. Acta Pharmacol Sin 2018; 39:825-844. [PMID: 29698389 DOI: 10.1038/aps.2018.33] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 02/19/2018] [Indexed: 12/13/2022]
Abstract
Nanotechnology has been applied extensively in drug delivery to improve the therapeutic outcomes of various diseases. Tremendous efforts have been focused on the development of novel nanoparticles and delineation of the physicochemical properties of nanoparticles in relation to their biological fate and functions. However, in the design and evaluation of these nanotechnology-based drug delivery systems, the pharmacology of delivered drugs and the (patho-)physiology of the host have received less attention. In this review, we discuss important pharmacological mechanisms, physiological characteristics, and pathological factors that have been integrated into the design of nanotechnology-enabled drug delivery systems and therapies. Firsthand examples are presented to illustrate the principles and advantages of such integrative design strategies for cancer treatment by exploiting 1) intracellular synergistic interactions of drug-drug and drug-nanomaterial combinations to overcome multidrug-resistant cancer, 2) the blood flow direction of the circulatory system to maximize drug delivery to the tumor neovasculature and cells overexpressing integrin receptors for lung metastases, 3) endogenous lipoproteins to decorate nanocarriers and transport them across the blood-brain barrier for brain metastases, and 4) distinct pathological factors in the tumor microenvironment to develop pH- and oxidative stress-responsive hybrid manganese dioxide nanoparticles for enhanced radiotherapy. Regarding the application in diabetes management, a nanotechnology-enabled closed-loop insulin delivery system was devised to provide dynamic insulin release at a physiologically relevant time scale and glucose levels. These examples, together with other research results, suggest that utilization of the interplay of pharmacology, (patho-)physiology and nanotechnology is a facile approach to develop innovative drug delivery systems and therapies with high efficiency and translational potential.
Collapse
|
62
|
Seiler A, Chen MA, Brown RL, Fagundes CP. Obesity, Dietary Factors, Nutrition, and Breast Cancer Risk. CURRENT BREAST CANCER REPORTS 2018; 10:14-27. [PMID: 30662586 PMCID: PMC6335046 DOI: 10.1007/s12609-018-0264-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW To synthesize the critical role of obesity-associated inflammation, dietary factors, and nutrition in determining breast cancer risk. RECENT FINDINGS Obesity-associated inflammation is strongly linked to breast cancer risk and progression, largely via two processes: inflammatory pathways and dysregulated metabolism. Cytokine production in excess adipose tissues creates a chronic inflammatory microenvironment, which favors tumor development. Lifestyle factors, including diet, have long been recognized as important determinants of breast cancer risk and mortality. SUMMARY Obesity increases the risk of developing breast cancer in both pre- and postmenopausal women and also negatively affects breast cancer recurrence and survival. Poor dietary habits characterized by the high intake of refined starches, sugar, and both saturated and trans-saturated fats, as well as the low intake of omega-3 fatty acids, natural antioxidants, and fiber, modulate inflammation and, thereby, appear to be linked to increased risk of breast cancer and mortality.
Collapse
Affiliation(s)
- Annina Seiler
- Department of Consultation-Liaison-Psychiatry and Psychosomatic Medicine, University Hospital Zurich, Haldenbachstrasse 18, 8091 Zurich, Switzerland
| | | | - Ryan L Brown
- Department of Psychology, Rice University, Houston, TX, USA
| | - Christopher P Fagundes
- Department of Psychology, Rice University, Houston, TX, USA
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
63
|
Yamauchi Y, Rogers MA. Sterol Metabolism and Transport in Atherosclerosis and Cancer. Front Endocrinol (Lausanne) 2018; 9:509. [PMID: 30283400 PMCID: PMC6157400 DOI: 10.3389/fendo.2018.00509] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/14/2018] [Indexed: 01/22/2023] Open
Abstract
Cholesterol is a vital lipid molecule for mammalian cells, regulating fluidity of biological membranes, and serving as an essential constituent of lipid rafts. Mammalian cells acquire cholesterol from extracellular lipoproteins and from de novo synthesis. Cholesterol biosynthesis generates various precursor sterols. Cholesterol undergoes metabolic conversion into oxygenated sterols (oxysterols), bile acids, and steroid hormones. Cholesterol intermediates and metabolites have diverse and important cellular functions. A network of molecular machineries including transcription factors, protein modifiers, sterol transporters/carriers, and sterol sensors regulate sterol homeostasis in mammalian cells and tissues. Dysfunction in metabolism and transport of cholesterol, sterol intermediates, and oxysterols occurs in various pathophysiological settings such as atherosclerosis, cancers, and neurodegenerative diseases. Here we review the cholesterol, intermediate sterol, and oxysterol regulatory mechanisms and intracellular transport machineries, and discuss the roles of sterols and sterol metabolism in human diseases.
Collapse
Affiliation(s)
- Yoshio Yamauchi
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
- *Correspondence: Yoshio Yamauchi
| | - Maximillian A. Rogers
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
64
|
The cholesterol metabolite 27 hydroxycholesterol facilitates breast cancer metastasis through its actions on immune cells. Nat Commun 2017; 8:864. [PMID: 29021522 PMCID: PMC5636879 DOI: 10.1038/s41467-017-00910-z] [Citation(s) in RCA: 291] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 08/07/2017] [Indexed: 12/29/2022] Open
Abstract
Obesity and elevated circulating cholesterol are risk factors for breast cancer recurrence, while the use of statins, cholesterol biosynthesis inhibitors widely used for treating hypercholesterolemia, is associated with improved disease-free survival. Here, we show that cholesterol mediates the metastatic effects of a high-fat diet via its oxysterol metabolite, 27-hydroxycholesterol. Ablation or inhibition of CYP27A1, the enzyme responsible for the rate-limiting step in 27-hydroxycholesterol biosynthesis, significantly reduces metastasis in relevant animal models of cancer. The robust effects of 27-hydroxycholesterol on metastasis requires myeloid immune cell function, and it was found that this oxysterol increases the number of polymorphonuclear-neutrophils and γδ-T cells at distal metastatic sites. The pro-metastatic actions of 27-hydroxycholesterol requires both polymorphonuclear-neutrophils and γδ-T cells, and 27-hydroxycholesterol treatment results in a decreased number of cytotoxic CD8+T lymphocytes. Therefore, through its actions on γδ-T cells and polymorphonuclear-neutrophils, 27-hydroxycholesterol functions as a biochemical mediator of the metastatic effects of hypercholesterolemia.High cholesterol is a risk factor for breast cancer recurrence. Here the authors show that cholesterol promotes breast cancer metastasis via its metabolite 27-hydroxycholesterol (27HC) that acts on immune myeloid cells residing at the distal metastatic sites, thus promoting an immune suppressive environment.
Collapse
|
65
|
Shapira N. The potential contribution of dietary factors to breast cancer prevention. Eur J Cancer Prev 2017; 26:385-395. [PMID: 28746163 PMCID: PMC5553235 DOI: 10.1097/cej.0000000000000406] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 06/21/2017] [Indexed: 12/30/2022]
Abstract
Breast cancer (BC), the leading cancer in women, is increasing in prevalence worldwide, concurrent with western metabolic epidemics, that is, obesity, metabolic syndrome, and diabetes, and shares major risk factors with these diseases. The corresponding potential for nutritional contributions toward BC prevention is reviewed and related to critical stages in the life cycle and their implications for carcinogenic and pathometabolic trajectories. BC initiation potentially involves diet-related pro-oxidative, inflammatory, and procarcinogenic processes, that interact through combined lipid/fatty acid peroxidation, estrogen metabolism, and related DNA-adduct/depurination/mutation formation. The pathometabolic trajectory is affected by high estrogen, insulin, and growth factor cascades and resultant accelerated proliferation/progression. Anthropometric risk factors - high birth weight, adult tallness, adiposity/BMI, and weight gain - are often reflective of these trends. A sex-based nutritional approach targets women's specific risk in western obesogenic environments, associated with increasing fatness, estrogen metabolism, n-6 : n-3 polyunsaturated fatty acid ratio, and n-6 polyunsaturated fatty acid conversion to proinflammatory/carcinogenic eicosanoids, and effects of timing of life events, for example, ages at menarche, full-term pregnancy, and menopause. Recent large-scale studies have confirmed the effectiveness of the evidence-based recommendations against BC risk, emphasizing low-energy density diets, highly nutritious plant-based regimes, physical activity, and body/abdominal adiposity management. Better understanding of dietary inter-relationships with BC, as applied to food intake, selection, combination, and processing/preparation, and recommended patterns, for example, Mediterranean, DASH, plant-based, low energy density, and low glycemic load, with high nutrient/phytonutrient density, would increase public motivation and authoritative support for early/timely prevention, optimally merging with other dietary/health goals, for lifelong BC prevention.
Collapse
Affiliation(s)
- Niva Shapira
- Department of Nutrition, School of Health Professions, Ashkelon Academic College, Ashkelon, Israel
| |
Collapse
|
66
|
Kimbung S, Chang CY, Bendahl PO, Dubois L, Thompson JW, McDonnell DP, Borgquist S. Impact of 27-hydroxylase (CYP27A1) and 27-hydroxycholesterol in breast cancer. Endocr Relat Cancer 2017; 24:339-349. [PMID: 28442559 DOI: 10.1530/erc-16-0533] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 04/25/2017] [Indexed: 11/08/2022]
Abstract
The impact of systemic 27-hydroxycholesterol (27HC) and intratumoral CYP27A1 expression on pathobiology and clinical response to statins in breast cancer needs clarification. 27HC is an oxysterol produced from cholesterol by the monooxygenase CYP27A1, which regulates intracellular cholesterol homeostasis. 27HC also acts as an endogenous selective estrogen receptor (ER) modulator capable of increasing breast cancer growth and metastasis. 27HC levels can be modulated by statins or direct inhibition of CYP27A1, thereby attenuating its pro-tumorigenic activities. Herein, the effect of statins on serum 27HC and tumor-specific CYP27A1 expression was evaluated in 42 breast cancer patients treated with atorvastatin within a phase II clinical trial. Further, the associations between CYP27A1 expression with other primary tumor pathological features and clinical outcomes were studied in two additional independent cohorts. Statin treatment effectively decreased serum 27HC and deregulated CYP27A1 expression in tumors. However, these changes were not associated with anti-proliferative responses to statin treatment. CYP27A1 was heterogeneously expressed among primary tumors, with high expression significantly associated with high tumor grade, ER negativity and basal-like subtype. High CYP27A1 expression was independently prognostic for longer recurrence-free and overall survival. Importantly, the beneficial effect of high CYP27A1 in ER-positive breast cancer seemed limited to women aged ≤50 years. These results establish a link between CYP27A1 and breast cancer pathobiology and prognosis and propose that the efficacy of statins in reducing serum lipids does not directly translate to anti-proliferative effects in tumors. Changes in other undetermined serum or tumor factors suggestively mediate the anti-proliferative effects of statins in breast cancer.
Collapse
Affiliation(s)
- Siker Kimbung
- Division of Oncology and PathologyDepartment of Clinical Sciences, Lund, Lund University, Sweden
| | - Ching-Yi Chang
- Department of Pharmacology and Cancer BiologyDuke University School of Medicine, Durham, NC, USA
| | - Pär-Ola Bendahl
- Division of Oncology and PathologyDepartment of Clinical Sciences, Lund, Lund University, Sweden
| | - Laura Dubois
- Duke Proteomics and Metabolomics ResourceDuke University School of Medicine, Durham, NC, USA
| | - J Will Thompson
- Department of Pharmacology and Cancer BiologyDuke University School of Medicine, Durham, NC, USA
- Duke Proteomics and Metabolomics ResourceDuke University School of Medicine, Durham, NC, USA
| | - Donald P McDonnell
- Department of Pharmacology and Cancer BiologyDuke University School of Medicine, Durham, NC, USA
| | - Signe Borgquist
- Division of Oncology and PathologyDepartment of Clinical Sciences, Lund, Lund University, Sweden
- Clinical Trial UnitClinical Studies Sweden, Forum South, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
67
|
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
|
68
|
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
|
69
|
Wallbillich JJ, Josyula S, Saini U, Zingarelli RA, Dorayappan KDP, Riley MK, Wanner RA, Cohn DE, Selvendiran K. High Glucose-Mediated STAT3 Activation in Endometrial Cancer Is Inhibited by Metformin: Therapeutic Implications for Endometrial Cancer. PLoS One 2017; 12:e0170318. [PMID: 28114390 PMCID: PMC5256996 DOI: 10.1371/journal.pone.0170318] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/03/2017] [Indexed: 12/14/2022] Open
Abstract
Objectives STAT3 is over-expressed in endometrial cancer, and diabetes is a risk factor for the development of type 1 endometrial cancer. We therefore investigated whether glucose concentrations influence STAT3 expression in type 1 endometrial cancer, and whether such STAT3 expression might be inhibited by metformin. Methods In Ishikawa (grade 1) endometrial cancer cells subjected to media with low, normal, or high concentrations of glucose, expression of STAT3 and its target proteins was evaluated by real-time quantitative PCR (qPCR). Ishikawa cells were treated with metformin and assessed with cell proliferation, survival, migration, and ubiquitin assays, as well as Western blot and qPCR. Expression of apoptosis proteins was evaluated with Western blot in Ishikawa cells transfected with a STAT3 overexpression plasmid and treated with metformin. A xenograft tumor model was used for studying the in vivo efficacy of metformin. Results Expression of STAT3 and its target proteins was increased in Ishikawa cells cultured in high glucose media. In vitro, metformin inhibited cell proliferation, survival and migration but induced apoptosis. Metformin reduced expression levels of pSTAT3 ser727, total STAT3, and its associated cell survival and anti-apoptotic proteins. Additionally, metformin treatment was associated with increased degradation of pSTAT3 ser727. No change in apoptotic protein expression was noticed with STAT3 overexpression in Ishikawa cells. In vivo, metformin treatment led to a decrease in tumor weight as well as reductions of STAT3, pSTAT3 ser727, its target proteins. Conclusions These results suggest that STAT3 expression in type 1 endometrial cancer is stimulated by a high glucose environment and inhibited by metformin.
Collapse
Affiliation(s)
- John J. Wallbillich
- Division of Gynecologic Oncology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Srirama Josyula
- Division of Gynecologic Oncology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Uksha Saini
- Division of Gynecologic Oncology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Roman A. Zingarelli
- Division of Gynecologic Oncology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Kalpana Deepa Priya Dorayappan
- Division of Gynecologic Oncology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Maria K. Riley
- Division of Gynecologic Oncology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Ross A. Wanner
- Division of Gynecologic Oncology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - David E. Cohn
- Division of Gynecologic Oncology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Karuppaiyah Selvendiran
- Division of Gynecologic Oncology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
70
|
Engin A. Obesity-associated Breast Cancer: Analysis of risk factors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:571-606. [PMID: 28585217 DOI: 10.1007/978-3-319-48382-5_25] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Several studies show that a significantly stronger association is obvious between increased body mass index (BMI) and higher breast cancer incidence. Furthermore, obese women are at higher risk of all-cause and breast cancer specific mortality when compared to non-obese women with breast cancer. In this context, increased levels of estrogens due to excessive aromatization activity of the adipose tissue, overexpression of pro-inflammatory cytokines, insulin resistance, hyperactivation of insulin-like growth factors (IGFs) pathways, adipocyte-derived adipokines, hypercholesterolemia and excessive oxidative stress contribute to the development of breast cancer in obese women. While higher breast cancer risk with hormone replacement therapy is particularly evident among lean women, in postmenopausal women who are not taking exogenous hormones, general obesity is a significant predictor for breast cancer. Moreover, increased plasma cholesterol leads to accelerated tumor formation and exacerbates their aggressiveness. In contrast to postmenopausal women, premenopausal women with high BMI are inversely associated with breast cancer risk. Nevertheless, life-style of women for breast cancer risk is regulated by avoiding the overweight and a high-fat diet. Estrogen-plus-progestin hormone therapy users for more than 5 years have elevated risks of both invasive ductal and lobular breast cancer. Additionally, these cases are more commonly node-positive and have a higher cancer-related mortality. Collectively, in this chapter, the impacts of obesity-related estrogen, cholesterol, saturated fatty acid, leptin and adiponectin concentrations, aromatase activity, leptin and insulin resistance on breast cancer patients are evaluated. Obesity-related prognostic factors of breast cancer also are discussed at molecular basis.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey. .,, Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
71
|
Nian X, Nagai Y, Jeffers C, N. Maxwell K, Zhang H. Dietary influence on estrogens and cytokines in breast cancer. AIMS MOLECULAR SCIENCE 2017. [DOI: 10.3934/molsci.2017.3.252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
72
|
Synthesis and Experimental Validation of New Designed Heterocyclic Compounds with Antiproliferative Activity versus Breast Cancer Cell Lines MCF-7 and MDA-MB-231. J CHEM-NY 2017. [DOI: 10.1155/2017/9729284] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Recent drug discovery efforts are highly focused towards identification, design, and synthesis of small molecules as anticancer agents. With this aim, we recently designed and synthesized novel compounds with high efficacy and specificity for the treatment of breast tumors. Based on the obtained results, we constructed a Volsurf+ (VS+) model using a dataset of 59 compounds able to predict the in vitro antitumor activity against MCF-7 cancer cell line for new derivatives. In the present paper, in order to further verify the robustness of this model, we report the results of the projection of more than 150 known molecules and 9 newly synthesized compounds. We predict their activity versus MCF-7 cell line and experimentally verify the in silico results for some promising chosen molecules in two human breast cell lines, MCF-7 and MDA-MB-231.
Collapse
|
73
|
Mutemberezi V, Guillemot-Legris O, Muccioli GG. Oxysterols: From cholesterol metabolites to key mediators. Prog Lipid Res 2016; 64:152-169. [PMID: 27687912 DOI: 10.1016/j.plipres.2016.09.002] [Citation(s) in RCA: 246] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/13/2016] [Accepted: 09/23/2016] [Indexed: 12/13/2022]
Abstract
Oxysterols are cholesterol metabolites that can be produced through enzymatic or radical processes. They constitute a large family of lipids (i.e. the oxysterome) involved in a plethora of physiological processes. They can act through GPCR (e.g. EBI2, SMO, CXCR2), nuclear receptors (LXR, ROR, ERα) and through transporters or regulatory proteins. Their physiological effects encompass cholesterol, lipid and glucose homeostasis. Additionally, they were shown to be involved in other processes such as immune regulatory functions and brain homeostasis. First studied as precursors of bile acids, they quickly emerged as interesting lipid mediators. Their levels are greatly altered in several pathologies and some oxysterols (e.g. 4β-hydroxycholesterol or 7α-hydroxycholestenone) are used as biomarkers of specific pathologies. In this review, we discuss the complex metabolism and molecular targets (including binding properties) of these bioactive lipids in human and mice. We also discuss the genetic mouse models currently available to interrogate their effects in pathophysiological settings. We also summarize the levels of oxysterols reported in two key organs in oxysterol metabolism (liver and brain), plasma and cerebrospinal fluid. Finally, we consider future opportunities and directions in the oxysterol field in order to gain a better insight and understanding of the complex oxysterol system.
Collapse
Affiliation(s)
- Valentin Mutemberezi
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Belgium
| | - Owein Guillemot-Legris
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Belgium.
| |
Collapse
|
74
|
Ma J, Fu G, Wu J, Han S, Zhang L, Yang M, Yu Y, Zhang M, Lin Y, Wang Y. 4-cholesten-3-one suppresses lung adenocarcinoma metastasis by regulating translocation of HMGB1, HIF1α and Caveolin-1. Cell Death Dis 2016; 7:e2372. [PMID: 27899819 PMCID: PMC5059879 DOI: 10.1038/cddis.2016.281] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/09/2016] [Accepted: 08/10/2016] [Indexed: 12/15/2022]
Abstract
Metastasis is a great challenge in lung adenocarcinoma (ADC) therapy. Cholesterol has been implicated in ADC metastasis. 4-cholesten-3-one, as cholesterol metabolite and analog, can substitute membrane cholesterol and increase membrane fluidity. In this study, we explored the possibility that 4-cholesten-3-one inhibited ADC metastasis. Low-dose 4-cholesten-3-one significantly restrained ADC cells migration and invasion with little effects on cells viabilities. Further investigation showed that 4-cholesten-3-one promoted ROS generation, which transiently activated AMPKα1, increased HIF1α expression, reduced Bcl-2 expression and caused autophagy. AMPKα1 knockdown partly suppressed 4-cholesten-3-one-induced autophagy but, neither prevented 4-cholesten-3-one-induced upregulation of HIF1α or downregulation of Bcl-2. 4-cholesten-3-one-induced autophagy facilitated the release of HMGB1 from nuclei to cytoplasm, blocking nuclear translocation of HIF1α and activation of MMP2 and MMP9. Also, 4-cholesten-3-one induced time-dependent phosphorylation of caveolin-1, Akt and NF-κB. With increasing treatment time, 4-cholesten-3-one accelerated caveolin-1 internalization, but reduced the phosphorylation of Akt and NF-κB, and inhibited the expression of snail and twist. These data suggested that 4-cholesten-3-one could be a potential candidate for anti-metastasis of lung adenocarcinoma.
Collapse
Affiliation(s)
- Jinben Ma
- Department of Anesthesiology, Shandong provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Guobin Fu
- Department of Oncology, Shandong provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Jing Wu
- Department of Anesthesiology, Shandong provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Shaoxian Han
- Department of thoracic surgery, Shandong chest Hospital, Jinan 250021, China
| | - Lishan Zhang
- Department of Hand and Foot Surgery, Shandong provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Ming Yang
- Department of Ultrasound, Shandong provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Yong Yu
- Department of Ultrasound, Shandong provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Mengyuan Zhang
- Department of Anesthesiology, Shandong provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Yanliang Lin
- Department of Center Laboratory, Shandong provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Yibing Wang
- Department of burn and plastic surgery, Shandong provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| |
Collapse
|
75
|
Chen CC, Hsu YP, Liu JC, Kao PF, Sung LC, Lin CF, Hao WR, Liu SH, Wu SY. Statins Dose-Dependently Exert Significant Chemopreventive Effects Against Various Cancers in Chronic Obstructive Pulmonary Disease Patients: A Population-Based Cohort Study. J Cancer 2016; 7:1892-1900. [PMID: 27698930 PMCID: PMC5039374 DOI: 10.7150/jca.15779] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 06/29/2016] [Indexed: 01/14/2023] Open
Abstract
PURPOSE: Chronic obstructive pulmonary disease (COPD) is associated with an increased cancer risk. We evaluated the chemopreventive effect of statins against all cancers in COPD patients and identified the statin with the strongest chemopreventive effect. PATIENTS AND METHODS: All patients diagnosed with COPD at health care facilities in Taiwan (n = 116,017) from January 1, 2001, to December 31, 2012, were recruited. Each patient was followed to assess the following protective and risk factors for all cancers: age; sex; comorbidities (diabetes, hypertension, dyslipidemia) and the Charlson comorbidity index [CCI]); urbanization level; monthly income; and nonstatin drug use. The index date of statins use was the date of COPD confirmation. Propensity scores (PSs) were derived using a logistic regression model to estimate the effect of statins by considering the covariates predicting intervention (statins) receipt. To examine the dose-response relationship, we categorized statin use into four groups in each cohort (<28 [statin nonusers], 28-90, 91-365, and >365 cumulative defined daily dose). RESULTS: After PS adjustment for age, sex, CCI, diabetes, hypertension, dyslipidemia, urbanization level, and monthly income, we analyzed the all-cancer risk. The adjusted hazard ratios (aHRs) for the all-cancer risk were lower among statin users than among statin nonusers (aHR = 0.46, 95% confidence interval: 0.43 to 0.50). The aHRs for the all-cancer risk were lower among patients using rosuvastatin, simvastatin, atorvastatin, pravastatin, and fluvastatin than among statin nonusers (aHRs = 0.42, 0.55, 0.59, 0.66, and 0.78, respectively). Sensitivity analysis indicated that statins dose-dependently reduced the all-cancer risk. CONCLUSION: Statins dose-dependently exert a significant chemopreventive effect against various cancers in COPD patients. In particular, rosuvastatin has the strongest chemopreventive effect.
Collapse
Affiliation(s)
- Chun-Chao Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yi-Ping Hsu
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Ju-Chi Liu
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Pai-Feng Kao
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Li-Chin Sung
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chao-Feng Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Wen-Rui Hao
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Szu-Yuan Wu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Radiation Oncology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Biotechnology, Hungkuang University, Taichung, Taiwan
| |
Collapse
|
76
|
Kong S, Ding C, Huang L, Bai Y, Xiao T, Guo J, Su Z. The effects of COST on the differentiation of 3T3-L1 preadipocytes and the mechanism of action. Saudi J Biol Sci 2016; 24:251-255. [PMID: 28149159 PMCID: PMC5272960 DOI: 10.1016/j.sjbs.2016.09.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 08/31/2016] [Accepted: 09/02/2016] [Indexed: 11/28/2022] Open
Abstract
The objectives of this study were to explore the effect of COST (one thousand Da molecular weight chitosan oligosaccharide) on the differentiation of 3T3-L1 preadipocytes and to determine the mechanism of action. 3T3-L1 preadipocytes were used as the target cells, and the induction of the methods for the differentiation of 3T3-L1 preadipocytes was based on classic cocktails. The MTT assay was used to filtrate the concentration of COST. On the 6th day of induced-differentiation, the differentiation of 3T3-L1 cells was detected by Oil Red O staining. The expression of PPARγ and C/EBPα mRNA was determined using real-time fluorescence quantitative PCR (Q-PCR). COST inhibited 3T3-L1 preadipocyte differentiation in a dose-dependent manner and decreased lipid accumulation. At the molecular level, the expression of the transcription factors, PPARγ and C/EBPα, was reduced by COST during adipogenesis. These results indicate that COST effectively inhibited the differentiation of 3T3-L1 preadipocytes. The mechanism is related to the down-regulation expression of PPARγ and C/EBPα.
Collapse
Affiliation(s)
- Shang Kong
- Key Research Center of Liver Regulation for Hyperlipidemia SATCM/Class III Laboratory of Metabolism SATCM, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chen Ding
- Key Research Center of Liver Regulation for Hyperlipidemia SATCM/Class III Laboratory of Metabolism SATCM, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lanlan Huang
- Key Research Center of Liver Regulation for Hyperlipidemia SATCM/Class III Laboratory of Metabolism SATCM, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yan Bai
- Guangzhou Boxabio Technology Ltd, Guangzhou Science City, China
| | - Tiancun Xiao
- Inorganic Chemistry Laboratory, Oxford University, South Parks Road, OX1 3QR, United Kingdom; Guangzhou Boxabio Technology Ltd, Guangzhou Science City, China
| | - Jiao Guo
- Key Research Center of Liver Regulation for Hyperlipidemia SATCM/Class III Laboratory of Metabolism SATCM, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhengquan Su
- Key Research Center of Liver Regulation for Hyperlipidemia SATCM/Class III Laboratory of Metabolism SATCM, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
77
|
Simigdala N, Gao Q, Pancholi S, Roberg-Larsen H, Zvelebil M, Ribas R, Folkerd E, Thompson A, Bhamra A, Dowsett M, Martin LA. Cholesterol biosynthesis pathway as a novel mechanism of resistance to estrogen deprivation in estrogen receptor-positive breast cancer. Breast Cancer Res 2016; 18:58. [PMID: 27246191 PMCID: PMC4888666 DOI: 10.1186/s13058-016-0713-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/26/2016] [Indexed: 02/07/2023] Open
Abstract
Background Therapies targeting estrogenic stimulation in estrogen receptor-positive (ER+) breast cancer (BC) reduce mortality, but resistance remains a major clinical problem. Molecular studies have shown few high-frequency mutations to be associated with endocrine resistance. In contrast, expression profiling of primary ER+ BC samples has identified several promising signatures/networks for targeting. Methods To identify common adaptive mechanisms associated with resistance to aromatase inhibitors (AIs), we assessed changes in global gene expression during adaptation to long-term estrogen deprivation (LTED) in a panel of ER+ BC cell lines cultured in 2D on plastic (MCF7, T47D, HCC1428, SUM44 and ZR75.1) or in 3D on collagen (MCF7) to model the stromal compartment. Furthermore, dimethyl labelling followed by LC-MS/MS was used to assess global changes in protein abundance. The role of target genes/proteins on proliferation, ER-mediated transcription and recruitment of ER to target gene promoters was analysed. Results The cholesterol biosynthesis pathway was the common upregulated pathway in the ER+ LTED but not the ER– LTED cell lines, suggesting a potential mechanism dependent on continued ER expression. Targeting the individual genes of the cholesterol biosynthesis pathway with siRNAs caused a 30–50 % drop in proliferation. Further analysis showed increased expression of 25-hydroxycholesterol (HC) in the MCF7 LTED cells. Exogenous 25-HC or 27-HC increased ER-mediated transcription and expression of the endogenous estrogen-regulated gene TFF1 in ER+ LTED cells but not in the ER– LTED cells. Additionally, recruitment of the ER and CREB-binding protein (CBP) to the TFF1 and GREB1 promoters was increased upon treatment with 25-HC and 27-HC. In-silico analysis of two independent studies of primary ER+ BC patients treated with neoadjuvant AIs showed that increased expression of MSMO1, EBP, LBR and SQLE enzymes, required for cholesterol synthesis and increased in our in-vitro models, was significantly associated with poor response to endocrine therapy. Conclusion Taken together, these data provide support for the role of cholesterol biosynthesis enzymes and the cholesterol metabolites, 25-HC and 27-HC, in a novel mechanism of resistance to endocrine therapy in ER+ BC that has potential as a therapeutic target. Electronic supplementary material The online version of this article (doi:10.1186/s13058-016-0713-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nikiana Simigdala
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Qiong Gao
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Sunil Pancholi
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | | | - Marketa Zvelebil
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Ricardo Ribas
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Elizabeth Folkerd
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK.,Academic Department of Biochemistry, Royal Marsden Hospital, London, SW3 6JJ, UK
| | - Andrew Thompson
- Proteomics Core Facility, Institute of Cancer Research, London, SW3 6JB, UK
| | - Amandeep Bhamra
- Proteomics Core Facility, Institute of Cancer Research, London, SW3 6JB, UK
| | - Mitch Dowsett
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK.,Academic Department of Biochemistry, Royal Marsden Hospital, London, SW3 6JJ, UK
| | - Lesley-Ann Martin
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK.
| |
Collapse
|
78
|
Kuzu OF, Noory MA, Robertson GP. The Role of Cholesterol in Cancer. Cancer Res 2016; 76:2063-70. [PMID: 27197250 DOI: 10.1158/0008-5472.can-15-2613] [Citation(s) in RCA: 478] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 02/08/2016] [Indexed: 12/19/2022]
Abstract
The roles played by cholesterol in cancer development and the potential of therapeutically targeting cholesterol homeostasis is a controversial area in the cancer community. Several epidemiologic studies report an association between cancer and serum cholesterol levels or statin use, while others suggest that there is not one. Furthermore, the Cancer Genome Atlas (TCGA) project using next-generation sequencing has profiled the mutational status and expression levels of all the genes in diverse cancers, including those involved in cholesterol metabolism, providing correlative support for a role of the cholesterol pathway in cancer development. Finally, preclinical studies tend to more consistently support the role of cholesterol in cancer, with several demonstrating that cholesterol homeostasis genes can modulate development. Because of space limitations, this review provides selected examples of the epidemiologic, TCGA, and preclinical data, focusing on alterations in cholesterol homeostasis and its consequent effect on patient survival. In melanoma, this focused analysis demonstrated that enhanced expression of cholesterol synthesis genes was associated with decreased patient survival. Collectively, the studies in melanoma and other cancer types suggested a potential role of disrupted cholesterol homeostasis in cancer development but additional studies are needed to link population-based epidemiological data, the TCGA database results, and preclinical mechanistic evidence to concretely resolve this controversy. Cancer Res; 76(8); 2063-70. ©2016 AACR.
Collapse
Affiliation(s)
- Omer F Kuzu
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Mohammad A Noory
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Gavin P Robertson
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania. Department of Pathology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania. Department of Dermatology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania. Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania. Penn State Hershey Melanoma Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania. Penn State Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania. Penn State Hershey Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania. The Foreman Foundation for Melanoma Research, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania.
| |
Collapse
|
79
|
Flote VG, Vettukattil R, Bathen TF, Egeland T, McTiernan A, Frydenberg H, Husøy A, Finstad SE, Lømo J, Garred Ø, Schlichting E, Wist EA, Thune I. Lipoprotein subfractions by nuclear magnetic resonance are associated with tumor characteristics in breast cancer. Lipids Health Dis 2016; 15:56. [PMID: 26970778 PMCID: PMC4789271 DOI: 10.1186/s12944-016-0225-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 03/08/2016] [Indexed: 12/25/2022] Open
Abstract
Background High-Density Lipoprotein (HDL)-cholesterol, has been associated with breast cancer development, but the association is under debate, and whether lipoprotein subfractions is associated with breast tumor characteristics remains unclear. Methods Among 56 women with newly diagnosed invasive breast cancer stage I/II, aged 35–75 years, pre-surgery overnight fasting serum concentrations of lipids were assessed, and body mass index (BMI) was measured. All breast tumors were immunohistochemically examined in the surgical specimen. Serum metabolomics of lipoprotein subfractions and their contents of cholesterol, free cholesterol, phospholipids, apolipoprotein-A1 and apolipoprotein-A2, were assessed using nuclear magnetic resonance. Principal component analysis, partial least square analysis, and uni- and multivariable linear regression models were used to study whether lipoprotein subfractions were associated with breast cancer tumor characteristics. Results The breast cancer patients had following means: age at diagnosis: 55.1 years; BMI: 25.1 kg/m2; total-Cholesterol: 5.74 mmol/L; HDL-Cholesterol: 1.78 mmol/L; Low-Density Lipoprotein (LDL)-Cholesterol: 3.45 mmol/L; triglycerides: 1.18 mmol/L. The mean tumor size was 16.4 mm, and the mean Ki67 hotspot index was 26.5 %. Most (93 %) of the patients had estrogen receptor (ER) positive tumors (≥1 % ER+), and 82 % had progesterone receptor (PgR) positive tumors (≥10 % PgR+). Several HDL subfraction contents were strongly associated with PgR expression: Apolipoprotein-A1 (β 0.46, CI 0.22–0.69, p < 0.001), HDL cholesterol (β 0.95, CI 0.51–1.39, p < 0.001), HDL free cholesterol (β 2.88, CI 1.28–4.48, p = 0.001), HDL phospholipids (β 0.70, CI 0.36–1.04, p < 0.001). Similar results were observed for the subfractions of HDL1-3. We observed inverse associations between HDL phospholipids and Ki67 (β -0.25, p = 0.008), and in particular between HDL1’s contents of cholesterol, phospholipids, apolipoprotein-A1, apolipoprotein-A2 and Ki67. No association was observed between lipoproteins and ER expression. Conclusion Our findings hypothesize associations between different lipoprotein subfractions, and PgR expression, and Ki 67 % in breast tumors. These findings may have clinical implications, but require confirmation in larger studies. Electronic supplementary material The online version of this article (doi:10.1186/s12944-016-0225-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vidar G Flote
- The Cancer Centre, Oslo University Hospital HF, N-0424, Oslo, Norway.
| | - Riyas Vettukattil
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Tone F Bathen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Thore Egeland
- Department of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, N-1432, Aas, Norway
| | - Anne McTiernan
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Hanne Frydenberg
- The Cancer Centre, Oslo University Hospital HF, N-0424, Oslo, Norway
| | - Anders Husøy
- The Cancer Centre, Oslo University Hospital HF, N-0424, Oslo, Norway
| | - Sissi E Finstad
- Norwegian Directorate of Health, PO Box 7000, St. Olavs plass, N-0130, Oslo, Norway
| | - Jon Lømo
- Department of Pathology, Oslo University Hospital, N-0424, Oslo, Norway
| | - Øystein Garred
- Department of Pathology, Oslo University Hospital, N-0424, Oslo, Norway
| | - Ellen Schlichting
- Department of Breast and Endocrine Surgery, Oslo University Hospital, N-0424, Oslo, Norway
| | - Erik A Wist
- The Cancer Centre, Oslo University Hospital HF, N-0424, Oslo, Norway
| | - Inger Thune
- The Cancer Centre, Oslo University Hospital HF, N-0424, Oslo, Norway.,Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, N-9037, Tromsø, Norway
| |
Collapse
|
80
|
MiR-33a Decreases High-Density Lipoprotein-Induced Radiation Sensitivity in Breast Cancer. Int J Radiat Oncol Biol Phys 2016; 95:791-9. [PMID: 27055396 DOI: 10.1016/j.ijrobp.2016.01.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 11/23/2015] [Accepted: 01/18/2016] [Indexed: 11/22/2022]
Abstract
PURPOSE We previously showed that high-density lipoprotein (HDL) radiosensitizes inflammatory breast cancer (IBC) cells in vitro and is associated with better local control after radiation therapy in IBC patients. The microRNA miR-33 family negatively regulates the adenosine triphosphate binding cassette transporter subfamily A member 1. We hypothesized that variations in miR-33a expression in IBC cancer cells versus non-IBC cells would correlate with radiation sensitivity following exposure to HDL in vitro. METHODS AND MATERIALS MiR-33a expression was analyzed by reverse transcriptase-polymerase chain reaction in 4 cell lines representing common clinical breast cancer subtypes. Overexpression and knockdown of miR-33a was demonstrated via transfection of an miR-33a mimic or an anti-miR-33a construct in high- and low-expressing miR-33a cell lines. Clonogenic survival in vitro in these cells was quantified at baseline and following HDL treatment. MiR-33a expression on distant relapse-free survival (DRFS) of 210 cases downloaded from the Oxford breast cancer dataset was determined. RESULTS Expression levels of miR-33a were lower in IBC cell lines and IBC tumor samples than in non-IBC cell lines and normal breast tissue. Cholesterol concentrations in the cell membranes were higher in IBC cells than in non-IBC cells. Clonogenic survival following 24 hours of HDL treatment was decreased in response to irradiation in the low-miR-33a-expressing cell lines SUM149 and KPL4, but survival following HDL treatment decreased in the high-miR-33a-expressing cell lines MDA-MB-231 and SUM159. In the high-miR-33a-expressing cell lines, anti-miR-33a transfection decreased radiation resistance in clonogenic assays. Conversely, in the low-miR-33a-expressing cell lines, the miR-33a mimic reversed the HDL-induced radiation sensitization. Breast cancer patients in the top quartile based on miR-33a expression had markedly lower rates of DRFS than the bottom quartile (P=.0228, log-rank test). For breast cancer patients treated with radiation, high miR-33a expression predicted worse overall survival (P=.06). CONCLUSIONS Our results reveal miR-33a negatively regulates HDL-induced radiation sensitivity in breast cancer.
Collapse
|
81
|
Abstract
It is considered that hypercholesterolemia is life-threatening and low cholesterol levels are a positive factor. However, taking into consideration the fact that cholesterol plays a key role in cell proliferation, it should be remembered that its low blood level may be linked to high cholesterol demands from neoplastic cells. The literature review analyzes the results of recent investigations of lipid metabolism in patients with hematologic cancers and their other types. All given investigations show a significant reduction in the serum levels of total cholesterol and high-density lipoproteins in patients with hematological disease at its onset. The data for other indicators of the lipid transport system are ambiguous. Such changes have been elucidated to be associated with the accumulation of cholesterol in the leukemia cells due to enhanced synthesis de novo, a more active absorption from circulation and blocked release of its surplus. If the disease runs a favorable course, lipid metabolic parameters become normalized and, in case of remission, correspond to those seen in healthy individuals. They continue to decline in patients with disease progression. This allows the consideration of cholesterol, its fractions, and apolipoproteins as biochemical prognostic markers in hematological cancer patients and as indicators for assessment of treatment results. In addition, there is evidence for the effect of chemotherapeutic agents on lipid metabolism. Recent attempts to elaborate new treatment strategies, by using the current knowledge on the role of lipid metabolism in cancers, are considered.
Collapse
Affiliation(s)
- S G Vladimirova
- Kirov Research Institute of Hematology and Blood Transfusion, Federal Biomedical Agency of Russia, Kirov, Russia
| | - L N Tarasova
- Kirov Research Institute of Hematology and Blood Transfusion, Federal Biomedical Agency of Russia, Kirov, Russia
| |
Collapse
|
82
|
Huang HL, Chen WC, Hsu HP, Cho CY, Hung YH, Wang CY, Lai MD. Argininosuccinate lyase is a potential therapeutic target in breast cancer. Oncol Rep 2015; 34:3131-9. [PMID: 26397737 DOI: 10.3892/or.2015.4280] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 07/23/2015] [Indexed: 11/06/2022] Open
Abstract
Arginine is a non-essential amino acid that modulates nitric oxide production and cancer homeostasis. In our previous study, we observed that blocking argininosuccinate lyase (ASL) attenuates tumor progression in liver cancer. However, the role of ASL in human breast cancer has been studied to a lesser degree. In the present study, we investigated the effect of targeting ASL in breast cancer. We found that ASL was induced by ER stress and was significantly upregulated in breast cancer tissues compared to that in the corresponding normal tissues. Downregulation of ASL inhibited the growth of breast cancer in vitro and in vivo. The level of cell cycle-related gene, cyclin A2, was reduced and was accompanied by a delay in G2/M transition. ASL shRNA-induced cell inhibition was rescued by exogenous cyclin A2. Furthermore, autophagy was observed in the cells expressing ASL shRNA, and inhibition of autophagy reduced cell growth, indicating that autophagy played a cell survival role in the ASL knockdown cells. Moreover, inhibition of ASL reduced NO content. Introduction of the NO donor partially restored the growth inhibition by ASL shRNA. Thus, the mechanism induced by ASL shRNA which occurred in human breast cancer may be attributed to a decrease in cyclin A2 and NO.
Collapse
Affiliation(s)
- Hau-Lun Huang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City 701, Taiwan, R.O.C
| | - Wei-Ching Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City 701, Taiwan, R.O.C
| | - Hui-Ping Hsu
- Department of Surgery, College of Medicine, National Cheng Kung University, Tainan City 701, Taiwan, R.O.C
| | - Chien-Yu Cho
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City 701, Taiwan, R.O.C
| | - Yu-Hsuan Hung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City 701, Taiwan, R.O.C
| | - Chih-Yang Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City 701, Taiwan, R.O.C
| | - Ming-Derg Lai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City 701, Taiwan, R.O.C
| |
Collapse
|
83
|
Xia X, Yin W, Zhang X, Yu X, Wang C, Xu S, Feng W, Yang H. PAX6 overexpression is associated with the poor prognosis of invasive ductal breast cancer. Oncol Lett 2015; 10:1501-1506. [PMID: 26622698 DOI: 10.3892/ol.2015.3434] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 04/28/2015] [Indexed: 12/21/2022] Open
Abstract
Paired box 6 (PAX6) plays a significant role in the development of human neuroectodermal epithelial tissues. Previous studies have suggested that the PAX6 promoter is hypermethylated in breast cancer and that it is involved in breast cancer cell proliferation. The present study aimed to investigate the expression of PAX6 in invasive breast cancer tissues, and to evaluate its prognostic significance. Immunohistochemistry (IHC) was used to detect PAX6 expression on a breast cancer tissue microarray containing tissues from 111 patients. Associations of PAX6 expression with staging and prognosis were analyzed. PAX6 was mainly expressed in the nucleus. The PAX6 staining intensity was not associated with age, histological grade, lymph node status, tumor size, or progesterone receptor and human epidermal growth factor receptor 2 expression (all P>0.05). A high level of PAX6 staining was more frequent in estrogen receptor (ER)-negative cases compared with ER-positive cases (43.9 vs. 25.7%; P=0.049). After a median follow-up time of 110 months, the patients with low PAX6 expression exhibited an improved survival rate compared with the patients with high PAX6 expression (P<0.001). Cox analysis showed a worse survival rate in the patients with high PAX6 staining (hazard ratio, 3.458; 95% confidence interval, 1.575-7.593; P=0.002). In conclusion, high tumor PAX6 staining intensity by IHC was associated with a poor prognosis in breast cancer patients.
Collapse
Affiliation(s)
- Xianghou Xia
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, P.R. China
| | - Wenjuan Yin
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, P.R. China
| | - Xiping Zhang
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, P.R. China
| | - Xingfei Yu
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, P.R. China
| | - Chen Wang
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, P.R. China
| | - Shenhua Xu
- Zhejiang Cancer Research Institute, Hangzhou, Zhejiang, P.R. China
| | - Weiliang Feng
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, P.R. China
| | - Hongjian Yang
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, P.R. China ; Department of Surgical Oncology, School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| |
Collapse
|
84
|
Bjarnadottir O, Kimbung S, Johansson I, Veerla S, Jönsson M, Bendahl PO, Grabau D, Hedenfalk I, Borgquist S. Global Transcriptional Changes Following Statin Treatment in Breast Cancer. Clin Cancer Res 2015; 21:3402-11. [PMID: 25840970 DOI: 10.1158/1078-0432.ccr-14-1403] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 03/29/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND Statins purportedly exert antitumoral effects, but the underlying mechanisms are currently not fully elucidated. The aim of this study was to explore potential statin-induced effects on global gene expression profiles in primary breast cancer. EXPERIMENTAL DESIGN This window-of-opportunity phase II trial enrolled 50 newly diagnosed breast cancer patients prescribed atorvastatin (80 mg/day) for 2 weeks presurgically. Pre- and posttreatment tumor samples were analyzed using Significance Analysis of Microarrays (SAM) to identify differentially expressed genes. Similarly, SAM and gene ontology analyses were applied to gene expression data derived from atorvastatin-treated breast cancer cell lines (MCF7, BT474, SKBR3, and MDAMB231) comparing treated and untreated cells. The Systematic Motif Analysis Retrieval Tool (SMART) was used to identify enriched transcription factor-binding sites. Literature Vector Analysis (LitVAn) identified gene module functionality, and pathway analysis was performed using GeneGo Pathways Software (MetaCore; https://portal.genego.com/). RESULTS Comparative analysis of gene expression profiles in paired clinical samples revealed 407 significantly differentially expressed genes (FDR = 0); 32 upregulated and 375 downregulated genes. Restricted filtration (fold change ≥1.49) resulted in 21 upregulated and 46 downregulated genes. Significantly upregulated genes included DUSP1, RHOB1, GADD45B, and RGS1. Pooled results from gene ontology, LitVAn and SMART analyses identified statin-induced effects on the apoptotic and MAPK pathways among others. Comparative analyses of gene expression profiles in breast cancer cell lines showed significant upregulation of the mevalonate and proapoptotic pathways following atorvastatin treatment. CONCLUSIONS We report potential statin-induced changes in global tumor gene expression profiles, indicating MAPK pathway inhibition and proapoptotic events.
Collapse
Affiliation(s)
- Olöf Bjarnadottir
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden. Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Siker Kimbung
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Ida Johansson
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Srinivas Veerla
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden. SciBlu genomics, Lund University, Sweden
| | - Mats Jönsson
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Pär-Ola Bendahl
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Dorthe Grabau
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Ingrid Hedenfalk
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Signe Borgquist
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden. Department of Oncology, Skåne University Hospital, Lund, Sweden.
| |
Collapse
|
85
|
|