1
|
Wang H, Liu B, Chen H, Xu P, Xue H, Yuan J. Dynamic changes of DNA methylation induced by benzo(a)pyrene in cancer. Genes Environ 2023; 45:21. [PMID: 37391844 DOI: 10.1186/s41021-023-00278-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/07/2023] [Indexed: 07/02/2023] Open
Abstract
Benzo(a)pyrene (BaP), the earliest and most significant carcinogen among polycyclic aromatic hydrocarbons (PAHs), has been found in foods, tobacco smoke, and automobiles exhaust, etc. Exposure to BaP induced DNA damage directly, or oxidative stress-related damage, resulting in cell apoptosis and carcinogenesis in human respiratory system, digestive system, reproductive system, etc. Moreover, BaP triggered genome-wide epigenetic alterations by methylation, which might cause disturbances in regulation of gene expression, and thereby induced cancer. It has been proved that BaP reduced genome-wide DNA methylation, and activated proto-oncogene by hypomethylation in the promoter region, but silenced tumor suppressor genes by promoter hypermethylation, resulting in cancer initiation and progression. Here we summarized the changes in DNA methylation in BaP exposure, and revealed the methylation of DNA plays a role in cancer development.
Collapse
Affiliation(s)
- Huizeng Wang
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Bingchun Liu
- Stem Cell Research Center, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Hong Chen
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Peixin Xu
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Huiting Xue
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010010, China.
| | - Jianlong Yuan
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China.
| |
Collapse
|
2
|
Lan A, Jin Y, Wang Y, Ding N, Wang Y, Dai Y, Jiang L, Tang Z, Peng Y, Liu S. Association of serum reproductive hormones changes after neoadjuvant chemotherapy with hormone receptors expression alterations and survival outcomes in breast cancer. Front Surg 2022; 9:947218. [PMID: 36117838 PMCID: PMC9470751 DOI: 10.3389/fsurg.2022.947218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
Purpose This study aimed to determine the effect of neoadjuvant chemotherapy (NAC) on circulating levels of reproductive hormones and evaluate the correlation of hormone changes after NAC with hormone receptors expression alterations and relapse-free survival (RFS) outcomes in breast cancer. Methods Information from 181 breast cancer patients who received NAC was retrospectively analyzed. For hormones parameters, the median and interquartile range (IQR) were provided at baseline and the end of NAC then was compared by Wilcoxon signed-rank test. Categorical variables were represented as numbers and percentages and were compared via two-sided chi-square and Fisher's tests. The RFS outcomes were compared between patients according to hormone changes using the log-rank test. Univariate and multivariate survival analyses with hazard ratios (HR) and 95% confidence intervals (95% CI) were carried out using Cox regression. Results Sex steroids including estradiol, progesterone, testosterone, and dehydroepiandrosterone sulfate (DHEAS) levels decreased significantly after NAC among both premenopausal and postmenopausal patients (all P < 0.05). Decreased estradiol levels were associated with reduced progesterone receptor (PR) expression (P = 0.030). In multivariate survival analysis, the non-decreased progesterone level was strongly associated with worse RFS (non-decreased vs. decreased, HR = 7.178, 95% CI 2.340–22.019, P = 0.001). Patients with decreased progesterone levels exhibited better 3-year RFS compared with those with non-decreased (87.6% vs. 58.3%, log-rank, P = 0.001). Conclusion Multiple reproductive hormone levels were influenced by NAC. The change in estradiol level had a positive connection with PR expression alteration. Furthermore, an inverse association between the change in progesterone level and RFS outcomes was found. These findings may provide a theoretical basis for pre-operative endocrine therapy combined with NAC in breast cancer patients.
Collapse
|
3
|
Kristanti AN, Aminah NS, Siswanto I, Manuhara YSW, Abdjan MI, Wardana AP, Aung EE, Takaya Y. Anticancer potential of β-sitosterol and oleanolic acid as through inhibition of human estrogenic 17beta-hydroxysteroid dehydrogenase type-1 based on an in silico approach. RSC Adv 2022; 12:20319-20329. [PMID: 35919602 PMCID: PMC9278416 DOI: 10.1039/d2ra03092f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/27/2022] [Indexed: 11/21/2022] Open
Abstract
The human estrogenic enzyme 17beta-hydroxysteroid dehydrogenase type-1 (HSD17B1) provides biosynthesis regulation of active estrogen in stimulating the development of breast cancer through cell proliferation. The β-sitosterol is classified as a steroid compound and is actually a type of triterpenoid compound that has a similar structure to a steroid. This similarity provides a great opportunity for the inhibitor candidate to bind to the HDS17B1 enzyme because of the template similarity on the active site. Several in silico approaches have been applied in this study to examine the potential of these two inhibitor candidates. Pharmacokinetic studies showed positive results by meeting several drug candidate criteria, such as drug-likeness, bioavailability, and ADMET properties. A combination of molecular docking and MD simulation showed good conformational interaction of the inhibitors and HSD17B1. Prediction of binding free energy (ΔG bind) using the Molecular Mechanics-Generalized Born Surface Area (MM-GBSA) approach shows ΔG bind (kcal mol-1) of C1-HSD17B1: -49.31 ± 0.23 and C2-HSD17B1: -33.54 ± 0.34. Meanwhile, decomposition energy analysis (ΔG residue bind) suggested several key residues that were also responsible for the interaction with inhibitors, such as C1-HSD17B1 (six residues: Leu96, Leu149, Pro187, Met193, Val225, and Phe226) and C2-HSD17B1 (four residues: Ile14, Gly94, Pro187, and Val188). Hopefully, the obtained results from this research could be considered for the mechanistic inhibition of the HSDS17B1 enzyme at molecular and atomistic levels.
Collapse
Affiliation(s)
- Alfinda Novi Kristanti
- Departement of Chemistry, Faculty of Science and Technology, Universitas Airlangga Surabaya 60115 Indonesia
- Biotechnology of Tropical Medicinal Plants Research Group, Universitas Airlangga Indonesia
| | - Nanik Siti Aminah
- Departement of Chemistry, Faculty of Science and Technology, Universitas Airlangga Surabaya 60115 Indonesia
- Biotechnology of Tropical Medicinal Plants Research Group, Universitas Airlangga Indonesia
| | - Imam Siswanto
- Departement of Chemistry, Faculty of Science and Technology, Universitas Airlangga Surabaya 60115 Indonesia
- Bioinformatic Laboratory, UCoE Research Center for Bio-Molecule Engineering, Universitas Airlangga Surabaya Indonesia
| | - Yosephine Sri Wulan Manuhara
- Biotechnology of Tropical Medicinal Plants Research Group, Universitas Airlangga Indonesia
- Department of Biology, Faculty of Science and Technology, Universitas Airlangga Surabaya 60115 Indonesia
| | - Muhammad Ikhlas Abdjan
- Departement of Chemistry, Faculty of Science and Technology, Universitas Airlangga Surabaya 60115 Indonesia
- PhD Student of Mathematics and Natural Sciences, Faculty of Science and Technology, Universitas Airlangga, Komplek Kampus C UNAIR Jl. Mulyorejo Surabaya 60115 Indonesia
| | - Andika Pramudya Wardana
- Departement of Chemistry, Faculty of Science and Technology, Universitas Airlangga Surabaya 60115 Indonesia
- PhD Student of Mathematics and Natural Sciences, Faculty of Science and Technology, Universitas Airlangga, Komplek Kampus C UNAIR Jl. Mulyorejo Surabaya 60115 Indonesia
| | - Ei Ei Aung
- Departement of Chemistry, Faculty of Science and Technology, Universitas Airlangga Surabaya 60115 Indonesia
- Departement of Chemistry, Yadanarbon University Amarapura Township Mandalay Myanmar
| | - Yoshiaki Takaya
- Faculty of Pharmacy, Meijo University 150 Yagotoyama, Tempaku Nagoya 468-8503 Japan
| |
Collapse
|
4
|
Luteolin Induced Hippocampal Neuronal Pyroptosis Inhibition by Regulation of miR-124-3p/TNF- α/TRAF6 Axis in Mice Affected by Breast-Cancer-Related Depression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2715325. [PMID: 35571739 PMCID: PMC9106465 DOI: 10.1155/2022/2715325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/23/2022] [Indexed: 11/18/2022]
Abstract
Background Breast-cancer-related depression (BCRD) is associated with an increased mortality rate among breast cancer (BC) survivors. Luteolin has many pharmacological effects, particularly in the treatment of BC. In this study, we aimed to explore the anti-BCRD activity of luteolin and its underlying functional mechanism. Methods A BCRD mouse model was induced by injecting 4T1 cells and corticosterone (COR). Behavioral test, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, Nissl staining, immunofluorescence, reverse-transcription quantitative PCR (RT-qPCR), and western blotting were used to study the effect of luteolin in mice with BCRD in vivo. A COR-induced neuron injury model was established in HT-22 cells in vitro. The role of miR-124-3p in the anti-BCRD effects of luteolin was studied using a miR-124-3p inhibitor. Results Luteolin significantly reduced the size and weight of the tumor, increased the mice entry frequency in the symmetrical sector, and reduced the duration of immobility in the tail suspension and forced swimming tests of mice affected by BCRD. Simultaneously, apoptosis of hippocampal neurons was inhibited, and the number of Nissl bodies increased with luteolin treatment. In addition, luteolin resulted in the upregulation of miR-124-3p expression in the hippocampus and downregulated the expression of tumor necrosis factor-α (TNF-α) and TNF receptor-associated factor 6 (TRAF6), as well as lowered the phosphorylation levels of nuclear factor-kappa B (NF-κB) and IkappaB (IκB). Luteolin also inhibited pyroptosis of hippocampal neurons in mice affected by BCRD, as revealed by the low protein levels of NOD-like receptor protein 3 (NLRP3), caspase-1, gasdermin D-N (GSDMD-N), interleukin (IL)-1β, and IL-18. However, the miR-124-3p inhibitor significantly reversed the therapeutic effect of luteolin on COR-induced HT-22 cells. Conclusion Our study demonstrated that the anti-BCRD function of luteolin was mediated by regulating the miR-124-3p/TNF-α/TRAF6-related pathway and inhibiting neuronal cell pyroptosis and subsequent inflammation. Therefore, luteolin may be a potential drug candidate in the treatments of BCRD.
Collapse
|
5
|
Gómez-Romero L, Alvarez-Suarez DE, Hernández-Lemus E, Ponce-Castañeda MV, Tovar H. The regulatory landscape of retinoblastoma: a pathway analysis perspective. ROYAL SOCIETY OPEN SCIENCE 2022; 9:220031. [PMID: 35620002 PMCID: PMC9114937 DOI: 10.1098/rsos.220031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 04/13/2022] [Indexed: 05/03/2023]
Abstract
Retinoblastoma (Rb) is a rare intraocular tumour in early childhood, with an approximate incidence of 1 in 18 000 live births. Experimental studies for Rb are complex due to the challenges associated with obtaining a normal retina to contrast with diseased tissue. In this work, we reanalyse a dataset that contains normal retina samples. We identified the individual genes whose expression is different in Rb in contrast with normal tissue, determined the pathways whose global expression pattern is more distant from the global expression observed in normal tissue, and finally, we identified which transcription factors regulate the highest number of differentially expressed genes (DEGs) and proposed as transcriptional master regulators (TMRs). The enrichment of DEGs in the phototransduction and retrograde endocannabinoid signalling pathways could be associated with abnormal behaviour of the processes leading to cellular differentiation and cellular proliferation. On the other hand, the TMRs nuclear receptor subfamily 5 group A member 2 and hepatocyte nuclear factor 4 gamma are involved in hepatocyte differentiation. Therefore, the enrichment of aberrant expression in these transcription factors could suggest an abnormal retina development that could be involved in Rb origin and progression.
Collapse
Affiliation(s)
- Laura Gómez-Romero
- Computational Genomics Division, National Institute of Genomic Medicine (INMEGEN), Mexico City, Mexico
| | - Diana E. Alvarez-Suarez
- Medical Research Unit in Infectious Diseases, Hospital de Pediatría, CMN SXXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
- Pharmacology Department, CINVESTAV, Mexico City, Mexico
| | - Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine (INMEGEN), Mexico City, Mexico
- Center for Complexity Sciences, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - M. Verónica Ponce-Castañeda
- Medical Research Unit in Infectious Diseases, Hospital de Pediatría, CMN SXXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Hugo Tovar
- Computational Genomics Division, National Institute of Genomic Medicine (INMEGEN), Mexico City, Mexico
| |
Collapse
|
6
|
Gao S, Dai Z, Xu H, Lai L. Pinpointing Cancer Sub-Type Specific Metabolic Tasks Facilitates Identification of Anti-cancer Targets. Front Med (Lausanne) 2022; 9:872024. [PMID: 35402442 PMCID: PMC8984102 DOI: 10.3389/fmed.2022.872024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/01/2022] [Indexed: 12/03/2022] Open
Abstract
Metabolic reprogramming is one of the hallmarks of tumorigenesis. Understanding the metabolic changes in cancer cells may provide attractive therapeutic targets and new strategies for cancer therapy. The metabolic states are not the same in different cancer types or subtypes, even within the same sample of solid tumors. In order to understand the heterogeneity of cancer cells, we used the Pareto tasks inference method to analyze the metabolic tasks of different cancers, including breast cancer, lung cancer, digestive organ cancer, digestive tract cancer, and reproductive cancer. We found that cancer subtypes haves different propensities toward metabolic tasks, and the biological significance of these metabolic tasks also varies greatly. Normal cells treat metabolic tasks uniformly, while different cancer cells focus on different pathways. We then integrated the metabolic tasks into the multi-objective genome-scale metabolic network model, which shows higher accuracy in the in silico prediction of cell states after gene knockout than the conventional biomass maximization model. The predicted potential single drug targets could potentially turn into biomarkers or drug design targets. We further implemented the multi-objective genome-scale metabolic network model to predict synthetic lethal target pairs of the Basal and Luminal B subtypes of breast cancer. By analyzing the predicted synthetic lethal targets, we found that mitochondrial enzymes are potential targets for drug combinations. Our study quantitatively analyzes the metabolic tasks of cancer and establishes cancer type-specific metabolic models, which opens a new window for the development of specific anti-cancer drugs and provides promising treatment plans for specific cancer subtypes.
Collapse
Affiliation(s)
- Shuaishi Gao
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Ziwei Dai
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Hanyu Xu
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Luhua Lai
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- Research Unit of Drug Design Method, Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Luhua Lai,
| |
Collapse
|
7
|
Pre-treatment circulating reproductive hormones levels predict pathological and survival outcomes in breast cancer submitted to neoadjuvant chemotherapy. Int J Clin Oncol 2022; 27:899-910. [PMID: 35239089 DOI: 10.1007/s10147-022-02141-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/03/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE This study aimed to evaluate the correlation of pre-treatment circulating reproductive hormones levels with pathological and survival outcomes in breast cancer patients received neoadjuvant chemotherapy (NAC). METHODS Information from 196 premenopausal and 137 postmenopausal breast cancer patients who received NAC were retrospectively analyzed. Treatment response to NAC, with odds ratios (OR) and 95% confidence intervals (95% CI) was estimated using logistic regression adjusted for key confounders. Survival outcomes with hazard ratios (HR) and 95% CI were estimated using Cox regression adjusted for key confounders. The Kaplan-Meier method was applied in the survival analysis. RESULTS Premenopausal patients with lower testosterone levels (OR = 0.996, 95% CI 0.992-0.999, P = 0.026), and postmenopausal patients with higher follicle-stimulating hormone (FSH) levels (OR = 1.045, 95% CI 1.014-1.077, P = 0.005) were likely to achieve pathological complete response (pCR). In multivariate survival analysis, the lowest tertile (T) progesterone was associated with worse overall survival (OS) in premenopausal patients (T2 vs T1, HR = 0.113, 95% CI 0.013-0.953, P = 0.045; T3 vs T1, HR = 0.109, 95% CI 0.013-0.916, P = 0.041). Premenopausal patients with the lowest tertile progesterone exhibited worse 3-year OS compared with those with higher tertiles (72.9% vs 97.4%, log-rank, P = 0.007). CONCLUSION Pre-treatment testosterone and FSH are significant independent predictors for pCR to NAC in premenopausal and postmenopausal patients, respectively. Low progesterone levels are correlated with worse OS in premenopausal patients. These findings may provide a theoretical basis for pre-operative endocrine therapy combined with NAC in breast cancer.
Collapse
|
8
|
Herman BE, Gardi J, Julesz J, Tömböly C, Szánti-Pintér E, Fehér K, Skoda-Földes R, Szécsi M. Steroidal ferrocenes as potential enzyme inhibitors of the estrogen biosynthesis. Biol Futur 2021; 71:249-264. [PMID: 34554507 DOI: 10.1007/s42977-020-00023-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 06/04/2020] [Indexed: 01/13/2023]
Abstract
The potential inhibitory effect of diverse triazolyl-ferrocene steroids on key enzymes of the estrogen biosynthesis was investigated. Test compounds were synthesized via copper-catalyzed cycloaddition of steroidal azides and ferrocenyl-alkynes using our efficient methodology published previously. Inhibition of human aromatase, steroid sulfatase (STS) and 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD1) activities was investigated with in vitro radiosubstrate incubations. Some of the test compounds were found to be potent inhibitors of the STS. A compound bearing ferrocenyl side chain on the C-2 displayed a reversible inhibition, whereas C-16 and C-17 derivatives displayed competitive irreversible binding mechanism toward the enzyme. 17α-Triazolyl-ferrocene derivatives of 17β-estradiol exerted outstanding inhibitory effect and experiments demonstrated a key role of the ferrocenyl moiety in the enhanced binding affinity. Submicromolar IC50 and Ki parameters enroll these compounds to the group of the most effective STS inhibitors published so far. STS inhibitory potential of the steroidal ferrocenes may lead to the development of novel compounds able to suppress in situ biosynthesis of 17β-estradiol in target tissues.
Collapse
Affiliation(s)
- Bianka Edina Herman
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, P. O. Box 427, Szeged, 6720, Hungary
| | - János Gardi
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, P. O. Box 427, Szeged, 6720, Hungary
| | - János Julesz
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, P. O. Box 427, Szeged, 6720, Hungary
| | - Csaba Tömböly
- Laboratory of Chemical Biology, Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Temesvári körút 62, P. O. Box 521, Szeged, 6726, Hungary
| | - Eszter Szánti-Pintér
- Department of Organic Chemistry, Institute of Chemistry, University of Pannonia, Egyetem utca 10, P. O. Box 158, Veszprém, 8200, Hungary
| | - Klaudia Fehér
- Department of Organic Chemistry, Institute of Chemistry, University of Pannonia, Egyetem utca 10, P. O. Box 158, Veszprém, 8200, Hungary
| | - Rita Skoda-Földes
- Department of Organic Chemistry, Institute of Chemistry, University of Pannonia, Egyetem utca 10, P. O. Box 158, Veszprém, 8200, Hungary.
| | - Mihály Szécsi
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, P. O. Box 427, Szeged, 6720, Hungary.
| |
Collapse
|
9
|
Paakinaho V, Palvimo JJ. Genome-wide crosstalk between steroid receptors in breast and prostate cancers. Endocr Relat Cancer 2021; 28:R231-R250. [PMID: 34137734 PMCID: PMC8345902 DOI: 10.1530/erc-21-0038] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/16/2021] [Indexed: 12/18/2022]
Abstract
Steroid receptors (SRs) constitute an important class of signal-dependent transcription factors (TFs). They regulate a variety of key biological processes and are crucial drug targets in many disease states. In particular, estrogen (ER) and androgen receptors (AR) drive the development and progression of breast and prostate cancer, respectively. Thus, they represent the main specific drug targets in these diseases. Recent evidence has suggested that the crosstalk between signal-dependent TFs is an important step in the reprogramming of chromatin sites; a signal-activated TF can expand or restrict the chromatin binding of another TF. This crosstalk can rewire gene programs and thus alter biological processes and influence the progression of disease. Lately, it has been postulated that there may be an important crosstalk between the AR and the ER with other SRs. Especially, progesterone (PR) and glucocorticoid receptor (GR) can reprogram chromatin binding of ER and gene programs in breast cancer cells. Furthermore, GR can take the place of AR in antiandrogen-resistant prostate cancer cells. Here, we review the current knowledge of the crosstalk between SRs in breast and prostate cancers. We emphasize how the activity of ER and AR on chromatin can be modulated by other SRs on a genome-wide scale. We also highlight the knowledge gaps in the interplay of SRs and their complex interactions with other signaling pathways and suggest how to experimentally fill in these gaps.
Collapse
Affiliation(s)
- Ville Paakinaho
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jorma J Palvimo
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Correspondence should be addressed to J J Palvimo:
| |
Collapse
|
10
|
Bleach R, Madden SF, Hawley J, Charmsaz S, Selli C, Sheehan KM, Young LS, Sims AH, Souček P, Hill AD, McIlroy M. Steroid Ligands, the Forgotten Triggers of Nuclear Receptor Action; Implications for Acquired Resistance to Endocrine Therapy. Clin Cancer Res 2021; 27:3980-3989. [PMID: 34016642 PMCID: PMC9401529 DOI: 10.1158/1078-0432.ccr-20-4135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/22/2021] [Accepted: 05/18/2021] [Indexed: 01/07/2023]
Abstract
PURPOSE There is strong epidemiologic evidence indicating that estrogens may not be the sole steroid drivers of breast cancer. We hypothesize that abundant adrenal androgenic steroid precursors, acting via the androgen receptor (AR), promote an endocrine-resistant breast cancer phenotype. EXPERIMENTAL DESIGN AR was evaluated in a primary breast cancer tissue microarray (n = 844). Androstenedione (4AD) levels were evaluated in serum samples (n = 42) from hormone receptor-positive, postmenopausal breast cancer. Levels of androgens, progesterone, and estradiol were quantified using LC/MS-MS in serum from age- and grade-matched recurrent and nonrecurrent patients (n = 6) before and after aromatase inhibitor (AI) therapy (>12 months). AR and estrogen receptor (ER) signaling pathway activities were analyzed in two independent AI-treated cohorts. RESULTS AR protein expression was associated with favorable progression-free survival in the total population (Wilcoxon, P < 0.001). Pretherapy serum samples from breast cancer patients showed decreasing levels of 4AD with age only in the nonrecurrent group (P < 0.05). LC/MS-MS analysis of an AI-sensitive and AI-resistant cohort demonstrated the ability to detect altered levels of steroids in serum of patients before and after AI therapy. Transcriptional analysis showed an increased ratio of AR:ER signaling pathway activities in patients failing AI therapy (t test P < 0.05); furthermore, 4AD mediated gene changes associated with acquired AI resistance. CONCLUSIONS This study highlights the importance of examining the therapeutic consequences of the steroid microenvironment and demonstrable receptor activation using indicative gene expression signatures.
Collapse
Affiliation(s)
- Rachel Bleach
- Endocrine Oncology Research, Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Stephen F Madden
- Data Science Centre, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - James Hawley
- Department of Biochemistry, Manchester University, NHS Foundation Trust, London, United Kingdom
| | - Sara Charmsaz
- Endocrine Oncology Research, Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Cigdem Selli
- Applied Bioinformatics of Cancer, Institute of Genetics and Cancer, University of Edinburgh Cancer Research Centre, Edinburgh, United Kingdom
| | | | - Leonie S Young
- Endocrine Oncology Research, Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Andrew H Sims
- Applied Bioinformatics of Cancer, Institute of Genetics and Cancer, University of Edinburgh Cancer Research Centre, Edinburgh, United Kingdom
| | - Pavel Souček
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
- Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
| | - Arnold D Hill
- Endocrine Oncology Research, Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Surgery, Beaumont Hospital, Dublin, Ireland
| | - Marie McIlroy
- Endocrine Oncology Research, Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland.
| |
Collapse
|
11
|
Bao G, Xu R, Wang X, Ji J, Wang L, Li W, Zhang Q, Huang B, Chen A, Zhang D, Kong B, Yang Q, Yuan C, Wang X, Wang J, Li X. Identification of lncRNA Signature Associated With Pan-Cancer Prognosis. IEEE J Biomed Health Inform 2021; 25:2317-2328. [PMID: 32991297 DOI: 10.1109/jbhi.2020.3027680] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Long noncoding RNAs (lncRNAs) have emerged as potential prognostic markers in various human cancers as they participate in many malignant behaviors. However, the value of lncRNAs as prognostic markers among diverse human cancers is still under investigation, and a systematic signature based on these transcripts that related to pan-cancer prognosis has yet to be reported. In this study, we proposed a framework to incorporate statistical power, biological rationale, and machine learning models for pan-cancer prognosis analysis. The framework identified a 5-lncRNA signature (ENSG00000206567, PCAT29, ENSG00000257989, LOC388282, and LINC00339) from TCGA training studies (n = 1,878). The identified lncRNAs are significantly associated (all P ≤ 1.48E-11) with overall survival (OS) of the TCGA cohort (n = 4,231). The signature stratified the cohort into low- and high-risk groups with significantly distinct survival outcomes (median OS of 9.84 years versus 4.37 years, log-rank P = 1.48E-38) and achieved a time-dependent ROC/AUC of 0.66 at 5 years. After routine clinical factors involved, the signature demonstrated better performance for long-term prognostic estimation (AUC of 0.72). Moreover, the signature was further evaluated on two independent external cohorts (TARGET, n = 1,122; CPTAC, n = 391; National Cancer Institute) which yielded similar prognostic values (AUC of 0.60 and 0.75; log-rank P = 8.6E-09 and P = 2.7E-06). An indexing system was developed to map the 5-lncRNA signature to prognoses of pan-cancer patients. In silico functional analysis indicated that the lncRNAs are associated with common biological processes driving human cancers. The five lncRNAs, especially ENSG00000206567, ENSG00000257989 and LOC388282 that never reported before, may serve as viable molecular targets common among diverse cancers.
Collapse
|
12
|
Valko-Rokytovská M, Očenáš P, Salayová A, Kostecká Z. Breast Cancer: Targeting of Steroid Hormones in Cancerogenesis and Diagnostics. Int J Mol Sci 2021; 22:ijms22115878. [PMID: 34070921 PMCID: PMC8199112 DOI: 10.3390/ijms22115878] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is the most common malignancy in women with high mortality. Sensitive and specific methods for the detection, characterization and quantification of endogenous steroids in body fluids or tissues are needed for the diagnosis, treatment and prognosis of breast cancer and many other diseases. At present, non-invasive diagnostic methods are gaining more and more prominence, which enable a relatively fast and painless way of detecting many diseases. Metabolomics is a promising analytical method, the principle of which is the study and analysis of metabolites in biological material. It represents a comprehensive non-invasive diagnosis, which has a high potential for use in the diagnosis and prognosis of cancers, including breast cancer. This short review focuses on the targeted metabolomics of steroid hormones, which play an important role in the development and classification of breast cancer. The most commonly used diagnostic tool is the chromatographic method with mass spectrometry detection, which can simultaneously determine several steroid hormones and metabolites in one sample. This analytical procedure has a high potential in effective diagnosis of steroidogenesis disorders. Due to the association between steroidogenesis and breast cancer progression, steroid profiling is an important tool, as well as in monitoring disease progression, improving prognosis, and minimizing recurrence.
Collapse
|
13
|
Aguilera-Buenosvinos I, Fernandez-Lazaro CI, Romanos-Nanclares A, Gea A, Sánchez-Bayona R, Martín-Moreno JM, Martínez-González MÁ, Toledo E. Dairy Consumption and Incidence of Breast Cancer in the 'Seguimiento Universidad de Navarra' (SUN) Project. Nutrients 2021; 13:nu13020687. [PMID: 33669972 PMCID: PMC7924827 DOI: 10.3390/nu13020687] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 01/21/2023] Open
Abstract
Dairy products might influence breast cancer (BC) risk. However, evidence is inconsistent. We sought to examine the association between dairy product consumption-and their subtypes-and incident BC in a Mediterranean cohort. The SUN ("Seguimiento Universidad de Navarra") Project is a Spanish dynamic ongoing cohort of university graduates. Dairy product consumption was estimated through a previously validated 136-item food frequency questionnaire (FFQ). Incident BC was reported in biennial follow-up questionnaires and confirmed with revision of medical records and consultation of the National Death Index. Hazard ratios (HR) and 95% confidence intervals (CI) were estimated with Cox regression models. Among 123,297 women-years of follow-up (10,930 women, median follow-up 12.1 years), we confirmed 119 incident BC cases. We found a nonlinear association between total dairy product consumption and BC incidence (pnonlinear = 0.048) and a significant inverse association for women with moderate total dairy product consumption (HRQ2vs.Q1 = 0.49 (95% CI 0.28-0.84); HRQ3vs.Q1 = 0.49 (95% CI 0.29-0.84) ptrend = 0.623) and with moderate low-fat dairy product consumption (HRQ2vs.Q1 = 0.58 (95% CI 0.35-0.97); HRQ3vs.Q1 = 0.55 (95% CI 0.32-0.92), ptrend = 0.136). In stratified analyses, we found a significant inverse association between intermediate low-fat dairy product consumption and premenopausal BC and between medium total dairy product consumption and postmenopausal BC. Thus, dairy products, especially low-fat dairy products, may be considered within overall prudent dietary patterns.
Collapse
Affiliation(s)
- Inmaculada Aguilera-Buenosvinos
- Departamento de Medicina Preventiva y Salud Pública, Universidad de Navarra, 31008 Pamplona, Spain; (I.A.-B.); (C.I.F.-L.); (A.R.-N.); (A.G.); (M.Á.M.-G.)
| | - Cesar Ignacio Fernandez-Lazaro
- Departamento de Medicina Preventiva y Salud Pública, Universidad de Navarra, 31008 Pamplona, Spain; (I.A.-B.); (C.I.F.-L.); (A.R.-N.); (A.G.); (M.Á.M.-G.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Andrea Romanos-Nanclares
- Departamento de Medicina Preventiva y Salud Pública, Universidad de Navarra, 31008 Pamplona, Spain; (I.A.-B.); (C.I.F.-L.); (A.R.-N.); (A.G.); (M.Á.M.-G.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Alfredo Gea
- Departamento de Medicina Preventiva y Salud Pública, Universidad de Navarra, 31008 Pamplona, Spain; (I.A.-B.); (C.I.F.-L.); (A.R.-N.); (A.G.); (M.Á.M.-G.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red Área de Fisiología de la Obesidad y la Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Rodrigo Sánchez-Bayona
- Department of Clinical Oncology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain;
| | - Jose M. Martín-Moreno
- Department of Preventive Medicine and Public Health, Medical School & INCLIVA, University of Valencia, 46010 Valencia, Spain;
| | - Miguel Ángel Martínez-González
- Departamento de Medicina Preventiva y Salud Pública, Universidad de Navarra, 31008 Pamplona, Spain; (I.A.-B.); (C.I.F.-L.); (A.R.-N.); (A.G.); (M.Á.M.-G.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red Área de Fisiología de la Obesidad y la Nutrición (CIBEROBN), 28029 Madrid, Spain
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
| | - Estefanía Toledo
- Departamento de Medicina Preventiva y Salud Pública, Universidad de Navarra, 31008 Pamplona, Spain; (I.A.-B.); (C.I.F.-L.); (A.R.-N.); (A.G.); (M.Á.M.-G.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red Área de Fisiología de la Obesidad y la Nutrición (CIBEROBN), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-948425600 (ext. 806224)
| |
Collapse
|
14
|
Grape Seed Procyanidins Inhibit the Growth of Breast Cancer MCF-7 Cells by Down-Regulating the EGFR/VEGF/MMP9 Pathway. Nat Prod Commun 2021. [DOI: 10.1177/1934578x21991691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Breast cancer is the most common invasive cancer in women and the second leading cause of cancer death in women. However, it is not clear about its effective treatments. As a potential anticancer agent, grape seed procyanidins (GSPs) have been shown to inhibit the proliferation of various cancer cells in vitro and in vivo. In this study, it was shown that GSPs significantly inhibit MCF-7 cell proliferation in a concentration/time-dependent manner. The flow cytometric data clearly demonstrated that GSPs cause cell cycle arrest in the G2/M phase, followed by cell apoptosis. Moreover, it also confirmed that growth inhibition mediated by treatment with GSPs is related to the induction of apoptosis due to p53 elevation, purportedly by inhibition of the epidermal growth factor receptor (EGFR)/vascular endothelial growth factor (VEGF)/matrix metalloproteinase 9 (MMP9) pathway. Taken together, these findings suggest that GSPs inhibit MCF-7 cells proliferation and induce cell apoptosis by suppressing EGFR/VEGF/MMP9 pathway.
Collapse
|
15
|
Mottinelli M, Sinreih M, Rižner TL, Leese MP, Potter BVL. N-Phenyl-1,2,3,4-tetrahydroisoquinoline: An Alternative Scaffold for the Design of 17β-Hydroxysteroid Dehydrogenase 1 Inhibitors. ChemMedChem 2020; 16:259-291. [PMID: 33151004 DOI: 10.1002/cmdc.202000762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Indexed: 11/08/2022]
Abstract
17β-Hydroxysteroid dehydrogenases catalyse interconversion at the C17 position between oxidized and reduced forms of steroidal nuclear receptor ligands. The type 1 enzyme, expressed in malignant cells, catalyses reduction of the less-active estrone to estradiol, and inhibitors have therapeutic potential in estrogen-dependent diseases such as breast and ovarian cancers and in endometriosis. Synthetic decoration of the nonsteroidal N-phenyl-1,2,3,4-tetrahydroisoquinoline (THIQ) template was pursued by using Pomeranz-Fritsch-Bobbitt, Pictet-Spengler and Bischler-Napieralski approaches to explore the viability of this scaffold as a steroid mimic. Derivatives were evaluated biologically in vitro as type 1 enzyme inhibitors in a bacterial cell homogenate as source of recombinant protein. Structure-activity relationships are discussed. THIQs possessing a 6-hydroxy group, lipophilic substitutions at the 1- or 4-positions in combination with N-4'-chlorophenyl substitution were most favourable for activity. Of these, one compound had an IC50 of ca. 350 nM as a racemate, testifying to the applicability of this novel approach.
Collapse
Affiliation(s)
- Marco Mottinelli
- Wolfson Laboratory of Medicinal Chemistry Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, BA2 7AY, Bath, UK.,Present address: Department of Medicinal Chemistry School of Pharmacy, University of Florida, 1345 Center Dr., Gainesville, FL 32611, USA
| | - Maša Sinreih
- Institute of Biochemistry Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia
| | - Tea L Rižner
- Institute of Biochemistry Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia
| | - Mathew P Leese
- Wolfson Laboratory of Medicinal Chemistry Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, BA2 7AY, Bath, UK
| | - Barry V L Potter
- Drug Discovery & Medicinal Chemistry Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK.,Wolfson Laboratory of Medicinal Chemistry Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, BA2 7AY, Bath, UK
| |
Collapse
|
16
|
Borghesi J, Caceres S, Mario LC, Alonso-Diez A, Silveira Rabelo AC, Illera MJ, Silvan G, Miglino MA, Favaron PO, Carreira ACO, Illera JC. Effects of doxorubicin associated with amniotic membrane stem cells in the treatment of canine inflammatory breast carcinoma (IPC-366) cells. BMC Vet Res 2020; 16:353. [PMID: 32972410 PMCID: PMC7513323 DOI: 10.1186/s12917-020-02576-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/15/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Tumours in mammary glands represent the most common neoplasia in bitches, as in humans. This high incidence results in part from the stimulation of sex hormones on these glands. Among mammary tumours, inflammatory carcinoma is the most aggressive, presenting a poor prognosis to surgical treatment and chemotherapy. One of the most widely used chemotherapy drugs for breast cancer treatment is doxorubicin (DOXO). Alternative therapies have been introduced in order to assist in these treatments; studies on treatments using stem cells have emerged, since they have anti-inflammatory and immunomodulatory properties. The aim of this study was to evaluate the effects of DOXO and canine amniotic membrane stem cells (AMCs) on the triple-negative canine inflammatory mammary carcinoma cell line IPC-366. METHODS Four experimental groups were analysed: a control group without treatment; Group I with DOXO, Group II with AMC and Group III with an association of DOXO and AMCs. We performed the MTT assay with DOXO in order to select the best concentration for the experiments. The growth curve was performed with all groups (I-III) in order to verify the potential of treatments to reduce the growth of IPC-366. For the cell cycle, all groups (I-III) were tested using propidium iodide. While in the flow cytometry, antibodies to progesterone receptor (PR), estrogen receptor (ER), PCNA, VEGF, IL-10 and TGF-β1 were used. For steroidogenic pathway hormones, an ELISA assay was performed. RESULTS The results showed that cells treated with 10 µg/mL DOXO showed a 71.64% reduction in cellular growth after 72 h of treatment. Reductions in the expression of VEGF and PCNA-3 were observed by flow cytometry in all treatments when compared to the control. The intracellular levels of ERs were also significantly increased in Group III (4.67% vs. 27.1%). Regarding to the levels of steroid hormones, significant increases in the levels of estradiol (E2) and estrone sulphate (S04E1) were observed in Groups I and III. On the other hand, Group II did not show differences in steroid hormone levels in relation to the control. We conclude that the association of DOXO with AMCs (Group III) promoted a reduction in cell growth and in the expression of proteins related to proliferation and angiogenesis in IPC-366 triple-negative cells. CONCLUSIONS This treatment promoted ER positive expression, suggesting that the accumulated oestrogen conducted these cells to a synergistic state, rendering these tumour cells responsive to ERs and susceptible to new hormonal cancer therapies.
Collapse
Affiliation(s)
- Jéssica Borghesi
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil.
| | - Sara Caceres
- Department of Animal Physiology, School of Veterinary Medicine, Complutense University of Madrid (UCM), Madrid, Spain
| | - Lara Carolina Mario
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Angela Alonso-Diez
- Department of Animal Medicine, Surgery and Pathology, School of Veterinary Medicine, Complutense University of Madrid (UCM), Madrid, Spain
| | - Ana Carolina Silveira Rabelo
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Maria J Illera
- Department of Animal Physiology, School of Veterinary Medicine, Complutense University of Madrid (UCM), Madrid, Spain
| | - Gema Silvan
- Department of Animal Physiology, School of Veterinary Medicine, Complutense University of Madrid (UCM), Madrid, Spain
| | - Maria Angélica Miglino
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Phelipe O Favaron
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Ana Claudia O Carreira
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil. .,NUCEL (Cell and Molecular Therapy Center), School of Medicine, University of Sao Paulo, Sao Paulo, Brazil.
| | - Juan Carlos Illera
- Department of Animal Physiology, School of Veterinary Medicine, Complutense University of Madrid (UCM), Madrid, Spain
| |
Collapse
|
17
|
R S, Mk K. Lead optimization of 4-(thio)-chromenone 6- O-sulfamate analogs using QSAR, molecular docking and DFT - a combined approach as steroidal sulfatase inhibitors. J Recept Signal Transduct Res 2020; 41:123-137. [PMID: 32705921 DOI: 10.1080/10799893.2020.1794004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Aromatase and steroidal sulfatase (STS) are steroidogenic enzyme that increases the concentration of estrogens in circulation, a primary factor leading to breast cancer. At molecular level, 87% of STS is expressed and an inhibitor targeting STS could decrease the level of estrogens. In an attempt to identify the chemical structural requirement targeting placental STS inhibition, 26 compounds with pIC50 ranging from 4.61 to 9.46 were subjected to computational studies including Quantitative Structural-Activity Relationship (QSAR), MolecularDocking followed by Density Functional Theory (DFT) studies. A robust and predictable model were developed with good R2 (0.834) and cross-validated correlation coefficient value Q2 LOO (0.786) explaining the relationship quantitatively. The regression graphs suggests that the STS inhibition was greatly dependent on the electro topological state of an atom, sum of the atom type E-state (SdssC), maximum E-states for strong hydrogen bond acceptors (maxHBa) and basic group count descriptor (BCUTp-1h). Furthermore, docking results showed favorable interactions of sulfamate analogs with catalytically important amino acid residues such as LEU74, VAL101, and VAL486. The interactions of the best active compound 3j when compared with standard Irosustat show similar binding energies. DFT studies further confirm the presence of HOMO orbital centered on chromenone ring further highlighting its importance for receptor ligand hydrophobic interaction. The study reveals that substitution of thio in chromenone nucleus and introduction of adamantyl substitution at second position are favorable in inhibiting the enzyme STS.
Collapse
Affiliation(s)
- Srimathi R
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRMIST, Kattankulathur, TamilNadu, India
| | - Kathiravan Mk
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRMIST, Kattankulathur, TamilNadu, India.,Dr. APJ Abdul Kalam Research Lab, SRM College of Pharmacy, SRM IST, Kattankulathur, Tamil Nadu, India
| |
Collapse
|
18
|
Jiang Y, Fan L. Evaluation of anticancer activities of Poria cocos ethanol extract in breast cancer: In vivo and in vitro, identification and mechanism. JOURNAL OF ETHNOPHARMACOLOGY 2020; 257:112851. [PMID: 32283190 DOI: 10.1016/j.jep.2020.112851] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/30/2020] [Accepted: 04/04/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Poria cocos Wolf (P. cocos), a well-known traditional East-Asian medicinal and edible fungus, is one of the most important components in Chinese medicine formulas like "Guizhi fuling wan" to treat hyperplasia of mammary glands and breast cancer. AIMING OF STUDY In this study, we attempted to verify the anticancer efficacy of the ethanol extract of P. cocos (PC) on the breast cancer as well as to investigate its most active compound and its underlying molecular mechanism in vivo and in vitro. MATERIALS AND METHODS The key anti-cancer components were separated and purified through chromatography and identified by spectral analyses. The in vivo anti-breast cancer efficacy and side effects of PC were evaluated in BALB/c nude mice that have been subcutaneously injected with breast cancer cells MDA-MB-231. Cytotoxicity, apoptosis and cell cycle arrest of PC were evaluated in vitro by cell viability assays and flow cytometry. The protein levels were examined via western blotting. RESULTS Pachymic acid (PA), separated and identified as the most active compound, induced the significant cytotoxicity on breast cancer cells MDA-MB-231(IC50 value, 2.13 ± 0.24 μg/mL) and was not active against the normal breast epithelium cells MCF-10A. The in vivo experiment revealed that PC could significantly inhibit the tumor development and the final mean tumor weight of the mice in the PC group (0.51 ± 0.12g) was significantly lower than that in the model group (1.22 ± 0.45g). Notably, compared to the first-line anticancer drug cisplatin, PC showed less side effects on the function of the vital organs and the muscle strength of the mice. Among in vitro study, PC significantly inhibited the cell growth of MDA-MB-231 by inducing cell apoptosis and cell cycle arrested at G0/G1 phase in a dose-dependent manner. The expression of cell cycle-associated cyclin D1, cyclin E, CDK2, and CDK4 were downregulated, while p53 and p21 expression were upregulated following the PA treatment. In addition, PA downregulated the apoptotic regulator Bcl-2, increased the expression of pro-apoptotic protein Bax, and promoted the release of cytochrome c and the activation of cleaved caspase-3, -9 and caspase -8 via mitochondria-mediated and death receptor-mediated signaling pathways. CONCLUSION This study verified the anticancer efficacy of PC on breast cancer in vivo and in vitro through induction of cell apoptosis and G0/G1 cell cycle arrest. The data also suggested that PA could be developed as an efficacious agent for breast cancer treatment with less side effects.
Collapse
Affiliation(s)
- Yu Jiang
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Liuping Fan
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
19
|
Liu Y, Fan D. The Preparation of Ginsenoside Rg5, Its Antitumor Activity against Breast Cancer Cells and Its Targeting of PI3K. Nutrients 2020; 12:nu12010246. [PMID: 31963684 PMCID: PMC7019936 DOI: 10.3390/nu12010246] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 02/06/2023] Open
Abstract
Ginsenosides have been reported to possess various pharmacological effects, including anticancer effects. Nevertheless, there are few reports about the antitumor activity and mechanisms of ginsenoside Rg5 against breast cancer cells. In the present study, the major ginsenoside Rb1 was transformed into the rare ginsenoside Rg5 through enzymatic bioconversion and successive acid-assisted high temperature and pressure processing. Ginsenosides Rb1, Rg3, and Rg5 were investigated for their antitumor effects against five human cancer cell lines via the MTT assay. Among them, Rg5 exhibited the greatest cytotoxicity against breast cancer. Moreover, Rg5 remarkably suppressed breast cancer cell proliferation through mitochondria-mediated apoptosis and autophagic cell death. LC3B-GFP/Lysotracker and mRFP-EGFP-LC3B were utilized to show that Rg5 induced autophagosome-lysosome fusion. Western blot assays further illustrated that Rg5 decreased the phosphorylation levels of PI3K, Akt, mTOR, and Bad and suppressed the PI3K/Akt signaling pathway in breast cancer. Moreover, Rg5-induced apoptosis and autophagy could be dramatically strengthened by the PI3K/Akt inhibitor LY294002. Finally, a molecular docking study demonstrated that Rg5 could bind to the active pocket of PI3K. Collectively, our results revealed that Rg5 could be a potential therapeutic agent for breast cancer treatment.
Collapse
Affiliation(s)
- Yannan Liu
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi’an 710069, Shaanxi, China;
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi’an 710069, Shaanxi, China
- Biotech. & Biomed. Reserch Institute, Northwest University, Taibai North Road 229, Xi’an 710069, Shaanxi, China
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi’an 710069, Shaanxi, China;
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi’an 710069, Shaanxi, China
- Biotech. & Biomed. Reserch Institute, Northwest University, Taibai North Road 229, Xi’an 710069, Shaanxi, China
- Correspondence:
| |
Collapse
|
20
|
Pietri E, Massa I, Bravaccini S, Ravaioli S, Tumedei MM, Petracci E, Donati C, Schirone A, Piacentini F, Gianni L, Nicolini M, Campadelli E, Gennari A, Saba A, Campi B, Valmorri L, Andreis D, Fabbri F, Amadori D, Rocca A. Phase II Study of Dehydroepiandrosterone in Androgen Receptor-Positive Metastatic Breast Cancer. Oncologist 2019; 24:743-e205. [PMID: 30591548 PMCID: PMC6656524 DOI: 10.1634/theoncologist.2018-0243] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/28/2018] [Indexed: 12/02/2022] Open
Abstract
LESSONS LEARNED The androgen receptor (AR) is present in most breast cancers (BC), but its exploitation as a therapeutic target has been limited.This study explored the activity of dehydroepiandrosterone (DHEA), a precursor being transformed into androgens within BC cells, in combination with an aromatase inhibitor (to block DHEA conversion into estrogens), in a two-stage phase II study in patients with AR-positive/estrogen receptor-positive/human epidermal growth receptor 2-negative metastatic BC.Although well tolerated, only 1 of 12 patients obtained a prolonged clinical benefit, and the study was closed after its first stage for poor activity. BACKGROUND Androgen receptors (AR) are expressed in most breast cancers, and AR-agonists have some activity in these neoplasms. We investigated the safety and activity of the androgen precursor dehydroepiandrosterone (DHEA) in combination with an aromatase inhibitor (AI) in patients with AR-positive metastatic breast cancer (MBC). METHODS A two-stage phase II study was conducted in two patient cohorts, one with estrogen receptor (ER)-positive (resistant to AIs) and the other with triple-negative MBC. DHEA 100 mg/day was administered orally. The combination with an AI aimed to prevent the conversion of DHEA into estrogens. The main endpoint was the clinical benefit rate. The triple-negative cohort was closed early. RESULTS Twelve patients with ER-positive MBC were enrolled. DHEA-related adverse events, reported in four patients, included grade 2 fatigue, erythema, and transaminitis, and grade 1 drowsiness and musculoskeletal pain. Clinical benefit was observed in one patient with ER-positive disease whose tumor had AR gene amplification. There was wide inter- and intra-patient variation in serum levels of DHEA and its metabolites. CONCLUSION DHEA showed excellent safety but poor activity in MBC. Although dose and patient selection could be improved, high serum level variability may hamper further DHEA development in this setting.
Collapse
Affiliation(s)
- Elisabetta Pietri
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Ilaria Massa
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Sara Bravaccini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Sara Ravaioli
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Maria Maddalena Tumedei
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Elisabetta Petracci
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Caterina Donati
- Oncology Pharmacy Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Alessio Schirone
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Federico Piacentini
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | | | - Mario Nicolini
- Oncology Day Hospital Unit, Cervesi Hospital, Cattolica, Italy
| | | | - Alessandra Gennari
- Medical Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Alessandro Saba
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Beatrice Campi
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Linda Valmorri
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Daniele Andreis
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Francesco Fabbri
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Dino Amadori
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Andrea Rocca
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
21
|
Liu Y, Fan D. Ginsenoside Rg5 induces apoptosis and autophagy via the inhibition of the PI3K/Akt pathway against breast cancer in a mouse model. Food Funct 2019; 9:5513-5527. [PMID: 30207362 DOI: 10.1039/c8fo01122b] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Breast cancer is the most frequently diagnosed cancer and has become the main cause of cancer-related death among women worldwide. Traditional chemotherapy for breast cancer has serious side effects for patients, such as the first-line drug docetaxel. Ginsenoside Rg5, a rare ginsenoside and the main ingredient extracted from fine black ginseng, has been proved to have anti-breast cancer efficacy in vitro. Here, the in vivo anti-breast cancer efficacy, side effects and potential molecular mechanisms of Rg5 were investigated on a BALB/c nude mouse model of human breast cancer. The tumor growth inhibition rate of high dose Rg5 (20 mg kg-1) was 71.4 ± 9.4%, similar to that of the positive control docetaxel (72.0 ± 9.1%). Compared to docetaxel, Rg5 showed fewer side effects in the treatment of breast cancer. Treatment with Rg5 induced apoptosis and autophagy in breast cancer tissues. Rg5 was proved to induce caspase-dependent apoptosis via the activation of the extrinsic death receptor and intrinsic mitochondrial signaling pathways. The autophagy induction was related to the formation of an autophagosome and accumulation of LC3BII, P62 and critical Atg proteins. Further studies showed that Rg5 in a dose-dependent manner induced apoptosis and autophagy through the inhibition of the PI3K/Akt signaling pathway as indicated by the reduced phosphorylation level of PI3K and Akt. Taken together, Rg5 could be a novel and promising clinical antitumor drug targeting breast cancer.
Collapse
Affiliation(s)
- Yannan Liu
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China.
| | | |
Collapse
|
22
|
Parsons TK, Pratt RN, Tang L, Wu Y. An active and selective molecular mechanism mediating the uptake of sex steroids by prostate cancer cells. Mol Cell Endocrinol 2018; 477:121-131. [PMID: 29928927 DOI: 10.1016/j.mce.2018.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/31/2018] [Accepted: 06/16/2018] [Indexed: 12/21/2022]
Abstract
Steroid hormones play important roles in normal physiological functions and diseases. Sex steroids hormones are important in the biology and treatment of sex hormone-related cancer such as prostate cancer and breast cancer. Cells may take up steroids using multiple mechanisms. The conventionally accepted hypothesis that steroids cross cell membrane through passive diffusion has not been tested rigorously. Experimental data suggested that cells may take up sex steroid using an active uptake mechanism. 3H-testosterone uptake by prostate cancer cells showed typical transporter-mediated uptake kinetic. Cells retained testosterone taken up from the medium. The uptake of testosterone was selective for certain steroid hormones but not others. Data also indicated that the active and selective uptake mechanism resided in cholesterol-rich membrane domains, and may involve ATP and membrane transporters. In summary, the present study provided strong evidence to support the existence of an active and selective molecular mechanism for sex steroid uptake.
Collapse
Affiliation(s)
- Todd K Parsons
- Department of Urology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Rachel N Pratt
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Li Tang
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Yue Wu
- Department of Urology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA.
| |
Collapse
|
23
|
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
|
24
|
Schiffer L, Arlt W, Storbeck KH. Intracrine androgen biosynthesis, metabolism and action revisited. Mol Cell Endocrinol 2018; 465:4-26. [PMID: 28865807 PMCID: PMC6565845 DOI: 10.1016/j.mce.2017.08.016] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/28/2017] [Accepted: 08/28/2017] [Indexed: 12/19/2022]
Abstract
Androgens play an important role in metabolic homeostasis and reproductive health in both men and women. Androgen signalling is dependent on androgen receptor activation, mostly by testosterone and 5α-dihydrotestosterone. However, the intracellular or intracrine activation of C19 androgen precursors to active androgens in peripheral target tissues of androgen action is of equal importance. Intracrine androgen synthesis is often not reflected by circulating androgens but rather by androgen metabolites and conjugates. In this review we provide an overview of human C19 steroid biosynthesis including the production of 11-oxygenated androgens, their transport in circulation and uptake into peripheral tissues. We conceptualise the mechanisms of intracrinology and review the intracrine pathways of activation and inactivation in selected human tissues. The contribution of liver and kidney as organs driving androgen inactivation and renal excretion are also highlighted. Finally, the importance of quantifying androgen metabolites and conjugates to assess intracrine androgen production is discussed.
Collapse
Affiliation(s)
- Lina Schiffer
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Karl-Heinz Storbeck
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; Department of Biochemistry, Stellenbosch University, Stellenbosch 7600, South Africa
| |
Collapse
|
25
|
Antitumor Mechanisms of Curcumae Rhizoma Based on Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:4509892. [PMID: 29636777 PMCID: PMC5832109 DOI: 10.1155/2018/4509892] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/27/2017] [Indexed: 01/13/2023]
Abstract
Curcumae Rhizoma, a traditional Chinese medication, is commonly used in both traditional treatment and modern clinical care. Its anticancer effects have attracted a great deal of attention, but the mechanisms of action remain obscure. In this study, we screened for the active compounds of Curcumae Rhizoma using a drug-likeness approach. Candidate protein targets with functions related to cancer were predicted by reverse docking and then checked by manual search of the PubMed database. Potential target genes were uploaded to the GeneMANIA server and DAVID 6.8 database for analysis. Finally, compound-target, target-pathway, and compound-target-pathway networks were constructed using Cytoscape 3.3. The results revealed that the anticancer activity of Curcumae Rhizoma potentially involves 13 active compounds, 33 potential targets, and 31 signaling pathways, thus constituting a “multiple compounds, multiple targets, and multiple pathways” network corresponding to the concept of systematic actions in TCM. These findings provide an overview of the anticancer action of Curcumae Rhizoma from a network perspective, as well as setting an example for future studies of other materials used in TCM.
Collapse
|