1
|
Alladio E, Trapani F, Castellino L, Massano M, Di Corcia D, Salomone A, Berrino E, Ponzone R, Marchiò C, Sapino A, Vincenti M. Enhancing breast cancer screening with urinary biomarkers and Random Forest supervised classification: A comprehensive investigation. J Pharm Biomed Anal 2024; 244:116113. [PMID: 38554554 DOI: 10.1016/j.jpba.2024.116113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/10/2024] [Accepted: 03/15/2024] [Indexed: 04/01/2024]
Abstract
OBJECTIVES Urinary sex hormones are investigated as potential biomarkers for the early detection of breast cancer, aiming to evaluate their relevance and applicability, in combination with supervised machine-learning data analysis, toward the ultimate goal of extensive screening. METHODS Sex hormones were determined on urine samples collected from 250 post-menopausal women (65 healthy - 185 with breast cancer, recruited among the clinical patients of Candiolo Cancer Institute FPO-IRCCS (Torino, Italy). Two analytical procedures based on UHPLC-MS/HRMS were developed and comprehensively validated to quantify 20 free and conjugated sex hormones from urine samples. The quantitative data were processed by seven machine learning algorithms. The efficiency of the resulting models was compared. RESULTS Among the tested models aimed to relate urinary estrogen and androgen levels and the occurrence of breast cancer, Random Forest (RF) proved to underscore all the other supervised classification approaches, including Partial Least Squares - Discriminant Analysis (PLS-DA), in terms of effectiveness and robustness. The final optimized model built on only five biomarkers (testosterone-sulphate, alpha-estradiol, 4-methoxyestradiol, DHEA-sulphate, and epitestosterone-sulphate) achieved an approximate 98% diagnostic accuracy on replicated validation sets. To balance the less-represented population of healthy women, a Synthetic Minority Oversampling TEchnique (SMOTE) data oversampling approach was applied. CONCLUSIONS By means of tunable hyperparameters optimization, the RF algorithm showed great potential for early breast cancer detection, as it provides clear biomarkers ranking and their relative efficiency, allowing to ground the final diagnostic model on a restricted selection five steroid biomarkers only, as desirable for noninvasive tests with wide screening purposes.
Collapse
Affiliation(s)
- Eugenio Alladio
- Department of Chemistry, University of Turin, Italy; Centro Regionale Antidoping, Orbassano, TO, Italy
| | - Fulvia Trapani
- Department of Chemistry, University of Turin, Italy; Centro Regionale Antidoping, Orbassano, TO, Italy
| | - Lorenzo Castellino
- Department of Chemistry, University of Turin, Italy; Centro Regionale Antidoping, Orbassano, TO, Italy
| | - Marta Massano
- Department of Chemistry, University of Turin, Italy; Centro Regionale Antidoping, Orbassano, TO, Italy
| | | | - Alberto Salomone
- Department of Chemistry, University of Turin, Italy; Centro Regionale Antidoping, Orbassano, TO, Italy
| | - Enrico Berrino
- Department of Medical Sciences, University of Turin, Turin, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | | | - Caterina Marchiò
- Department of Medical Sciences, University of Turin, Turin, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Anna Sapino
- Department of Medical Sciences, University of Turin, Turin, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Marco Vincenti
- Department of Chemistry, University of Turin, Italy; Centro Regionale Antidoping, Orbassano, TO, Italy.
| |
Collapse
|
2
|
Davis SJ, Arscott SA, Goltz S, Muir C, Binkley N, Tanumihardjo SA. Urinary 2- to 16α-hydroxyestrone ratio did not change with cruciferous vegetable intake in premenopausal women. INT J VITAM NUTR RES 2024; 94:177-186. [PMID: 37335576 DOI: 10.1024/0300-9831/a000785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
The mass ratio of urinary 2-hydroxyestrone to 16-α-hydroxyestrone (2:16) is hypothesized as a biomarker of breast cancer risk in premenopausal women, with higher ratios being theoretically protective. Cruciferous vegetable intake has been associated with higher urinary 2:16 in some studies. We investigated whether a whole-food supplement made from dried Brussels sprouts and kale would increase urinary 2:16 in comparison with placebo or cruciferous vegetables in women. This randomized, parallel arm, placebo-controlled, partly blinded study included 78 healthy premenopausal women (38-50 y) with screening urinary 2:16 ≤3.0. Subjects received either six capsules containing 550 mg dried Brussels sprouts and kale per capsule, 40 g daily alternating broccoli or Brussels sprouts, or placebo for eight weeks. Urinary 2:16 and creatinine were measured at baseline, four, and eight weeks. Intent-to-treat repeated measures-ANOVA with multiple imputation (n=100) for missing values identified no treatment effect (P=0.9) or treatment-by-time interaction (P=0.6); however, a significant time effect was noted (P=0.02). Per-protocol analyses including complete cases found no treatment effect (P=1) or treatment-by-time interaction (P=0.6); however, the significant time effect remained (P=0.03). Restricting analysis to subjects with >80% compliance maintained the time effect (P=0.02). Using Pearson correlations, android-pattern and android:gynoid fat were predictive of change (P≤0.05). In conclusion, neither cruciferous supplements nor an added vegetable serving altered urinary 2:16 in premenopausal women with eight weeks treatment. This ratio did vary with time, which is important for designing future trials.
Collapse
Affiliation(s)
- Stephanie J Davis
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison (UW), Madison, USA
| | - Sara A Arscott
- Standard Process Inc., Palmyra, Wisconsin, USA
- Department of Family Medicine and Community Health, University of Wisconsin-Madison (UW), Madison, USA
| | - Shellen Goltz
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison (UW), Madison, USA
| | - Cassidy Muir
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison (UW), Madison, USA
| | - Neil Binkley
- Osteoporosis Clinical Research Program, University of Wisconsin-Madison (UW), Madison, USA
| | - Sherry A Tanumihardjo
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison (UW), Madison, USA
| |
Collapse
|
3
|
Guo Z, Yang B, Zhu J, Lou S, Hao H, Lu W. Light-activated, dual-mode fluorescence and colorimetric detection of estradiol with high fidelity based on aptamer's special recognition. Food Chem 2024; 436:137702. [PMID: 37844513 DOI: 10.1016/j.foodchem.2023.137702] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/26/2023] [Accepted: 10/07/2023] [Indexed: 10/18/2023]
Abstract
The intake of estradiol residue from food will lead to health problems, so the rapid and reliable detection of estradiol residue is essential. Multi-mode assays are inherently self-correcting and self-validating, providing more reliable, interference-resistant, high-fidelity results. Here, we developed a dual-mode method to achieve a rapid, reliable, and sensitive detection of estradiol. The binding of thioflavin T to the cavity sites of estradiol aptamer not only generates a strong fluorescence signal, but also provides light-activated oxidase activity to produce a blue oxidation product. But the specific binding between aptamer and estradiol will compete with the above process. Thus a dual-mode fluorescence and colorimetric detection of estradiol was realized with a detection limit of 0.15 μM or 0.27 μM. Besides, the dual-mode method showed a good selectivity for estradiol detection. The method could detect estradiol in meat food, showing a good application potential to monitor endocrine-disrupting chemical.
Collapse
Affiliation(s)
- Zihua Guo
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 410005, PR China
| | - Bin Yang
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 410005, PR China.
| | - Jian Zhu
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 410005, PR China
| | - Shuyan Lou
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 410005, PR China
| | - Huimin Hao
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 410005, PR China
| | - Weiyi Lu
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 410005, PR China
| |
Collapse
|
4
|
Zhao H, Yang H, Li Z, Ge Z, Zhou M, Li L, He J. Urine levels of estrogen and its metabolites in premenopausal middle-aged women with different degrees of obesity and their correlation with glucose and lipid metabolism. Heliyon 2023; 9:e22362. [PMID: 38058651 PMCID: PMC10696012 DOI: 10.1016/j.heliyon.2023.e22362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 12/08/2023] Open
Abstract
Objectives To determine the levels of estrogen and estrogen metabolites in the urine of premenopausal women with obesity and their correlation with glucose and lipid metabolism. Methords 135 premenopausal women were selected from the same area. According to the body mass index (BMI), they were divided into four different groups. High performance liquid chromatography-mass spectrometry (HPLC/MS) was adopted to detect the concentrations of estrogen and estrogen metabolites in the urine. The influencing factors of BMI were analyzed, the correlation between the urinary degrees of estrogen and estrogen metabolites and glucose and lipid metabolism levels was assessed. Results (1) The concentrations of 17β-estradiol (E2), estrone (E1), 16α-hydroxyestrone (16α-OHE1) and 2-hydroxyestrone (2-OHE1) gradually increased with increasing BMI (p < 0.05). (2) Stepwise regression analysis displayed that the concentrations of E2, 16α-OHE1 and 2-OHE1 in urine were significantly positively correlated with BMI (p < 0.05). (3) The concentrations of E2, E1, 16α-OHE1, 2-OHE1 and 16α-OHE1/2-OHE1 in urine were greatly positively related to fasting insulin (FIN), Triglyceride (TG), Total Cholesterol (TC) and Low-density lipoprotein (LDL) (p < 0.05). And they were greatly negatively related to High-density lipoprotein (HDL) (p < 0.05). Conclusions Early screening can reflect the degree of obesity and glucose and lipid metabolism disorders in premenopausal middle-aged women, thereby providing guidance for improving the prognosis of obese women.
Collapse
Affiliation(s)
- Huanhuan Zhao
- Department of Obstetrics and Gynecology, Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei Province, China
| | - Hongfang Yang
- Department of Obstetrics and Gynecology, Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei Province, China
| | - Zhiwei Li
- School of Chemistry and Pharmaceutical Engineering, Shijiazhuang University of Science and Technology, Shijiazhuang, 050018, Hebei Province, China
| | - Zhonghuan Ge
- Department of Laboratory, Nanpi County People's Hospital, Cangzhou Nanpi County, 061550, Hebei Province, China
| | - Mei Zhou
- Department of Laboratory, Nanpi County People's Hospital, Cangzhou Nanpi County, 061550, Hebei Province, China
| | - Li Li
- Department of Obstetrics and Gynecology, Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei Province, China
| | - Jing He
- Department of Obstetrics and Gynecology, Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei Province, China
| |
Collapse
|
5
|
Nahmias-Blank D, Maimon O, Meirovitz A, Sheva K, Peretz-Yablonski T, Elkin M. Excess body weight and postmenopausal breast cancer: Emerging molecular mechanisms and perspectives. Semin Cancer Biol 2023; 96:26-35. [PMID: 37739109 DOI: 10.1016/j.semcancer.2023.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
Postmenopausal, obese women have a significantly higher risk of developing estrogen receptor-positive (ER+) breast tumors, that are resistant to therapies and are associated with higher recurrence and death rates. The global prevalence of overweight/obese women has reached alarming proportions and with postmenopausal ER+ breast carcinoma (BC) having the highest incidence among the three obesity-related cancers in females (i.e., breast, endometrial and ovarian), this is of significant concern. Elucidation of the precise molecular mechanisms underlying the pro-cancerous action of obesity in ER+BC is therefore critical for disease prevention and novel treatment initiatives. Interestingly, accumulating data has shown opposing relationships between obesity and cancer in either pre- or post-menopausal women. Excess body weight is associated with an increased risk of breast cancer in postmenopausal women and a decreased risk in pre-menopausal women. Moreover, excess adiposity during early life appears to be protective against postmenopausal breast cancer, including both ER+ and ER negative BC subtypes. Overall, estrogen-dependent mechanisms have been implicated as the main driving force in obesity-related breast tumorigenesis. In the present review we discuss the epidemiologic and mechanistic aspects of association between obesity and breast tumors after menopause, mainly in the context of hormone dependency. Molecular and cellular events underlying this association present as potential avenues for both therapeutic intervention as well as the prevention of BC-promoting processes linked to excess adiposity, which is proving to be vital in an increasingly obese global population.
Collapse
Affiliation(s)
- Daniela Nahmias-Blank
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ofra Maimon
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Amichay Meirovitz
- Legacy Heritage Oncology Center and Dr. Larry Norton Institute, Soroka University Medical Center, Be'er Sheva 84101, Israel
| | - Kim Sheva
- Legacy Heritage Oncology Center and Dr. Larry Norton Institute, Soroka University Medical Center, Be'er Sheva 84101, Israel
| | - Tamar Peretz-Yablonski
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel; Hebrew University Medical School, Jerusalem 91120, Israel
| | - Michael Elkin
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel; Hebrew University Medical School, Jerusalem 91120, Israel.
| |
Collapse
|
6
|
Ghosh D. Structures and Functions of Human Placental Aromatase and Steroid Sulfatase, Two Key Enzymes in Estrogen Biosynthesis. Steroids 2023; 196:109249. [PMID: 37207843 DOI: 10.1016/j.steroids.2023.109249] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/21/2023]
Abstract
Cytochrome P450 aromatase (AROM) and steroid sulfatase (STS) are the two key enzymes for the biosynthesis of estrogens in human, and maintenance of the critical balance between androgens and estrogens. Human AROM, an integral membrane protein of the endoplasmic reticulum, is a member of the cytochrome P450 superfamily. It is the only enzyme to catalyze the conversion of androgens with non-aromatic A-rings to estrogens characterized by the aromatic A-ring. Human STS, also an integral membrane protein of the endoplasmic reticulum, is a Ca2+-dependent enzyme that catalyzes the hydrolysis of sulfate esters of estrone and dehydroepiandrosterone to the unconjugated steroids, the precursors of the most potent forms of estrogens and androgens, namely, 17β-estradiol, 16α,17β-estriol, testosterone and dihydrotestosterone. Expression of these steroidogenic enzymes locally within organs and tissues of the endocrine, reproductive, and central nervous systems is the key for maintaining high levels of the reproductive steroids. The enzymes have been drug targets for the prevention and treatment of diseases associated with steroid hormone excesses, especially in breast, endometrial and prostate malignancies. Both enzymes have been the subjects of vigorous research for the past six decades. In this article, we review the important findings on their structure-function relationships, specifically, the work that began with unravelling of the closely guarded secrets, namely, the 3-D structures, active sites, mechanisms of action, origins of substrate specificity and the basis of membrane integration. Remarkably, these studies were conducted on the enzymes purified in their pristine forms from human placenta, the discarded and their most abundant source. The purification, assay, crystallization, and structure determination methodologies are described. Also reviewed are their functional quaternary organizations, post-translational modifications and the advancements made in the structure-guided inhibitor design efforts. Outstanding questions that still remain open are summarized in closing.
Collapse
Affiliation(s)
- Debashis Ghosh
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210.
| |
Collapse
|
7
|
Harnsoongnoen S, Loutchanwoot P, Srivilai P. Sensing High 17β-Estradiol Concentrations Using a Planar Microwave Sensor Integrated with a Microfluidic Channel. BIOSENSORS 2023; 13:bios13050541. [PMID: 37232902 DOI: 10.3390/bios13050541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/05/2023] [Accepted: 05/10/2023] [Indexed: 05/27/2023]
Abstract
The global issue of pollution caused by endocrine-disrupting chemicals (EDCs) has been gaining increasing attention. Among the EDCs of environmental concern, 17β-estradiol (E2) can produce the strongest estrogenic effects when it enters the organism exogenously through various routes and has the potential to cause harm, including malfunctions of the endocrine system and development of growth and reproductive disorders in humans and animals. Additionally, in humans, supraphysiological levels of E2 have been associated with a range of E2-dependent disorders and cancers. To ensure environmental safety and prevent potential risks of E2 to human and animal health, it is crucial to develop rapid, sensitive, low cost and simple approaches for detecting E2 contamination in the environment. A planar microwave sensor for E2 sensing is presented based on the integration of a microstrip transmission line (TL) loaded with a Peano fractal geometry with a narrow slot complementary split-ring resonator (PF-NSCSRR) and a microfluidic channel. The proposed technique offers a wide linear range for detecting E2, ranging from 0.001 to 10 mM, and can achieve high sensitivity with small sample volumes and simple operation methods. The proposed microwave sensor was validated through simulations and empirical measurements within a frequency range of 0.5-3.5 GHz. The E2 solution was delivered to the sensitive area of the sensor device via a microfluidic polydimethylsiloxane (PDMS) channel with an area of 2.7 mm2 and sample value of 1.37 µL and measured by a proposed sensor. The injection of E2 into the channel resulted in changes in the transmission coefficient (S21) and resonance frequency (Fr), which can be used as an indicator of E2 levels in solution. The maximum quality factor of 114.89 and the maximum sensitivity based on S21 and Fr at a concentration of 0.01 mM were 1746.98 dB/mM and 40 GHz/mM, respectively. Upon comparing the proposed sensor with the original Peano fractal geometry with complementary split-ring (PF-CSRR) sensors without a narrow slot, several parameters were evaluated, including sensitivity, quality factor, operating frequency, active area, and sample volume. The results showed that the proposed sensor exhibited an increased sensitivity of 6.08% and had a 40.72% higher quality factor, while the operating frequency, active area, and sample volume showed decreases of 1.71%, 25%, and 28.27%, respectively. The materials under tests (MUTs) were analyzed and categorized into groups using principal component analysis (PCA) with a K-mean clustering algorithm. The proposed E2 sensor has a compact size and simple structure that can be easily fabricated with low-cost materials. With the small sample volume requirement, fast measurement with a wide dynamic range, and a simple protocol, this proposed sensor can also be applied to measure high E2 levels in environmental, human, and animal samples.
Collapse
Affiliation(s)
- Supakorn Harnsoongnoen
- The Biomimicry for Sustainable Agriculture, Health, Environment and Energy Research Unit, Department of Physics, Faculty of Science, Mahasarakham University, Kantarawichai District, Maha Sarakham 44150, Thailand
| | - Panida Loutchanwoot
- Department of Biology, Faculty of Science, Mahasarakham University, Kantarawichai District, Maha Sarakham 44150, Thailand
| | - Prayook Srivilai
- Department of Biology, Faculty of Science, Mahasarakham University, Kantarawichai District, Maha Sarakham 44150, Thailand
| |
Collapse
|
8
|
Tian M, Wu S, Wang YX, Liu L, Zhang J, Shen H, Lu Y, Bao H, Huang Q. Associations of environmental phthalate exposure with male steroid hormone synthesis and metabolism: An integrated epidemiology and toxicology study. JOURNAL OF HAZARDOUS MATERIALS 2022; 436:129213. [PMID: 35739735 DOI: 10.1016/j.jhazmat.2022.129213] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
Humans are simultaneously and constantly exposed to various lipophilic chain phthalate acid esters. The association of urinary phthalate metabolites with altered male steroid hormone synthesis and metabolism was examined using epidemiology and toxicology studies. We measured 8 phthalate metabolites [monomethyl phthalate (MMP), monoethyl phthalate (MEP), mono-n-butyl phthalate (MBP), mono-benzyl phthalate (MBzP), mono-n-octylphthalate (MOP), mono-(2-ethylhexyl) phthalate (MEHP), mono-(2-ethyl-5-hydroxyhexyl) phthalate (MEHHP) and mono (2-ethyl-5-oxohexyl) phthalate (MEOHP)] and two sex hormones [testosterone (T) and estradiol (E2)] in single serum and repeated spot urine samples among 451 reproductive-age males. Moreover, in vitro experiments with Leydig cell MLTC-1 steroidogenesis and liver cell HepG2 efflux in response to mixed and individual phthalates were designed to simulate real-world scenarios of human exposure. As a joint mixture, the phthalate metabolite was inversely associated with serum T and E2 concentrations but positively associated with urinary T and E2 concentrations. Combined with in vitro experiments, DEHP metabolites were identified as the predominant contributor to the decline in hormone synthesis, and ATP-binding cassette (ABC) gene activation might be involved in hormone excretion. Exposure to environmentally relevant phthalates was associated with both altered steroid synthesis and excretion, which provides additional insights into the endocrine-disrupting potential of phthalates.
Collapse
Affiliation(s)
- Meiping Tian
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China.
| | - Shuangshan Wu
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Yi-Xin Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Liangpo Liu
- School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Jie Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Heqing Shen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yanyang Lu
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Huaqiong Bao
- NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning Science and Technology Research Institute, Chongqing 400020, China
| | - Qingyu Huang
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China.
| |
Collapse
|
9
|
Drummond AE, Swain CTV, Brown KA, Dixon-Suen SC, Boing L, van Roekel EH, Moore MM, Gaunt TR, Milne RL, English DR, Martin RM, Lewis SJ, Lynch BM. Linking Physical Activity to Breast Cancer via Sex Steroid Hormones, Part 2: The Effect of Sex Steroid Hormones on Breast Cancer Risk. Cancer Epidemiol Biomarkers Prev 2022; 31:28-37. [PMID: 34670801 PMCID: PMC7612577 DOI: 10.1158/1055-9965.epi-21-0438] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/10/2021] [Accepted: 10/07/2021] [Indexed: 11/25/2022] Open
Abstract
We undertook a systematic review and appraised the evidence for an effect of circulating sex steroid hormones and sex hormone-binding globulin (SHBG) on breast cancer risk in pre- and postmenopausal women. Systematic searches identified prospective studies relevant to this review. Meta-analyses estimated breast cancer risk for women with the highest compared with the lowest level of sex hormones, and the DRMETA Stata package was used to graphically represent the shape of these associations. The ROBINS-E tool assessed risk of bias, and the GRADE system appraised the strength of evidence. In premenopausal women, there was little evidence that estrogens, progesterone, or SHBG were associated with breast cancer risk, whereas androgens showed a positive association. In postmenopausal women, higher estrogens and androgens were associated with an increase in breast cancer risk, whereas higher SHBG was inversely associated with risk. The strength of the evidence quality ranged from low to high for each hormone. Dose-response relationships between sex steroid hormone concentrations and breast cancer risk were most notable for postmenopausal women. These data support the plausibility of a role for sex steroid hormones in mediating the causal relationship between physical activity and the risk of breast cancer.See related reviews by Lynch et al., p. 11 and Swain et al., p. 16.
Collapse
Affiliation(s)
- Ann E Drummond
- Cancer Epidemiology Division, Cancer Council Victoria, Victoria, Australia
| | | | - Kristy A Brown
- Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Suzanne C Dixon-Suen
- Cancer Epidemiology Division, Cancer Council Victoria, Victoria, Australia
- Institute for Physical Activity and Nutrition, Deakin University, Geelong, Victoria, Australia
| | - Leonessa Boing
- Laboratory of Research in Leisure and Physical Activity, Santa Catarina State University, Florianópolis, Brazil
| | - Eline H van Roekel
- Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Melissa M Moore
- Medical Oncology, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Tom R Gaunt
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Roger L Milne
- Cancer Epidemiology Division, Cancer Council Victoria, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Australia
| | - Dallas R English
- Cancer Epidemiology Division, Cancer Council Victoria, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Richard M Martin
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
- NIHR Biomedical Research Centre at University Hospitals Bristol and Weston NHS Foundation Trust and the University of Bristol, Bristol, United Kingdom
| | - Sarah J Lewis
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Brigid M Lynch
- Cancer Epidemiology Division, Cancer Council Victoria, Victoria, Australia.
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
10
|
Pacyga DC, Gardiner JC, Flaws JA, Li Z, Calafat AM, Korrick SA, Schantz SL, Strakovsky RS. Maternal phthalate and phthalate alternative metabolites and urinary biomarkers of estrogens and testosterones across pregnancy. ENVIRONMENT INTERNATIONAL 2021; 155:106676. [PMID: 34116379 PMCID: PMC8292204 DOI: 10.1016/j.envint.2021.106676] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/02/2021] [Accepted: 05/26/2021] [Indexed: 05/06/2023]
Abstract
BACKGROUND/OBJECTIVES Pregnant women are ubiquitously exposed to phthalates from food packaging materials and personal care products. Phthalates alter estrogen and testosterone concentrations in experimental models, but their ability to impact these hormones in human pregnancy is not well characterized. METHODS We recruited women ages 18-40 into the Illinois Kids Development Study (I-KIDS) in early pregnancy. Participants provided up to 5 first-morning urine samples across pregnancy (8-40 weeks gestation) that we pooled for quantification of 19 phthalate or phthalate alternative metabolites. Either individual (ng/mL) or molar sums (nmol/mL) of metabolites were used as exposure biomarkers. We summed urinary concentrations (ng/mL) of eight major estrogen (SumEstrogens) and two major testosterone (SumTestosterones) metabolites measured at median 13, 28, and 34 weeks gestation. We also estimated the ratio of estrogens-to-androgens. Linear mixed-effects models assessed relationships of phthalates/alternatives as continuous measures or as concentration quartiles with SumEstrogens, SumTestosterones, and the Estrogen/Androgen ratio in 434 women. In our models, we controlled for age, race, education, parity, smoking in the first trimester, pre-pregnancy body mass index, diet quality, conception season, fetal sex, and gestational age at hormone assessment. We also explored whether gestational age at hormone assessment or fetal sex modified these associations. All biomarkers and outcomes were specific gravity-adjusted, and continuous exposures and outcomes were also natural log-transformed. RESULTS Most participants were non-Hispanic white (80.9%), college educated (82.2%), and had urinary phthalate/alternative metabolite concentrations similar to those of reproductive-aged U.S. women. Overall, select phthalate metabolites were positively associated with SumEstrogens and SumTestosterones, but negatively associated with the Estrogen/Androgen ratio. For example, SumEstrogens was 5.1% (95%CI: 1.8, 8.5) higher with every 2-fold increase in sum of di(2-ethylhexyl) phthalate metabolites, while SumTestosterones was 7.9% (95%CI: 1.0, 15.3) higher and Estrogen/Androgen ratio was -7.7% (95%CI: -13.6, -1.4) lower with every 2-fold increase in monoethyl phthalate. However, phthalate alternatives were only positively associated with SumEstrogens, which was 2.4% (95%CI: 0.4, 4.5) and 3.2% (95%CI: 0.7, 5.8) higher with every 2-fold increase in sum of di(isononyl) cyclohexane-1,2-dicarboxylate metabolites and sum of di(2-ethylhexyl) terephthalate metabolites, respectively. Gestational age- and fetal sex-specific associations were only consistently observed for associations of phthalates/alternatives with SumEstrogens, where associations were strongest in mid-to-late pregnancy in women carrying females. CONCLUSION Phthalates/alternatives may impact gestational hormones, with potential for gestational age- and fetal sex-specific associations. Whether maternal urinary estrogens and testosterones mediate associations of phthalates/alternatives with pregnancy and fetal outcomes merits further investigation.
Collapse
Affiliation(s)
- Diana C Pacyga
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
| | - Joseph C Gardiner
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
| | - Jodi A Flaws
- Department of Comparative Biosciences, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Zhong Li
- Roy J. Carver Biotechnology Center, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Antonia M Calafat
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA
| | - Susan A Korrick
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Susan L Schantz
- Department of Comparative Biosciences, University of Illinois, Urbana-Champaign, IL 61801, USA; Beckman Institute, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Rita S Strakovsky
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
11
|
Starek-Świechowicz B, Budziszewska B, Starek A. Endogenous estrogens-breast cancer and chemoprevention. Pharmacol Rep 2021; 73:1497-1512. [PMID: 34462889 PMCID: PMC8599256 DOI: 10.1007/s43440-021-00317-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer is the most common female malignancy and the second leading cause of cancer related deaths. It is estimated that about 40% of all cancer in women is hormonally mediated. Both estrogens and androgens play critical roles in the initiation and development of breast cancer. Estrogens influence normal physiological growth, proliferation, and differentiation of breast tissues, as well as the development and progression of breast malignancy. Breast cancer is caused by numerous endo- and exogenous risk factors. The paper presents estrogen metabolism, in particular 17β-estradiol and related hormones. The mechanisms of estrogen carcinogenesis include the participation of estrogen receptors, the genotoxic effect of the estrogen metabolites, and epigenetic processes that are also presented. The role of reactive oxygen species in breast cancer has been described. It called attention to a role of numerous signaling pathways in neoplastic transformation. Chemoprotective agents, besides other phytoestrogens, classical antioxidants, synthetic compounds, and their mechanisms of action have been shown.
Collapse
Affiliation(s)
- Beata Starek-Świechowicz
- Department of Biochemical Toxicology, Chair of Toxicology, Medical College, Jagiellonian University, Medyczna 9, 30-688, Kraków, Poland.
| | - Bogusława Budziszewska
- Department of Biochemical Toxicology, Chair of Toxicology, Medical College, Jagiellonian University, Medyczna 9, 30-688, Kraków, Poland.,Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Andrzej Starek
- Department of Biochemical Toxicology, Chair of Toxicology, Medical College, Jagiellonian University, Medyczna 9, 30-688, Kraków, Poland
| |
Collapse
|
12
|
Garzia NA, Cushing-Haugen K, Kensler TW, Tamimi RM, Harris HR. Adolescent and early adulthood inflammation-associated dietary patterns in relation to premenopausal mammographic density. Breast Cancer Res 2021; 23:71. [PMID: 34233736 PMCID: PMC8261986 DOI: 10.1186/s13058-021-01449-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 06/23/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Adolescence and early adulthood has been identified as a critical time window for establishing breast cancer risk. Mammographic density is an independent risk factor for breast cancer that may be influenced by diet, but there has been limited research conducted on the impact of diet on mammographic density. Thus, we sought to examine the association between adolescent and early adulthood inflammatory dietary patterns, which have previously been associated with breast cancer risk, and premenopausal mammographic density among women in the Nurses' Health Study II (NHSII). METHODS This study included control participants with premenopausal mammograms from an existing breast cancer case-control study nested within the NHSII who completed a Food Frequency Questionnaire in 1998 about their diet during high school (HS-FFQ) (n = 685) and/or a Food Frequency Questionnaire in 1991 (Adult-FFQ) when they were 27-44 years old (n = 1068). Digitized analog film mammograms were used to calculate the percent density, absolute dense, and non-dense areas. Generalized linear models were fit to evaluate the associations of a pro-inflammatory dietary pattern and the Alternative Healthy Eating Index (AHEI, an anti-inflammatory dietary pattern) with each breast density measure. RESULTS Significant associations were observed between an adolescent pro-inflammatory dietary pattern and mammographic density in some age-adjusted models; however, these associations did not remain after adjustment for BMI and other breast cancer risk factors. No associations were observed with the pro-inflammatory pattern or with the AHEI pattern in adolescence or early adulthood in fully adjusted models. CONCLUSIONS To our knowledge, this is the first study to evaluate the dietary patterns during adolescence and early adulthood in relation to mammographic density phenotypes. Our findings do not support an association between adolescent and early adulthood diet and breast density in mid-adulthood that is independent of BMI or other breast cancer risk factors.
Collapse
Affiliation(s)
- Nichole A Garzia
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. North, Seattle, WA, 98109-1024, USA.
- Department of Epidemiology, School of Public Health, University of Washington, 3980 15th Ave. NE, Seattle, WA, 98195-002, USA.
| | - Kara Cushing-Haugen
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. North, Seattle, WA, 98109-1024, USA
| | - Thomas W Kensler
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. North, Seattle, WA, 98109-1024, USA
| | - Rulla M Tamimi
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115-6028, USA
- Department of Population Health Sciences, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065-4805, USA
| | - Holly R Harris
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. North, Seattle, WA, 98109-1024, USA
- Department of Epidemiology, School of Public Health, University of Washington, 3980 15th Ave. NE, Seattle, WA, 98195-002, USA
| |
Collapse
|
13
|
Fujii T, Ogasawara M, Kamishikiryo J, Morita T. β-Estradiol Enhanced Secretion of Lipoprotein Lipase from Mouse Mammary Tumor FM3A Cells. Biol Pharm Bull 2021; 43:1407-1412. [PMID: 32879215 DOI: 10.1248/bpb.b20-00408] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The role of β-estradiol (E2) in lipoprotein metabolism in mammary tumors is unclear, therefore, we investigated the effect of E2 on the secretion of lipoprotein lipase (LPL) from mouse mammary tumor FM3A cells. E2-treated cells increased the secretion of active LPL from FM3A cells in a time- and dose-dependent manner. Activity of mitogen-activated protein kinase (MAPK) was increased in the tumor cells treated with E2, and enhanced secretion of LPL was suppressed by MAPK kinase 1/2 inhibitor, PD98059, extracellular signal-regulated kinase (ERK) 1/2 inhibitor, FR180204, p38 MAPK inhibitor, SB202190, and phosphatidyl inositol 3-kinase (PI3K) inhibitor, LY294002. In addition, the effect of E2 on LPL secretion was markedly suppressed by an inhibitor of mammalian target of rapamycin complex (mTORC) 1 and 2, KU0063794, but were not by a mTORC1 inhibitor, rapamycin. Furthermore, a small interfering RNA (siRNA)-mediated decrease in the expression of rapamycin-insensitive companion of mTOR (Rictor), a pivotal component of mTORC2, suppressed secretion of LPL by E2. These results suggest that the stimulatory secretion of LPL by E2 from the tumor cells is closely associated with an activation of mTORC2 rather than mTORC1 possibly via the MAPK cascade.
Collapse
Affiliation(s)
- Tomoyasu Fujii
- Department of Biochemistry, Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University
| | - Mizuho Ogasawara
- Department of Biochemistry, Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University.,Department of Pharmacy, Kochi Health Sciences Center
| | - Jun Kamishikiryo
- Department of Biochemistry, Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University
| | - Tetsuo Morita
- Department of Biochemistry, Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University
| |
Collapse
|
14
|
Timblin GA, Tharp KM, Ford B, Winchester JM, Wang J, Zhu S, Khan RI, Louie SK, Iavarone AT, Ten Hoeve J, Nomura DK, Stahl A, Saijo K. Mitohormesis reprogrammes macrophage metabolism to enforce tolerance. Nat Metab 2021; 3:618-635. [PMID: 34031590 PMCID: PMC8162914 DOI: 10.1038/s42255-021-00392-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 04/15/2021] [Indexed: 02/03/2023]
Abstract
Macrophages generate mitochondrial reactive oxygen species and mitochondrial reactive electrophilic species as antimicrobials during Toll-like receptor (TLR)-dependent inflammatory responses. Whether mitochondrial stress caused by these molecules impacts macrophage function is unknown. Here, we demonstrate that both pharmacologically driven and lipopolysaccharide (LPS)-driven mitochondrial stress in macrophages triggers a stress response called mitohormesis. LPS-driven mitohormetic stress adaptations occur as macrophages transition from an LPS-responsive to LPS-tolerant state wherein stimulus-induced pro-inflammatory gene transcription is impaired, suggesting tolerance is a product of mitohormesis. Indeed, like LPS, hydroxyoestrogen-triggered mitohormesis suppresses mitochondrial oxidative metabolism and acetyl-CoA production needed for histone acetylation and pro-inflammatory gene transcription, and is sufficient to enforce an LPS-tolerant state. Thus, mitochondrial reactive oxygen species and mitochondrial reactive electrophilic species are TLR-dependent signalling molecules that trigger mitohormesis as a negative feedback mechanism to restrain inflammation via tolerance. Moreover, bypassing TLR signalling and pharmacologically triggering mitohormesis represents a new anti-inflammatory strategy that co-opts this stress response to impair epigenetic support of pro-inflammatory gene transcription by mitochondria.
Collapse
Affiliation(s)
- Greg A Timblin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA.
| | - Kevin M Tharp
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Breanna Ford
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
- Novartis-Berkeley Center for Proteomics and Chemistry Technologies and Department of Chemistry, University of California, Berkeley, Berkeley, CA, USA
| | - Janet M Winchester
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Jerome Wang
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Stella Zhu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Rida I Khan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Shannon K Louie
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Anthony T Iavarone
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley, CA, USA
- QB3/Chemistry Mass Spectrometry Facility, University of California, Berkeley, Berkeley, CA, USA
| | - Johanna Ten Hoeve
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging and UCLA Metabolomics Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Daniel K Nomura
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
- Novartis-Berkeley Center for Proteomics and Chemistry Technologies and Department of Chemistry, University of California, Berkeley, Berkeley, CA, USA
| | - Andreas Stahl
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Kaoru Saijo
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
15
|
Xu Z, Sun T, He H, Liu W, Fan L, Zhao L, Wu X, Han Z, Zhang Y, Wang Q, Ning B, Gao Z. Simultaneous detection of diethylstilbestrol and estradiol residues with a single immunochromatographic assay strip. Food Sci Nutr 2021; 9:1824-1830. [PMID: 33747491 PMCID: PMC7958558 DOI: 10.1002/fsn3.2127] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/18/2020] [Accepted: 12/26/2020] [Indexed: 01/17/2023] Open
Abstract
An immunochromatographic assay (ICA) based on competitive format was developed and validated for simultaneously rapid and sensitive detection of diethylstilbestrol (DES) and estradiol (E2) in milk and tissue samples. For this purpose, two monoclonal antibodies raised against those two estrogens were conjugated to gold nanoparticles and were applied to the conjugate pads of the test strip. The competitors of the DES-BSA/E2-BSA conjugates were immobilized onto a nitrocellulose membrane at two detection zones to form T1 and T2, respectively. The immunochromatographic assay had a visual detection limit of DES at 30 ng/g in milk powder, 25 ng/g in liquid milk, and 25 ng/g in shrimp tissue, respectively, and the results can be judged within 7-10 min. The visual detection limit of E2 was 75 ng/g in milk powder, 65 ng/g in liquid milk, and 60 ng/g in shrimp tissue, respectively, and the results can be judged within 3-4 min. It had advantages in easy operation without requiring sophisticated equipment and specialized skills. By testing thirty milk and shrimp tissue samples from the local market, the method was compared with the HPLC-MS / MS method, and there was no statistical difference between the two methods. Furthermore, the immunochromatographic assay had good specificity, simple procedure, and low cost. This protocol was well suited for the food safety monitoring and early warning.
Collapse
Affiliation(s)
- Zehua Xu
- Tianjin Institute of Environmental and Operational MedicineTianjinChina
| | - Tieqiang Sun
- Tianjin Institute of Environmental and Operational MedicineTianjinChina
| | - Hongwei He
- Tianjin Institute of Environmental and Operational MedicineTianjinChina
| | - Wentao Liu
- Tianjin Institute of Environmental and Operational MedicineTianjinChina
| | - Longxing Fan
- Tianjin Institute of Environmental and Operational MedicineTianjinChina
| | - Lingdi Zhao
- Tianjin Institute of Environmental and Operational MedicineTianjinChina
| | - Xinglin Wu
- Tianjin Institute of Environmental and Operational MedicineTianjinChina
| | - Zhenyu Han
- Tianjin Institute of Environmental and Operational MedicineTianjinChina
| | - Yingcun Zhang
- Tianjin Institute of Environmental and Operational MedicineTianjinChina
| | - Qiangqiang Wang
- Tianjin Institute of Environmental and Operational MedicineTianjinChina
| | - Baoan Ning
- Tianjin Institute of Environmental and Operational MedicineTianjinChina
| | - Zhixian Gao
- Tianjin Institute of Environmental and Operational MedicineTianjinChina
| |
Collapse
|
16
|
Zhao F, Hao Z, Zhong Y, Xu Y, Guo M, Zhang B, Yin X, Li Y, Zhou X. Discovery of breast cancer risk genes and establishment of a prediction model based on estrogen metabolism regulation. BMC Cancer 2021; 21:194. [PMID: 33632172 PMCID: PMC7905915 DOI: 10.1186/s12885-021-07896-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 02/09/2021] [Indexed: 11/18/2022] Open
Abstract
Background Multiple common variants identified by genome-wide association studies have shown limited evidence of the risk of breast cancer in Chinese individuals. In this study, we aimed to uncover the relationship between estrogen levels and the genetic polymorphism of estrogen metabolism-related enzymes in breast cancer (BC) and establish a risk prediction model composed of estrogen-metabolizing enzyme genes and GWAS-identified breast cancer-related genes based on a polygenic risk score. Methods Unrelated BC patients and healthy subjects were recruited for analysis of estrogen levels and single nucleotide polymorphisms (SNPs) in genes encoding estrogen metabolism-related enzymes. The polygenic risk score (PRS) was used to explore the combined effect of multiple genes, which was calculated using a Bayesian approach. An independent sample t-test was used to evaluate the differences between PRS scores of BC and healthy subjects. The discriminatory accuracy of the models was compared using the area under the receiver operating characteristic (ROC) curve. Results The estrogen homeostasis profile was disturbed in BC patients, with parent estrogens (E1, E2) and carcinogenic catechol estrogens (2/4-OHE1, 2-OHE2, 4-OHE2) significantly accumulating in the serum of BC patients. We then established a PRS model to evaluate the role of SNPs in multiple genes. PRS model 1 (M1) was established from SNPs in 6 GWAS-identified high risk genes. On the basis of M1, we added SNPs from 7 estrogen metabolism enzyme genes to establish PRS model 2 (M2). The independent sample t-test results showed that there was no difference between BC and healthy subjects in M1 (P = 0.17); however, there was a significant difference between BC and healthy subjects in M2 (P = 4.9*10− 5). The ROC curve results showed that the accuracy of M2 (AUC = 62.18%) in breast cancer risk identification was better than that of M1 (AUC = 54.56%). Conclusion Estrogen and related metabolic enzyme gene polymorphisms are closely related to BC. The model constructed by adding estrogen metabolic enzyme gene SNPs has a good predictive ability for breast cancer risk, and the accuracy is greatly improved compared with that of the PRS model that only includes GWAS-identified gene SNPs.
Collapse
Affiliation(s)
- Feng Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, College of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China.,Department of Pharmacy, The First People's Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, China
| | - Zhixiang Hao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, College of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Yanan Zhong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, College of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Yinxue Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, College of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Meng Guo
- Department of Thyroid and Breast Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Bei Zhang
- Department of Obstetrics and Gynecology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, College of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Ying Li
- Department of Thyroid and Breast Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xueyan Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, College of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China.
| |
Collapse
|
17
|
Markers of Local and Systemic Estrogen Metabolism in Endometriosis. Reprod Sci 2020; 28:1001-1011. [PMID: 33216295 DOI: 10.1007/s43032-020-00383-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 11/01/2020] [Indexed: 12/21/2022]
Abstract
Estrogen metabolites (EMs) can work independently from their parent hormones. We hypothesize that in endometriosis, estrogen is metabolized preferentially along hormonally active pathways. We recruited 62 women with endometriosis (proven laparoscopically and histologically) and 52 control women (normal findings with laparoscopy) among patients undergoing surgery for pelvic pain and/or infertility during the proliferative phase of the menstrual cycle. Urinary samples were collected preoperatively. Biopsies from eutopic endometrium of control women and women with endometriosis were collected during surgery. EMs in urine and endometrial tissues were extracted and determined using Liquid Chromatography-Electrospray Ionization Tandem Mass Spectrometry (LC-ESI-MS/MS). These included: 2-hydroxyestrone (2OHE1), 16-α hydroxyestrone (16α-OHE1), 2OHE1/16α-OHE1 ratio, 4-hydroxyestrone (4OHE1), 2-hydroxyestradiol (2OHE2), and 4-hydroxyestradiol (4OHE2). Eutopic endometrium of endometriosis patients, as compared to control endometrium, contained significantly higher level of 4OHE1 (0.03 (IQR: 0.03-0.265) versus 0.03 (IQR: 0.03-0.03) μg/g, respectively, P = 0.005), 2-OHE2 (0.241 (IQR: 0.1-0.960) versus 0.1 (IQR: 0.1-0.1) μg/g, respectively, P < 0.001), and 4-OHE2 (0.225 (IQR: 0.22-1.29) versus 0.0.2 (IQR: 0.2-0.2) μg/g, respectively, P < 0.001). Only 2OHE1 showed higher concentration in urine of women with endometriosis than controls (9.9 (IQR: 3.64-14.88) versus 4.5 (IQR: 1.37-17.00) μg/mg creatinine, respectively, P = 0.042). Eutopic endometrium of women with endometriosis metabolizes estrogen preferentially to the biologically active 2OHE2, and potentially genotoxic 4OHE1 and 4OHE2 metabolites. This contributes to further understanding of endometriosis etiology, its link to ovarian cancer, and could help identifying an endometrial biomarker of the disease.
Collapse
|
18
|
Houghton LC, Howland RE, Wei Y, Ma X, Kehm RD, Chung WK, Genkinger JM, Santella RM, Hartmann MF, Wudy SA, Terry MB. The Steroid Metabolome and Breast Cancer Risk in Women with a Family History of Breast Cancer: The Novel Role of Adrenal Androgens and Glucocorticoids. Cancer Epidemiol Biomarkers Prev 2020; 30:89-96. [PMID: 32998947 DOI: 10.1158/1055-9965.epi-20-0471] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/09/2020] [Accepted: 09/26/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND No study has comprehensively examined how the steroid metabolome is associated with breast cancer risk in women with familial risk. METHODS We examined 36 steroid metabolites across the spectrum of familial risk (5-year risk ranged from 0.14% to 23.8%) in pre- and postmenopausal women participating in the New York site of the Breast Cancer Family Registry (BCFR). We conducted a nested case-control study with 62 cases/124 controls individually matched on menopausal status, age, and race. We measured metabolites using GC-MS in urine samples collected at baseline before the onset of prospectively ascertained cases. We used conditional logistic regression to estimate odds ratios (OR) and 95% confidence intervals (CI) per doubling in hormone levels. RESULTS The average proportion of total steroid metabolites in the study sample were glucocorticoids (61%), androgens (26%), progestogens (11%), and estrogens (2%). A doubling in glucocorticoids (aOR = 2.7; 95% CI = 1.3-5.3) and androgens (aOR = 1.6; 95% CI = 1.0-2.7) was associated with increased breast cancer risk. Specific glucocorticoids (THE, THF αTHF, 6β-OH-F, THA, and α-THB) were associated with 49% to 161% increased risk. Two androgen metabolites (AN and 11-OH-AN) were associated with 70% (aOR = 1.7; 95% CI = 1.1-2.7) and 90% (aOR = 1.9; 95% CI = 1.2-3.1) increased risk, respectively. One intermediate metabolite of a cortisol precursor (THS) was associated with 65% (OR = 1.65; 95% CI = 1.0-2.7) increased risk. E1 and E2 estrogens were associated with 20% and 27% decreased risk, respectively. CONCLUSIONS Results suggest that glucocorticoids and 11-oxygenated androgens are positively associated with breast cancer risk across the familial risk spectrum. IMPACT If replicated, our findings suggest great potential of including steroids into existing breast cancer risk assessment tools.
Collapse
Affiliation(s)
- Lauren C Houghton
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York. .,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Renata E Howland
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York
| | - Ying Wei
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, New York
| | - Xinran Ma
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York
| | - Rebecca D Kehm
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York
| | - Wendy K Chung
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York.,Departments of Pediatrics and Medicine, Columbia University Medical Center, New York, New York
| | - Jeanine M Genkinger
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Regina M Santella
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York.,Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, New York
| | - Michaela F Hartmann
- Steroid Research and Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics in Pediatric Endocrinology, Division of Pediatric Endocrinology and Diabetology, Justus Liebig University, Giessen, Germany
| | - Stefan A Wudy
- Steroid Research and Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics in Pediatric Endocrinology, Division of Pediatric Endocrinology and Diabetology, Justus Liebig University, Giessen, Germany
| | - Mary Beth Terry
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| |
Collapse
|
19
|
Darville LNF, Cline JK, Rozmeski C, Martinez YC, Rich S, Eschrich SA, Egan KM, Yaghjyan L, Koomen JM. LC-HRMS of derivatized urinary estrogens and estrogen metabolites in postmenopausal women. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1154:122288. [PMID: 32769047 DOI: 10.1016/j.jchromb.2020.122288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 07/01/2020] [Accepted: 07/25/2020] [Indexed: 12/21/2022]
Abstract
In order to undertake an epidemiologic study relating levels of parent estrogens (estrone and estradiol) and estrogen metabolites (EMs) to other breast cancer risk factors, we have optimized methods for EM quantification with ultra high performance liquid chromatography-high resolution mass spectrometry (UHPLC-HRMS). A two-step approach was adopted; the first step comprised method development and evaluation of the method performance. The second step consisted of applying this method to quantify estrogens in postmenopausal women and determine if the observed patterns are consistent with the existing literature and prior knowledge of estrogen metabolism. First, 1-methylimidazole-2-sulfonyl chloride (MIS) was used to derivatize endogenous estrogens and estrogen metabolites in urine from study participants. Since C18 reversed phase columns have not been able to separate all the structurally related EMs, we used a C18-pentafluorophenyl (PFP) column. The parent estrogens and EMs were baseline resolved with distinct retention times on this C18-PFP column using a 30 min gradient. This method was used to quantify the parent estrogens and 13 EMs in urine samples collected in an initial pilot study involving males as well as pre- and peri-menopausal females to assess a range of EM levels in urine samples and enable comparison to the previous literature for assay evaluation. Detection limits ranged from 1 - 20 pg/mL depending on the EM. We evaluated matrix effects and interference as well as the intra- and inter-batch reproducibility including hydrolysis, extraction, derivatization and LC-MS analysis using charcoal-stripped human urine as a matrix. Methods were then applied to the measurement of estrogens in urine samples from 169 postmenopausal women enrolled in an epidemiological study to examine relationships between breast cancer risk, the intestinal microbiome, and urinary EMs. The results from our cohort are comparable to previous reports on urinary EMs in postmenopausal women and enabled thorough evaluation of the method.
Collapse
Affiliation(s)
- Lancia N F Darville
- H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States.
| | - Jayden K Cline
- H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Carrie Rozmeski
- H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Yessica C Martinez
- H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Shannan Rich
- University of Florida, Gainesville, FL, United States
| | - Steven A Eschrich
- H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Kathleen M Egan
- H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States.
| | | | - John M Koomen
- H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| |
Collapse
|
20
|
Zhao F, Wang X, Wang Y, Zhang J, Lai R, Zhang B, Zhou X. The function of uterine UDP-glucuronosyltransferase 1A8 (UGT1A8) and UDP-glucuronosyltransferase 2B7 (UGT2B7) is involved in endometrial cancer based on estrogen metabolism regulation. Hormones (Athens) 2020; 19:403-412. [PMID: 32592099 DOI: 10.1007/s42000-020-00213-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 05/20/2020] [Indexed: 01/03/2023]
Abstract
BACKGROUND The progression of endometrial cancer (EC) is closely related to estrogen levels. UDP-glucuronosyltransferases (UGTs) are an essential class of phase II metabolizing enzymes that play a pivotal role in detoxifying steroid hormone. PURPOSE In this study, we aimed to uncover the role of UGTs in estrogen metabolism and the pathogenesis of EC. METHODS A total of 100 unrelated EC patients (mean age 52.15 ± 10.04 y) and 100 healthy subjects (mean age 50.26 ± 8.80 y) were recruited for analysis of the UGT gene polymorphism and estrogen level. In six cases of EC, EC-adjacent tissues and cancer tissues were collected for detection of UGT expression. RESULTS Our results showed that the estrogen homeostasis profile was disturbed in EC patients, with carcinogenic catechol estrogens (4-OHE1, 2-OHE1, 2-OHE2) significantly accumulated in the serum of these patients. Also, levels of estrogen-glucuronides were decreased significantly, and the expression of UGT1A8 and UGT2B7 in uterine tissues was downregulated in EC patients. Consistent with this, we observed that the distribution of genotypes and allele frequencies in UGT1A8 rs1042597 and UGT2B7 rs7439366 was significantly different between EC patients and healthy volunteers. CONCLUSION These results indicated that UGT1A8 and UGT2B7 may contribute to the estrogen signaling pathway and the pathogenesis of EC.
Collapse
Affiliation(s)
- Feng Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Xi Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Yan Wang
- Department of Pharmacy, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jingbo Zhang
- Department of Obstetrics and Gynecology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, China
| | - Ran Lai
- Department of Pharmacy, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Bei Zhang
- Department of Obstetrics and Gynecology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, China
| | - Xueyan Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China.
| |
Collapse
|
21
|
Abstract
Aromatase CYP19A1 catalyzes the synthesis of estrogens in endocrine, reproductive and central nervous systems. Higher levels of 17β-estradiol (E2) are associated with malignancies and diseases of the breast, ovary and endometrium, while low E2 levels increase the risk for osteoporosis, cardiovascular diseases and cognitive disorders. E2, the transcriptional activator of the estrogen receptors, is also known to be involved in non-genomic signaling as a neurotransmitter/neuromodulator, with recent evidence for rapid estrogen synthesis (RES) within the synaptic terminal. Although regulation of brain aromatase activity by phosphorylation/dephosphorylation has been suggested, it remains obscure in the endocrine and reproductive systems. RES and overabundance of estrogens could stimulate the genomic and non-genomic signaling pathways, and genotoxic effects of estrogen metabolites. Here, by utilizing biochemical, cellular, mass spectrometric, and structural data we unequivocally demonstrate phosphorylation of human placental aromatase and regulation of its activity. We report that human aromatase has multiple phosphorylation sites, some of which are consistently detectable. Phosphorylation of the residue Y361 at the reductase-coupling interface significantly elevates aromatase activity. Other sites include the active site residue S478 and several at the membrane interface. We present the evidence that two histidine residues are phosphorylated. Furthermore, oxidation of two proline residues near the active site may have implications in regulation. Taken together, the results demonstrate that aromatase activity is regulated by phosphorylation and possibly other post-translational modifications. Protein level regulation of aromatase activity not only represents a paradigm shift in estrogen-mediated biology, it could also explain unresolved clinical questions such as aromatase inhibitor resistance.
Collapse
|
22
|
Wang T, Nichols HB, Nyante SJ, Bradshaw PT, Moorman PG, Kabat GC, Parada H, Khankari NK, Teitelbaum SL, Terry MB, Santella RM, Neugut AI, Gammon MD. Urinary Estrogen Metabolites and Long-Term Mortality Following Breast Cancer. JNCI Cancer Spectr 2020; 4:pkaa014. [PMID: 32455334 PMCID: PMC7236781 DOI: 10.1093/jncics/pkaa014] [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: 09/18/2019] [Revised: 12/11/2019] [Accepted: 02/26/2020] [Indexed: 12/09/2022] Open
Abstract
Background Estrogen metabolite concentrations of 2-hydroxyestrone (2-OHE1) and 16-hydroxyestrone (16-OHE1) may be associated with breast carcinogenesis. However, no study has investigated their possible impact on mortality after breast cancer. Methods This population-based study was initiated in 1996–1997 with spot urine samples obtained shortly after diagnosis (mean = 96 days) from 683 women newly diagnosed with first primary breast cancer and 434 age-matched women without breast cancer. We measured urinary concentrations of 2-OHE1 and 16-OHE1 using an enzyme-linked immunoassay. Vital status was determined via the National Death Index (n = 244 deaths after a median of 17.7 years of follow-up). We used multivariable-adjusted Cox proportional hazards to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for the estrogen metabolites-mortality association. We evaluated effect modification using likelihood ratio tests. All statistical tests were two-sided. Results Urinary concentrations of the 2-OHE1 to 16-OHE1 ratio (>median of 1.8 vs ≤median) were inversely associated with all-cause mortality (HR = 0.74, 95% CI = 0.56 to 0.98) among women with breast cancer. Reduced hazard was also observed for breast cancer mortality (HR = 0.73, 95% CI = 0.45 to 1.17) and cardiovascular diseases mortality (HR = 0.76, 95% CI = 0.47 to 1.23), although the 95% confidence intervals included the null. Similar findings were also observed for women without breast cancer. The association with all-cause mortality was more pronounced among breast cancer participants who began chemotherapy before urine collection (n = 118, HR = 0.42, 95% CI = 0.22 to 0.81) than among those who had not (n = 559, HR = 0.98, 95% CI = 0.72 to 1.34; Pinteraction = .008). Conclusions The urinary 2-OHE1 to 16-OHE1 ratio may be inversely associated with long-term all-cause mortality, which may depend on cancer treatment status at the time of urine collection.
Collapse
Affiliation(s)
- Tengteng Wang
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Hazel B Nichols
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Sarah J Nyante
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA.,Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | | | - Patricia G Moorman
- Department of Community and Family Medicine, Duke University, Durham, NC, USA
| | | | - Humberto Parada
- Division of Epidemiology and Biostatistics, San Diego State University, San Diego, CA, USA
| | - Nikhil K Khankari
- Division of Epidemiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Susan L Teitelbaum
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary Beth Terry
- Department of Epidemiology, Columbia University, New York, NY, USA
| | - Regina M Santella
- Department of Environmental Health Sciences, Columbia University, New York, NY, USA
| | - Alfred I Neugut
- Department of Epidemiology, Columbia University, New York, NY, USA.,Department of Medicine, Columbia University, New York, NY, USA
| | - Marilie D Gammon
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
23
|
Trabert B, Bauer DC, Buist DSM, Cauley JA, Falk RT, Geczik AM, Gierach GL, Hada M, Hue TF, Lacey JV, LaCroix AZ, Tice JA, Xu X, Dallal CM, Brinton LA. Association of Circulating Progesterone With Breast Cancer Risk Among Postmenopausal Women. JAMA Netw Open 2020; 3:e203645. [PMID: 32329771 PMCID: PMC7182797 DOI: 10.1001/jamanetworkopen.2020.3645] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
IMPORTANCE The role of endogenous progesterone in the development of breast cancer remains largely unexplored to date, primarily owing to assay sensitivity limitations and low progesterone concentrations in postmenopausal women. Recently identified progesterone metabolites may provide insights as experimental data suggest that 5α-dihydroprogesterone (5αP) concentrations reflect cancer-promoting properties and 3α-dihydroprogesterone (3αHP) concentrations reflect cancer-inhibiting properties. OBJECTIVE To evaluate the association between circulating progesterone and progesterone metabolite levels and breast cancer risk. DESIGN, SETTING, AND PARTICIPANTS Using a sensitive liquid chromatography-tandem mass spectrometry assay, prediagnostic serum levels of progesterone and progesterone metabolites were quantified in a case-cohort study nested within the Breast and Bone Follow-up to the Fracture Intervention Trial (n = 15 595). Participation was limited to women not receiving exogenous hormone therapy at the time of blood sampling (1992-1993). Incident breast cancer cases (n = 405) were diagnosed during 12 follow-up years and a subcohort of 495 postmenopausal women were randomly selected within 10-year age and clinical center strata. Progesterone assays were completed in July 2017; subsequent data analyses were conducted between July 15, 2017, and December 20, 2018. EXPOSURES Circulating concentrations of pregnenolone, progesterone, and their major metabolites. MAIN OUTCOMES AND MEASURES Development of breast cancer, with hazard ratios (HRs) and 95% CIs was estimated using Cox proportional hazards regression adjusted for key confounders, including estradiol. Evaluation of hormone ratios and effect modification were planned a priori. RESULTS The present study included 405 incident breast cancer cases and a subcohort of 495 postmenopausal women; the mean (SD) age at the time of the blood draw was 67.2 (6.2) years. Progesterone concentrations were a mean (SD) of 4.6 (1.7) ng/dL. Women with higher circulating progesterone levels were at an increased risk for breast cancer per SD increase in progesterone levels (HR, 1.16; 95% CI, 1.00-1.35; P = .048). The association with progesterone was linear in a 5-knot spline and stronger for invasive breast cancers (n = 267) (HR, 1.24; 95% CI, 1.07-1.43; P = .004). Among women in the lowest quintile (Q1) of circulating estradiol (<6.30 pg/mL) elevated progesterone concentrations were associated with reduced breast cancer risk per SD increase in progesterone levels (HR, 0.38; 95% CI, 0.15-0.95; P = .04) and increased risk among women in higher quintiles of estradiol (Q2-Q5; ≥6.30 pg/mL) (HR, 1.18; 95% CI, 1.04-1.35; P = .01; P = .04 for interaction). CONCLUSIONS AND RELEVANCE In this case-cohort study of postmenopausal women, elevated circulating progesterone levels were associated with a 16% increase in the risk of breast cancer. Additional research should be undertaken to assess how postmenopausal breast cancer risk is associated with both endogenous progesterone and progesterone metabolites and their interactions with estradiol.
Collapse
Affiliation(s)
- Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Doug C. Bauer
- Department of Medicine and Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Diana S. M. Buist
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington
| | - Jane A. Cauley
- Graduate School of Public Health Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Roni T. Falk
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Ashley M. Geczik
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Gretchen L. Gierach
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Manila Hada
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Trisha F. Hue
- Department of Medicine and Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - James V. Lacey
- Division of Cancer Etiology, Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, California
| | - Andrea Z. LaCroix
- Division of Epidemiology, Department of Family and Preventive Medicine, University of California, San Diego
| | - Jeffrey A. Tice
- Department of Medicine and Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Xia Xu
- Leidos Biomedical Research Inc, Frederick, Maryland
| | - Cher M. Dallal
- School of Public Health, University of Maryland College Park
| | - Louise A. Brinton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
24
|
Assessing Endogenous and Exogenous Hormone Exposures and Breast Development in a Migrant Study of Bangladeshi and British Girls. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17041185. [PMID: 32069802 PMCID: PMC7068451 DOI: 10.3390/ijerph17041185] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/17/2020] [Accepted: 02/08/2020] [Indexed: 11/16/2022]
Abstract
Timing of breast development (or thelarche) and its endogenous and exogenous determinants may underlie global variation in breast cancer incidence. The study objectives were to characterize endogenous estrogen levels and bisphenol A (BPA) exposure using a migrant study of adolescent girls and test whether concentrations explained differences in thelarche by birthplace and growth environment. Estrogen metabolites (EM) and BPA-glucuronide (BPA-G) were quantified in urine spot samples using liquid chromatography tandem mass spectrometry (LC-MS/MS) from a cross-sectional study of Bangladeshi, first- and second-generation Bangladeshi migrants to the UK, and white British girls aged 5–16 years (n = 348). Thelarche status at the time of interview was self-reported and defined equivalent to Tanner Stage ≥2. We compared geometric means (and 95% confidence interval (CIs)) of EM and BPA-G using linear regression and assessed whether EM and BPA-G explained any of the association between exposure to the UK and the age at thelarche using hazard ratios and 95% confidence intervals. Average EM decreased with exposure to the UK, whereas BPA-G increased and was significantly higher among white British (0.007 ng/mL, 95% CI: 0.0024–0.0217) and second-generation British-Bangladeshi girls (0.009 ng/mL, 95% CI: 0.0040–0.0187) compared to Bangladeshi girls (0.002 ng/mL, 95% CI: 0.0018–0.0034). Two of four EM ratios (16-pathway/parent and parent/all pathways) were significantly associated with thelarche. The relationship between exposure to the UK and thelarche did not change appreciably after adding EM and BPA-G to the models. While BPA-G is often considered a ubiquitous exposure, our findings suggest it can vary based on birthplace and growth environment, with increasing levels for girls who were born in or moved to the UK. Our study did not provide statistically significant evidence that BPA-G or EM concentrations explained earlier thelarche among girls who were born or raised in the UK.
Collapse
|
25
|
van der Berg C, Venter G, van der Westhuizen FH, Erasmus E. Development and validation of LC-ESI-MS/MS methods for quantification of 27 free and conjugated estrogen-related metabolites. Anal Biochem 2019; 590:113531. [PMID: 31805274 DOI: 10.1016/j.ab.2019.113531] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/27/2019] [Accepted: 11/30/2019] [Indexed: 10/25/2022]
Abstract
An imbalance in the estrogen metabolism has been associated with an increased risk of breast cancer development. Evaluation of the estrogen biotransformation capacity requires monitoring of various estrogen metabolites. Up to now, only some estrogen metabolites could be measured in urine. However, in order to offer tailor made nutritional support or therapies, a complete estrogen metabolite profile is required in order to identify specific deficiencies in this pathway for each patient individually. Here, we focused on this need to quantify as many as possible of the estrogen-related metabolites excreted in urine. The method was developed to quantify 27 estrogen-related metabolites in small urine quantities. This entailed sample clean-up with a multi-step solid phase extraction procedure, derivatisation of the metabolites in the less water-soluble fraction through dansylation, and analyses using liquid chromatography-tandem mass spectrometry (LC-MS/MS). The metabolites accurately quantified by the method devised included parent estrogens, hydroxylated and methylated forms, metabolites of the 16α-hydroxyestrogen pathway, sulphate and glucuronide conjugated forms, precursors and a related steroid hormone. This method was validated and enabled quantification in the high picograms and low nanograms per millilitre range. Finally, analyses of urine samples confirmed detection and quantification of each of the metabolites.
Collapse
Affiliation(s)
- Carien van der Berg
- Human Metabolomics, North-West University (Potchefstroom Campus), Potchefstroom, 2531, South Africa.
| | - Gerda Venter
- Human Metabolomics, North-West University (Potchefstroom Campus), Potchefstroom, 2531, South Africa
| | | | - Elardus Erasmus
- Human Metabolomics, North-West University (Potchefstroom Campus), Potchefstroom, 2531, South Africa.
| |
Collapse
|
26
|
Bozzolino C, Vaglio S, Amante E, Alladio E, Gerace E, Salomone A, Vincenti M. Individual and cyclic estrogenic profile in women: Structure and variability of the data. Steroids 2019; 150:108432. [PMID: 31279660 DOI: 10.1016/j.steroids.2019.108432] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 06/11/2019] [Accepted: 06/18/2019] [Indexed: 11/24/2022]
Abstract
The concentration of estrogens in the body fluids of women is highly variable, due to the menstrual cycle, circadian oscillations, and other physiological and pathological causes. To date, only the cyclic fluctuations of the principal estrogens (estradiol and estrone) have been studied, with limited outcome of general significance. Aim of the present study was to examine in detail the cyclic variability of a wide estrogens' panel and to interpret it by multivariate statistics. Four estrogens (17α-estradiol, 17β-estradiol, estrone, estriol) and eleven of their metabolites (4-methoxyestrone, 2-methoxyestrone, 16α-hydroxyestrone, 4-hydroxyestrone, 2-hydroxyestrone, 4-methoxyestradiol, 2-methoxyestradiol, 4-hydroxyestradiol, 2-hydroxyestradiol, estriol, 16-epiestriol, and 17-epiestriol) were determined in urine by a gas chromatography - mass spectrometry method, which was developed by design of experiments and fully validated according to ISO 17025 requirements. Then, urine samples collected every morning for a complete menstrual cycle from 9 female volunteers aged 24-35 years (1 parous) were analysed. The resulting three-dimensional data (subjects × days × estrogens) were interpreted using several statistical tools. Parallel Factor Analysis compared the estrogen profiles in order to explore the cyclic and inter-individual variability of each analyte. Principal Component Analysis (PCA) provided clear separation of the sampling days along the cycle, allowing discrimination among the luteal, ovulation, and follicular phases. The scores obtained from PCA were used to build a Linear Discriminant Analysis classification model which enhanced the recognition of the three cycle's phases, yielding an overall classification non-error rate equal to 90%. These statistical models may find prospective application in fertility studies and the investigation of endocrinology disorders and other hormone-dependent diseases.
Collapse
Affiliation(s)
- Cristina Bozzolino
- Dipartimento di Chimica, Università degli Studi di Torino, via P. Giuria 7, 10125 Torino, Italy
| | - Sara Vaglio
- Dipartimento di Chimica, Università degli Studi di Torino, via P. Giuria 7, 10125 Torino, Italy
| | - Eleonora Amante
- Dipartimento di Chimica, Università degli Studi di Torino, via P. Giuria 7, 10125 Torino, Italy; Centro Regionale Antidoping e di Tossicologia "A. Bertinaria", regione Gonzole 10/1, 10043 Orbassano, TO, Italy
| | - Eugenio Alladio
- Dipartimento di Chimica, Università degli Studi di Torino, via P. Giuria 7, 10125 Torino, Italy; Centro Regionale Antidoping e di Tossicologia "A. Bertinaria", regione Gonzole 10/1, 10043 Orbassano, TO, Italy.
| | - Enrico Gerace
- Centro Regionale Antidoping e di Tossicologia "A. Bertinaria", regione Gonzole 10/1, 10043 Orbassano, TO, Italy
| | - Alberto Salomone
- Dipartimento di Chimica, Università degli Studi di Torino, via P. Giuria 7, 10125 Torino, Italy; Centro Regionale Antidoping e di Tossicologia "A. Bertinaria", regione Gonzole 10/1, 10043 Orbassano, TO, Italy
| | - Marco Vincenti
- Dipartimento di Chimica, Università degli Studi di Torino, via P. Giuria 7, 10125 Torino, Italy; Centro Regionale Antidoping e di Tossicologia "A. Bertinaria", regione Gonzole 10/1, 10043 Orbassano, TO, Italy
| |
Collapse
|
27
|
Miao S, Yang F, Wang Y, Shao C, Zava DT, Ding Q, Shi YE. 4-Hydroxy estrogen metabolite, causing genomic instability by attenuating the function of spindle-assembly checkpoint, can serve as a biomarker for breast cancer. Am J Transl Res 2019; 11:4992-5007. [PMID: 31497216 PMCID: PMC6731443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/17/2019] [Indexed: 06/10/2023]
Abstract
Sex hormone metabolism is altered during mammary gland tumorigenesis, and different metabolites may have different effects on mammary epithelial cells. This study aimed to evaluate associations between urinary sexual metabolite levels and breast cancer risk among premenopausal women of Mainland China. The molecular metabolism of the cancer-related metabolites was also explored based on the clinical data. The sex hormone metabolites in the urine samples of patients with breast cancer versus normal healthy women were analyzed comprehensively. Among many alterations of sex hormone metabolisms, 4-hydroxy estrogen (4-OH-E) metabolite was found to be significantly increased in the urine samples of patients with breast cancer compared with the normal healthy controls. This was the most important risk factor for breast cancer. Several experiments were conducted in vitro and in vivo to probe this mechanism. 4-Hydroxyestradiol (4-OH-E2) was found to induce malignant transformation of breast cells and tumorigenesis in nude mice. At the molecular level, 4-OH-E2 compromised the function of spindle-assembly checkpoint and rendered resistance to the anti-microtubule drug. Further, transgenic mice with high expression of CYP1B1, a key enzyme of 4-hydroxy metabolites, were established and stimulated with estrogen. Cancerous tissue was found to appear in the mammary gland of transgenic mice.
Collapse
Affiliation(s)
- Suyu Miao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210000, China
- Jiangsu Breast Disease Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210000, China
| | - Fengming Yang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210000, China
| | - Ying Wang
- Jiangsu Breast Disease Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210000, China
| | - Chuchu Shao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210000, China
| | | | - Qiang Ding
- Jiangsu Breast Disease Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210000, China
| | - Yuenian Eric Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210000, China
| |
Collapse
|
28
|
Pu H, Huang Z, Sun DW, Fu H. Recent advances in the detection of 17β-estradiol in food matrices: A review. Crit Rev Food Sci Nutr 2019; 59:2144-2157. [PMID: 31084362 DOI: 10.1080/10408398.2019.1611539] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pollution of endocrine disrupting chemicals has become a global issue. As one of the hormonally active compounds, 17β-estradiol produces the strongest estrogenic effect when it enters the organism exogenously including food intakes, bringing potential harmfulness such as malfunction of the endocrine system. Therefore, in order to assure food safety and avoid potential risks of 17β-estradiol to humans, it is of great significance to develop rapid, sensitive and selective approaches for the detection of 17β-estradiol in food matrices. In this review, the harmfulness and main sources of 17β-estradiol are firstly introduced, followed by the description of the principles and applications of different approaches for 17β-estradiol detection including high performance liquid chromatography, electrochemistry, Raman spectroscopy, fluorescence and colorimetry. Particularly, applications in detecting 17β-estradiol in food matrices over the years of 2010-2018 are discussed. Finally, advantages and limitations of these detection methods are highlighted and perspectives on future developments in the detection methods for 17β-estradiol are also proposed. Although many detection approaches can achieve trace or ultratrace detection of 17β-estradiol, further studies should be focused on the development of in-situ and real-time methods to monitor and evaluate 17β-estradiol for food safety.
Collapse
Affiliation(s)
- Hongbin Pu
- a School of Food Science and Engineering , South China University of Technology , Guangzhou , China.,b Academy of Contemporary Food Engineering , South China University of Technology, Guangzhou Higher Education Mega Center , Guangzhou , China.,c Engineering and Technological Research Centre of Guangdong Province on Intelligent Sensing and Process Control of Cold Chain Foods , Guangzhou Higher Education Mega Center , Guangzhou , China
| | - Zhibin Huang
- a School of Food Science and Engineering , South China University of Technology , Guangzhou , China.,b Academy of Contemporary Food Engineering , South China University of Technology, Guangzhou Higher Education Mega Center , Guangzhou , China.,c Engineering and Technological Research Centre of Guangdong Province on Intelligent Sensing and Process Control of Cold Chain Foods , Guangzhou Higher Education Mega Center , Guangzhou , China
| | - Da-Wen Sun
- a School of Food Science and Engineering , South China University of Technology , Guangzhou , China.,b Academy of Contemporary Food Engineering , South China University of Technology, Guangzhou Higher Education Mega Center , Guangzhou , China.,c Engineering and Technological Research Centre of Guangdong Province on Intelligent Sensing and Process Control of Cold Chain Foods , Guangzhou Higher Education Mega Center , Guangzhou , China.,d Food Refrigeration and Computerized Food Technology (FRCFT), Agriculture and Food Science Centre , University College Dublin, National University of Ireland , Belfield , Dublin 4 , Ireland
| | - Haohua Fu
- e Tang Renshen Group Co., Ltd , Zhuzhou , China
| |
Collapse
|
29
|
Transition metal complexes based aptamers as optical diagnostic tools for disease proteins and biomolecules. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2018.09.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
30
|
Zhu J, Liao M, Yao Z, Liang W, Li Q, Liu J, Yang H, Ji Y, Wei W, Tan A, Liang S, Chen Y, Lin H, Zhu X, Huang S, Tian J, Tang R, Wang Q, Mo Z. Breast cancer in postmenopausal women is associated with an altered gut metagenome. MICROBIOME 2018; 6:136. [PMID: 30081953 PMCID: PMC6080540 DOI: 10.1186/s40168-018-0515-3] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 07/10/2018] [Indexed: 05/12/2023]
Abstract
BACKGROUND Increasing evidence suggests that gut microbiota play a role in the pathogenesis of breast cancer. The composition and functional capacity of gut microbiota associated with breast cancer have not been studied systematically. METHODS We performed a comprehensive shotgun metagenomic analysis of 18 premenopausal breast cancer patients, 25 premenopausal healthy controls, 44 postmenopausal breast cancer patients, and 46 postmenopausal healthy controls. RESULTS Microbial diversity was higher in breast cancer patients than in controls. Relative species abundance in gut microbiota did not differ significantly between premenopausal breast cancer patients and premenopausal controls. In contrast, relative abundance of 45 species differed significantly between postmenopausal patients and postmenopausal controls: 38 species were enriched in postmenopausal patients, including Escherichia coli, Klebsiella sp_1_1_55, Prevotella amnii, Enterococcus gallinarum, Actinomyces sp. HPA0247, Shewanella putrefaciens, and Erwinia amylovora, and 7 species were less abundant in postmenopausal patients, including Eubacterium eligens and Lactobacillus vaginalis. Acinetobacter radioresistens and Enterococcus gallinarum were positively but weakly associated with expression of high-sensitivity C-reactive protein; Shewanella putrefaciens and Erwinia amylovora were positively but weakly associated with estradiol levels. Actinomyces sp. HPA0247 negatively but weakly correlated with CD3+CD8+ T cell numbers. Further characterization of metagenome functional capacity indicated that the gut metagenomes of postmenopausal breast cancer patients were enriched in genes encoding lipopolysaccharide biosynthesis, iron complex transport system, PTS system, secretion system, and beta-oxidation. CONCLUSION The composition and functions of the gut microbial community differ between postmenopausal breast cancer patients and healthy controls. The gut microbiota may regulate or respond to host immunity and metabolic balance. Thus, while cause and effect cannot be determined, there is a reproducible change in the microbiota of treatment-naive patients relative to matched controls.
Collapse
Affiliation(s)
- Jia Zhu
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Department of Breast Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Ming Liao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Ziting Yao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Wenying Liang
- Clabee Genomics, Urban Garden Building, Bookstore Road, Luohu District, Shenzhen, 518000, Guangdong, China
| | - Qibin Li
- Clabee Genomics, Urban Garden Building, Bookstore Road, Luohu District, Shenzhen, 518000, Guangdong, China
| | - Jianlun Liu
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Huawei Yang
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yinan Ji
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Wei Wei
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Aihua Tan
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Department of Chemotherapy, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Siyuan Liang
- Department of Colorectal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yang Chen
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Haisong Lin
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiujuan Zhu
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Shengzhu Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jiarong Tian
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Ruiqiang Tang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Qiuyan Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Zengnan Mo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
31
|
Smy L, Straseski JA. Measuring estrogens in women, men, and children: Recent advances 2012-2017. Clin Biochem 2018; 62:11-23. [PMID: 29800559 DOI: 10.1016/j.clinbiochem.2018.05.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 05/22/2018] [Indexed: 12/11/2022]
Abstract
The measurement of estrogens is important for diagnosing and monitoring the health of women, men, and children. For example, for postmenopausal women or women undergoing treatment for breast cancer with aromatase inhibitors, the measurement of extremely low concentrations of estrogens in serum, especially estradiol, is problematic but essential for proper medical care. Achieving superb analytical sensitivity and specificity has been and continues to be a challenge for the clinical laboratory, but is a challenge that is being taken seriously. Focusing on publications from 2012 to 2017, this review will provide an overview of recent research in the development of methods to accurately and precisely measure estrogens, including a variety of estrogen metabolites. Additionally, the latest in clinical research involving estrogen measurement in women, men, and children will be presented to provide an update on the association of estrogens with diseases or conditions such as breast cancer, precocious puberty, infertility, and pregnancy. This research update will provide context as to why estrogen measurement is important and why laboratories are working hard to support the recommendations made by the Endocrine Society regarding estrogen measurement.
Collapse
Affiliation(s)
- Laura Smy
- Department of Pathology, University of Utah Health Sciences Center, Salt Lake City, UT 84108, USA
| | - Joely A Straseski
- Department of Pathology, University of Utah Health Sciences Center, Salt Lake City, UT 84108, USA.
| |
Collapse
|
32
|
Aigaje Espinosa EK, Qiu J, Jarošová R, Castiaux A, Swain GM. HPLC−EC Analysis of Estrogenic Compounds: A Comparison of Diamond and Tetrahedral Amorphous Carbon Electrode Performance. ELECTROANAL 2018. [DOI: 10.1002/elan.201800261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Elizabeth K. Aigaje Espinosa
- Faculty of Chemical Engineering and Agro-Industry; Ladrón de Guevara E11-253, Escuela Politécnica Nacional Quito 170525 Ecuador
- Department of Chemistry; Michigan State University; East Lansing, MI 48824 USA
| | - Joy Qiu
- High School Honors Science Program (HSHSP); Michigan State University; East Lansing, MI 48824 USA
| | - Romana Jarošová
- Department of Chemistry; Michigan State University; East Lansing, MI 48824 USA
- Charles University; Department of Analytical Chemistry; Prague 128 3 Czech Republic
| | - Andre Castiaux
- Department of Chemistry; Michigan State University; East Lansing, MI 48824 USA
| | - Greg M. Swain
- Department of Chemistry; Michigan State University; East Lansing, MI 48824 USA
| |
Collapse
|
33
|
Houghton LC, Sisti JS, Hankinson SE, Xie J, Xu X, Hoover RN, Eliassen AH, Ziegler RG. Estrogen Metabolism in Premenopausal Women Is Related to Early Life Body Fatness. Cancer Epidemiol Biomarkers Prev 2018; 27:585-593. [PMID: 29511040 PMCID: PMC5932230 DOI: 10.1158/1055-9965.epi-17-0595] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/04/2017] [Accepted: 02/19/2018] [Indexed: 12/15/2022] Open
Abstract
Background: Estrogen metabolism in premenopausal women may be related to early life body fatness.Methods: Premenopausal women participating in the Nurses' Health Study II recalled their body fatness at ages 5, 10, and 20 years using a validated 9-level pictogram. Fifteen estrogens and estrogen metabolites (EM) were measured using LC/MS-MS in luteal phase urines from 603 women ages 32-54 years. Geometric means of individual EM, metabolic pathway groups, and pathway ratios were examined by body fatness categories using linear mixed models.Results: Body fatness at each age was inversely associated with adult concentrations of all EM combined, parent estrogens (estrone, estradiol), and the 2-hydroxylation pathway. Women in the top (vs. bottom) category of body fatness at age 10 had 21% lower levels of all EM (Ptrend = 0.003), 24% lower parent estrogens (Ptrend = 0.002), and 36% lower 2-pathway (Ptrend = 0.0003). Body fatness at age 10 was inversely associated with 2-catechols (35% lower, Ptrend = 0.0004) and 2-methylated catechols (30% lower, Ptrend = 0.002). After adjusting for premenopausal body mass index (BMI), these associations remained inverse but were attenuated; only parent estrogens remained statistically significant (21% lower, Ptrend = 0.01). Body fatness at ages 5 and 20 were similarly, but more weakly, associated with estrogen pathways.Conclusions: Estimates of body fatness during early life were inversely associated with premenopausal levels of all EM combined, parent estrogens, and 2-pathway estrogen metabolites. These relationships were not fully explained by adult BMI.Impact: These findings inform investigations of diseases linked to early life body fatness and estrogen metabolism. Cancer Epidemiol Biomarkers Prev; 27(5); 585-93. ©2018 AACR.
Collapse
Affiliation(s)
- Lauren C Houghton
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York.
| | - Julia S Sisti
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Susan E Hankinson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts, Amherst, Massachusetts
| | | | - Xia Xu
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Robert N Hoover
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York
| | - A Heather Eliassen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Regina G Ziegler
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York
| |
Collapse
|
34
|
Liu Y, Tamimi RM, Colditz GA, Bertrand KA. Alcohol consumption across the life course and mammographic density in premenopausal women. Breast Cancer Res Treat 2018; 167:529-535. [PMID: 28952004 PMCID: PMC5792299 DOI: 10.1007/s10549-017-4517-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 09/18/2017] [Indexed: 10/18/2022]
Abstract
PURPOSE Consumption of alcoholic beverages during adolescence and early adulthood has been consistently associated with higher breast cancer risk. The influence of alcohol consumption early in life on mammographic breast density, a marker of breast cancer risk, is inconclusive. This study examined associations of alcohol consumption across the life course with premenopausal mammographic density. METHODS The study population included 1211 premenopausal women in the Nurses' Health Study II without cancer, who recalled their alcohol consumption at age 15 through enrollment in 1989 (baseline), and had mammograms available. Recent alcohol consumption was updated over follow-up. Percent and absolute measures of mammographic density were quantified on digitized film mammograms. Generalized linear regression was used to assess associations. RESULTS There were no notable differences in any of the three density measures for alcohol consumption at any age (15-17, 18-22, 23-30, and 31-mammogram). Neither alcohol consumption before first pregnancy nor after first pregnancy was significantly associated with any of the three density measures. CONCLUSIONS Moderate alcohol consumption during different age intervals during adolescence and early adulthood was not associated with mammographic density in premenopausal women.
Collapse
Affiliation(s)
- Ying Liu
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO, USA
| | - Rulla M Tamimi
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Graham A Colditz
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO, USA
| | - Kimberly A Bertrand
- Slone Epidemiology Center at Boston University, 72 East Concord Street, L-7, Boston, MA, 02118, USA.
| |
Collapse
|
35
|
Mahabir S, Pfeiffer R, Xu X, Baer DJ, Taylor PR. Effects of low-to-moderate alcohol supplementation on urinary estrogen metabolites in postmenopausal women in a controlled feeding study. Cancer Med 2017; 6:2419-2423. [PMID: 28879665 PMCID: PMC5633545 DOI: 10.1002/cam4.1153] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 06/19/2017] [Accepted: 07/11/2017] [Indexed: 01/13/2023] Open
Abstract
Heavy alcohol drinking is associated with increased breast cancer risk, but associations with low‐to‐moderate alcohol consumption are less clear and the biological mechanisms are not well defined. The objective of this study was to evaluate the effects of 8 weeks of low (15 g/d) and moderate (30 g/d) alcohol ingestion on concentrations of 15 urinary estrogen metabolites (EMs) in postmenopausal women (n = 51) in a controlled feeding study with a randomized crossover design. Compared to no alcohol, 15 g/day for 8 weeks had no effect on urinary EMs. However, compared to no alcohol, 30 g/day for 8 weeks decreased urinary 2‐hydroestrone (2‐OHE1) by 3.3% (P = 0.055) and increased 16‐epiestriol (16‐EpiE3) by 26.6% (P = 0.037). Trends for reduced urinary 2‐OHE1 (P = 0.045), reduced ratio of 2‐OH:16OH pathways (P = 0.008), and increased 16‐EpiE3 (P = 0.035) were observed as alcohol ingestion increased from 0 g to 15 g to 30 g/d. Moderate alcohol consumption for 8 weeks had modest effects on urinary concentrations of 2‐OHE1 and 16‐EpiE3 among postmenopausal women in a carefully controlled feeding study.
Collapse
Affiliation(s)
- Somdat Mahabir
- Environmental Epidemiology Branch, Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute (NCI), Rockville, Maryland.,Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI, Rockville, Maryland
| | - Ruth Pfeiffer
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, NCI, Rockville, Maryland
| | - Xia Xu
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - David J Baer
- Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland
| | - Philip R Taylor
- Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI, Rockville, Maryland
| |
Collapse
|
36
|
Abstract
The advent of mass spectrometry into the clinical laboratory has led to an improvement in clinical management of several endocrine diseases. Liquid chromatography tandem mass spectrometry found some of its first clinical applications in the diagnosis of inborn errors of metabolism, in quantitative steroid analysis, and in drug analysis laboratories. Mass spectrometry assays offer analytical sensitivity and specificity that is superior to immunoassays for many analytes. This article highlights several areas of clinical endocrinology that have witnessed the use of liquid chromatography tandem mass spectrometry to improve clinical outcomes.
Collapse
Affiliation(s)
- Siva S Ketha
- Department of Cardiovascular Diseases, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Ravinder J Singh
- Department of Pathology and Laboratory Medicine, Mayo Clinic, 200 2nd Street, Rochester, MN, 55905, USA
| | - Hemamalini Ketha
- Department of Pathology, University Hospital, University of Michigan Hospital and Health Systems, 1500 East Medical Center Drive, Room 2F432, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
37
|
Dai X, Hua T, Hong T. Integrated diagnostic network construction reveals a 4-gene panel and 5 cancer hallmarks driving breast cancer heterogeneity. Sci Rep 2017; 7:6827. [PMID: 28754978 PMCID: PMC5533795 DOI: 10.1038/s41598-017-07189-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/23/2017] [Indexed: 12/26/2022] Open
Abstract
Breast cancer encompasses a group of heterogeneous diseases, each associated with distinct clinical implications. Dozens of molecular biomarkers capable of categorizing tumors into clinically relevant subgroups have been proposed which, though considerably contribute in precision medicine, complicate our understandings toward breast cancer subtyping and its clinical translation. To decipher the networking of markers with diagnostic roles on breast carcinomas, we constructed the diagnostic networks by incorporating 6 publically available gene expression datasets with protein interaction data retrieved from BioGRID on previously identified 1015 genes with breast cancer subtyping roles. The Greedy algorithm and mutual information were used to construct the integrated diagnostic network, resulting in 37 genes enclosing 43 interactions. Four genes, FAM134B, KIF2C, ALCAM, KIF1A, were identified having comparable subtyping efficacies with the initial 1015 genes evaluated by hierarchical clustering and cross validations that deploy support vector machine and k nearest neighbor algorithms. Pathway, Gene Ontology, and proliferation marker enrichment analyses collectively suggest 5 primary cancer hallmarks driving breast cancer differentiation, with those contributing to uncontrolled proliferation being the most prominent. Our results propose a 37-gene integrated diagnostic network implicating 5 cancer hallmarks that drives breast cancer heterogeneity and, in particular, a 4-gene panel with clinical diagnostic translation potential.
Collapse
Affiliation(s)
- Xiaofeng Dai
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China.
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China.
| | - Tongyan Hua
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Tingting Hong
- Department of medical oncology, the affiliated hospital of Jiangnan University, the fourth people's hospital of Wuxi, Wuxi, China
| |
Collapse
|
38
|
Kupcová E, Reiffová K. Dispersive liquid-liquid microextraction as an effective preanalytical step for the determination of estradiol in human urine. J Sep Sci 2017; 40:2620-2628. [DOI: 10.1002/jssc.201700123] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/10/2017] [Accepted: 04/14/2017] [Indexed: 11/08/2022]
Affiliation(s)
- Elena Kupcová
- Department of Analytical Chemistry, Faculty of Science; Pavol Jozef Šafárik University in Košice; Košice Slovakia
| | - Katarína Reiffová
- Department of Analytical Chemistry, Faculty of Science; Pavol Jozef Šafárik University in Košice; Košice Slovakia
| |
Collapse
|
39
|
Sood D, Johnson N, Jain P, Siskos AP, Bennett M, Gilham C, Busana MC, Peto J, Dos-Santos-Silva I, Keun HC, Fletcher O. CYP3A7*1C allele is associated with reduced levels of 2-hydroxylation pathway oestrogen metabolites. Br J Cancer 2017; 116:382-388. [PMID: 28072767 PMCID: PMC5294487 DOI: 10.1038/bjc.2016.432] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 11/24/2016] [Accepted: 12/01/2016] [Indexed: 11/29/2022] Open
Abstract
Background: Endogenous sex hormones are well-established risk factors for breast cancer; the contribution of specific oestrogen metabolites (EMs) and/or ratios of specific EMs is less clear. We have previously identified a CYP3A7*1C allele that is associated with lower urinary oestrone (E1) levels in premenopausal women. The purpose of this analysis was to determine whether this allele was associated with specific pathway EMs. Methods: We measured successfully 12 EMs in mid-follicular phase urine samples from 30 CYP3A7*1C carriers and 30 non-carriers using HPLC-MS/MS. Results: In addition to having lower urinary E1 levels, CYP3A7*1C carriers had significantly lower levels of four of the 2-hydroxylation pathway EMs that we measured (2-hydroxyestrone, P=1.1 × 10−12; 2-hydroxyestradiol, P=2.7 × 10−7; 2-methoxyestrone, P=1.9 × 10−12; and 2-methoxyestradiol, P=0.0009). By contrast, 16α-hydroxylation pathway EMs were slightly higher in carriers and significantly so for 17-epiestriol (P=0.002). Conclusions: The CYP3A7*1C allele is associated with a lower urinary E1 levels, a more pronounced reduction in 2-hydroxylation pathway EMs and a lower ratio of 2-hydroxylation:16α-hydroxylation EMs in premenopausal women. To further characterise the association between parent oestrogens, EMs and subsequent risk of breast cancer, characterisation of additional genetic variants that influence oestrogen metabolism and large prospective studies of a broad spectrum of EMs will be required.
Collapse
Affiliation(s)
- Deepti Sood
- Faculty of Medicine, Department of Surgery and Cancer, Imperial College, London SW7 2AZ, UK
| | - Nichola Johnson
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, 237 Fulham Road, London SW7 3RP, UK
| | - Pooja Jain
- Faculty of Medicine, Department of Surgery and Cancer, Imperial College, London SW7 2AZ, UK
| | - Alexandros P Siskos
- Faculty of Medicine, Department of Surgery and Cancer, Imperial College, London SW7 2AZ, UK
| | - Mark Bennett
- Department of Life Sciences, Imperial College, London SW7 2AZ, UK
| | - Clare Gilham
- Department of Non-Communicable Disease Epidemiology, The London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Marta Cecilia Busana
- Department of Non-Communicable Disease Epidemiology, The London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Julian Peto
- Department of Non-Communicable Disease Epidemiology, The London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Isabel Dos-Santos-Silva
- Department of Non-Communicable Disease Epidemiology, The London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Hector C Keun
- Faculty of Medicine, Department of Surgery and Cancer, Imperial College, London SW7 2AZ, UK
| | - Olivia Fletcher
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, 237 Fulham Road, London SW7 3RP, UK
| |
Collapse
|
40
|
Kim S, Campbell J, Yoo W, Taylor JA, Sandler DP. Systemic Levels of Estrogens and PGE 2 Synthesis in Relation to Postmenopausal Breast Cancer Risk. Cancer Epidemiol Biomarkers Prev 2016; 26:383-388. [PMID: 27864342 DOI: 10.1158/1055-9965.epi-16-0556] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/23/2016] [Accepted: 10/18/2016] [Indexed: 12/14/2022] Open
Abstract
Background: Prostaglandin E2 (PGE2) induces aromatase expression in adipose tissue, leading to increased estrogen production that may promote the development and progression of breast cancer. However, few studies have simultaneously investigated systemic levels of PGE2 and estrogen in relation to postmenopausal breast cancer risk.Methods: Here, we determined urinary estrogen metabolites (EM) using mass spectrometry in a case-cohort study (295 incident breast cancer cases and 294 subcohort members), and using linear regression estimated the effect of urinary levels of a major PGE2 metabolite (PGE-M) on EMs. HRs for the risk of developing breast cancer in relation to PGE-M and EMs were compared between Cox regression models with and without mutual adjustment.Results: PGE-M was a significant predictor of estrone (E1), but not estradiol (E2) levels in multivariable analysis. Elevated E2 levels were associated with an increased risk of developing breast cancer [HRQ5vs.Q1, 1.54; 95% confidence interval (CI), 1.01-2.35], and this association remained unchanged after adjustment for PGE-M (HRQ5vs.Q1, 1.52; 95% CI, 0.99-2.33). Similarly, elevated levels of PGE-M were associated with increased risk of developing breast cancer (HRQ4vs.Q1, 2.01; 95% CI, 1.01-4.29), and this association was only nominally changed after consideration of E1 or E2 levels.Conclusions: Urinary levels of PGE-M and estrogens were independently associated with future risk of developing breast cancer among these postmenopausal women.Impact: Increased breast cancer risk associated with PGE-M might not be fully explained by the estrogens-breast cancer association alone but also by additional effects related to inflammation. Cancer Epidemiol Biomarkers Prev; 26(3); 383-8. ©2016 AACR.
Collapse
Affiliation(s)
- Sangmi Kim
- Medical College of Georgia, Department of Medicine, Section of Hematology/Oncology, Augusta University Georgia Cancer Center, Augusta, Georgia.
| | - Jeff Campbell
- Medical College of Georgia, Department of Medicine, Section of Hematology/Oncology, Augusta University Georgia Cancer Center, Augusta, Georgia
| | - Wonsuk Yoo
- Institute of Public and Preventive Health - Data Coordinating Center, Augusta University, Augusta, Georgia
| | - Jack A Taylor
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| |
Collapse
|
41
|
Harrelson JP, Lee MW. Expanding the view of breast cancer metabolism: Promising molecular targets and therapeutic opportunities. Pharmacol Ther 2016; 167:60-73. [DOI: 10.1016/j.pharmthera.2016.07.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/21/2016] [Indexed: 12/23/2022]
|
42
|
Gastrointestinal microbiome and breast cancer: correlations, mechanisms and potential clinical implications. Breast Cancer 2016; 24:220-228. [PMID: 27709424 DOI: 10.1007/s12282-016-0734-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 09/21/2016] [Indexed: 02/05/2023]
Abstract
Gastrointestinal microbiome plays as a symbiont which provides protection effect against invading pathogens, aids in the immune system development, nutrient reclamation and absorption as well as molecule breakdown. And it may avert carcinogenesis through these biological activities. By now, studies have been carried out to elaborate the association between gastrointestinal microbiome and breast cancer. It has been implicated that breast cancer was substantially associated with estrogen-dependent and estrogen-independent functions of gastrointestinal microbiome. Evidence from animal experiments also confirmed mammary tumor-related changes in microbial community. The possible mechanisms involve estrogen metabolism, immune regulation, obese status and so forth. Based on the current evidence, cues on future management strategies of breast cancer such as antibiotics and dietary interventions are proposed. In conclusion, large-scale clinical studies and bench-based researches are needed to validate the associations and elaborate the mechanisms, so as to reduce the risk of breast cancer and improve the outcomes of those already diagnosed.
Collapse
|
43
|
Moore SC, Matthews CE, Ou Shu X, Yu K, Gail MH, Xu X, Ji BT, Chow WH, Cai Q, Li H, Yang G, Ruggieri D, Boyd-Morin J, Rothman N, Hoover RN, Gao YT, Zheng W, Ziegler RG. Endogenous Estrogens, Estrogen Metabolites, and Breast Cancer Risk in Postmenopausal Chinese Women. J Natl Cancer Inst 2016; 108:djw103. [PMID: 27193440 DOI: 10.1093/jnci/djw103] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 03/09/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The role of estrogen metabolism in determining breast cancer risk and differences in breast cancer rates between high-incidence and low-incidence nations is poorly understood. METHODS We measured urinary concentrations of estradiol and estrone (parent estrogens) and 13 estrogen metabolites formed by irreversible hydroxylation at the C-2, C-4, or C-16 positions of the steroid ring in a nested case-control study of 399 postmenopausal invasive breast cancer case participants and 399 matched control participants from the population-based Shanghai Women's Health Study cohort. Odds ratios (ORs) and 95% confidence intervals (CIs) of breast cancer by quartiles of metabolic pathway groups, pathway ratios, and individual estrogens/estrogen metabolites were estimated by multivariable conditional logistic regression. Urinary estrogen/estrogen metabolite measures were compared with those of postmenopausal non-hormone-using Asian Americans, a population with three-fold higher breast cancer incidence rates. All statistical tests were two-sided. RESULTS Urinary concentrations of parent estrogens were strongly associated with breast cancer risk (ORQ4vsQ1 = 1.94, 95% CI = 1.21 to 3.12, Ptrend = .01). Of the pathway ratios, the 2-pathway:total estrogens/estrogen metabolites and 2-pathway:parent estrogens were inversely associated with risk (ORQ4vsQ1 = 0.57, 95% CI = 0.35 to 0.91, Ptrend = .03, and ORQ4vsQ1 = 0.61, 95% CI = 0.37 to 0.99, Ptrend = .04, respectively). After adjusting for parent estrogens, these associations remained clearly inverse but lost statistical significance (ORQ4vsQ1 = 0.65, 95% CI = 0.39 to 1.06, Ptrend = .12 and ORQ4vsQ1 = 0.76, 95% CI = 0.44 to 1.32, Ptrend = .28). The urinary concentration of all estrogens/estrogen metabolites combined in Asian American women was triple that in Shanghai women. CONCLUSIONS Lower urinary parent estrogen concentrations and more extensive 2-hydroxylation were each associated with reduced postmenopausal breast cancer risk in a low-risk nation. Markedly higher total estrogen/estrogen metabolite concentrations in postmenopausal United States women (Asian Americans) than in Shanghai women may partly explain higher breast cancer rates in the United States.
Collapse
Affiliation(s)
- Steven C Moore
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| | - Charles E Matthews
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| | - Xiao Ou Shu
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| | - Kai Yu
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| | - Mitchell H Gail
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| | - Xia Xu
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| | - Bu-Tian Ji
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| | - Wong-Ho Chow
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| | - Qiuyin Cai
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| | - Honglan Li
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| | - Gong Yang
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| | - David Ruggieri
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| | - Jennifer Boyd-Morin
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| | - Nathaniel Rothman
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| | - Robert N Hoover
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| | - Yu-Tang Gao
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| | - Wei Zheng
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| | - Regina G Ziegler
- Affiliations of authors: Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (SCM, CEM, KY, MHG, BTJ, NR, RNH, RGZ); Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN (XOS, QC, GY, WZ); Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD (XX); Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China (HL, YTG); Department of Epidemiology, Division of OVP, Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX (WHC); Information Management Services, Inc., Rockville, MD (DR, JBM)
| |
Collapse
|
44
|
Maskarinec G, Beckford F, Morimoto Y, Franke AA, Stanczyk FZ. Association of estrogen measurements in serum and urine of premenopausal women. Biomark Med 2016; 9:417-24. [PMID: 25985172 DOI: 10.2217/bmm.15.10] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIMS We evaluated conjugated and unconjugated urinary estrogen metabolites as surrogate biomarkers for serum levels of unconjugated E1 and E2 in premenopausal women. MATERIALS & METHODS Repeated blood and urine samples were analyzed for estrogens and their metabolites using radioimmunoassays and liquid chromatography/mass spectrometry. RESULTS The strongest correlation (r = 0.39) was observed between serum E1 and urinary E1 and E2. The correlations of urinary E2 (r = 0.35), E1 (r = 0.29), all E2 metabolites (r = 0.30), all E1 metabolites (r = 0.23) and total estrogens (r = 0.26) with serum E2 were only moderate although statistically significant. All correlations were substantially stronger for Whites than Asians. CONCLUSION Urinary E2 emerged as the best predictor for serum E1 and E2, but the large intra-subject variability in urinary estrogen levels limits its use as a biomarker.
Collapse
Affiliation(s)
- Gertraud Maskarinec
- 1University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI 96813, USA
| | - Fanchon Beckford
- 1University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI 96813, USA
| | - Yukiko Morimoto
- 1University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI 96813, USA
| | - Adrian A Franke
- 1University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI 96813, USA
| | | |
Collapse
|
45
|
Hartman TJ, Sisti JS, Hankinson SE, Xu X, Eliassen AH, Ziegler R. Alcohol Consumption and Urinary Estrogens and Estrogen Metabolites in Premenopausal Women. HORMONES & CANCER 2016; 7:65-74. [PMID: 26728472 PMCID: PMC4729640 DOI: 10.1007/s12672-015-0249-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/16/2015] [Indexed: 01/23/2023]
Abstract
In a cross-sectional analysis, we evaluated the associations of usual total alcohol and wine intake with a comprehensive profile of mid-luteal phase urinary estrogens and estrogen metabolites (referred to jointly as EM) in a sample of 603 premenopausal women participating in the Nurses' Health Study II (NHSII). A total of 15 individual EM (pmol/mg creatinine) were measured by a liquid chromatography/tandem mass spectrometry (LC-MS/MS) method with high accuracy and reproducibility. We used linear mixed models to calculate the adjusted geometric means of individual EM, EM grouped by metabolic pathways, and pathway ratios by category of alcohol intake with non-drinkers of alcohol as the referent. Total alcohol intake was not associated with total EM but was positively associated with estradiol (26% higher among women consuming >15 g/day vs. non-drinkers; P trend = 0.03). Wine consumption was positively associated with a number of EM measures including estradiol (22% higher among women consuming ≥ 5 drinks/week vs. non-drinkers, P trend < 0.0001). In conclusion, the total alcohol intake was positively and significantly associated with urinary estradiol levels. Some differences in urinary estrogen metabolites were observed with wine drinking, when compared with non-drinkers. This study strengthens the evidence that alcohol consumption might play a role in breast cancer and other estrogen-related conditions. Additional studies of premenopausal women are needed to further explore the association of alcohol, particularly the specific types of alcohol, on patterns of estrogen metabolism in blood, urine, and tissue.
Collapse
Affiliation(s)
- Terryl J Hartman
- Department of Epidemiology, Rollins School of Public Health & Winship Cancer Institute, Emory University, 1518 Clifton Road NE, CNR #3035, Atlanta, GA, 30322, USA.
| | - Julia S Sisti
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Susan E Hankinson
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, USA
| | - Xia Xu
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - A Heather Eliassen
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Regina Ziegler
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
46
|
Bertrand KA, Burian RA, Eliassen AH, Willett WC, Tamimi RM. Adolescent intake of animal fat and red meat in relation to premenopausal mammographic density. Breast Cancer Res Treat 2016; 155:385-93. [PMID: 26791521 DOI: 10.1007/s10549-016-3679-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 01/04/2016] [Indexed: 11/24/2022]
Abstract
Adolescence is hypothesized to be a time period of particular susceptibility to breast cancer risk factors. Red meat and fat intake during high school was positively associated with risk of breast cancer among premenopausal women in the Nurses' Health Study II (NHSII). High mammographic density is a strong predictor of breast cancer risk but there is limited research on dietary factors associated with breast density. To test the hypothesis that high intake of animal fat or red meat during adolescence is associated with mammographic density, we analyzed data from premenopausal women in the NHSII. Participants recalled adolescent diet on a high school food frequency questionnaire. We assessed absolute and percent mammographic density on digitized analog film mammograms for 687 premenopausal women with no history of cancer. We used generalized linear regression to quantify associations of adolescent animal fat and red meat intake with mammographic density, adjusting for age, body mass index, and other predictors of mammographic density. Adolescent animal fat intake was significantly positively associated with premenopausal mammographic density, with a mean percent density of 39.2 % in the lowest quartile of adolescent animal fat intake versus 43.1 % in the highest quartile (p trend: 0.03). A non-significant positive association was also observed for adolescent red meat intake (p trend: 0.14). These findings suggest that higher adolescent animal fat intake is weakly associated with percent mammographic density in premenopausal women.
Collapse
Affiliation(s)
- Kimberly A Bertrand
- Slone Epidemiology Center at Boston University, 1010 Commonwealth Avenue, Boston, MA, 02215, USA.
| | - Rosemarie A Burian
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA
| | - A Heather Eliassen
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| | - Walter C Willett
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| | - Rulla M Tamimi
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| |
Collapse
|
47
|
Na W, Park JW, An JH, Jang J. Size-controllable ultrathin carboxylated polypyrrole nanotube transducer for extremely sensitive 17β-estradiol FET-type biosensors. J Mater Chem B 2016; 4:5025-5034. [DOI: 10.1039/c6tb00897f] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Size-controllable aptamer conjugated ultrathin carboxylated polypyrrole nanotubes (A-UCPPyNTs) were successfully fabricated as transducers in 17β-estradiol field-effect transistor (FET)-type biosensors which has extremely high sensitivity (∼1 fM) and unique selectivity.
Collapse
Affiliation(s)
- Wonjoo Na
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul 151-742
- Korea
| | - Jin Wook Park
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul 151-742
- Korea
| | - Ji Hyun An
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul 151-742
- Korea
| | - Jyongsik Jang
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul 151-742
- Korea
| |
Collapse
|
48
|
Schmitz KH, Williams NI, Kontos D, Domchek S, Morales KH, Hwang WT, Grant LL, DiGiovanni L, Salvatore D, Fenderson D, Schnall M, Galantino ML, Stopfer J, Kurzer MS, Wu S, Adelman J, Brown JC, Good J. Dose-response effects of aerobic exercise on estrogen among women at high risk for breast cancer: a randomized controlled trial. Breast Cancer Res Treat 2015; 154:309-18. [PMID: 26510851 PMCID: PMC6196733 DOI: 10.1007/s10549-015-3604-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 10/09/2015] [Indexed: 10/22/2022]
Abstract
UNLABELLED Medical and surgical interventions for elevated breast cancer risk (e.g., BRCA1/2 mutation, family history) focus on reducing estrogen exposure. Women at elevated risk may be interested in less aggressive approaches to risk reduction. For example, exercise might reduce estrogen, yet has fewer serious side effects and less negative impact than surgery or hormonal medications. Randomized controlled trial. Increased risk defined by risk prediction models or BRCA mutation status. Eligibility: Age 18-50, eumenorrheic, non-smokers, and body mass index (BMI) between 21 and 50 kg/m(2). 139 were randomized. Treadmill exercise: 150 or 300 min/week, five menstrual cycles. Control group maintained exercise <75 min/week. PRIMARY OUTCOME Area under curve (AUC) for urinary estrogen. Secondary measures: urinary progesterone, quantitative digitized breast dynamic contrast-enhanced magnetic resonance imaging background parenchymal enhancement. Mean age 34 years, mean BMI 26.8 kg/m(2). A linear dose-response relationship was observed such that every 100 min of exercise is associated with 3.6 % lower follicular phase estrogen AUC (linear trend test, p = 0.03). No changes in luteal phase estrogen or progesterone levels. There was also a dose-response effect noted: for every 100 min of exercise, there was a 9.7 % decrease in background parenchymal enhancement as measured by imaging (linear trend test, p = 0.009). Linear dose-response effect observed to reduce follicular phase estrogen exposure measured via urine and hormone sensitive breast tissue as measured by imaging. Future research should explore maintenance of effects and extent to which findings are repeatable in lower risk women. Given the high benefit to risk ratio, clinicians can inform young women at increased risk that exercise may blunt estrogen exposure while considering whether to try other preventive therapies.
Collapse
Affiliation(s)
- Kathryn H Schmitz
- Perelman School of Medicine, University of Pennsylvania, 8th Floor Blockley Hall, 423 Guardian Dr., Philadelphia, PA, 19104-6021, USA.
| | - Nancy I Williams
- Department of Kinesiology, Pennsylvania State University, State College, USA
| | - Despina Kontos
- Perelman School of Medicine, University of Pennsylvania, 8th Floor Blockley Hall, 423 Guardian Dr., Philadelphia, PA, 19104-6021, USA
| | - Susan Domchek
- Perelman School of Medicine, University of Pennsylvania, 8th Floor Blockley Hall, 423 Guardian Dr., Philadelphia, PA, 19104-6021, USA
| | - Knashawn H Morales
- Perelman School of Medicine, University of Pennsylvania, 8th Floor Blockley Hall, 423 Guardian Dr., Philadelphia, PA, 19104-6021, USA
| | - Wei-Ting Hwang
- Perelman School of Medicine, University of Pennsylvania, 8th Floor Blockley Hall, 423 Guardian Dr., Philadelphia, PA, 19104-6021, USA
| | - Lorita L Grant
- Perelman School of Medicine, University of Pennsylvania, 8th Floor Blockley Hall, 423 Guardian Dr., Philadelphia, PA, 19104-6021, USA
| | - Laura DiGiovanni
- Perelman School of Medicine, University of Pennsylvania, 8th Floor Blockley Hall, 423 Guardian Dr., Philadelphia, PA, 19104-6021, USA
| | - Domenick Salvatore
- Perelman School of Medicine, University of Pennsylvania, 8th Floor Blockley Hall, 423 Guardian Dr., Philadelphia, PA, 19104-6021, USA
| | - Desire' Fenderson
- Perelman School of Medicine, University of Pennsylvania, 8th Floor Blockley Hall, 423 Guardian Dr., Philadelphia, PA, 19104-6021, USA
| | - Mitchell Schnall
- Perelman School of Medicine, University of Pennsylvania, 8th Floor Blockley Hall, 423 Guardian Dr., Philadelphia, PA, 19104-6021, USA
| | - Mary Lou Galantino
- Perelman School of Medicine, University of Pennsylvania, 8th Floor Blockley Hall, 423 Guardian Dr., Philadelphia, PA, 19104-6021, USA
| | - Jill Stopfer
- Perelman School of Medicine, University of Pennsylvania, 8th Floor Blockley Hall, 423 Guardian Dr., Philadelphia, PA, 19104-6021, USA
| | - Mindy S Kurzer
- Department of Nutrition, University of Minnesota, Minneapolis, USA
| | - Shandong Wu
- Department of Radiology, University of Pittsburgh, Pittsburgh, USA
| | - Jessica Adelman
- Perelman School of Medicine, University of Pennsylvania, 8th Floor Blockley Hall, 423 Guardian Dr., Philadelphia, PA, 19104-6021, USA
| | - Justin C Brown
- Perelman School of Medicine, University of Pennsylvania, 8th Floor Blockley Hall, 423 Guardian Dr., Philadelphia, PA, 19104-6021, USA
| | - Jerene Good
- Perelman School of Medicine, University of Pennsylvania, 8th Floor Blockley Hall, 423 Guardian Dr., Philadelphia, PA, 19104-6021, USA
| |
Collapse
|
49
|
Oh H, Smith-Warner SA, Tamimi RM, Wang M, Xu X, Hankinson SE, Fuhrman BJ, Ziegler RG, Eliassen AH. Dietary Fat and Fiber Intakes Are Not Associated with Patterns of Urinary Estrogen Metabolites in Premenopausal Women. J Nutr 2015; 145:2109-16. [PMID: 26180245 PMCID: PMC4548163 DOI: 10.3945/jn.115.212779] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 04/02/2015] [Accepted: 06/18/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Interindividual differences in the bioavailability of potentially carcinogenic estrogen and estrogen metabolites (EMs) may play a role in the risk of breast cancer. OBJECTIVE We examined whether dietary intakes of fiber and fat influence premenopausal EM profiles through effects on estrogen synthesis, metabolism, or excretion. METHODS We conducted a cross-sectional analysis of 598 premenopausal women who participated in a reproducibility study (n = 109) or served as controls in a nested case-control study of breast cancer (n = 489) within the Nurses' Health Study II. Dietary intakes of fiber and fat were assessed via semiquantitative food frequency questionnaires in 1995 and 1999. Midluteal urine samples were collected between 1996 and 1999 and EMs were quantified with the use of HPLC-tandem mass spectrometry. Linear mixed models were used to estimate creatinine-adjusted geometric means for individual EMs and their pathway groups across categories of dietary intake while controlling for total energy intake and potential confounders. RESULTS Higher total dietary fiber intake (>25 g/d vs. ≤15 g/d) was associated with significantly higher concentrations of 4-methoxyestradiol (50% difference, P-difference = 0.01, P-trend = 0.004) and lower concentrations of 17-epiestriol (-27% difference, P-difference = 0.03, P-trend = 0.03), but was not associated with any other EMs. The associations did not vary by fiber intake from different sources. Total fat intake (>35% energy vs. ≤25% energy) was suggestively positively associated with 17-epiestriol (22.6% difference, P-difference = 0.14, P-trend = 0.06); the association was significant for polyunsaturated fatty acid (37% difference, P-difference = 0.01, P-trend = 0.01) and trans fat (36.1% difference, P-difference = 0.01, P-trend = 0.01) intakes. CONCLUSION Fiber and fat intakes were not strongly associated with patterns of estrogen metabolism in premenopausal women. Our data suggest estrogen metabolism is not a major mechanism through which dietary fiber and fat may affect breast or other hormone-related cancer risks.
Collapse
Affiliation(s)
- Hannah Oh
- Department of Epidemiology, Department of Nutrition, and Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA;
| | | | - Rulla M Tamimi
- Department of Epidemiology, Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Molin Wang
- Department of Epidemiology, Department of Biostatistics, Harvard School of Public Health, Boston, MA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Xia Xu
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Susan E Hankinson
- Department of Epidemiology, Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Division of Biostatistics and Epidemiology, University of Massachusetts, Amherst, MA
| | - Barbara J Fuhrman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD; and Department of Epidemiology, Fay W Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Regina G Ziegler
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD; and
| | - A Heather Eliassen
- Department of Epidemiology, Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
50
|
Sisti JS, Hankinson SE, Caporaso NE, Gu F, Tamimi RM, Rosner B, Xu X, Ziegler R, Eliassen AH. Caffeine, coffee, and tea intake and urinary estrogens and estrogen metabolites in premenopausal women. Cancer Epidemiol Biomarkers Prev 2015; 24:1174-83. [PMID: 26063478 PMCID: PMC4526325 DOI: 10.1158/1055-9965.epi-15-0246] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/22/2015] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Prior studies have found weak inverse associations between breast cancer and caffeine and coffee intake, possibly mediated through their effects on sex hormones. METHODS High-performance liquid chromatography/tandem mass spectrometry was used to quantify levels of 15 individual estrogens and estrogen metabolites (EM) among 587 premenopausal women in the Nurses' Health Study II with mid-luteal phase urine samples and caffeine, coffee, and/or tea intakes from self-reported food frequency questionnaires. Multivariate linear mixed models were used to estimate geometric means of individual EM, pathways, and ratios by intake categories, and P values for tests of linear trend. RESULTS Compared with women in the lowest quartile of caffeine consumption, those in the top quartile had higher urinary concentrations of 16α-hydroxyestrone (28% difference; Ptrend = 0.01) and 16-epiestriol (13% difference; Ptrend = 0.04), and a decreased parent estrogens/2-, 4-, 16-pathway ratio (Ptrend = 0.03). Coffee intake was associated with higher 2-catechols, including 2-hydroxyestradiol (57% difference, ≥4 cups/day vs. ≤6 cups/week; Ptrend = 0.001) and 2-hydroxyestrone (52% difference; Ptrend = 0.001), and several ratio measures. Decaffeinated coffee was not associated with 2-pathway metabolism, but women in the highest (vs. lowest) category of intake (≥2 cups/day vs. ≤1-3 cups/month) had significantly lower levels of two 16-pathway metabolites, estriol (25% difference; Ptrend = 0.01) and 17-epiestriol (48% difference; Ptrend = 0.0004). Tea intake was positively associated with 17-epiestriol (52% difference; Ptrend = 0.01). CONCLUSION Caffeine and coffee intake were both associated with profiles of estrogen metabolism in premenopausal women. IMPACT Consumption of caffeine and coffee may alter patterns of premenopausal estrogen metabolism.
Collapse
Affiliation(s)
- Julia S Sisti
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.
| | - Susan E Hankinson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts, Amherst, Massachusetts
| | - Neil E Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Fangyi Gu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Rulla M Tamimi
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Bernard Rosner
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Xia Xu
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Regina Ziegler
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - A Heather Eliassen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|