1
|
Gong G, Wan W, Liu X, Yin J. Apelin-13, a regulator of autophagy, apoptosis and inflammation in multifaceted bone protection. Int Immunopharmacol 2023; 117:109991. [PMID: 37012875 DOI: 10.1016/j.intimp.2023.109991] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/12/2023]
Abstract
Apelin/APJ is widely distributed in various tissues in the body and participates in the regulation of physiological and pathological mechanisms such as autophagy, apoptosis, inflammation, and oxidative stress. Apelin-13 is an adipokine family member with multiple biological roles and has been shown to be involved in the development and progression of bone diseases. In the process of osteoporosis and fracture healing, Apelin-13 plays an osteoprotective role by regulating the autophagy and apoptosis of BMSCs, and promotes the osteogenic differentiation of BMSCs. In addition, Apelin-13 also attenuates the progression of arthritis by regulating the inflammatory response of macrophages. In conclusion, Apelin-13 has an important connection with bone protection, which provides a new strategy for the clinical treatment of bone-related diseases.
Collapse
Affiliation(s)
- Ge Gong
- Department of Geriatrics, Jinling Hospital, Medical School of Nanjing University, Nanjing 211002, China
| | - Wenhui Wan
- Department of Geriatrics, Jinling Hospital, Medical School of Nanjing University, Nanjing 211002, China
| | - Xinhui Liu
- Department of Orthopedics, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China.
| | - Jian Yin
- Department of Orthopedics, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China.
| |
Collapse
|
2
|
The effects of alone and combination tamoxifen, raloxifene and estrogen on lipid profile and atherogenic index of ovariectomized type 2 diabetic rats. Life Sci 2020; 263:118573. [PMID: 33058909 DOI: 10.1016/j.lfs.2020.118573] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/29/2020] [Accepted: 10/04/2020] [Indexed: 12/13/2022]
|
3
|
Beneficial effects of tamoxifen on leptin sensitivity in young mice fed a high fat diet: Role of estrogen receptor α and cytokines. Life Sci 2020; 246:117384. [PMID: 32061672 DOI: 10.1016/j.lfs.2020.117384] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 01/26/2020] [Accepted: 01/28/2020] [Indexed: 01/02/2023]
|
4
|
Belcher SM, Cline JM, Conley J, Groeters S, Jefferson WN, Law M, Mackey E, Suen AA, Williams CJ, Dixon D, Wolf JC. Endocrine Disruption and Reproductive Pathology. Toxicol Pathol 2019; 47:1049-1071. [PMID: 31833458 PMCID: PMC8008741 DOI: 10.1177/0192623319879903] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
During the past 20 years, investigations involving endocrine active substances (EAS) and reproductive toxicity have dominated the landscape of ecotoxicological research. This has occurred in concert with heightened awareness in the scientific community, general public, and governmental entities of the potential consequences of chemical perturbation in humans and wildlife. The exponential growth of experimentation in this field is fueled by our expanding knowledge into the complex nature of endocrine systems and the intricacy of their interactions with xenobiotic agents. Complicating factors include the ever-increasing number of novel receptors and alternate mechanistic pathways that have come to light, effects of chemical mixtures in the environment versus those of single EAS laboratory exposures, the challenge of differentiating endocrine disruption from direct cytotoxicity, and the potential for transgenerational effects. Although initially concerned with EAS effects chiefly in the thyroid glands and reproductive organs, it is now recognized that anthropomorphic substances may also adversely affect the nervous and immune systems via hormonal mechanisms and play substantial roles in metabolic diseases, such as type 2 diabetes and obesity.
Collapse
Affiliation(s)
| | - J. Mark Cline
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | | | | | | | - Mac Law
- North Carolina State College of Veterinary Medicine, Raleigh, NC, USA
| | - Emily Mackey
- Michigan State University, East Lansing, MI, USA
| | | | | | | | | |
Collapse
|
5
|
Mao X, Dong B, Gao M, Ruan G, Huang M, Braicu EI, Sehouli J, Sun P. Dual targeting of estrogen receptor α and estrogen-related receptor α: a novel endocrine therapy for endometrial cancer. Onco Targets Ther 2019; 12:6757-6767. [PMID: 31686835 PMCID: PMC6709363 DOI: 10.2147/ott.s216146] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 07/16/2019] [Indexed: 11/24/2022] Open
Abstract
Background Endometrial cancer (EC) is a hormone dependent carcinoma that may involve complex molecular mechanisms. Endocrine therapy by blocking the estrogen and estrogen receptor α (ERα) has been effective in breast cancer, while it is still controversial in EC. Recently, estrogen-related receptor α (ERRα) was proven to be another endocrine therapy target. Methods The anti-tumor effect of selective estrogen receptor modulators (SERMs) and XCT790 (XCT) used alone or in combination were evaluated in both of ERα-positive (ERα+) and ERα-negative (ERα-) EC cells. ERα and ERRα mRNA were tested by qPCR, while the protein was detected by Western blot. The proliferation was tested by MTS and cell cycle, apoptosis rate were analyzed by flow cytometry. Results A relatively high dose (10 μM) of tamoxifen (TAM) suppressed the expression of ERα and ERRα in two types of EC cells. However, 10 μM raloxifene (RAL) exhibited no effect on ERα and ERRα, while 10 μM XCT down regulated ERRα specifically, but not ERα in all EC cells. When dual targeting on ERα and ERRα by combining TAM with XCT, the proliferation inhibitory effect and apoptosis reached the strongest in all EC cells (P<0.05). Moreover, the inhibitory effect of proliferation was attributed significantly to the G0/G1 arrest (P<0.05). Interestingly, the apoptosis induced by combining TAM with XCT were obviously higher in ERα+ EC cells than ERα- EC cells (P<0.05). Conclusion Taken together, the results indicate that dual targeting on ERα and ERRα represents a better anti-tumor effect, which provides a novel endocrine based therapy strategy for EC.
Collapse
Affiliation(s)
- XiaoDan Mao
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, People's Republic of China
| | - Binhua Dong
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, People's Republic of China
| | - Min Gao
- Department of Gynecology Oncology, Peking University Cancer Hospital, Beijing 100142, People's Republic of China
| | - GuanYu Ruan
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, People's Republic of China
| | - MeiMei Huang
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, People's Republic of China
| | - Elena Ioana Braicu
- Department of Gynecology, Campus Virchow-Klinikum, Charité Universitätmedizin Berlin, Berlin D-13353, Germany
| | - Jalid Sehouli
- Department of Gynecology, Campus Virchow-Klinikum, Charité Universitätmedizin Berlin, Berlin D-13353, Germany
| | - PengMing Sun
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, People's Republic of China
| |
Collapse
|
6
|
Azizian H, Khaksari M, Asadikaram G, Sepehri G, Najafipour H. Therapeutic effects of tamoxifen on metabolic parameters and cytokines modulation in rat model of postmenopausal diabetic cardiovascular dysfunction: Role of classic estrogen receptors. Int Immunopharmacol 2018; 65:190-198. [PMID: 30316077 DOI: 10.1016/j.intimp.2018.10.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 09/29/2018] [Accepted: 10/05/2018] [Indexed: 12/25/2022]
Abstract
In postmenopausal women, the risk of diabetic cardiovascular disease drastically increases compared with that of premenopausal women. In the present study we surveyed the effects of Tamoxifen (TAM) and 17-β-estradiol (E2) on diabetic cardiovascular dysfunction. Female wistar rats were divided into six groups: sham-control, Diabetes, Ovariectomized (OVX) + Diabetes, OVX + Diabetes + Vehicle, OVX + Diabetes + E2, OVX + Diabetes + TAM. Type 2 diabetes was induced by High Fat Diet and low doses of STZ. E2 and TAM were administrated every four days for four weeks. Results show that, TAM or E2 reduces cardiac weight, atherogenic and cardiac risk indices. Mean arterial blood pressure (MABP) increased in diabetes group, while TAM and E2 prevented MABP increment. Also, fasting blood glucose was decreased by TAM and E2. Significant decrement in the level of IL-10 was observed in diabetes group and this effect was abolished by TAM and E2. Also, treatment with TAM and E2 resulted in improved inflammatory balance in favor of anti-inflammation. Although diabetes resulted in, increment of TC and LDL, TAM and E2 reduced lipids profile. Furthermore, treatment with TAM prevented the reduction of estrogen receptors (ERs) α and β protein levels, but its effect on the ERβ protein level was higher. Our results indicated that TAM protects against diabetic cardiovascular dysfunction and is a good candidate for E2 substitution.
Collapse
Affiliation(s)
- Hossein Azizian
- Department of Physiology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research, and Physiology Research Centers, Kerman University of Medical Sciences, Kerman, Iran.
| | - Gholamreza Asadikaram
- Department of Biochemistry, and Metabolism & Endocrinology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholamreza Sepehri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman Univerity of Medical Sciences, Kerman, Iran
| | - Hamid Najafipour
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
7
|
Silva LAS, Felix FB, Araujo JMD, Souza EV, Camargo EA, Grespan R. Agonistic activity of tamoxifen, a selective estrogen-receptor modulator (SERM), on arthritic ovariectomized mice. ACTA ACUST UNITED AC 2017; 51:e6799. [PMID: 29160416 PMCID: PMC5685064 DOI: 10.1590/1414-431x20176799] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 09/05/2017] [Indexed: 11/22/2022]
Abstract
Arthritis is positively associated with the decline of sex hormones, especially estrogen. Tamoxifen (TMX) is a selective estrogen receptor modulator, possessing agonist or antagonistic activity in different tissues. Thus, the objective of this study was to investigate the effect of TMX on the zymosan-induced arthritis model. Female Swiss normal and ovariectomized (OVX) mice were divided into groups and treated for five days with TMX (0.3, 0.9 or 2.7 mg/kg) or 17-β-estradiol (E2, 50 µg/kg). On the fifth day, arthritis was induced and 4 h later, leukocyte migration into joint cavities was evaluated. The neutrophil migration in OVX animals, but not in normal mice, treated with TMX (all tested doses) was significantly decreased compared with mice that received the vehicle (P≤0.05). Similarly, this effect was also demonstrated in the E2-treated group. Therefore, the present study demonstrates that TMX presented agonist effects in inhibiting neutrophil migration and preventing arthritis progression in OVX mice.
Collapse
Affiliation(s)
- L A S Silva
- Programa de Pós-Graduação em Ciências Fisiológicas, Departamento de Fisiologia, Universidade Federal de Sergipe, São Cristóvão, SE, Brasil
| | - F B Felix
- Programa de Pós-Graduação em Ciências Fisiológicas, Departamento de Fisiologia, Universidade Federal de Sergipe, São Cristóvão, SE, Brasil
| | - J M D Araujo
- Programa de Pós-Graduação em Ciências Fisiológicas, Departamento de Fisiologia, Universidade Federal de Sergipe, São Cristóvão, SE, Brasil
| | - E V Souza
- Programa de Pós-Graduação em Ciências Fisiológicas, Departamento de Fisiologia, Universidade Federal de Sergipe, São Cristóvão, SE, Brasil
| | - E A Camargo
- Programa de Pós-Graduação em Ciências Fisiológicas, Departamento de Fisiologia, Universidade Federal de Sergipe, São Cristóvão, SE, Brasil
| | - R Grespan
- Programa de Pós-Graduação em Ciências Fisiológicas, Departamento de Fisiologia, Universidade Federal de Sergipe, São Cristóvão, SE, Brasil
| |
Collapse
|
8
|
Effects of ospemifene on the female reproductive and urinary tracts: translation from preclinical models into clinical evidence. Menopause 2016; 22:786-96. [PMID: 25423325 PMCID: PMC4481022 DOI: 10.1097/gme.0000000000000365] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Objective Treatment of menopausal symptoms by compounds with tissue-selective estrogen agonist/antagonist effects, often called selective estrogen receptor modulators, has been researched as an alternative to the use of estrogen therapy. These structurally diverse molecules elicit tissue-dependent responses in hormone-responsive tissues and organs, exhibiting variations in estrogenic activity in preclinical models of postmenopausal reproductive tissues that may improve postmenopausal women’s health (eg, prevention and treatment of breast cancer, osteoporosis, and vulvar and vaginal atrophy). Methods This literature review investigates whether preclinical data predicted the clinical effects of ospemifene on female reproductive and urinary tract tissues and compares these findings with the specific vaginal effects of other estrogen receptor agonists/antagonists (tamoxifen, raloxifene, and bazedoxifene) in preclinical and clinical studies. Lasofoxifene, although not currently available, is included because of its unique effects on vaginal tissue. Results The response of endometrial and vaginal tissues to estrogen receptor agonists/antagonists can be differentiated using transvaginal ultrasound, endometrial histopathology, cytologic examination of vaginal smears, assessment of physical changes in the vagina, and relief of symptoms associated with vulvar and vaginal atrophy (such as dyspareunia). Conclusions Available evidence indicates that ospemifene has unique effects on tissue, leading to a favorable long-term profile for the relief of vulvar and vaginal atrophy compared with other estrogen receptor agonists/antagonists (eg, tamoxifen, raloxifene, and bazedoxifene) with no short-term concerns about endometrial safety (based on endometrial hyperplasia, carcinoma, endometrial spotting, and endometrial bleeding).
Collapse
|
9
|
Integrated Bioinformatics, Environmental Epidemiologic and Genomic Approaches to Identify Environmental and Molecular Links between Endometriosis and Breast Cancer. Int J Mol Sci 2015; 16:25285-322. [PMID: 26512648 PMCID: PMC4632802 DOI: 10.3390/ijms161025285] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/10/2015] [Accepted: 10/12/2015] [Indexed: 12/29/2022] Open
Abstract
We present a combined environmental epidemiologic, genomic, and bioinformatics approach to identify: exposure of environmental chemicals with estrogenic activity; epidemiologic association between endocrine disrupting chemical (EDC) and health effects, such as, breast cancer or endometriosis; and gene-EDC interactions and disease associations. Human exposure measurement and modeling confirmed estrogenic activity of three selected class of environmental chemicals, polychlorinated biphenyls (PCBs), bisphenols (BPs), and phthalates. Meta-analysis showed that PCBs exposure, not Bisphenol A (BPA) and phthalates, increased the summary odds ratio for breast cancer and endometriosis. Bioinformatics analysis of gene-EDC interactions and disease associations identified several hundred genes that were altered by exposure to PCBs, phthalate or BPA. EDCs-modified genes in breast neoplasms and endometriosis are part of steroid hormone signaling and inflammation pathways. All three EDCs–PCB 153, phthalates, and BPA influenced five common genes—CYP19A1, EGFR, ESR2, FOS, and IGF1—in breast cancer as well as in endometriosis. These genes are environmentally and estrogen responsive, altered in human breast and uterine tumors and endometriosis lesions, and part of Mitogen Activated Protein Kinase (MAPK) signaling pathways in cancer. Our findings suggest that breast cancer and endometriosis share some common environmental and molecular risk factors.
Collapse
|
10
|
Jen SH, Wei MPS, Yin ACY. The Combinatory Effects of Glabridin and Tamoxifen on Ishikawa and MCF-7 Cell Lines. Nat Prod Commun 2015. [DOI: 10.1177/1934578x1501000922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Estrogen replacement therapy is commonly used to replace the loss of estrogen in post-menopausal women. However, it is not suitable to be used in women taking tamoxifen as both of the drugs increase the risk of endometrial cancer. This project aimed to study the potential of using the natural compound glabridin in combination with tamoxifen as a drug for estrogen replacement therapy. Ishikawa and MCF-7 cells were used to investigate the estrogenic activities stimulated by the combination of tamoxifen and glabridin through ALP and MTT assays. The expressions of the ESR1 and bcl-2 genes have also been determined using RT-PCR. The results indicated that the combination of 1×10−5M tamoxifen and 1×10−6M glabridin exhibited estrogenic activities and suppressed cell growth in both cell lines. The relative expressions of ESR1 and bcl-2 genes indicated that the estrogenicity expressed by the combinatory drug was regulated by estrogen receptor α; however, the reduction in cell proliferation was not modulated by bcl-2 anti-apoptotic proteins. These results suggested that the combination of tamoxifen and glabridin has potential to be used as an estrogen replacement drug with a reduced risk of endometrial cancer that has arisen from the intake of tamoxifen.
Collapse
Affiliation(s)
- Soe Hui Jen
- School of Biosciences, Taylor's University Lakeside Campus, Jalan Taylor's, 47500 Subang Jaya, Selangor, Malaysia
| | - Melissa Poh Su Wei
- School of Biosciences, Taylor's University Lakeside Campus, Jalan Taylor's, 47500 Subang Jaya, Selangor, Malaysia
| | - Adeline Chia Yoke Yin
- School of Biosciences, Taylor's University Lakeside Campus, Jalan Taylor's, 47500 Subang Jaya, Selangor, Malaysia
| |
Collapse
|
11
|
Wood CE, Branstetter D, Jacob AP, Cline JM, Register TC, Rohrbach K, Huang LY, Borgerink H, Dougall WC. Progestin effects on cell proliferation pathways in the postmenopausal mammary gland. Breast Cancer Res 2014; 15:R62. [PMID: 23938070 PMCID: PMC3978455 DOI: 10.1186/bcr3456] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 07/22/2013] [Indexed: 11/25/2022] Open
Abstract
Introduction Menopausal hormone therapies vary widely in their effects on breast cancer risk, and the mechanisms underlying these differences are unclear. The primary goals of this study were to characterize the mammary gland transcriptional profile of estrogen + progestin therapy in comparison with estrogen-alone or tibolone and investigate pathways of cell proliferation in a postmenopausal primate model. Methods Ovariectomized female cynomolgus macaque monkeys were randomized into the following groups: placebo (Con), oral conjugated equine estrogens (CEE), CEE with medroxyprogesterone acetate (MPA) (CEE + MPA), and tibolone given at a low or high dose (Lo or Hi Tib). All study treatment doses represented human clinical dose equivalents and were administered in the diet over a period of 2 years. Results Treatment with CEE + MPA had the greatest effect on global mRNA profiles and markers of mammary gland proliferation compared to CEE or tibolone treatment. Changes in the transcriptional patterns resulting from the addition of MPA to CEE were related to increased growth factors and decreased estrogen receptor (ER) signaling. Specific genes induced by CEE + MPA treatment included key members of prolactin receptor (PRLR)/signal transducer and activator of transcription 5 (STAT5), epidermal growth factor receptor (EGFR), and receptor activator of nuclear factor kappa B (RANK)/receptor activator of nuclear factor kappa B ligand (RANKL) pathways that were highly associated with breast tissue proliferation. In contrast, tibolone did not affect breast tissue proliferation but did elicit a mixed pattern of ER agonist activity. Conclusion Our findings indicate that estrogen + progestin therapy results in a distinct molecular profile compared to estrogen-alone or tibolone therapy, including upregulation of key growth factor targets associated with mammary carcinogenesis in mouse models. These changes may contribute to the promotional effects of estrogen + progestin therapy on breast cancer risk.
Collapse
|
12
|
Glass K, Girvan M. Annotation enrichment analysis: an alternative method for evaluating the functional properties of gene sets. Sci Rep 2014; 4:4191. [PMID: 24569707 PMCID: PMC3935204 DOI: 10.1038/srep04191] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 01/28/2014] [Indexed: 12/18/2022] Open
Abstract
Gene annotation databases (compendiums maintained by the scientific community that describe the biological functions performed by individual genes) are commonly used to evaluate the functional properties of experimentally derived gene sets. Overlap statistics, such as Fishers Exact test (FET), are often employed to assess these associations, but don't account for non-uniformity in the number of genes annotated to individual functions or the number of functions associated with individual genes. We find FET is strongly biased toward over-estimating overlap significance if a gene set has an unusually high number of annotations. To correct for these biases, we develop Annotation Enrichment Analysis (AEA), which properly accounts for the non-uniformity of annotations. We show that AEA is able to identify biologically meaningful functional enrichments that are obscured by numerous false-positive enrichment scores in FET, and we therefore suggest it be used to more accurately assess the biological properties of gene sets.
Collapse
Affiliation(s)
- Kimberly Glass
- 1] Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Department of Biostatistics, Harvard School of Public Health, Boston, MA, USA [3] Department of Physics, University of Maryland, College Park, MD, USA
| | - Michelle Girvan
- 1] Department of Physics, University of Maryland, College Park, MD, USA [2] Institute for Physical Science and Technology, University of Maryland, College Park, MD, USA [3] Santa Fe Institute, Santa Fe, NM
| |
Collapse
|
13
|
Hernandez-Ramon EE, Sandoval NA, John K, Cline JM, Wood CE, Woodward RA, Poirier MC. Tamoxifen-DNA adduct formation in monkey and human reproductive organs. Carcinogenesis 2014; 35:1172-6. [PMID: 24501327 DOI: 10.1093/carcin/bgu029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The estrogen analog tamoxifen (TAM), used for adjuvant therapy of breast cancer, induces endometrial and uterine tumors in breast cancer patients. Proliferation stimulus of the uterine endometrium is likely involved in tumor induction, but genotoxicity may also play a role. Formation of TAM-DNA adducts in human tissues has been reported but remains controversial. To address this issue, we examined TAM-DNA adducts in uteri from two species of monkeys, Erythrocebus patas (patas) and Macaca fascicularis (macaque), and in human endometrium and myometrium. Monkeys were given 3-4 months of chronic TAM dosing scaled to be equivalent to the daily human dose. In the uteri, livers and brains from the patas (n = 3), and endometrium from the macaques (n = 4), TAM-DNA adducts were measurable by TAM-DNA chemiluminescence immunoassay. Average TAM-DNA adduct values for the patas uteri (23 adducts/10(8) nucleotides) were similar to those found in endometrium of the macaques (19 adducts/10(8) nucleotides). Endometrium of macaques exposed to both TAM and low-dose estradiol (n = 5) averaged 34 adducts/10(8) nucleotides. To examine TAM-DNA persistence in the patas, females (n = 3) were exposed to TAM for 3 months and to no drug for an additional month, resulting in low or non-detectable TAM-DNA in livers and uteri. Human endometrial and myometrial samples from women receiving (n = 8) and not receiving (n = 8) TAM therapy were also evaluated. Women receiving TAM therapy averaged 10.3 TAM-DNA adducts/10(8) nucleotides, whereas unexposed women showed no detectable TAM-DNA. The data indicate that genotoxicity, in addition to estrogen agonist effects, may contribute to TAM-induced human endometrial cancer.
Collapse
Affiliation(s)
- Elena E Hernandez-Ramon
- Carcinogen-DNA Interactions Section, LCBG, CCR, National Cancer Institute, NIH, Building 37, Room 4032, NIH 37 Convent Drive, MSC-4255, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Endometrial profile of bazedoxifene acetate alone and in combination with conjugated equine estrogens in a primate model. Menopause 2014; 20:777-84. [PMID: 23793168 DOI: 10.1097/gme.0b013e31827ce57a] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Concerns of breast cancer risk in postmenopausal women taking combined estrogen + progestin therapy have generated interest in the use of selective estrogen receptor modulators (SERMs) as potential progestin alternatives. Endometrial proliferation and cancer risk are major concerns, however, for estrogens and certain types of SERMs when given alone. The primary aim of this study was to evaluate the endometrial profile of bazedoxifene acetate (BZA), a third-generation SERM, alone and in combination with conjugated equine estrogens (CEE) in a postmenopausal primate model. METHODS Ninety-eight ovariectomized cynomolgus monkeys (Macaca fascicularis) were randomized to receive no hormone treatment (controls), BZA 20 mg, CEE 0.45 mg, or the combination of BZA 20 mg + CEE 0.45 mg once daily for 20 months in a parallel-arm study design. The primary outcome measure was endometrial epithelial proliferation. RESULTS BZA + CEE and BZA treatment resulted in significantly less endometrial epithelial area and Ki67 expression compared with CEE (P < 0.001 for all). The prevalence of endometrial hyperplasia and other estrogen-induced morphologic changes in the BZA + CEE and BZA groups was not significantly different from controls. The addition of BZA to CEE completely inhibited the expression of estrogen receptor-α-regulated genes (TFF1 and PGR), whereas BZA alone had no effect. BZA + CEE and BZA treatment also resulted in lower estrogen receptor-α protein expression in the endometrium compared with the control and CEE groups (P < 0.05 for all). CONCLUSIONS BZA given at a clinically relevant dose inhibits estrogen effects on the endometrium and lacks uterotropic effects when given alone.
Collapse
|
15
|
Xu J, Liu L, Zheng X, You C, Li Q. Expression and inhibition of ADAMDEC1 in craniopharyngioma cells. Neurol Res 2013; 34:701-6. [PMID: 22776555 DOI: 10.1179/1743132812y.0000000067] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Jianguo Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Liang Liu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaomei Zheng
- Department of NeurologyPeople's Hospital of Luzhou City, China
| | - Chao You
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qiang Li
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Serrano D, Lazzeroni M, Gandini S, Macis D, Johansson H, Gjerde J, Lien E, Feroce I, Pruneri G, Sandri M, Bassi F, Brenelli F, Luini A, Cazzaniga M, Varricchio C, Guerrieri-Gonzaga A, DeCensi A, Bonanni B. A randomized phase II presurgical trial of weekly low-dose tamoxifen versus raloxifene versus placebo in premenopausal women with estrogen receptor-positive breast cancer. Breast Cancer Res 2013; 15:R47. [PMID: 23786776 PMCID: PMC4053157 DOI: 10.1186/bcr3439] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 06/20/2013] [Indexed: 11/27/2022] Open
Abstract
Introduction We previously demonstrated that 1 or 5 mg per day of tamoxifen (T) given for four weeks before surgery reduces Ki-67 in breast cancer (BC) patients to the same extent as the standard 20 mg/d. Given the long half-life of T, a weekly dose (10 mg per week (w)) may be worth testing. Also, raloxifene (R) has shown Ki-67 reduction in postmenopausal patients in a preoperative setting, but data in premenopausal women are limited. We conducted a randomized trial testing T 10 mg/w vs. R 60 mg/d vs. placebo in a presurgical model. Methods Out of 204 screened subjects, 57 were not eligible, 22 refused to participate and 125 were included in the study. The participants were all premenopausal women with estrogen receptor-positive BC. They were randomly assigned to either T 10mg/w or R 60 mg/d or placebo for six weeks before surgery. The primary endpoint was tissue change of Ki-67. Secondary endpoints were modulation of estrogen and progesterone receptors and several other circulating biomarkers. Results Ki-67 was not significantly modulated by either treatment. In contrast, both selective estrogen receptor modulators (SERMs) significantly modulated circulating IGF-I/IGFBP-3 ratio, cholesterol, fibrinogen and antithrombin III. Estradiol was increased with both SERMs. Within the tamoxifen arm, CYP2D6 polymorphism analysis showed a higher concentration of N-desTamoxifen, one of the tamoxifen metabolites, in subjects with reduced CYP2D6 activity. Moreover, a reduction of Ki-67 and a marked increase of sex hormone-binding globulin (SHBG) were observed in the active phenotype. Conclusions A weekly dose of tamoxifen and a standard dose of raloxifene did not inhibit tumor cell proliferation, measured as Ki-67 expression, in premenopausal BC patients. However, in the tamoxifen arm women with an extensive phenotype for CYP2D6 reached a significant Ki-67 modulation.
Collapse
|
17
|
Effects of bazedoxifene acetate with and without conjugated equine estrogens on the breast of postmenopausal monkeys. Menopause 2013; 19:1242-52. [PMID: 23103754 DOI: 10.1097/gme.0b013e318252e46d] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Concerns about increased breast cancer risk with estrogen and progestin therapy have led to an increased interest in progestin alternatives. The main objective of this study was to determine if bazedoxifene acetate (BZA), a new selective estrogen receptor modulator, will antagonize the proliferative and transcriptional effects of conjugated equine estrogens (CEE) in the breast. METHODS As part of a 20-month preclinical trial, 95 ovariectomized cynomolgus macaques (Macaca fascicularis) were randomized to receive no treatment or treatment with BZA (20 mg/d), CEE (0.45 mg/d), or BZA and CEE in combination (women's daily equivalent doses). The data presented here include breast effects after 6 months of treatment. Endpoints included histomorphometry, histopathological evaluations, gene microarray assays, polymerase chain reaction quantification of specific estrogen receptor α (ER-α) activity markers, and immunohistochemical detection of sex steroid receptors, and the proliferation marker Ki67. RESULTS BZA + CEE and BZA resulted in significantly less total epithelial density, lobular enlargement, and Ki67 immunolabeling in the terminal ducts compared with CEE alone (P < 0.05 for all). The addition of BZA to CEE antagonized the expression of ER-α-regulated genes such as GREB1 and TFF1 (P < 0.01 for both), whereas BZA alone had minimal effects on ER-α-mediated transcriptional activity. BZA and BZA + CEE did not significantly up-regulate genes related to cell cycle progression and proliferation. BZA with and without CEE also resulted in less lobular and terminal duct ER-α immunolabeling compared with control and CEE (P < 0.0001 for all). CONCLUSIONS These findings demonstrate that BZA given at a clinically relevant dose is an estrogen antagonist in the breast, supporting the idea that CEE + BZA may provide a lower breast cancer risk profile compared with traditional estrogen + progestin therapies.
Collapse
|
18
|
Bhukhai K, Suksen K, Bhummaphan N, Janjorn K, Thongon N, Tantikanlayaporn D, Piyachaturawat P, Suksamrarn A, Chairoungdua A. A phytoestrogen diarylheptanoid mediates estrogen receptor/Akt/glycogen synthase kinase 3β protein-dependent activation of the Wnt/β-catenin signaling pathway. J Biol Chem 2012; 287:36168-78. [PMID: 22936801 DOI: 10.1074/jbc.m112.344747] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Estrogen promotes growth in many tissues by activating Wnt/β-catenin signaling. Recently, ASPP 049, a diarylheptanoid isolated from Curcuma comosa Roxb., has been identified as a phytoestrogen. This investigation determined the involvement of Wnt/β-catenin signaling in the estrogenic activity of this diarylheptanoid in transfected HEK 293T and in mouse preosteoblastic (MC3T3-E1) cells using a TOPflash luciferase assay and immunofluorescence. ASPP 049 rapidly activated T-cell-specific transcription factor/lymphoid enhancer binding factor-mediated transcription activity and induced β-catenin accumulation in the nucleus. Interestingly, the effects of ASPP 049 on the transcriptional activity and induction and accumulation of β-catenin protein in the nucleus of MC3T3-E1 cells were greater compared with estradiol. Activation of β-catenin in MC3T3-E1 cells was inhibited by ICI 182,780, suggesting that an estrogen receptor is required. In addition, ASPP 049 induced phosphorylations at serine 473 of Akt and serine 9 of GSK-3β. Moreover, ASPP 049 also induced proliferation and expressions of Wnt target genes Axin2 and Runx2 in MC3T3-E1 cells. In addition, ASPP 049 increased alkaline phosphatase expression, and activity that was abolished by DKK-1, a blocker of the Wnt/β-catenin receptor. Taken together, these results suggest that ASPP 049 from C. comosa induced osteoblastic cell proliferation and differentiation through ERα-, Akt-, and GSK-3β-dependent activation of β-catenin signaling. Our findings provide a scientific rationale for using C. comosa as a dietary supplement to prevent bone loss in postmenopausal women.
Collapse
Affiliation(s)
- Kanit Bhukhai
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
|
20
|
Copy number variants in pharmacogenetic genes. Trends Mol Med 2011; 17:244-51. [PMID: 21388883 DOI: 10.1016/j.molmed.2011.01.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 01/14/2011] [Accepted: 01/14/2011] [Indexed: 01/07/2023]
Abstract
Variation in drug efficacy and toxicity remains an important clinical concern. Presently, single nucleotide polymorphisms (SNPs) only explain a portion of this problem, even in situations where the pharmacological trait is clearly heritable. The Human CNV Project identified copy number variations (CNVs) across approximately 12% of the human genome, and these CNVs were considered causes of diseases. Although the contribution of CNVs to the pathogenesis of many common diseases is questionable, CNVs play a clear role in drug-related genes by altering drug metabolizing and drug response. In this review, we provide a comprehensive evaluation of the clinical relevance of CNVs to drug efficacy, toxicity, and disease prevalence in world populations, and discuss the implication of using CNVs as a diagnostic tool in clinical intervention.
Collapse
|
21
|
de Almeida Chuffa LG, de Souza RB, Frei F, de Fátima Paccola Mesquita S, Camargo ICC. Nandrolone Decanoate and Physical Effort: Histological and Morphometrical Assessment in Adult Rat Uterus. Anat Rec (Hoboken) 2010; 294:335-41. [DOI: 10.1002/ar.21314] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Accepted: 10/09/2010] [Indexed: 11/12/2022]
|
22
|
Cognitive changes associated with endocrine therapy for breast cancer. Maturitas 2010; 67:209-14. [PMID: 20688441 DOI: 10.1016/j.maturitas.2010.07.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2010] [Revised: 07/09/2010] [Accepted: 07/09/2010] [Indexed: 01/22/2023]
Abstract
Endocrine therapy in the setting of breast cancer has undoubtedly advanced clinical outcomes in this disease, but treatment with endocrine therapy is accompanied by a wide spectrum of side effects. It is of prime importance to understand and characterize these toxicities to facilitate clinical decision-making. Somewhat surprisingly, there is a relative paucity of data pertaining to cognitive changes associated with endocrine therapy. In this article we review cognitive associated with two classes of endocrine therapy: (1) selective estrogen receptor modulators (SERMs; tamoxifen and raloxifene) and (2) aromatase inhibitors (AIs; anastrozole, letrozole, and exemestane). Companion studies to the Multiple Outcome of Raloxifene Evaluation (MORE), the Study of Tamoxifen and Raloxifene (STAR) and National Surgical Adjuvant Breast and Bowel Project (NSABP) P-1 trials provide relevant data to understand the effect of SERMs on cognition. In contrast, substudies of the Arimidex, Tamoxifen Alone or in Combination (ATAC), Tamoxifen and Exemestane Adjuvant Multinational (TEAM) and Breast International Group (BIG) 1-98 trials juxtapose cognitive effects of AIs against those of tamoxifen. These and other studies are examined herein to provide a comprehensive overview of the effect of endocrine therapy on cognition.
Collapse
|