1
|
Sun G, Yang Y, Liu J, Gao Z, Xu T, Chai J, Xu J, Fan Z, Xiao T, Jia Q, Li M. Cancer stem cells in esophageal squamous cell carcinoma. Pathol Res Pract 2022; 237:154043. [DOI: 10.1016/j.prp.2022.154043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/21/2022] [Accepted: 07/26/2022] [Indexed: 02/07/2023]
|
2
|
Tassi E, Lai EY, Li L, Solis G, Chen Y, Kietzman WE, Ray PE, Riegel AT, Welch WJ, Wilcox CS, Wellstein A. Blood Pressure Control by a Secreted FGFBP1 (Fibroblast Growth Factor-Binding Protein). Hypertension 2018; 71:160-167. [PMID: 29158353 PMCID: PMC5730494 DOI: 10.1161/hypertensionaha.117.10268] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 10/06/2017] [Accepted: 10/13/2017] [Indexed: 12/29/2022]
Abstract
Fibroblast growth factors (FGFs) participate in organ development and tissue maintenance, as well as the control of vascular function. The paracrine-acting FGFs are stored in the extracellular matrix, and their release is controlled by a secreted FGF-binding protein (FGF-BP, FGFBP1, and BP1) that modulates FGF receptor signaling. A genetic polymorphism in the human FGFBP1 gene was associated with higher gene expression and an increased risk of familial hypertension. Here, we report on the effects of inducible BP1 expression in a transgenic mouse model. Induction of BP1 expression in adult animals leads to a sustained rise in mean arterial pressure by >30 mm Hg. The hypertensive effect of BP1 expression is prevented by candesartan, an angiotensin II (AngII) receptor antagonist, or by tempol, an inhibitor of reactive oxygen species. In vivo, BP1 expression sensitizes peripheral resistance vessels to AngII constriction by 20-fold but does not alter adrenergic vasoconstriction. FGF receptor kinase inhibition reverses the sensitization to AngII. Also, constriction of isolated renal afferent arterioles by AngII is enhanced after BP1 expression and blocked by FGF receptor kinase inhibition. Furthermore, AngII-mediated constriction of renal afferent arterioles is abolished in FGF2-/- mice but can be restored by add-back of FGF2 plus BP1 proteins. In contrast to AngII, adrenergic constriction is not affected in the FGF2-/- model. Proteomics and gene expression analysis of kidney tissues after BP1 induction show that MAPK (mitogen-activated protein kinase) signaling via MKK4 (MAPK kinase 4), p38, and JNK (c-Jun N-terminal kinase) integrates the crosstalk of the FGF receptor and AngII pathways and thus impact vascular tone and blood pressure.
Collapse
Affiliation(s)
- Elena Tassi
- From the Lombardi Cancer Center (E.T., W.E.K., A.T.R., A.W.) and Division of Nephrology and Hypertension (E.Y.L., L.L., G.S., Y.C., W.J.W., C.S.W.), Georgetown University, Washington, DC; Department of Physiology, Zhejiang University, Hangzhou, China (E.Y.L.); and Children's National Medical Center, George Washington University, DC (P.E.R.)
| | - En Yin Lai
- From the Lombardi Cancer Center (E.T., W.E.K., A.T.R., A.W.) and Division of Nephrology and Hypertension (E.Y.L., L.L., G.S., Y.C., W.J.W., C.S.W.), Georgetown University, Washington, DC; Department of Physiology, Zhejiang University, Hangzhou, China (E.Y.L.); and Children's National Medical Center, George Washington University, DC (P.E.R.)
| | - Lingli Li
- From the Lombardi Cancer Center (E.T., W.E.K., A.T.R., A.W.) and Division of Nephrology and Hypertension (E.Y.L., L.L., G.S., Y.C., W.J.W., C.S.W.), Georgetown University, Washington, DC; Department of Physiology, Zhejiang University, Hangzhou, China (E.Y.L.); and Children's National Medical Center, George Washington University, DC (P.E.R.)
| | - Glenn Solis
- From the Lombardi Cancer Center (E.T., W.E.K., A.T.R., A.W.) and Division of Nephrology and Hypertension (E.Y.L., L.L., G.S., Y.C., W.J.W., C.S.W.), Georgetown University, Washington, DC; Department of Physiology, Zhejiang University, Hangzhou, China (E.Y.L.); and Children's National Medical Center, George Washington University, DC (P.E.R.)
| | - Yifan Chen
- From the Lombardi Cancer Center (E.T., W.E.K., A.T.R., A.W.) and Division of Nephrology and Hypertension (E.Y.L., L.L., G.S., Y.C., W.J.W., C.S.W.), Georgetown University, Washington, DC; Department of Physiology, Zhejiang University, Hangzhou, China (E.Y.L.); and Children's National Medical Center, George Washington University, DC (P.E.R.)
| | - William E Kietzman
- From the Lombardi Cancer Center (E.T., W.E.K., A.T.R., A.W.) and Division of Nephrology and Hypertension (E.Y.L., L.L., G.S., Y.C., W.J.W., C.S.W.), Georgetown University, Washington, DC; Department of Physiology, Zhejiang University, Hangzhou, China (E.Y.L.); and Children's National Medical Center, George Washington University, DC (P.E.R.)
| | - Patricio E Ray
- From the Lombardi Cancer Center (E.T., W.E.K., A.T.R., A.W.) and Division of Nephrology and Hypertension (E.Y.L., L.L., G.S., Y.C., W.J.W., C.S.W.), Georgetown University, Washington, DC; Department of Physiology, Zhejiang University, Hangzhou, China (E.Y.L.); and Children's National Medical Center, George Washington University, DC (P.E.R.)
| | - Anna T Riegel
- From the Lombardi Cancer Center (E.T., W.E.K., A.T.R., A.W.) and Division of Nephrology and Hypertension (E.Y.L., L.L., G.S., Y.C., W.J.W., C.S.W.), Georgetown University, Washington, DC; Department of Physiology, Zhejiang University, Hangzhou, China (E.Y.L.); and Children's National Medical Center, George Washington University, DC (P.E.R.)
| | - William J Welch
- From the Lombardi Cancer Center (E.T., W.E.K., A.T.R., A.W.) and Division of Nephrology and Hypertension (E.Y.L., L.L., G.S., Y.C., W.J.W., C.S.W.), Georgetown University, Washington, DC; Department of Physiology, Zhejiang University, Hangzhou, China (E.Y.L.); and Children's National Medical Center, George Washington University, DC (P.E.R.)
| | - Christopher S Wilcox
- From the Lombardi Cancer Center (E.T., W.E.K., A.T.R., A.W.) and Division of Nephrology and Hypertension (E.Y.L., L.L., G.S., Y.C., W.J.W., C.S.W.), Georgetown University, Washington, DC; Department of Physiology, Zhejiang University, Hangzhou, China (E.Y.L.); and Children's National Medical Center, George Washington University, DC (P.E.R.)
| | - Anton Wellstein
- From the Lombardi Cancer Center (E.T., W.E.K., A.T.R., A.W.) and Division of Nephrology and Hypertension (E.Y.L., L.L., G.S., Y.C., W.J.W., C.S.W.), Georgetown University, Washington, DC; Department of Physiology, Zhejiang University, Hangzhou, China (E.Y.L.); and Children's National Medical Center, George Washington University, DC (P.E.R.).
| |
Collapse
|
3
|
Alkner S, Bendahl PO, Ehinger A, Lövgren K, Rydén L, Fernö M. Prior Adjuvant Tamoxifen Treatment in Breast Cancer Is Linked to Increased AIB1 and HER2 Expression in Metachronous Contralateral Breast Cancer. PLoS One 2016; 11:e0150977. [PMID: 26959415 PMCID: PMC4784945 DOI: 10.1371/journal.pone.0150977] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 02/22/2016] [Indexed: 01/22/2023] Open
Abstract
Aim The estrogen receptor coactivator Amplified in Breast Cancer 1 (AIB1) has been associated with an improved response to adjuvant tamoxifen in breast cancer, but also with endocrine treatment resistance. We hereby use metachronous contralateral breast cancer (CBC) developed despite prior adjuvant tamoxifen for the first tumor as an “in vivo”-model for tamoxifen resistance. AIB1-expression in the presumable resistant (CBC after prior tamoxifen) and naïve setting (CBC without prior tamoxifen) is compared and correlated to prognosis after CBC. Methods From a well-defined population-based cohort of CBC-patients we have constructed a unique tissue-microarray including >700 patients. Results CBC developed after adjuvant tamoxifen more often had a HER2-positive/triple negative-subtype and a high AIB1-expression (37% vs. 23%, p = 0.009), than if no prior endocrine treatment had been administered. In patients with an estrogen receptor (ER) positive CBC, a high AIB1-expression correlated to an inferior prognosis. However, these patients seemed to respond to tamoxifen, but only if endocrine therapy had not been administered for BC1. Conclusions Metachronous CBC developed after prior endocrine treatment has a decreased ER-expression and an increased HER2-expression. This is consistent with endocrine treatment escape mechanisms previously suggested, and indicates metachronous CBC to be a putative model for studies of treatment resistance “in vivo”. The increased AIB1-expression in CBC developed after prior tamoxifen suggests a role of AIB1 in endocrine treatment resistance. In addition, we found indications that the response to tamoxifen in CBC with a high AIB1-expression seem to differ depending on previous exposure to this drug. A different function for AIB1 in the tamoxifen treatment naïve vs. resistant setting is suggested, and may explain previously conflicting results where a high AIB1-expression has been correlated to both a good response to adjuvant tamoxifen and tamoxifen resistance.
Collapse
Affiliation(s)
- Sara Alkner
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Medicon Village, SE-223 63, Lund, Sweden
- Skåne Clinic of Oncology, Skåne University Hospital Lund, SE-222 41, Lund, Sweden
- * E-mail:
| | - Pär-Ola Bendahl
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Medicon Village, SE-223 63, Lund, Sweden
| | - Anna Ehinger
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Medicon Village, SE-223 63, Lund, Sweden
- Department of Pathology and Cytology, Blekinge County Hospital, SE-371 85, Karlskrona, Sweden
| | - Kristina Lövgren
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Medicon Village, SE-223 63, Lund, Sweden
| | - Lisa Rydén
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Medicon Village, SE-223 63, Lund, Sweden
- Clinic of Surgery, Skåne University Hospital Lund, SE-222 41, Lund, Sweden
| | - Mårten Fernö
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Medicon Village, SE-223 63, Lund, Sweden
| |
Collapse
|
4
|
Ryan N, Chorley B, Tice RR, Judson R, Corton JC. Moving Toward Integrating Gene Expression Profiling Into High-Throughput Testing: A Gene Expression Biomarker Accurately Predicts Estrogen Receptor α Modulation in a Microarray Compendium. Toxicol Sci 2016; 151:88-103. [PMID: 26865669 DOI: 10.1093/toxsci/kfw026] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Microarray profiling of chemical-induced effects is being increasingly used in medium- and high-throughput formats. Computational methods are described here to identify molecular targets from whole-genome microarray data using as an example the estrogen receptor α (ERα), often modulated by potential endocrine disrupting chemicals. ERα biomarker genes were identified by their consistent expression after exposure to 7 structurally diverse ERα agonists and 3 ERα antagonists in ERα-positive MCF-7 cells. Most of the biomarker genes were shown to be directly regulated by ERα as determined by ESR1 gene knockdown using siRNA as well as through chromatin immunoprecipitation coupled with DNA sequencing analysis of ERα-DNA interactions. The biomarker was evaluated as a predictive tool using the fold-change rank-based Running Fisher algorithm by comparison to annotated gene expression datasets from experiments using MCF-7 cells, including those evaluating the transcriptional effects of hormones and chemicals. Using 141 comparisons from chemical- and hormone-treated cells, the biomarker gave a balanced accuracy for prediction of ERα activation or suppression of 94% and 93%, respectively. The biomarker was able to correctly classify 18 out of 21 (86%) ER reference chemicals including "very weak" agonists. Importantly, the biomarker predictions accurately replicated predictions based on 18 in vitro high-throughput screening assays that queried different steps in ERα signaling. For 114 chemicals, the balanced accuracies were 95% and 98% for activation or suppression, respectively. These results demonstrate that the ERα gene expression biomarker can accurately identify ERα modulators in large collections of microarray data derived from MCF-7 cells.
Collapse
Affiliation(s)
- Natalia Ryan
- *Oak Ridge Institute for Science and Education (ORISE) Integrated Systems Toxicology Division, US-EPA
| | | | - Raymond R Tice
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences
| | - Richard Judson
- National Center for Computational Toxicology, US-EPA, Research Triangle Park, North Carolina 27711
| | | |
Collapse
|
5
|
Obiorah IE, Jordan VC. Differences in the rate of oestrogen-induced apoptosis in breast cancer by oestradiol and the triphenylethylene bisphenol. Br J Pharmacol 2015; 171:4062-72. [PMID: 24819221 DOI: 10.1111/bph.12762] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 04/16/2014] [Accepted: 04/25/2014] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND AND PURPOSE Triphenylethylene (TPE)-like compounds were the first agents to be used in the treatment of metastatic breast cancer in postmenopausal women. Although structurally related to the anti-oestrogen, 4-hydroxytamoxifen, TPEs possess oestrogenic properties in fully oestrogenized breast cancer cells but do not induce apoptosis with short-term treatment in long-term oestrogen-deprived breast cancer cells. This study determined the differential effects of bisphenol, a TPE, on growth and apoptosis based on the modulation of the shape of the ligand-oestrogen receptor complex. EXPERIMENTAL APPROACH Apoptotic flow cytometric studies were used to evaluate apoptosis over time. Proliferation of the breast cancer cells was assessed using DNA quantification and cell cycle analysis. Real-time PCR was performed to quantify mRNA levels of apoptotic genes. Regulation of cell cycle and apoptotic genes was determined using PCR-based arrays. KEY RESULTS Bisphenol induced an up-regulation of cell cycle genes similar to those induced by 17β oestradiol (E2 ). Unlike the changes induced by E2 that occur after 24 h, the apoptosis evoked by bisphenol occurred after 4 days, with quantifiable apoptotic changes noted at 6 days. A prolonged up-regulation of endoplasmic reticulum stress and inflammatory stress response genes was observed with subsequent activation of apoptosis-related genes in the second week of treatment with bisphenol. CONCLUSIONS AND IMPLICATIONS The bisphenol: ERα complex induces delayed biological effects on the growth and apoptosis of breast cancer cells. Both the shape of the complex and the duration of treatment control the initiation of apoptosis.
Collapse
Affiliation(s)
- I E Obiorah
- Tumor Biology Training Program, Georgetown University, Washington, DC, USA
| | | |
Collapse
|
6
|
Abstract
The successful use of high-dose synthetic estrogens to treat postmenopausal metastatic breast cancer is the first effective 'chemical therapy' proven in clinical trial to treat any cancer. This review documents the clinical use of estrogen for breast cancer treatment or estrogen replacement therapy (ERT) in postmenopausal hysterectomized women, which can either result in breast cancer cell growth or breast cancer regression. This has remained a paradox since the 1950s until the discovery of the new biology of estrogen-induced apoptosis at the end of the 20th century. The key to triggering apoptosis with estrogen is the selection of breast cancer cell populations that are resistant to long-term estrogen deprivation. However, estrogen-independent growth occurs through trial and error. At the cellular level, estrogen-induced apoptosis is dependent upon the presence of the estrogen receptor (ER), which can be blocked by nonsteroidal or steroidal antiestrogens. The shape of an estrogenic ligand programs the conformation of the ER complex, which, in turn, can modulate estrogen-induced apoptosis: class I planar estrogens (e.g., estradiol) trigger apoptosis after 24 h, whereas class II angular estrogens (e.g., bisphenol triphenylethylene) delay the process until after 72 h. This contrasts with paclitaxel, which causes G2 blockade with immediate apoptosis. The process is complete within 24 h. Estrogen-induced apoptosis is modulated by glucocorticoids and cSrc inhibitors, but the target mechanism for estrogen action is genomic and not through a nongenomic pathway. The process is stepwise through the creation of endoplasmic reticulum stress and inflammatory responses, which then initiate an unfolded protein response. This, in turn, initiates apoptosis through the intrinsic pathway (mitochondrial) with the subsequent recruitment of the extrinsic pathway (death receptor) to complete the process. The symmetry of the clinical and laboratory studies now permits the creation of rules for the future clinical application of ERT or phytoestrogen supplements: a 5-year gap is necessary after menopause to permit the selection of estrogen-deprived breast cancer cell populations to cause them to become vulnerable to apoptotic cell death. Earlier treatment with estrogen around menopause encourages growth of ER-positive tumor cells, as the cells are still dependent on estrogen to maintain replication within the expanding population. An awareness of the evidence that the molecular events associated with estrogen-induced apoptosis can be orchestrated in the laboratory in estrogen-deprived breast cancers now supports the clinical findings regarding the treatment of metastatic breast cancer following estrogen deprivation, decreases in mortality following long-term antihormonal adjuvant therapy, and the results of treatment with ERT and ERT plus progestin in the Women's Health Initiative for women over the age of 60. Principles have emerged for understanding and applying physiological estrogen therapy appropriately by targeting the correct patient populations.
Collapse
Affiliation(s)
- V Craig Jordan
- Departments of Breast Medical Oncology and Molecular and Cellular OncologyMD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
7
|
HEGER ZBYNEK, GUMULEC JAROMIR, CERNEI NATALIA, TMEJOVA KATERINA, KOPEL PAVEL, BALVAN JAN, MASARIK MICHAL, ZITKA ONDREJ, BEKLOVA MIROSLAVA, ADAM VOJTECH, KIZEK RENE. 17β-estradiol-containing liposomes as a novel delivery system for the antisense therapy of ER-positive breast cancer: An in vitro study on the MCF-7 cell line. Oncol Rep 2014; 33:921-9. [DOI: 10.3892/or.2014.3627] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 07/11/2014] [Indexed: 11/06/2022] Open
|
8
|
Ghasemi O, Ma Y, Lindsey ML, Jin YF. Using systems biology approaches to understand cardiac inflammation and extracellular matrix remodeling in the setting of myocardial infarction. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2014; 6:77-91. [PMID: 24741709 DOI: 10.1002/wsbm.1248] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Inflammation and extracellular matrix (ECM) remodeling are important components regulating the response of the left ventricle to myocardial infarction (MI). Significant cellular- and molecular-level contributors can be identified by analyzing data acquired through high-throughput genomic and proteomic technologies that provide expression levels for thousands of genes and proteins. Large-scale data provide both temporal and spatial information that need to be analyzed and interpreted using systems biology approaches in order to integrate this information into dynamic models that predict and explain mechanisms of cardiac healing post-MI. In this review, we summarize the systems biology approaches needed to computationally simulate post-MI remodeling, including data acquisition, data analysis for biomarker classification and identification, data integration to build dynamic models, and data interpretation for biological functions. An example for applying a systems biology approach to ECM remodeling is presented as a reference illustration.
Collapse
|
9
|
Sengupta S, Obiorah I, Maximov PY, Curpan R, Jordan VC. Molecular mechanism of action of bisphenol and bisphenol A mediated by oestrogen receptor alpha in growth and apoptosis of breast cancer cells. Br J Pharmacol 2014; 169:167-78. [PMID: 23373633 DOI: 10.1111/bph.12122] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 12/21/2012] [Accepted: 01/08/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Oestrogen receptor alpha (ERα) binds to different ligand which can function as complete/partial oestrogen-agonist or antagonist. This depends on the chemical structure of the ligands which modulates the transcriptional activity of the oestrogen-responsive genes by altering the conformation of the liganded-ERα complex. This study determined the molecular mechanism of oestrogen-agonistic/antagonistic action of structurally similar ligands, bisphenol (BP) and bisphenol A (BPA) on cell proliferation and apoptosis of ERα + ve breast cancer cells. EXPERIMENTAL APPROACH DNA was measured to assess the proliferation and apoptosis of breast cancer cells. RT-PCR and ChIP assays were performed to quantify the transcripts of TFF1 gene and recruitment of ERα and SRC3 at the promoter of TFF1 gene respectively. Molecular docking was used to delineate the binding modes of BP and BPA with the ERα. PCR-based arrays were used to study the regulation of the apoptotic genes. KEY RESULTS BP and BPA induced the proliferation of breast cancer cells; however, unlike BPA, BP failed to induce apoptosis. BPA consistently acted as an agonist in our studies but BP exhibited mixed agonistic/antagonistic properties. Molecular docking revealed agonistic and antagonistic mode of binding for BPA and BP respectively. BPA treatment resembled E2 treatment in terms of PCR-based regulation of apoptotic genes whereas BP was similar to 4OHT treatment. CONCLUSIONS AND IMPLICATIONS The chemical structure of ERα ligand determines the agonistic or antagonistic biological responses by the virtue of their binding mode, conformation of the liganded-ERα complex and the context of the cellular function.
Collapse
Affiliation(s)
- S Sengupta
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | | | | | | | | |
Collapse
|
10
|
Obiorah I, Sengupta S, Curpan R, Jordan VC. Defining the conformation of the estrogen receptor complex that controls estrogen-induced apoptosis in breast cancer. Mol Pharmacol 2014; 85:789-99. [PMID: 24608856 DOI: 10.1124/mol.113.089250] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Development of acquired antihormone resistance exposes a vulnerability in breast cancer: estrogen-induced apoptosis. Triphenylethylenes (TPEs), which are structurally similar to 4-hydroxytamoxifen (4OHT), were used for mechanistic studies of estrogen-induced apoptosis. These TPEs all stimulate growth in MCF-7 cells, but unlike the planar estrogens they block estrogen-induced apoptosis in the long-term estrogen-deprived MCF7:5C cells. To define the conformation of the TPE:estrogen receptor (ER) complex, we employed a previously validated assay using the induction of transforming growth factor α (TGFα) mRNA in situ in MDA-MB 231 cells stably transfected with wild-type ER (MC2) or D351G ER mutant (JM6). The assays discriminate ligand fit in the ER based on the extremes of published crystallography of planar estrogens or TPE antiestrogens. We classified the conformation of planar estrogens or angular TPE complexes as "estrogen-like" or "antiestrogen-like" complexes, respectively. The TPE:ER complexes did not readily recruit the coactivator steroid receptor coactivator-3 (SRC3) or ER to the PS2 promoter in MCF-7 and MCF7:5C cells, and molecular modeling showed that they prefer to bind to the ER in an antagonistic fashion, i.e., helix 12 not sealing the ligand binding domain (LBD) effectively, and therefore reduce critical SRC3 binding. The fully activated ER complex with helix 12 sealing the LBD is suggested to be the appropriate trigger to initiate rapid estrogen-induced apoptosis.
Collapse
Affiliation(s)
- Ifeyinwa Obiorah
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia (I.O., S.S., V.C.J.); and Institute of Chemistry, Romanian Academy, Timisoara, Romania (R.C.)
| | | | | | | |
Collapse
|
11
|
Scientific rationale for postmenopause delay in the use of conjugated equine estrogens among postmenopausal women that causes reduction in breast cancer incidence and mortality. Menopause 2014; 20:372-82. [PMID: 23921472 DOI: 10.1097/gme.0b013e31828865a5] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
High-dose synthetic estrogens were the first successful chemical therapy used in the treatment of metastatic breast cancer in postmenopausal women, and this approach became the standard of care in postmenopausal women with metastatic breast cancer between the 1950s and the end of the 1970s. The most recent analysis of the Women's Health Initiative estrogen-alone trial in hysterectomized women revealed a persistently significant decrease in the incidence of breast cancer and breast cancer mortality. Although estrogens are known to induce the proliferation of breast cancer cells, we have shown that physiologic concentrations induce apoptosis in breast cancer cells with long-term estrogen deprivation. We have developed laboratory models that illustrate the new biology of estrogen-induced apoptosis or growth to explain the effects of estrogen therapy. The key to the success of estrogen therapy lies in a sufficient period of withdrawal of physiologic estrogens (5-10 y) and the subsequent regrowth of nascent breast tumor cells that survive under estrogen-deprived conditions. These nascent tumors are now vulnerable to estrogen-induced apoptosis.
Collapse
|
12
|
Pinto G, Alhaiek AAM, Amadi S, Qattan AT, Crawford M, Radulovic M, Godovac-Zimmermann J. Systematic nucleo-cytoplasmic trafficking of proteins following exposure of MCF7 breast cancer cells to estradiol. J Proteome Res 2014; 13:1112-27. [PMID: 24422525 DOI: 10.1021/pr4012359] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We have used a proteomics subcellular spatial razor approach to look at changes in total protein abundance and in protein distribution between the nucleus and cytoplasm following exposure of MCF7 breast cancer cells to estradiol. The dominant response of MCF7 cells to estrogen stimulation involves dynamic changes in protein subcellular spatial distribution rather than changes in total protein abundance. Of the 3604 quantitatively monitored proteins, only about 2% show substantial changes in total abundance (>2-fold), whereas about 20% of the proteins show substantial changes in local abundance and/or redistribution of their subcellular location, with up to 16-fold changes in their local concentration in the nucleus or the cytoplasm. We propose that dynamic redistribution of the subcellular location of multiple proteins in response to stimuli is a fundamental characteristic of cells and suggest that perturbation of cellular spatial control may be an important feature of cancer.
Collapse
Affiliation(s)
- Gabriella Pinto
- Proteomics and Molecular Cell Dynamics, Center for Nephrology, Division of Medicine, School of Life and Medical Sciences, University College London , Royal Free Campus, Rowland Hill Street, London NW3 2PF, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
13
|
Pick H, Terrettaz S, Baud O, Laribi O, Brisken C, Vogel H. Monitoring proliferative activities of hormone-like odorants in human breast cancer cells by gene transcription profiling and electrical impedance spectroscopy. Biosens Bioelectron 2013; 50:431-6. [DOI: 10.1016/j.bios.2013.06.052] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 06/21/2013] [Accepted: 06/24/2013] [Indexed: 12/22/2022]
|
14
|
Li JV, Chien CD, Garee JP, Xu J, Wellstein A, Riegel AT. Transcriptional repression of AIB1 by FoxG1 leads to apoptosis in breast cancer cells. Mol Endocrinol 2013; 27:1113-27. [PMID: 23660594 DOI: 10.1210/me.2012-1353] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The oncogene nuclear receptor coactivator amplified in breast cancer 1 (AIB1) is a transcriptional coactivator that is overexpressed in various types of human cancers. However, the molecular mechanisms controlling AIB1 expression in the majority of cancers remain unclear. In this study, we identified a novel interacting protein of AIB1, forkhead-box protein G1 (FoxG1), which is an evolutionarily conserved forkhead-box transcriptional corepressor. We show that FoxG1 expression is low in breast cancer cell lines and that low levels of FoxG1 are correlated with a worse prognosis in breast cancer. We also demonstrate that transient overexpression of FoxG1 can suppress endogenous levels of AIB1 mRNA and protein in MCF-7 breast cancer cells. Exogenously expressed FoxG1 in MCF-7 cells also leads to apoptosis that can be rescued in part by AIB1 overexpression. Using chromatin immunoprecipitation, we determined that FoxG1 is recruited to a region of the AIB1 gene promoter previously characterized to be responsible for AIB1-induced, positive autoregulation of transcription through the recruitment of an activating, multiprotein complex, involving AIB1, E2F transcription factor 1, and specificity protein 1. Increased FoxG1 expression significantly reduces the recruitment of AIB1, E2F transcription factor 1 and E1A-binding protein p300 to this region of the endogenous AIB1 gene promoter. Our data imply that FoxG1 can function as a pro-apoptotic factor in part through suppression of AIB1 coactivator transcription complex formation, thereby reducing the expression of the AIB1 oncogene.
Collapse
Affiliation(s)
- Jordan V Li
- Department of Pharmacology, Lombardi Cancer Center, Georgetown University, Research Building E307, 3970 Reservoir Road Northwest, Washington, DC 20007-2197, USA
| | | | | | | | | | | |
Collapse
|
15
|
Alkner S, Bendahl P, Grabau D, Malmström P, Fernö M, Rydén L. The role of AIB1 and PAX2 in primary breast cancer: validation of AIB1 as a negative prognostic factor. Ann Oncol 2013; 24:1244-52. [DOI: 10.1093/annonc/mds613] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
16
|
Santen RJ, Yue W, Heitjan DF. Occult breast tumor reservoir: biological properties and clinical significance. Discov Oncol 2013; 4:195-207. [PMID: 23632998 DOI: 10.1007/s12672-013-0145-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 04/11/2013] [Indexed: 12/31/2022] Open
Abstract
Small, occult, undiagnosed breast cancers are found at autopsy in up to 15.6 % of women dying from unrelated causes with an average of 7 % from eight separate studies. The mammographic detection threshold of breast tumors ranges from 0.88 to 1.66 cm in diameter based on the patient's age. Tumor growth rates, expressed as "effective doubling times," vary from 10 to >700 days. We previously reported two models, based on iterative analysis of these parameters, to describe the biologic behavior of undiagnosed, occult breast tumors. Our models facilitate interpretation of the Women's Health Initiative (WHI) and antiestrogen breast cancer prevention studies. A nude mouse xenograft model was used to validate our assumption that breast tumors grow in a log-linear fashion. We then used our previously reported occult tumor growth (OTG) and computer-simulated tumor growth models to analyze various clinical trial data. Parameters used in the OTG model included a 200-day effective doubling time, 7 % prevalence of occult tumors, and 1.16 cm detection threshold. These models had been validated by comparing predicted with observed incidence of breast cancer in eight different populations of women. Our model suggests that menopausal hormone therapy with estrogens plus a progestogen (E + P) in the WHI trial primarily promoted the growth of pre-existing, occult lesions and minimally initiated de novo tumors. We provide a potential explanation for the lack of an increase in breast cancer incidence in the subgroup of women in the WHI who had not received E + P prior to randomization. This result may have reflected a leftward skew in the distribution of occult tumor doublings and insufficient time for stimulated tumors to reach the detection threshold. Our model predicted that estrogen alone reduced the incidence of breast cancer as a result of apoptosis. Understanding of the biology of occult tumors suggests that breast cancer "prevention" with antiestrogens or aromatase inhibitors represents early treatment rather than a reduction in de novo tumor formation. Our models suggest that occult, undiagnosed tumors are prevalent, grow slowly, and are the biologic targets of a hormone therapy in menopausal women and of antiestrogen therapy for prevention.
Collapse
Affiliation(s)
- Richard J Santen
- Department of Internal Medicine, Division of Endocrinology, University of Virginia, Charlottesville, VA 22908-1416, USA.
| | | | | |
Collapse
|
17
|
McDaniel RE, Maximov PY, Jordan VC. Estrogen-mediated mechanisms to control the growth and apoptosis of breast cancer cells: a translational research success story. VITAMINS AND HORMONES 2013; 93:1-49. [PMID: 23810002 DOI: 10.1016/b978-0-12-416673-8.00007-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The treatment and prevention of solid tumors have proved to be a major challenge for medical science. The paradigms for success in the treatment of childhood leukemia, Hodgkin's disease, Burkett's lymphoma, and testicular carcinoma with cytotoxic chemotherapy did not translate to success in solid tumors--the majority of cancers that kill. In contrast, significant success has accrued for patients with breast cancer with antihormone treatments (tamoxifen or aromatase inhibitors) that are proved to enhance survivorship, and remarkably, there are now two approved prevention strategies using either tamoxifen or raloxifene. This was considered impossible 40 years ago. We describe the major clinical advances with nonsteroidal antiestrogens that evolved into selective estrogen receptor modulators (SERMs) which successfully exploited the ER target selectively inside a woman's body. The standard paradigm that estrogen stimulates breast cancer growth has been successfully exploited for over 4 decades with therapeutic strategies that block (tamoxifen, raloxifene) or reduce (aromatase inhibitors) circulating estrogens in patients to stop breast tumor growth. But this did not explain why high-dose estrogen treatment that was the standard of care to treat postmenopausal breast cancer for 3 decades before tamoxifen caused tumor regression. This paradox was resolved with the discovery that breast cancer resistance to long-term estrogen deprivation causes tumor regression with physiologic estrogen through apoptosis. The new biology of estrogen action has been utilized to explain the findings in the Women's Health Initiative that conjugated equine estrogen alone given to postmenopausal women, average age 68, will produce a reduction of breast cancer incidence and mortality compared to no treatment. Estrogen is killing nascent breast cancer cells in the ducts of healthy postmenopausal women. The modulation of the ER using multifunctional medicines called SERMs has provided not only significant improvements in women's health and survivorship not anticipated 40 years ago but also has been the catalyst to enhance our knowledge of estrogen's apoptotic action that can be further exploited in the future.
Collapse
Affiliation(s)
- Russell E McDaniel
- Department of Oncology, Georgetown University, Lombardi Comprehensive Cancer Center, Washington, District of Columbia, USA
| | | | | |
Collapse
|
18
|
Becnel LB, McKenna NJ. Minireview: progress and challenges in proteomics data management, sharing, and integration. Mol Endocrinol 2012; 26:1660-74. [PMID: 22902541 DOI: 10.1210/me.2012-1180] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The proteome represents the identity, expression levels, interacting partners, and posttranslational modifications of proteins expressed within any given cell. Proteomic studies aim to census the quantitative and qualitative factors regulating the biological relationships of proteins acting in concert as functional cellular networks. In the field of endocrinology, proteomics has been of considerable value in determining the function and mechanism of action of endocrine signaling molecules in the cell membrane, cytoplasm, and nucleus and for the discovery of proteins as candidates for clinical biomarkers. The volume of data that can be generated by proteomics methodologies, up to gigabytes of data within a few hours, brings with it its own logistical hurdles and presents significant challenges to realizing the full potential of these datasets. In this minireview, we describe selected current proteomics methodologies and their application in basic and translational endocrinology before focusing on mass spectrometry as a model for current progress and challenges in data analysis, management, sharing, and integration.
Collapse
Affiliation(s)
- Lauren B Becnel
- Department of Medicine, Hematology and Oncology, Baylor College of Medicine, 1 Baylor Plaza MS-BCM305, Houston, Texas 77030, USA.
| | | |
Collapse
|
19
|
Santen RJ, Yue W, Heitjan DF. Modeling of the growth kinetics of occult breast tumors: role in interpretation of studies of prevention and menopausal hormone therapy. Cancer Epidemiol Biomarkers Prev 2012; 21:1038-48. [PMID: 22586072 PMCID: PMC4589189 DOI: 10.1158/1055-9965.epi-12-0043] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Autopsy studies report a reservoir of small, occult, undiagnosed breast cancers in up to 15.6% of women dying from unrelated causes. The effective doubling times (EDT) of these occult neoplasms range from 70 to 350 days and mammographic detection threshold diameters from 0.88 to 1.66 cm. Modeling of the biologic behavior of these occult tumors facilitates interpretation of tamoxifen breast cancer prevention and menopausal hormone therapy studies. METHODS We used iterative and mathematical techniques to develop a model of occult tumor growth (OTG) whose parameters included prevalence, EDT, and detection threshold. The model was validated by comparing predicted with observed incidence of breast cancer in several populations. RESULTS Iterative analysis identified a 200-day EDT, 7% prevalence and 1.16 cm detection threshold as optimal parameters for an OTG model as judged by comparison with Surveillance Epidemiology and End Results (SEER) population incidence rates in the United States. We validated the model by comparing predicted incidence rates with those observed in five separate population databases, in three long-term contralateral breast cancer detection studies, and with data from a computer-simulated tumor growth (CSTG) model. Our model strongly suggests that breast cancer prevention with anti-estrogens or aromatase inhibitors represents early treatment not prevention. In addition, menopausal hormone therapy does not primarily induce de novo tumors but promotes the growth of occult lesions. CONCLUSIONS Our OGTG model suggests that occult, undiagnosed tumors are prevalent, grow slowly, and are the biologic targets of anti-estrogen therapy for prevention and hormone therapy for menopausal women.
Collapse
Affiliation(s)
- Richard J Santen
- Department of Internal Medicine, Division of Endocrinology, University of Virginia, Charlottesville, VA 22908, USA.
| | | | | |
Collapse
|
20
|
Xu Y, Liu X, Guo F, Ning Y, Zhi X, Wang X, Chen S, Yin L, Li X. Effect of estrogen sulfation by SULT1E1 and PAPSS on the development of estrogen-dependent cancers. Cancer Sci 2012; 103:1000-9. [PMID: 22380844 DOI: 10.1111/j.1349-7006.2012.02258.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 01/11/2012] [Accepted: 02/26/2012] [Indexed: 12/21/2022] Open
Abstract
Estrogens are involved in the complex regulation of cell proliferation and apoptosis of hormone sensitive tumors including breast and endometrial cancers. Sulfation is the main pathway for estrogen metabolism, which is believed to be involved in the inactivation of estrogens in target tissues. SULT1E1 and PAPSS (PAPSS1 and PAPSS2) are responsible for the estrogen sulfation by providing catalyzing enzyme and universal sulfate donor. The present study showed the expression patterns of SULT1E1 and PAPSS in the breast and endometrial tissues by tissue array analysis and the assessment of clinical samples. The estrogen sulfation enzymes were comparatively higher in the tumorous tissues than their adjacent normal tissues. SULT1E1 overexpression inhibited the tumorigenesis in subcutaneous xenograft model. By CCK-8 assay and flow cytometry assay, overexpression of SULT1E1 and PAPSS1 by adenovirus blocked the estrogen pro-proliferating effect and promoted cell apoptosis induced by H(2)O(2) in MCF-7 cells. By real-time reverse transcription-polymerase chain reaction and western-blot assays, overexpression of SULT1E1 and PAPSS1 suppressed cell growth and triggered apoptosis by downregulating the levels of c-myc, cyclin D1 and bcl-2, meanwhile, upregulating bax expression. In conclusion, the discrepancies in expressions of SULT1E1 and PAPSS between breast and endometrial tumorous tissues and their adjacent normal tissues were prominent. Overexpression of SULT1E1 and PAPSS1 retarded MCF-7 cells growth in vivo and in vitro by arresting cell cycles and inducing apoptosis. Thus, targeting SULT1E1 and PAPSS expressions might be an important approach for estrogen-dependent cancers.
Collapse
Affiliation(s)
- Yali Xu
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Greve B, Kelsch R, Spaniol K, Eich HT, Götte M. Flow cytometry in cancer stem cell analysis and separation. Cytometry A 2012; 81:284-93. [PMID: 22311742 DOI: 10.1002/cyto.a.22022] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 12/21/2011] [Accepted: 01/07/2012] [Indexed: 12/19/2022]
Abstract
In recent years, a special type of cancer cell--the cancer stem cell (CSC)--has been identified and characterized for different tumors. CSCs may be responsible for the recurrence of a tumor following a primarily successful therapy and are thought to bear a high metastatic potential. For the development of efficient treatment strategies, the establishment of reliable methods for the identification and effective isolation of CSCs is imperative. Similar to their stem cell counterparts in bone marrow or small intestine, different cluster of differentiation surface antigens have been characterized, thus enabling researchers to identify them within the tumor bulk and to determine their degree of differentiation. In addition, functional properties characteristic of stem cells can be measured. Side population analysis is based on the stem cell-specific activity of certain ATP-binding cassette transporter proteins, which are able to transport fluorescent dyes out of the cells. Furthermore, the stem cell-specific presence of aldehyde dehydrogenase isoform 1 can be used for CSC labeling. However, the flow cytometric analysis of these CSC functional features requires specific technical adjustments. This review focuses on the principles and strategies of the flow cytometric analysis of CSCs and provides an overview of current protocols as well as technical requirements and pitfalls. A special focus is set on side population analysis and analysis of ALDH activity. Flow cytometry-based sorting principles and future flow cytometric applications for CSC analysis are also discussed.
Collapse
Affiliation(s)
- Burkhard Greve
- Department of Radiotherapy, University Hospital, 48149 Münster, Germany.
| | | | | | | | | |
Collapse
|
22
|
Sweeney EE, McDaniel RE, Maximov PY, Fan P, Jordan VC. Models and Mechanisms of Acquired Antihormone Resistance in Breast Cancer: Significant Clinical Progress Despite Limitations. Horm Mol Biol Clin Investig 2012; 9:143-163. [PMID: 23308083 PMCID: PMC3539798 DOI: 10.1515/hmbci-2011-0004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Translational research for the treatment and prevention of breast cancer depends upon the four Ms: models, molecules, and mechanisms in order to create medicines. The process, to target the estrogen receptor (ER) in estrogen-dependent breast cancer, has yielded significant advances in patient survivorship and the first approved medicines (tamoxifen and raloxifene) to reduce the incidence of any cancer in high- or low-risk women. This review focuses on the critical role of the few ER-positive cell lines (MCF-7, T47D, BT474, ZR-75) that continue to advance our understanding of the estrogen-regulated biology of breast cancer. More importantly, the model cell lines have provided an opportunity to document the development and evolution of acquired antihormone resistance. The description of this evolutionary process that occurs in micrometastatic disease during up to a decade of adjuvant therapy would not be possible in the patient. The use of the MCF-7 breast cancer cell line in particular has been instrumental in discovering a vulnerability of ER-positive breast cancer exhaustively treated with antihormone therapy. Physiologic estradiol acts as an apoptotic trigger to cause tumor regression. These unanticipated findings in the laboratory have translated to clinical advances in our knowledge of the paradoxical role of estrogen in the life and death of breast cancer.
Collapse
Affiliation(s)
- Elizabeth E Sweeney
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | | | | | | | | |
Collapse
|
23
|
Estrogen induces apoptosis in estrogen deprivation-resistant breast cancer through stress responses as identified by global gene expression across time. Proc Natl Acad Sci U S A 2011; 108:18879-86. [PMID: 22011582 DOI: 10.1073/pnas.1115188108] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In laboratory studies, acquired resistance to long-term antihormonal therapy in breast cancer evolves through two phases over 5 y. Phase I develops within 1 y, and tumor growth occurs with either 17β-estradiol (E(2)) or tamoxifen. Phase II resistance develops after 5 y of therapy, and tamoxifen still stimulates growth; however, E(2) paradoxically induces apoptosis. This finding is the basis for the clinical use of estrogen to treat advanced antihormone-resistant breast cancer. We interrogated E(2)-induced apoptosis by analysis of gene expression across time (2-96 h) in MCF-7 cell variants that were estrogen-dependent (WS8) or resistant to estrogen deprivation and refractory (2A) or sensitive (5C) to E(2)-induced apoptosis. We developed a method termed differential area under the curve analysis that identified genes uniquely regulated by E(2) in 5C cells compared with both WS8 and 2A cells and hence, were associated with E(2)-induced apoptosis. Estrogen signaling, endoplasmic reticulum stress (ERS), and inflammatory response genes were overrepresented among the 5C-specific genes. The identified ERS genes indicated that E(2) inhibited protein folding, translation, and fatty acid synthesis. Meanwhile, the ERS-associated apoptotic genes Bcl-2 interacting mediator of cell death (BIM; BCL2L11) and caspase-4 (CASP4), among others, were induced. Evaluation of a caspase peptide inhibitor panel showed that the CASP4 inhibitor z-LEVD-fmk was the most active at blocking E(2)-induced apoptosis. Furthermore, z-LEVD-fmk completely prevented poly (ADP-ribose) polymerase (PARP) cleavage, E(2)-inhibited growth, and apoptotic morphology. The up-regulated proinflammatory genes included IL, IFN, and arachidonic acid-related genes. Functional testing showed that arachidonic acid and E(2) interacted to superadditively induce apoptosis. Therefore, these data indicate that E(2) induced apoptosis through ERS and inflammatory responses in advanced antihormone-resistant breast cancer.
Collapse
|
24
|
Wangefjord S, Manjer J, Gaber A, Nodin B, Eberhard J, Jirström K. Cyclin D1 expression in colorectal cancer is a favorable prognostic factor in men but not in women in a prospective, population-based cohort study. Biol Sex Differ 2011; 2:10. [PMID: 21888663 PMCID: PMC3179695 DOI: 10.1186/2042-6410-2-10] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 09/03/2011] [Indexed: 11/13/2022] Open
Abstract
Background Although colorectal cancer (CRC) is generally not considered to be a hormone-dependent malignancy, several sex-related differences in incidence, molecular characteristics and survival have been reported. Epidemiological studies have consistently shown that increased exposure to female sex hormones is associated with a lower risk of CRC in women, and cyclin D1, an important downstream effector in estrogen-mediated signaling, is commonly activated in CRC. In this study, we analyzed the prognostic significance of cyclin D1 expression in CRC, with particular reference to sex-related differences, in tumors from a large, prospective, population-based cohort. Methods Using tissue microarrays and immunohistochemistry, the fraction and intensity of cyclin D1 expression was evaluated in 527 incident CRC cases from the Malmö Diet and Cancer Study. The χ2 and Spearman's rho (ρ) tests were used for comparison of cyclin D1 expression and relevant clinicopathological characteristics. Kaplan-Meier analysis and Cox proportional hazards modeling were used to assess the effect of cyclin D1 expression on cancer-specific survival (CSS) in univariate and multivariate analysis, adjusted for established prognostic factors. Results Cyclin D1 intensity was significantly lower in male compared with female CRC (P = 0.018). In the full cohort, cyclin D1 expression was associated with a significantly prolonged CSS (hazard ratio (HR) = 0.69; 95% CI 0.49 to 0.96, P = 0.026) but subgroup analysis according to gender revealed a strongly accentuated prognostic effect of cyclin D1 in male CRC (HR = 0.48; 95% CI 0.31 to 0.74, P < 0.001), which was in contrast to female CRC, where cyclin D1 was not prognostic (HR = 1.05; 95% CI 0.62 to 1.78, P = 0.864) (Pinteraction = 0.024). The prognostic value of cyclin D1 was not retained in multivariate analysis, either in the full cohort or in male CRC. Conclusions Cyclin D1 expression is strongly associated with prolonged survival in male CRC. These findings not only support an important role for cyclin D1 in colorectal carcinogenesis, but also add further weight to the accumulating evidence that CRC is indeed a hormone-dependent malignancy, for which prognostic and treatment-predictive molecular biomarkers should be evaluated differently in women and men.
Collapse
Affiliation(s)
- Sakarias Wangefjord
- Department of Clinical Sciences, Division of Pathology, Lund University, Skåne University Hospital, 221 85 Lund, Sweden.
| | | | | | | | | | | |
Collapse
|