99 results on '"Frasor J"'
Search Results
2. Mouse Embryo Development in Three Media Supplemented with Synthetic Serum: PP-16-349
- Author
-
Frasor, J, Sherbahn, R, Sant'Anna, T, and Rawlins, R G
- Published
- 1997
3. Crystal Structure of the ER-alpha Ligand-binding Domain (Y537S) in Complex with an N-ethyl, 4-methoxybenzyl OBHS-N derivative
- Author
-
Nwachukwu, J.C., primary, Srinivasan, S., additional, Bruno, N.E., additional, Dharmarajan, V., additional, Goswami, D., additional, Kastrati, I., additional, Novick, S., additional, Nowak, J., additional, Zhou, H.B., additional, Boonmuen, N., additional, Zhao, Y., additional, Min, J., additional, Frasor, J., additional, Katzenellenbogen, B.S., additional, Griffin, P.R., additional, Katzenellenbogen, J.A., additional, and Nettles, K.W., additional
- Published
- 2016
- Full Text
- View/download PDF
4. Crystal Structure of the ER-alpha Ligand-binding Domain (Y537S) in Complex with an N-methyl Substituted OBHS-N derivative
- Author
-
Nwachukwu, J.C., primary, Srinivasan, S., additional, Bruno, N.E., additional, Dharmarajan, V., additional, Goswami, D., additional, Kastrati, I., additional, Novick, S., additional, Nowak, J., additional, Zhou, H.B., additional, Boonmuen, N., additional, Zhao, Y., additional, Min, J., additional, Frasor, J., additional, Katzenellenbogen, B.S., additional, Griffin, P.R., additional, Katzenellenbogen, J.A., additional, and Nettles, K.W., additional
- Published
- 2016
- Full Text
- View/download PDF
5. Crystal Structure of the ER-alpha Ligand-binding Domain (Y537S) in Complex with an N-methyl, 2-chlorobenzyl OBHS-N derivative
- Author
-
Nwachukwu, J.C., primary, Srinivasan, S., additional, Bruno, N.E., additional, Dharmarajan, V., additional, Goswami, D., additional, Kastrati, I., additional, Novick, S., additional, Nowak, J., additional, Zhou, H.B., additional, Boonmuen, N., additional, Zhao, Y., additional, Min, J., additional, Frasor, J., additional, Katzenellenbogen, B.S., additional, Griffin, P.R., additional, Katzenellenbogen, J.A., additional, and Nettles, K.W., additional
- Published
- 2016
- Full Text
- View/download PDF
6. Crystal Structure of the ER-alpha Ligand-binding Domain (Y537S) in Complex with an N-trifluoroethyl 4-chlorobenzyl OBHS-N derivative
- Author
-
Nwachukwu, J.C., primary, Srinivasan, S., additional, Bruno, N.E., additional, Dharmarajan, V., additional, Goswami, D., additional, Kastrati, I., additional, Novick, S., additional, Nowak, J., additional, Zhou, H.B., additional, Boonmuen, N., additional, Zhao, Y., additional, Min, J., additional, Frasor, J., additional, Katzenellenbogen, B.S., additional, Griffin, P.R., additional, Katzenellenbogen, J.A., additional, and Nettles, K.W., additional
- Published
- 2016
- Full Text
- View/download PDF
7. Crystal Structure of the ER-alpha Ligand-binding Domain (Y537S) in Complex with an N-ethyl, 4-chlorobenzyl OBHS-N derivative
- Author
-
Nwachukwu, J.C., primary, Srinivasan, S., additional, Bruno, N.E., additional, Dharmarajan, V., additional, Goswami, D., additional, Kastrati, I., additional, Novick, S., additional, Nowak, J., additional, Zhou, H.B., additional, Boonmuen, N., additional, Zhao, Y., additional, Min, J., additional, Frasor, J., additional, Katzenellenbogen, B.S., additional, Griffin, P.R., additional, Katzenellenbogen, J.A., additional, and Nettles, K.W., additional
- Published
- 2016
- Full Text
- View/download PDF
8. Crystal Structure of the ER-alpha Ligand-binding Domain (Y537S) in Complex with an N-trifluoroethyl OBHS-N derivative
- Author
-
Nwachukwu, J.C., primary, Srinivasan, S., additional, Bruno, N.E., additional, Dharmarajan, V., additional, Goswami, D., additional, Kastrati, I., additional, Novick, S., additional, Nowak, J., additional, Zhou, H.B., additional, Boonmuen, N., additional, Zhao, Y., additional, Min, J., additional, Frasor, J., additional, Katzenellenbogen, B.S., additional, Griffin, P.R., additional, Katzenellenbogen, J.A., additional, and Nettles, K.W., additional
- Published
- 2016
- Full Text
- View/download PDF
9. Crystal Structure of the ER-alpha Ligand-binding Domain (Y537S) in Complex with an N-ethyl, alpha-naphthyl OBHS-N derivative
- Author
-
Nwachukwu, J.C., primary, Srinivasan, S., additional, Bruno, N.E., additional, Dharmarajan, V., additional, Goswami, D., additional, Kastrati, I., additional, Novick, S., additional, Nowak, J., additional, Zhou, H.B., additional, Boonmuen, N., additional, Zhao, Y., additional, Min, J., additional, Frasor, J., additional, Katzenellenbogen, B.S., additional, Griffin, P.R., additional, Katzenellenbogen, J.A., additional, and Nettles, K.W., additional
- Published
- 2016
- Full Text
- View/download PDF
10. PHLDA1 expression is controlled by an estrogen receptor-NFκB-miR-181 regulatory loop and is essential for formation of ER+ mammospheres
- Author
-
Kastrati, I, primary, Canestrari, E, additional, and Frasor, J, additional
- Published
- 2014
- Full Text
- View/download PDF
11. Differential roles for signal transducers and activators of transcription 5a and 5b in PRL stimulation of ERalpha and ERbeta transcription
- Author
-
Frasor, J., Park, K., Byers, M., Telleria, C., Kitamura, T., Yu-Lee, L.Y., Djiane, J., Park-Sarge, O.K., Gibori, G., Unité de biologie cellulaire et moléculaire, and Institut National de la Recherche Agronomique (INRA)
- Subjects
œstrogène ,RAT ,PROMOTEUR DU GENE ,phosphorylation ,gène ,[SDV]Life Sciences [q-bio] ,oestradiol ,rattus rattus ,gestation ,pcr ,clonage ,mutation ,récepteur ,ComputingMilieux_MISCELLANEOUS ,prolactine - Abstract
International audience
- Published
- 2001
12. BindSDb: A binding-information spatial database.
- Author
-
Roqueiro, D., Frasor, J., and Yang Dai
- Published
- 2010
- Full Text
- View/download PDF
13. Prolactin regulation of estrogen receptor expression
- Author
-
FRASOR, J, primary and GIBORI, G, additional
- Published
- 2003
- Full Text
- View/download PDF
14. PRL-Induced ER Gene Expression Is Mediated by Janus Kinase 2 (Jak2) While Signal Transducer and Activator of Transcription 5b (Stat5b) Phosphorylation Involves Jak2 and a Second Tyrosine Kinase
- Author
-
Frasor, J., primary
- Published
- 2001
- Full Text
- View/download PDF
15. Expression of Prolactin and Its Receptor in the Baboon Uterus during the Menstrual Cycle and Pregnancy
- Author
-
Frasor, J., primary
- Published
- 1999
- Full Text
- View/download PDF
16. Molecular Cloning and Characterization of the Rat Ovarian 20α-Hydroxysteroid Dehydrogenase Gene
- Author
-
Zhong, L., primary, Ou, J., additional, Barkai, U., additional, Mao, J.F., additional, Frasor, J., additional, and Gibori, G., additional
- Published
- 1998
- Full Text
- View/download PDF
17. Fertilization and early embryology: Comparison of mouse embryo development in open and microdrop co-culture systems
- Author
-
Sherbahn, R., primary, Frasor, J., additional, Radwanska, E., additional, Binor, Z., additional, Wood-Molo, M., additional, Hibner, M., additional, Mack, S., additional, and Rawlins, R. G., additional
- Published
- 1996
- Full Text
- View/download PDF
18. Estrogenic effects of herbal medicines from Costa Rica used for the management of menopausal symptoms.
- Author
-
Doyle BJ, Frasor J, Bellows LE, Locklear TD, Perez A, Gomez-Laurito J, Mahady GB, Doyle, Brian J, Frasor, Jonna, Bellows, Lauren E, Locklear, Tracie D, Perez, Alice, Gomez-Laurito, Jorge, and Mahady, Gail B
- Published
- 2009
- Full Text
- View/download PDF
19. The relationship of human granulosa-lutein cell proliferative index to follicular diameter and serum estradiol.
- Author
-
Attaran, Marjan, Frasor, Jonna, Mascha, Edward, Radwanska, Ewa, Rawlins, Richard G., Attaran, M, Frasor, J, Mascha, E, Radwanska, E, and Rawlins, R G
- Published
- 1998
- Full Text
- View/download PDF
20. Comparison of mouse embryo development in open and microdrop co-culture systems.
- Author
-
Sherbahn, R, Frasor, J, Radwanska, E, Binor, Z, Wood-Molo, M, Hibner, M, Mack, S, and Rawlins, R G
- Abstract
Co-culture with numerous cell lines has been shown to improve in-vitro embryo development. It is usually performed in open culture without an oil overlay, or in relatively large volumes of medium (e.g. 0.5 ml) under oil. We compared the efficacy of open and microdrop co-culture systems using human endometrial and tubal cell lines and mouse zygotes. Although the mean pH values of the media from the tubal cell cultures (both open and oil-covered) decreased significantly over 5 days of culture, this did not appear to impair embryo development. Both co-culture and microdrop culture significantly improved blastocyst and hatching blastocyst formation rates. The combination of the two techniques (microdrop and co-culture) demonstrated the highest blastocyst formation and hatching blastocyst formation rates, as well as the highest mean cell numbers in hatching blastocysts. Co-culture in a microdrop is a superior system for mouse embryo culture. [ABSTRACT FROM AUTHOR]
- Published
- 1996
- Full Text
- View/download PDF
21. Evaluation of co-culture and alternative culture systems for promoting in-vitro development of mouse embryos.
- Author
-
Piekos, M W, Frasor, J, Mack, S, Binor, Z, Soltes, B, Molo, M W, Radwanska, E, and Rawlins, R G
- Abstract
The goal of this study was to compare mouse embryo development in a defined synthetic medium (human tubal fluid) against the same medium supplemented with a defined synthetic serum (SS), co-culture on human tubal epithelium (TECC), and culture on human fibronectin (FN) with and without SS. After 48 h, TECC, SS and FN + SS cultures demonstrated accelerated development with > 70% achieving > or = 8-cell stage. After 72 h, these culture conditions also significantly increased the proportion of embryos reaching the blastocyst stage but only TECC significantly increased the number of hatching blastocysts. Nuclei of the trophectoderm of unhatched and hatched blastocysts were stained with propidium iodide before fixing and labelling both the trophectoderm and inner cell mass with bisbenzimide. Blastocysts from the TECC contained a significantly higher total cell number (TCN) and trophectoderm and inner cell mass cell numbers than all other groups. These findings indicate equivalent improvements in mouse embryo development to the blastocyst stage in response to TECC, SS and FN and an enhanced number of cells and rate of hatching found only with TECC.
- Published
- 1995
22. Transcriptomic analysis of TNFα- and 17β-estradiol (17βE2)-treated MCF-7 cells
- Author
-
Frasor, J, primary
- Full Text
- View/download PDF
23. ASCT2 is a major contributor to serine uptake in cancer cells.
- Author
-
Conger KO, Chidley C, Ozgurses ME, Zhao H, Kim Y, Semina SE, Burns P, Rawat V, Lietuvninkas L, Sheldon R, Ben-Sahra I, Frasor J, Sorger PK, DeNicola GM, and Coloff JL
- Subjects
- Humans, Glutamine metabolism, Cell Line, Tumor, Estrogen Receptor alpha metabolism, Neoplasms metabolism, Neoplasms pathology, Neoplasms genetics, Animals, Biological Transport, Female, MCF-7 Cells, Amino Acid Transport System ASC metabolism, Amino Acid Transport System ASC genetics, Serine metabolism, Minor Histocompatibility Antigens metabolism, Minor Histocompatibility Antigens genetics
- Abstract
The non-essential amino acid serine is a critical nutrient for cancer cells due to its diverse biosynthetic functions. While some tumors can synthesize serine de novo, others are auxotrophic and therefore reliant on serine uptake. Importantly, despite several transporters being known to be capable of transporting serine, the transporters that mediate serine uptake in cancer cells are not known. Here, we characterize the amino acid transporter ASCT2 (SLC1A5) as a major contributor to serine uptake in cancer cells. ASCT2 is well known as a glutamine transporter in cancer, and our work demonstrates that serine and glutamine compete for uptake through ASCT2. We further show that ASCT2-mediated serine uptake is essential for purine nucleotide biosynthesis and that estrogen receptor α (ERα) promotes serine uptake by directly activating SLC1A5 transcription. Collectively, our work defines an additional important role for ASCT2 as a serine transporter in cancer and evaluates ASCT2 as a potential therapeutic target., Competing Interests: Declaration of interests P.K.S. is a member of the SAB or BOD for Applied Biomath, RareCyte. Nanostring, Glencoe Software, and Montai; he is consultant for Merck. None of these activities impact the content of this manuscript., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
24. ASCT2 is the primary serine transporter in cancer cells.
- Author
-
Conger KO, Chidley C, Ozgurses ME, Zhao H, Kim Y, Semina SE, Burns P, Rawat V, Sheldon R, Ben-Sahra I, Frasor J, Sorger PK, DeNicola GM, and Coloff JL
- Abstract
The non-essential amino acid serine is a critical nutrient for cancer cells due to its diverse biosynthetic functions. While some tumors can synthesize serine de novo , others are auxotrophic for serine and therefore reliant on the uptake of exogenous serine. Importantly, however, the transporter(s) that mediate serine uptake in cancer cells are not known. Here, we characterize the amino acid transporter ASCT2 (coded for by the gene SLC1A5 ) as the primary serine transporter in cancer cells. ASCT2 is well-known as a glutamine transporter in cancer, and our work demonstrates that serine and glutamine compete for uptake through ASCT2. We further show that ASCT2-mediated serine uptake is essential for purine nucleotide biosynthesis and that ERα promotes serine uptake by directly activating SLC1A5 transcription. Together, our work defines an additional important role for ASCT2 as a serine transporter in cancer and evaluates ASCT2 as a potential therapeutic target in serine metabolism., Competing Interests: Declaration of competing interests PKS is a member of the SAB or BOD for Applied Biomath, RareCyte. Nanostring, Glencoe Software and Montai; he is consultant for Merck. None of these activities impact the content of this manuscript. The other authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
25. Identification of metabolic pathways contributing to ER + breast cancer disparities using a machine-learning pipeline.
- Author
-
Santaliz-Casiano A, Mehta D, Danciu OC, Patel H, Banks L, Zaidi A, Buckley J, Rauscher GH, Schulte L, Weller LR, Taiym D, Liko-Hazizi E, Pulliam N, Friedewald SM, Khan S, Kim JJ, Gradishar W, Hegerty S, Frasor J, Hoskins KF, and Madak-Erdogan Z
- Subjects
- Humans, Female, United States, Epigenesis, Genetic, Ethnicity, Metabolic Networks and Pathways, White, Breast Neoplasms pathology
- Abstract
African American (AA) women in the United States have a 40% higher breast cancer mortality rate than Non-Hispanic White (NHW) women. The survival disparity is particularly striking among (estrogen receptor positive) ER
+ breast cancer cases. The purpose of this study is to examine whether there are racial differences in metabolic pathways typically activated in patients with ER+ breast cancer. We collected pretreatment plasma from AA and NHW ER+ breast cancer cases (AA n = 48, NHW n = 54) and cancer-free controls (AA n = 100, NHW n = 48) to conduct an untargeted metabolomics analysis using gas chromatography mass spectrometry (GC-MS) to identify metabolites that may be altered in the different racial groups. Unpaired t-test combined with multiple feature selection and prediction models were employed to identify race-specific altered metabolic signatures. This was followed by the identification of altered metabolic pathways with a focus in AA patients with breast cancer. The clinical relevance of the identified pathways was further examined in PanCancer Atlas breast cancer data set from The Cancer Genome Atlas Program (TCGA). We identified differential metabolic signatures between NHW and AA patients. In AA patients, we observed decreased circulating levels of amino acids compared to healthy controls, while fatty acids were significantly higher in NHW patients. By mapping these metabolites to potential epigenetic regulatory mechanisms, this study identified significant associations with regulators of metabolism such as methionine adenosyltransferase 1A (MAT1A), DNA Methyltransferases and Histone methyltransferases for AA individuals, and Fatty acid Synthase (FASN) and Monoacylglycerol lipase (MGL) for NHW individuals. Specific gene Negative Elongation Factor Complex E (NELFE) with histone methyltransferase activity, was associated with poor survival exclusively for AA individuals. We employed a comprehensive and novel approach that integrates multiple machine learning and statistical methods, coupled with human functional pathway analyses. The metabolic profile of plasma samples identified may help elucidate underlying molecular drivers of disproportionately aggressive ER+ tumor biology in AA women. It may ultimately lead to the identification of novel therapeutic targets. To our knowledge, this is a novel finding that describes a link between metabolic alterations and epigenetic regulation in AA breast cancer and underscores the need for detailed investigations into the biological underpinnings of breast cancer health disparities., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
26. Identification of a novel ER-NFĸB-driven stem-like cell population associated with relapse of ER+ breast tumors.
- Author
-
Semina SE, Alejo LH, Chopra S, Kansara NS, Kastrati I, Sartorius CA, and Frasor J
- Subjects
- Animals, Humans, Female, Antineoplastic Agents, Hormonal therapeutic use, Receptors, Estrogen genetics, Receptors, Estrogen metabolism, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local drug therapy, MCF-7 Cells, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Breast Neoplasms pathology, Mammary Neoplasms, Animal genetics
- Abstract
Background: Up to 40% of patients with estrogen receptor-positive (ER+) breast cancer experience relapse. This can be attributed to breast cancer stem cells (BCSCs), which are known to be involved in therapy resistance, relapse, and metastasis. Therefore, there is an urgent need to identify genes/pathways that drive stem-like cell properties in ER+ breast tumors., Methods: Using single-cell RNA sequencing and various bioinformatics approaches, we identified a unique stem-like population and established its clinical relevance. With follow-up studies, we validated our bioinformatics findings and confirmed the role of ER and NFĸB in the promotion of stem-like properties in breast cancer cell lines and patient-derived models., Results: We identified a novel quiescent stem-like cell population that is driven by ER and NFĸB in multiple ER+ breast cancer models. Moreover, we found that a gene signature derived from this stem-like population is expressed in primary ER+ breast tumors, endocrine therapy-resistant and metastatic cell populations and predictive of poor patient outcome., Conclusions: These findings indicate a novel role for ER and NFĸB crosstalk in BCSCs biology and understanding the mechanism by which these pathways promote stem properties can be exploited to improve outcomes for ER+ breast cancer patients at risk of relapse., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
27. Emerging Roles of Ceramides in Breast Cancer Biology and Therapy.
- Author
-
Pal P, Atilla-Gokcumen GE, and Frasor J
- Subjects
- Biology, Ceramides metabolism, Humans, Phosphates, Sphingolipids metabolism, Sphingosine metabolism, Neoplasms metabolism, Sphingomyelins
- Abstract
One of the classic hallmarks of cancer is the imbalance between elevated cell proliferation and reduced cell death. Ceramide, a bioactive sphingolipid that can regulate this balance, has long been implicated in cancer. While the effects of ceramide on cell death and therapeutic efficacy are well established, emerging evidence indicates that ceramide turnover to downstream sphingolipids, such as sphingomyelin, hexosylceramides, sphingosine-1-phosphate, and ceramide-1-phosphate, is equally important in driving pro-tumorigenic phenotypes, such as proliferation, survival, migration, stemness, and therapy resistance. The complex and dynamic sphingolipid network has been extensively studied in several cancers, including breast cancer, to find key sphingolipidomic alterations that can be exploited to develop new therapeutic strategies to improve patient outcomes. Here, we review how the current literature shapes our understanding of how ceramide synthesis and turnover are altered in breast cancer and how these changes offer potential strategies to improve breast cancer therapy.
- Published
- 2022
- Full Text
- View/download PDF
28. Estrogen Receptor-Regulated Gene Signatures in Invasive Breast Cancer Cells and Aggressive Breast Tumors.
- Author
-
Smart E, Semina SE, Alejo LH, Kansara NS, and Frasor J
- Abstract
Most metastatic breast cancers arise from estrogen receptor α (ER)-positive disease, and yet the role of ER in promoting metastasis is unclear. Here, we used an ER+ breast cancer cell line that is highly invasive in an ER- and IKKβ-dependent manner. We defined two ER-regulated gene signatures that are specifically regulated in the subpopulations of invasive cells. The first consists of proliferation-associated genes, which is a known function of ER, which actually suppress rather than enhance invasion. The second signature consists of genes involved in essential biological processes, such as organelle assembly and vesicle trafficking. Importantly, the second subpopulation-specific signature is associated with aggressive disease and poor patient outcome, independently of proliferation. These findings indicate a complex interplay between ER-driven proliferation and invasion, and they define new ER-regulated gene signatures that are predictive of aggressive ER+ breast cancer.
- Published
- 2022
- Full Text
- View/download PDF
29. Endocrine Therapy-Resistant Breast Cancer Cells Are More Sensitive to Ceramide Kinase Inhibition and Elevated Ceramide Levels Than Therapy-Sensitive Breast Cancer Cells.
- Author
-
Pal P, Millner A, Semina SE, Huggins RJ, Running L, Aga DS, Tonetti DA, Schiff R, Greene GL, Atilla-Gokcumen GE, and Frasor J
- Abstract
ET resistance is a critical problem for estrogen receptor-positive (ER+) breast cancer. In this study, we have investigated how alterations in sphingolipids promote cell survival in ET-resistant breast cancer. We have performed LC-MS-based targeted sphingolipidomics of tamoxifen-sensitive and -resistant MCF-7 breast cancer cell lines. Follow-up studies included treatments of cell lines and patient-derived xenograft organoids (PDxO) with small molecule inhibitors; cytometric analyses to measure cell death, proliferation, and apoptosis; siRNA-mediated knockdown; RT-qPCR and Western blot for gene and protein expression; targeted lipid analysis; and lipid addback experiments. We found that tamoxifen-resistant cells have lower levels of ceramides and hexosylceramides compared to their tamoxifen-sensitive counterpart. Upon perturbing the sphingolipid pathway with small molecule inhibitors of key enzymes, we identified that CERK is essential for tamoxifen-resistant breast cancer cell survival, as well as a fulvestrant-resistant PDxO. CERK inhibition induces ceramide-mediated cell death in tamoxifen-resistant cells. Ceramide-1-phosphate (C1P) partially reverses CERK inhibition-induced cell death in tamoxifen-resistant cells, likely through lowering endogenous ceramide levels. Our findings suggest that ET-resistant breast cancer cells maintain lower ceramide levels as an essential pro-survival mechanism. Consequently, ET-resistant breast cancer models have a unique dependence on CERK as its activity can inhibit de novo ceramide production.
- Published
- 2022
- Full Text
- View/download PDF
30. Ceramide-1-Phosphate Is Involved in Therapy-Induced Senescence.
- Author
-
Millner A, Running L, Colon-Rosa N, Aga DS, Frasor J, and Atilla-Gokcumen GE
- Subjects
- Cell Cycle Checkpoints, Cellular Senescence, Phosphates, Signal Transduction, Ceramides metabolism, Ceramides pharmacology, Sphingolipids metabolism, Sphingolipids pharmacology
- Abstract
Sphingolipids are key signaling lipids and their dysregulation has been associated with various cellular processes. We have previously shown significant changes in sphingolipids in therapy-induced senescence, a state of cell cycle arrest as a response to chemotherapy, including the accumulation of ceramides, and provided evidence suggesting that ceramide processing is important for this process. Herein, we conducted a focused small molecule inhibitor screen targeting the sphingolipid pathway, which highlighted a new lipid regulator of therapy-induced senescence. Among the inhibitors tested, the inhibition of ceramide kinase by NVP-231 reduced the levels of senescent cells. Ceramide kinase knockdown exhibited similar effects, strongly supporting the involvement of ceramide kinase during this process. We showed that ceramide-1-phosphate was upregulated in therapy-induced senescence and that NVP-231 reduced ceramide-1-phosphate levels in different cell line models of therapy-induced senescence. Finally, ceramide-1-phosphate addition to NVP-231-treated cells reversed the effects of NVP-231 during senescence. Overall, our results identify a previously unknown lipid player in therapy-induced senescence and highlight a potential targetable enzyme to reduce the levels of therapy-induced senescent cells.
- Published
- 2022
- Full Text
- View/download PDF
31. Selective pressure of endocrine therapy activates the integrated stress response through NFκB signaling in a subpopulation of ER positive breast cancer cells.
- Author
-
Semina SE, Pal P, Kansara NS, Huggins RJ, Alarid ET, Greene GL, and Frasor J
- Subjects
- Antineoplastic Agents, Hormonal pharmacology, Antineoplastic Agents, Hormonal therapeutic use, Drug Resistance, Neoplasm genetics, Female, Humans, NF-kappa B genetics, NF-kappa B metabolism, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local genetics, Signal Transduction, Tamoxifen therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology
- Abstract
Background: While estrogen receptor (ER) positive breast tumors generally respond well to endocrine therapy (ET), up to 40% of patients will experience relapse, either while on endocrine therapy or after ET is completed. We previously demonstrated that the selective pressure of tamoxifen activates the NFκB pathway in ER + patient tumors, breast cancer cell lines, and breast cancer xenograft tumors, and that this activation allows for survival of a subpopulation of NFκB + cells that contribute to cell regrowth and tumor relapse after ET withdrawal. However, the mechanisms contributing to the expansion of an NFκB + cell population on ET are unknown., Methods: Here, we utilized single-cell RNA sequencing and bioinformatics approaches to characterize the NFκB + cell population and its clinical relevance. Follow-up studies were conducted to validate our findings and assess the function of the integrated stress response pathway in breast cancer cell lines and patient-derived models., Results: We found that the NFκB + population that arises in response to ET is a preexisting population is enriched under the selective pressure of ET. Based on the preexisting NFκB + cell population, we developed a gene signature and found that it is predictive of tumor relapse when expressed in primary ER + tumors and is retained in metastatic cell populations. Moreover, we identified that the integrated stress response (ISR), as indicated by increased phosphorylation of eIF2α, occurs in response to ET and contributes to clonogenic growth under the selective pressure of ET., Conclusions: Taken together, our findings suggest that a cell population with active NFκB and ISR signaling can survive and expand under the selective pressure of ET and that targeting this population may be a viable therapeutic strategy to improve patient outcome by eliminating cells that survive ET. Understanding the mechanisms by which breast cancer cells survive the selective pressure of ET may improve relapse rates and overall outcome for patients with ER + breast tumors., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
32. Intestinal estrogen receptor beta suppresses colon inflammation and tumorigenesis in both sexes.
- Author
-
Hases L, Indukuri R, Birgersson M, Nguyen-Vu T, Lozano R, Saxena A, Hartman J, Frasor J, Gustafsson JÅ, Katajisto P, Archer A, and Williams C
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation, Female, Humans, Intestinal Mucosa pathology, Male, Mice, Mice, Inbred C57BL, NF-kappa B physiology, Sex Characteristics, Tumor Necrosis Factor-alpha pharmacology, Colitis prevention & control, Colorectal Neoplasms prevention & control, Estrogen Receptor beta physiology
- Abstract
Estrogen hormones protect against colorectal cancer (CRC) and a preventative role of estrogen receptor beta (ERβ) on CRC has been supported using full knockout animals. However, it is unclear through which cells or organ ERβ mediates this effect. To investigate the functional role of intestinal ERβ during colitis-associated CRC we used intestine-specific ERβ knockout mice treated with azoxymethane and dextran sodium sulfate, followed by ex vivo organoid culture to corroborate intrinsic effects. We explored genome-wide impact on TNFα signaling using human CRC cell lines and chromatin immunoprecipitation assay to mechanistically characterize the regulation of ERβ. Increased tumor formation in males and tumor size in females was noted upon intestine-specific ERβ knockout, accompanied by enhanced local expression of TNFα, deregulation of key NFκB targets, and increased colon ulceration. Unexpectedly, we noted especially strong effects in males. We corroborated that intestinal ERβ protects against TNFα-induced damage intrinsically, and characterized an underlying genome-wide signaling mechanism in CRC cell lines whereby ERβ binds to cis-regulatory chromatin areas of key NFκB regulators. Our results support a protective role of intestinal ERβ against colitis-associated CRC, proposing new therapeutic strategies., (Copyright © 2020 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
33. Update on the Role of NFκB in Promoting Aggressive Phenotypes of Estrogen Receptor-Positive Breast Cancer.
- Author
-
Smart E, Semina SE, and Frasor J
- Subjects
- Animals, Breast Neoplasms genetics, Breast Neoplasms metabolism, Disease Progression, Drug Resistance, Neoplasm genetics, Female, Gene Expression Regulation, Neoplastic, Humans, NF-kappa B genetics, Neoplasm Invasiveness, Phenotype, Receptors, Estrogen genetics, Signal Transduction genetics, Breast Neoplasms pathology, NF-kappa B physiology, Receptors, Estrogen metabolism
- Abstract
The majority of breast cancers are diagnosed as estrogen receptor-positive (ER+) and respond well to ER-targeted endocrine therapy. Despite the initial treatability of ER+ breast cancer, this subtype still accounts for the majority of deaths. This is partly due to the changing molecular characteristics of tumors as they progress to aggressive, metastatic, and frequently therapy resistant disease. In these advanced tumors, targeting ER alone is often less effective, as other signaling pathways become active, and ER takes on a redundant or divergent role. One signaling pathway whose crosstalk with ER has been widely studied is the nuclear factor kappa B (NFκB) signaling pathway. NFκB is frequently implicated in ER+ tumor progression to an aggressive disease state. Although ER and NFκB frequently co-repress each other, it has emerged that the 2 pathways can positively converge to play a role in promoting endocrine resistance, metastasis, and disease relapse. This will be reviewed here, paying particular attention to new developments in the field. Ultimately, finding targeted therapies that remain effective as tumors progress remains one of the biggest challenges for the successful treatment of ER+ breast cancer. Although early attempts to therapeutically block NFκB activity frequently resulted in systemic toxicity, there are some effective options. The drugs parthenolide and dimethyl fumarate have both been shown to effectively inhibit NFκB, reducing tumor aggressiveness and reversing endocrine therapy resistance. This highlights the need to revisit targeting NFκB in the clinic to potentially improve outcome for patients with ER+ breast cancer., (© Endocrine Society 2020. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
34. The NF-κB Pathway Promotes Tamoxifen Tolerance and Disease Recurrence in Estrogen Receptor-Positive Breast Cancers.
- Author
-
Kastrati I, Joosten SEP, Semina SE, Alejo LH, Brovkovych SD, Stender JD, Horlings HM, Kok M, Alarid ET, Greene GL, Linn SC, Zwart W, and Frasor J
- Subjects
- Animals, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms metabolism, Epithelial-Mesenchymal Transition drug effects, Female, Gene Expression Profiling methods, Gene Expression Regulation, Neoplastic drug effects, Humans, MCF-7 Cells, Mice, NF-kappa B metabolism, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local metabolism, Neoplasm Transplantation, Signal Transduction drug effects, Tamoxifen pharmacology, Breast Neoplasms pathology, Drug Resistance, Neoplasm, Estrogen Receptor alpha metabolism, Gene Regulatory Networks drug effects, Neoplasm Recurrence, Local pathology, Tamoxifen administration & dosage
- Abstract
The purpose of this study was to identify critical pathways promoting survival of tamoxifen-tolerant, estrogen receptor α positive (ER
+ ) breast cancer cells, which contribute to therapy resistance and disease recurrence. Gene expression profiling and pathway analysis were performed in ER+ breast tumors of patients before and after neoadjuvant tamoxifen treatment and demonstrated activation of the NF-κB pathway and an enrichment of epithelial-to mesenchymal transition (EMT)/stemness features. Exposure of ER+ breast cancer cell lines to tamoxifen, in vitro and in vivo , gives rise to a tamoxifen-tolerant population with similar NF-κB activity and EMT/stemness characteristics. Small-molecule inhibitors and CRISPR/Cas9 knockout were used to assess the role of the NF-κB pathway and demonstrated that survival of tamoxifen-tolerant cells requires NF-κB activity. Moreover, this pathway was essential for tumor recurrence following tamoxifen withdrawal. These findings establish that elevated NF-κB activity is observed in breast cancer cell lines under selective pressure with tamoxifen in vitro and in vivo , as well as in patient tumors treated with neoadjuvant tamoxifen therapy. This pathway is essential for survival and regrowth of tamoxifen-tolerant cells, and, as such, NF-κB inhibition offers a promising approach to prevent recurrence of ER+ tumors following tamoxifen exposure. IMPLICATIONS: Understanding initial changes that enable survival of tamoxifen-tolerant cells, as mediated by NF-κB pathway, may translate into therapeutic interventions to prevent resistance and relapse, which remain major causes of breast cancer lethality., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
35. Cytoplasmic ERα and NFκB Promote Cell Survival in Mouse Mammary Cancer Cell Lines.
- Author
-
Smart E, Alejo LH, and Frasor J
- Subjects
- Animals, Antineoplastic Agents, Hormonal pharmacology, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Line, Tumor, Cell Proliferation physiology, Cell Survival physiology, Cytoplasm metabolism, Dimethyl Fumarate pharmacology, Disease Models, Animal, Estrogen Receptor alpha antagonists & inhibitors, Female, Humans, MCF-7 Cells, Mammary Neoplasms, Experimental drug therapy, Mammary Neoplasms, Experimental pathology, Mice, NF-kappa B antagonists & inhibitors, Oxidative Stress physiology, Selective Estrogen Receptor Modulators pharmacology, Tamoxifen pharmacology, Estrogen Receptor alpha metabolism, Mammary Neoplasms, Experimental metabolism, NF-kappa B metabolism
- Abstract
There is a desperate need in the field for mouse mammary tumors and cell lines that faithfully mimic estrogen receptor (ER) expression and activity found in human breast cancers. We found that several mouse mammary cancer cell lines express ER but fail to demonstrate classical estrogen-driven proliferation or transcriptional activity. We investigated whether these cell lines may be used to model tamoxifen resistance by using small molecule inhibitors to signaling pathways known to contribute to resistance. We found that the combination of NFκB inhibition and ER antagonists significantly reduced cell proliferation in vitro, as well as growth of syngeneic tumors. Surprisingly, we found that ER was localized to the cytoplasm, regardless of any type of treatment. Based on this, we probed extra-nuclear functions of ER and found that co-inhibition of ER and NFκB led to an increase in oxidative stress and apoptosis. Together, these findings suggest that cytoplasmic ER and NFκB may play redundant roles in protecting mammary cancer cells from oxidative stress and cell death. Although this study has not identified a mouse model with classical ER activity, cytoplasmic ER has been described in a small subset of human breast tumors, suggesting that these findings may be relevant for some breast cancer patients.
- Published
- 2020
- Full Text
- View/download PDF
36. Removal of Serum Lipids and Lipid-Derived Metabolites to Investigate Breast Cancer Cell Biology.
- Author
-
Brovkovych V, Aldrich A, Li N, Atilla-Gokcumen GE, and Frasor J
- Subjects
- Apoptosis drug effects, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Cholesterol blood, Cholesterol isolation & purification, Estrogens pharmacology, Female, Humans, MCF-7 Cells, Phosphatidylcholines blood, Phosphatidylcholines isolation & purification, Tandem Mass Spectrometry, Breast Neoplasms metabolism, Cell Proliferation drug effects, Estradiol pharmacology, Lipids blood, Lipids isolation & purification, Silicates chemistry
- Abstract
The use of cultured cells has been instrumental in studying biochemical, molecular, and cellular processes. The composition of serum that cells are maintained in can have a profound impact on important cellular checkpoints. Cell growth and apoptosis are analyzed in an estrogen receptor positive breast cancer cell line in the presence of serum that have been treated to remove steroids or lipids, as well-described in the literature. It is shown that maintaining cells in the presence of charcoal-dextran-treated serum causes reduced growth rate, which can be reversed by the addition of estradiol. Silica-treated-serum also slows down cell growth and induces apoptosis. In order to investigate the role of lipids in these phenotypes, the levels of a wide range of lipids in different sera are investigated. It is shown that silica-treatment significantly depletes phosphatidylcholines and cholesterol. It is also shown that lipogenesis is stimulated when cells are cultured with silica-treated-serum and this is reversed by the addition of exogenous lipids, which also restores growth rate and apoptosis. The results show that cultured cells are sensitive to different serum, most likely due to the differences in levels of structural and signaling metabolites present in their growth environment., (© 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2019
- Full Text
- View/download PDF
37. Editorial for Special Issue on "Alternative nuclear receptor ligands".
- Author
-
Moore TW and Frasor J
- Subjects
- Animals, Humans, Ligands, Protein Binding drug effects, Receptors, Cytoplasmic and Nuclear chemistry, Signal Transduction drug effects, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology, Transcription Factors chemistry, Transcription Factors metabolism, Receptors, Cytoplasmic and Nuclear metabolism
- Published
- 2019
- Full Text
- View/download PDF
38. Structurally Diverse Histone Deacetylase Photoreactive Probes: Design, Synthesis, and Photolabeling Studies in Live Cells and Tissue.
- Author
-
Aboukhatwa SM, Hanigan TW, Taha TY, Neerasa J, Ranjan R, El-Bastawissy EE, Elkersh MA, El-Moselhy TF, Frasor J, Mahmud N, McLachlan A, and Petukhov PA
- Subjects
- Animals, Cells, Cultured, Fluorescent Dyes chemical synthesis, Fluorescent Dyes chemistry, Histone Deacetylase Inhibitors chemical synthesis, Histone Deacetylase Inhibitors chemistry, Humans, Isoenzymes antagonists & inhibitors, Isoenzymes metabolism, Liver diagnostic imaging, Mice, Mice, 129 Strain, Photoaffinity Labels chemical synthesis, Photoaffinity Labels chemistry, Drug Design, Fluorescent Dyes pharmacology, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylases metabolism, Optical Imaging, Photoaffinity Labels pharmacology
- Abstract
Histone deacetylase (HDAC) activity is modulated in vivo by post-translational modifications and formation of multiprotein complexes. Novel chemical tools to study how these factors affect engagement of HDAC isoforms by HDAC inhibitors (HDACi) in cells and tissues are needed. In this study, a synthetic strategy to access chemically diverse photoreactive probes (PRPs) was developed and used to prepare seven novel HDAC PRPs 9-15. The class I HDAC isoform engagement by PRPs was determined in biochemical assays and photolabeling experiments in live SET-2, HepG2, HuH7, and HEK293T cell lines and in mouse liver tissue. Unlike the HDAC protein abundance and biochemical activity against recombinant HDACs, the chemotype of the PRPs and the type of cells were key in defining the engagement of HDAC isoforms in live cells. Our findings suggest that engagement of HDAC isoforms by HDACi in vivo may be substantially modulated in a cell- and tissue-type-dependent manner., (© 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2019
- Full Text
- View/download PDF
39. Fatostatin induces pro- and anti-apoptotic lipid accumulation in breast cancer.
- Author
-
Brovkovych V, Izhar Y, Danes JM, Dubrovskyi O, Sakallioglu IT, Morrow LM, Atilla-Gokcumen GE, and Frasor J
- Abstract
Given the dependence of cancers on de novo lipogenesis, we tested the effect of fatostatin, a small molecule thought to target this pathway by blocking activation of SREBP transcription factors, in breast cancer cell lines and xenograft tumors. We found that estrogen receptor (ER) positive cells were more sensitive to fatostatin than ER negative cells and responded with cell cycle arrest and apoptosis. Surprisingly, we found that rather than inhibiting lipogenesis, fatostatin caused an accumulation of lipids as a response to endoplasmic reticulum stress rather than inhibition of SREBP activity. In particular, ceramide and dihydroceramide levels increased and contributed to the apoptotic effects of fatostatin. In addition, an accumulation of triacylglycerides (TAGs), particularly those containing polyunsaturated fatty acids (PUFAs), was also observed as a result of elevated diacylglycerol transferase activity. Blocking PUFA-TAG production enhanced the apoptotic effect of fatostatin, suggesting that these lipids play a protective role and limit fatostatin response. Together, these findings indicate that the ability of breast cancer cells to respond to fatostatin depends on induction of endoplasmic reticulum stress and subsequent ceramide accumulation, and that limiting production of PUFA-TAGs may be therapeutically beneficial in specific tumor subtypes.
- Published
- 2018
- Full Text
- View/download PDF
40. Using Tumor Explants for Imaging Mass Spectrometry Visualization of Unlabeled Peptides and Small Molecules.
- Author
-
David BP, Dubrovskyi O, Speltz TE, Wolff JJ, Frasor J, Sanchez LM, and Moore TW
- Abstract
Matrix assisted laser desorption ionization time-of-flight (MALDI-TOF) imaging mass spectrometry has emerged as a powerful, label-free technique to visualize penetration of small molecules in vivo and in vitro , including in 3D cell culture spheroids; however, some spheroids do not grow sufficiently large to provide enough area for imaging mass spectrometry. Here, we describe an ex vivo method for visualizing unlabeled peptides and small molecules in tumor explants, which can be divided into pieces of desired size, thus circumventing the size limitations of many spheroids. As proof-of-concept, a small molecule drug (4-hydroxytamoxifen), as well as a peptide drug (cyclosporin A) and peptide chemical probe, can be visualized after in vitro incubation with tumor explants so that this technique may provide a solution to robing cell penetration by unlabeled peptides., Competing Interests: The authors declare no competing financial interest.
- Published
- 2018
- Full Text
- View/download PDF
41. A Cell-Permeable Stapled Peptide Inhibitor of the Estrogen Receptor/Coactivator Interaction.
- Author
-
Speltz TE, Danes JM, Stender JD, Frasor J, and Moore TW
- Subjects
- Breast Neoplasms pathology, Cell Nucleus metabolism, Cell Proliferation drug effects, Female, Humans, Molecular Dynamics Simulation, Receptors, Estrogen metabolism, Transcription, Genetic drug effects, Breast Neoplasms drug therapy, Cell-Penetrating Peptides pharmacology, Receptors, Estrogen antagonists & inhibitors
- Abstract
We and others have proposed that coactivator binding inhibitors, which block the interaction of estrogen receptor and steroid receptor coactivators, may represent a potential class of new breast cancer therapeutics. The development of coactivator binding inhibitors has been limited, however, because many of the current molecules which are active in in vitro and biochemical assays are not active in cell-based assays. Our goal in this work was to prepare a coactivator binding inhibitor active in cellular models of breast cancer. To accomplish this, we used molecular dynamics simulations to convert a high-affinity stapled peptide with poor cell permeability into R4K1, a cell-penetrating stapled peptide. R4K1 displays high binding affinity for estrogen receptor α, inhibits the formation of estrogen receptor/coactivator complexes, and distributes throughout the cell with a high percentage of nuclear localization. R4K1 represses native gene transcription mediated by estrogen receptor α and inhibits proliferation of estradiol-stimulated MCF-7 cells. Using RNA-Seq, we demonstrate that almost all of the effects of R4K1 on global gene transcription are estrogen-receptor-associated. This chemical probe provides a significant proof-of-concept for preparing cell-permeable stapled peptide inhibitors of the estrogen receptor/coactivator interaction.
- Published
- 2018
- Full Text
- View/download PDF
42. Coactivation of Estrogen Receptor and IKKβ Induces a Dormant Metastatic Phenotype in ER-Positive Breast Cancer.
- Author
-
El-Shennawy L, Dubrovskyi O, Kastrati I, Danes JM, Zhang Y, Whiteley HE, Creighton CJ, and Frasor J
- Subjects
- Animals, Breast Neoplasms pathology, Cell Line, Tumor, Cell Proliferation, Female, Humans, Mice, Mice, Nude, Phenotype, Signal Transduction, Breast Neoplasms genetics, I-kappa B Kinase metabolism, Receptors, Estrogen metabolism
- Abstract
A growing body of evidence suggests that the inflammatory NFκB pathway is associated with the progression of ER
+ tumors to more aggressive stages. However, it is unknown whether NFκB is a driver or a consequence of aggressive ER+ disease. To investigate this question, we developed breast cancer cell lines expressing an inducible, constitutively active form of IκB kinase β (CA-IKKβ), a key kinase in the canonical NFκB pathway. We found that CA-IKKβ blocked E2-dependent cell proliferation in vitro and tumor growth in vivo in a reversible manner, suggesting that IKKβ may contribute to tumor dormancy and recurrence of ER+ disease. Moreover, coactivation of ER and IKKβ promoted cell migration and invasion in vitro and drove experimental metastasis in vivo Gene expression profiling revealed a strong association between ER and CA-IKKβ-driven gene expression and clinically relevant invasion and metastasis gene signatures. Mechanistically, the invasive phenotype appeared to be driven by an expansion of a basal/stem-like cell population rather than EMT. Taken together, our findings suggest that coactivation of ER and the canonical NFκB pathway promotes a dormant, metastatic phenotype in ER+ breast cancer and implicates IKKβ as a driver of certain features of aggressive ER+ breast cancer. Significance: The canonical NFκB pathway promotes expansion of stem/basal-like cells and a dormant, metastatic phenotype in ER+ breast cancer cells. Cancer Res; 78(4); 974-84. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
43. A Protective Role for Triacylglycerols during Apoptosis.
- Author
-
Li N, Sancak Y, Frasor J, and Atilla-Gokcumen GE
- Subjects
- Antibiotics, Antineoplastic pharmacology, Biomarkers metabolism, Cell Membrane chemistry, Cell Membrane drug effects, Cell Membrane metabolism, Cell Survival drug effects, Doxorubicin pharmacology, Etoposide pharmacology, Fatty Acids, Nonesterified adverse effects, Fatty Acids, Unsaturated analysis, HCT116 Cells, Humans, Intracellular Membranes chemistry, Intracellular Membranes drug effects, Intracellular Membranes metabolism, Lipid Droplets chemistry, Lipid Droplets drug effects, Lipid Droplets metabolism, Lipid Peroxidation drug effects, MCF-7 Cells, Oxidative Stress drug effects, Staurosporine pharmacology, Topoisomerase II Inhibitors pharmacology, Triglycerides chemistry, Apoptosis drug effects, Fatty Acids, Unsaturated metabolism, Models, Biological, Triglycerides metabolism
- Abstract
Triacylglycerols (TAGs) are one of the major constituents of the glycerolipid family. Their main role in cells is to store excess fatty acids, and they are mostly found within lipid droplets. TAGs contain acyl chains that vary in length and degree of unsaturation, resulting in hundreds of chemically distinct species. We have previously reported that TAGs containing polyunsaturated fatty acyl chains (PUFA-TAGs) accumulate via activation of diacylglycerol acyltransferases during apoptosis. In this work, we show that accumulation of PUFA-TAGs is a general phenomenon during this process. We further show that the accumulated PUFA-TAGs are stored in lipid droplets. Because membrane-residing PUFA phospholipids can undergo oxidation and form reactive species under increased levels of oxidative stress, we hypothesized that incorporation of PUFAs into PUFA-TAGs and their localization within lipid droplets during apoptosis limit the toxicity during this process. Indeed, exogenous delivery of a polyunsaturated fatty acid resulted in a profound accumulation of PUFA phospholipids and rendered cells more sensitive to oxidative stress, causing reduced viability. Overall, our results support the concept that activation of TAG biosynthesis protects cells from lipid peroxide-induced membrane damage under increased levels of oxidative stress during apoptosis. As such, targeting triacylglycerol biosynthesis in cancer cells might represent a new approach to promoting cell death during apoptosis.
- Published
- 2018
- Full Text
- View/download PDF
44. Histone deacetylase inhibitor-based chromatin precipitation for identification of targeted genomic loci.
- Author
-
Hanigan TW, Danes JM, Taha TY, Frasor J, and Petukhov PA
- Abstract
Histone deacetylase (HDAC) catalyzes the removal of acetyl marks from histones, effectively regulating gene expression. Genome wide chromatin immunoprecipitation (ChIP) studies have shown HDACs are present on numerous active and repressed genes. However, HDAC inhibitors (HDACi) only regulate a small subset of this population in a cell type dependent fashion. To determine genomic locations directly targeted by HDACi, we developed a chromatin precipitation method using a photoreactive HDAC inhibitor probe (photomate). We validate this method by analyzing several canonical HDACi regulated genes, CDKN1A and FOSL1, and compare it to traditional ChIP using HDAC1 antibodies. We show that HDACi target HDACs bound at the promoter regions but not gene bodies, differing from HDAC1 antibody-based ChIP in the case of CDKN1A. This approach is anticipated to be useful for genome wide studies to identify the subset of genes directly regulated by an HDACi in a given cell type., Competing Interests: Competing interests: The authors have declared that no competing interests exist.
- Published
- 2018
- Full Text
- View/download PDF
45. Design, Synthesis, Molecular Modeling, and Biological Evaluation of Novel Amine-based Histone Deacetylase Inhibitors.
- Author
-
Abdelkarim H, Neelarapu R, Madriaga A, Vaidya AS, Kastrati I, Karumudi B, Wang YT, Taha TY, Thatcher GRJ, Frasor J, and Petukhov PA
- Subjects
- Amines chemical synthesis, Amines chemistry, Antineoplastic Agents chemical synthesis, Antineoplastic Agents chemistry, Cell Line, Tumor, Cell Proliferation drug effects, Crystallography, X-Ray, Dose-Response Relationship, Drug, Drug Screening Assays, Antitumor, Histone Deacetylase Inhibitors chemical synthesis, Histone Deacetylase Inhibitors chemistry, Humans, Models, Molecular, Molecular Structure, Structure-Activity Relationship, Amines pharmacology, Antineoplastic Agents pharmacology, Drug Design, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylases metabolism
- Abstract
Histone deacetylases (HDACs) are promising drug targets for a variety of therapeutic applications. Herein we describe the design, synthesis, biological evaluation in cellular models of cancer, and preliminary drug metabolism and pharmacokinetic studies (DMPK) of a series of secondary and tertiary N-substituted 7-aminoheptanohydroxamic acid-based HDAC inhibitors. Introduction of an amino group with one or two surface binding groups (SBGs) yielded a successful strategy to develop novel and potent HDAC inhibitors. The secondary amines were found to be generally more potent than the corresponding tertiary amines. Docking studies suggested that the SBGs of tertiary amines cannot be favorably accommodated at the gorge region of the binding site. The secondary amines with naphthalen-2-ylmethyl, 5-phenylthiophen-2-ylmethyl, and 1H-indol-2-ylmethyl (2 j) substituents exhibited the highest potency against class I HDACs: HDAC1 IC
50 39-61 nm, HDAC2 IC50 260-690 nm, HDAC3 IC50 25-68 nm, and HDAC8 IC50 320-620 nm. The cytotoxicity of a representative set of secondary and tertiary N-substituted 7-aminoheptanoic acid hydroxyamide-based inhibitors against HT-29, SH-SY5Y, and MCF-7 cancer cells correlated with their inhibition of HDAC1, 2, and 3 and was found to be similar to or better than that of suberoylanilide hydroxamic acid (SAHA). Compounds in this series increased the acetylation of histones H3 and H4 in a time-dependent manner. DMPK studies indicated that secondary amine 2 j is metabolically stable and has plasma and brain concentrations >23- and >1.6-fold higher than the IC50 value for class I HDACs, respectively. Overall, the secondary and tertiary N-substituted 7-aminoheptanoic acid hydroxyamide-based inhibitors exhibit excellent lead- and drug-like properties and therapeutic capacity for cancer applications., (© 2017 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.)- Published
- 2017
- Full Text
- View/download PDF
46. Divergent JNK Phosphorylation of HDAC3 in Triple-Negative Breast Cancer Cells Determines HDAC Inhibitor Binding and Selectivity.
- Author
-
Hanigan TW, Aboukhatwa SM, Taha TY, Frasor J, and Petukhov PA
- Subjects
- Cell Line, Tumor, Female, Histone Deacetylase Inhibitors chemistry, Histone Deacetylase Inhibitors metabolism, Histone Deacetylases chemistry, Histone Deacetylases genetics, Humans, Hydroxamic Acids chemistry, Hydroxamic Acids metabolism, Isoenzymes chemistry, Isoenzymes genetics, Isoenzymes metabolism, MCF-7 Cells, Phosphorylation, Protein Binding, Recombinant Proteins biosynthesis, Recombinant Proteins chemistry, Recombinant Proteins isolation & purification, Triple Negative Breast Neoplasms metabolism, Triple Negative Breast Neoplasms pathology, Histone Deacetylases metabolism, JNK Mitogen-Activated Protein Kinases metabolism
- Abstract
Histone deacetylase (HDAC) catalytic activity is regulated by formation of co-regulator complexes and post-translational modification. Whether these mechanisms are transformed in cancer and how this affects the binding and selectivity of HDAC inhibitors (HDACis) is unclear. In this study, we developed a method that identified a 3- to 16-fold increase in HDACi selectivity for HDAC3 in triple-negative breast cancer (TNBC) cells in comparison with luminal subtypes that was not predicted by current practice measurements with recombinant proteins. We found this increase was caused by c-Jun N-terminal kinase (JNK) phosphorylation of HDAC3, was independent of HDAC3 complex composition or subcellular localization, and was associated with a 5-fold increase in HDAC3 enzymatic activity. This study points to HDAC3 and the JNK axes as targets in TNBC, highlights how HDAC phosphorylation affects HDACi binding and selectivity, and outlines a method to identify changes in individual HDAC isoforms catalytic activity, applicable to any disease state., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
47. Scaffold dependent histone deacetylase (HDAC) inhibitor induced re-equilibration of the subcellular localization and post-translational modification state of class I HDACs.
- Author
-
Hanigan TW, Taha TY, Aboukhatwa SM, Frasor J, and Petukhov PA
- Subjects
- Acetylation drug effects, Cytosol drug effects, Cytosol enzymology, Histone Deacetylase Inhibitors chemistry, Histones metabolism, Humans, MCF-7 Cells, Microscopy, Confocal, Mitogens pharmacology, Models, Biological, Phosphorylation drug effects, Protein Transport drug effects, Subcellular Fractions drug effects, Subcellular Fractions enzymology, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylases metabolism, Protein Processing, Post-Translational drug effects
- Abstract
The mechanism of action of histone deacetylase inhibitors (HDACi) is mainly attributed to the inhibition of the deacetylase catalytic activity for their histone substrates. In this study, we analyzed the abundance of class I HDACs in the cytosolic, nuclear soluble and chromatin bound cellular fractions in breast cancer cells after HDACi treatment. We found that potent N-hydroxy propenamide-based HDACi induced a concentration dependent decrease in the HDAC1 associated with chromatin and a lasting concomitant increase in cytoplasmic HDAC1 while maintaining total protein expression. No such change occurred with HDAC2 or 8, however, an increase in cytoplasmic non-phosphorylated HDAC3 was also observed. The subcellular re-equilibration of HDAC1 was subsequent to the accumulation of acetylated histones and might be cell cycle dependent. This study suggests that the biological activity of a subset of N-hydroxy propenamide-based HDACi may stem from direct competition with histone substrates of HDACs as well as from spatial separation from their substrates in the nucleus and/or change in post-translational modification status of HDACs.
- Published
- 2017
- Full Text
- View/download PDF
48. A Novel Strategy to Co-target Estrogen Receptor and Nuclear Factor κB Pathways with Hybrid Drugs for Breast Cancer Therapy.
- Author
-
Kastrati I, Siklos MI, Brovkovych SD, Thatcher GRJ, and Frasor J
- Subjects
- Breast Neoplasms genetics, Breast Neoplasms pathology, Female, Fumarates administration & dosage, Humans, MCF-7 Cells, Molecular Targeted Therapy, NF-kappa B antagonists & inhibitors, Raloxifene Hydrochloride administration & dosage, Receptors, Estrogen antagonists & inhibitors, Selective Estrogen Receptor Modulators administration & dosage, Signal Transduction drug effects, Tamoxifen administration & dosage, Antineoplastic Agents, Hormonal administration & dosage, Breast Neoplasms drug therapy, NF-kappa B genetics, Receptors, Estrogen genetics
- Abstract
Nearly 75% of breast tumors express estrogen receptor (ER), and will be treated with endocrine therapy, such as selective estrogen receptor modulator (SERM), tamoxifen, or aromatase inhibitors. Despite their proven success, as many as 40-50% of ER+ tumors fail to respond to endocrine therapy and eventually recur as aggressive, metastatic cancers. Therefore, preventing and/or overcoming endocrine resistance in ER+ tumors remains a major clinical challenge. Deregulation or activation of the nuclear factor κB (NFκB) pathway has been implicated in endocrine resistance and poor patient outcome in ER+ tumors. As a consequence, one option to improve on existing anti-cancer treatment regimens may be to introduce additional anti-NFκB activity to endocrine therapy drugs. Our approach was to design and test SERM-fumarate co-targeting hybrid drugs capable of simultaneously inhibiting both ER, via the SERM, raloxifene, and the NFκB pathway, via fumarate, in breast cancer cells. We find that the hybrid drugs display improved anti-NFκB pathway inhibition compared to either raloxifene or fumarate. Despite some loss in potency against the ER pathway, these hybrid drugs maintain anti-proliferative activity in ER+ breast cancer cells. Furthermore, these drugs prevent clonogenic growth and mammosphere formation of ER+ breast cancer cells. As a proof-of-principle, the simultaneous inhibition of ER and NFκB via a single bifunctional hybrid drug may represent a viable approach to improve the anti-inflammatory activity and prevent therapy resistance of ER-targeted anti-cancer drugs.
- Published
- 2017
- Full Text
- View/download PDF
49. Racial disparity in survival from estrogen and progesterone receptor-positive breast cancer: implications for reducing breast cancer mortality disparities.
- Author
-
Rauscher GH, Silva A, Pauls H, Frasor J, Bonini MG, and Hoskins K
- Subjects
- Black or African American, Aged, Biomarkers, Tumor metabolism, Breast Neoplasms ethnology, Breast Neoplasms metabolism, Chicago epidemiology, Female, Hispanic or Latino, Humans, Kaplan-Meier Estimate, Middle Aged, Proportional Hazards Models, Receptors, Estrogen metabolism, Receptors, Progesterone metabolism, Breast Neoplasms mortality, Health Status Disparities
- Abstract
Introduction: Non-Latina black breast cancer patients experience a shorter survival from breast cancer than their non-Latina white counterparts. We compared breast cancer-specific survival for the subset of black and white patients with estrogen and/or progesterone receptor-positive tumors that are generally targeted with endocrine therapy., Methods: Using data collected from a population-based cohort of breast cancer patients from Chicago, IL, Kaplan-Meier survival curves and hazard functions were generated and proportional hazards models were estimated to determine the black/white disparity in time to death from breast cancer while adjusting for age at diagnosis, patient characteristics, treatment-related variables, and tumor grade and stage., Results: In regression models, hazard of breast cancer death among ER/PR-positive patients was at least 4 times higher for black than for white patients in all models tested. Notably, even after adjusting for stage at diagnosis, tumor grade, and treatment variables (including initiation of systemic adjuvant therapies), the hazard ratio for death from ER/PR-positive breast cancer between black and white women was 4.39 (95% CI 1.76, 10.9, p = 0.001)., Conclusions: We observed a racial disparity in breast cancer survival for patients diagnosed with ER/PR-positive tumors that did not appear to be due to differences in tumor stage, grade, or therapy initiation in black patients, suggesting that there may be racial differences in the molecular characteristics of hormone receptor-positive tumors, such that ER/PR-positive tumors in black patients may be less responsive to standard treatments.
- Published
- 2017
- Full Text
- View/download PDF
50. Corrigendum: Full antagonism of the estrogen receptor without a prototypical ligand side chain.
- Author
-
Srinivasan S, Nwachukwu JC, Bruno NE, Dharmarajan V, Goswami D, Kastrati I, Novick S, Nowak J, Cavett V, Zhou HB, Boonmuen N, Zhao Y, Min J, Frasor J, Katzenellenbogen BS, Griffin PR, Katzenellenbogen JA, and Nettles KW
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.