1
|
Van Moortel L, Verhee A, Thommis J, Houtman R, Melchers D, Delhaye L, Van Leene C, Hellemans M, Gevaert K, Eyckerman S, De Bosscher K. Selective Modulation of the Human Glucocorticoid Receptor Compromises GR Chromatin Occupancy and Recruitment of p300/CBP and the Mediator Complex. Mol Cell Proteomics 2024; 23:100741. [PMID: 38387774 PMCID: PMC10957501 DOI: 10.1016/j.mcpro.2024.100741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 02/13/2024] [Accepted: 02/17/2024] [Indexed: 02/24/2024] Open
Abstract
Exogenous glucocorticoids are frequently used to treat inflammatory disorders and as adjuncts for the treatment of solid cancers. However, their use is associated with severe side effects and therapy resistance. Novel glucocorticoid receptor (GR) ligands with a patient-validated reduced side effect profile have not yet reached the clinic. GR is a member of the nuclear receptor family of transcription factors and heavily relies on interactions with coregulator proteins for its transcriptional activity. To elucidate the role of the GR interactome in the differential transcriptional activity of GR following treatment with the selective GR agonist and modulator dagrocorat compared to classic (ant)agonists, we generated comprehensive interactome maps by high-confidence proximity proteomics in lung epithelial carcinoma cells. We found that dagrocorat and the antagonist RU486 both reduced GR interaction with CREB-binding protein/p300 and the mediator complex compared to the full GR agonist dexamethasone. Chromatin immunoprecipitation assays revealed that these changes in GR interactome were accompanied by reduced GR chromatin occupancy with dagrocorat and RU486. Our data offer new insights into the role of differential coregulator recruitment in shaping ligand-specific GR-mediated transcriptional responses.
Collapse
Affiliation(s)
- Laura Van Moortel
- VIB-UGent Center for Medical Biotechnology, VIB Institute, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Annick Verhee
- VIB-UGent Center for Medical Biotechnology, VIB Institute, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jonathan Thommis
- VIB-UGent Center for Medical Biotechnology, VIB Institute, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | | | | | - Louis Delhaye
- VIB-UGent Center for Medical Biotechnology, VIB Institute, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Chloé Van Leene
- VIB-UGent Center for Medical Biotechnology, VIB Institute, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Madeleine Hellemans
- VIB-UGent Center for Medical Biotechnology, VIB Institute, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB-UGent Inflammation Research Center, VIB Institute, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Kris Gevaert
- VIB-UGent Center for Medical Biotechnology, VIB Institute, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Sven Eyckerman
- VIB-UGent Center for Medical Biotechnology, VIB Institute, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.
| | - Karolien De Bosscher
- VIB-UGent Center for Medical Biotechnology, VIB Institute, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.
| |
Collapse
|
2
|
Chilakamarthi U, Mahadik NS, Bhattacharyya T, Gangadhar PS, Giribabu L, Banerjee R. Glucocorticoid receptor mediated sensitization of colon cancer to photodynamic therapy induced cell death. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 251:112846. [PMID: 38237432 DOI: 10.1016/j.jphotobiol.2024.112846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/30/2023] [Accepted: 01/12/2024] [Indexed: 02/10/2024]
Abstract
Photodynamic therapy (PDT) is a clinically approved, non-invasive alternate cancer therapy. A synthetic glucocorticoid (GC), dexamethasone (Dex) has previously been demonstrated to sensitize cancer cells to chemotherapy. However, to the best of our knowledge, the sensitization effect of GCs on PDT has not yet been investigated. We hypothesized that glucocorticoid receptor (GR) targeting can selectively make cancer cells more sensitive to PDT treatment, as PDT induces hypoxia wherein GR-activity gets enhanced. In addition, Dex was reported to act against the PDT-induced cell survival pathways like HIF-1α, NRF2, NF-κB, STAT3 etc. Thus, both the treatments can complement each other and may result in increasing the effectiveness of combination therapy. Hence, in this study, we developed liposomal formulations of our previously reported PDT agent P-Nap, either alone (D1P-Nap) or in combination with Dex (D1XP-Nap) to elucidate the sensitization effect. Interestingly, our RT-PCR results in hypoxic conditions showed down-regulation of HIF-1α and over expression of GR-activated genes for glucose-6-phosphatase (G6Pase) and PEPCK enzymes, indicating prominent GR-transactivation. We also observed higher phototoxicity in CT26.WT cells treated with D1XP-Nap PDT under hypoxic conditions as compared to normoxic conditions. These effects were reversed when cells were pre-treated with RU486, a competitive inhibitor of GCs. Moreover, our in vivo findings of subcutaneous tumor model of Balb/C mice for colon cancer revealed a significant decrease in tumor volume as well as considerable enhancement in the survivability of PDT treated tumor-bearing mice when Dex was present in the formulation. A high Bax/Bcl-xL ratio, high p53 expression, enhanced E-cadherin expression and down-regulation of pro-tumorigenic transcription factors NF-κB and c-Myc were found in tumor lysates from mice treated with D1XP-Nap under PDT, indicating GR-mediated sensitization of the tumor to PDT-induced cell death and enhancement of life-span for tumor bearing mice.
Collapse
Affiliation(s)
- Ushasri Chilakamarthi
- Department of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India.
| | - Namita S Mahadik
- Department of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Gaziabad 201002, U.P., India
| | - Tithi Bhattacharyya
- Department of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Gaziabad 201002, U.P., India
| | - Palivela Siva Gangadhar
- Academy of Scientific and Innovative Research (AcSIR), Gaziabad 201002, U.P., India; Polymers and Functional Materials Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Lingamallu Giribabu
- Academy of Scientific and Innovative Research (AcSIR), Gaziabad 201002, U.P., India; Polymers and Functional Materials Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Rajkumar Banerjee
- Department of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Gaziabad 201002, U.P., India.
| |
Collapse
|
3
|
Misra R, Sivaranjani A, Saleem S, Dash BR. Copper Nanoclusters as Novel Podium for Cancer Detection, Imaging, and Therapy Applications. Crit Rev Ther Drug Carrier Syst 2024; 41:51-80. [PMID: 37938190 DOI: 10.1615/critrevtherdrugcarriersyst.2023044994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Nanoclusters (NCs) are meticulously small, kinetically stable, crystalline materials which hold immense potential as multifaceted catalysts for a broad range of biomedical applications. Metal NCs are atomically precise and exist within the range of Fermi wavelength of electrons. They are highly advantageous as functional materials as their physicochemical properties can be customized to meet specific requirements. Copper NCs (CuNCs) are emerging as an efficient substitute to the other existing metal NCs. The synthesis of CuNCs is highly methodical, fast, cost effective and does not involve any complicated manipulation. On the contrary to gold and silver NCs, copper is a vital trace element for humans that can be excreted easily out the body. Further, the relatively inexpensiveness and easy availability of copper aids in potential nanotechnological applications in large quantity. As such, CuNCs have attracted great interest among the research community recently. The modern developments in the strategy, synthesis, surface modifications, and use of CuNCs in diagnosis of disease, imaging and treatment have been discussed in the present review. Approaches to regulate and augment the emission of CuNCs, challenges and drawbacks have also been considered. This review brings to light the multifarious applications of CuNCs and their potential as emerging theranostic agents. It is anticipated that the visions and directions for translating existing developments in CuNCs from the laboratory to the clinic can be further improved and enhanced.
Collapse
Affiliation(s)
- Ranjita Misra
- Department of Biotechnology, School of Sciences, Jain University, Bangalore, Karnataka, India
| | - A Sivaranjani
- Advanced Institute for Wildlife Conservation, Chennai, Tamil Nadu, India
| | - Suraiya Saleem
- Department of Biotechnology, Indian Institute of Technology, Madras, Chennai 600036, Tamil Nadu, India
| | - Bignya Rani Dash
- Department of Chemistry, Indian Institute of Technology, Madras, Chennai 600036, Tamil Nadu, India
| |
Collapse
|
4
|
Mozaffari MS, Abdelsayed R. Expression Profiles of GILZ and SGK-1 in Potentially Malignant and Malignant Human Oral Lesions. FRONTIERS IN ORAL HEALTH 2022; 2:675288. [PMID: 35048019 PMCID: PMC8757717 DOI: 10.3389/froh.2021.675288] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 08/13/2021] [Indexed: 02/06/2023] Open
Abstract
Glucocorticoid-induced leucine zipper and serum-glucocorticoid-regulated kinase-1 (SGK-1) are major glucocorticoid-inducible proteins. Recent studies indicate the local production of cortisol in oral mucosa, which can impact the tissue generation of glucocorticoid-induced leucine zipper (GILZ) and SGK-1. Furthermore, GILZ and SGK-1 play pathogenic roles in a variety of cancers, but their status in potentially malignant (e.g., epithelial dysplasia) or malignant oral lesions remains unknown. This study tested the hypothesis that expression profiles of GILZ and SGK-1, along with the phosphorylated (active) form of SGK-1 (pSGK-1), are different in epithelial dysplasia than squamous cell carcinoma. Accordingly, archived paraffin-embedded biopsy samples were subjected to immunohistochemistry to establish tissue localization and the profile of proteins of interest, while hematoxylin-eosin stained tissues were used for histopathological assessment. Based on histopathological examinations, tissue specimens were categorized as displaying mild-moderate or severe epithelial dysplasia and squamous cell carcinoma; benign keratosis specimens served as controls. All the tissue specimens showed staining for SGK-1 and pSGK-1; however, while SGK-1 staining was primarily cytoplasmic, pSGK-1 was mainly confined to the cell membrane. On the other hand, all the tissue specimens displayed primarily nuclear staining for GILZ. A semi-quantitative analysis of immunohistochemistry staining indicates increased GILZ expression in epithelial dysplasia but reversal in squamous cell carcinoma to a level seen for benign keratosis. On the other hand, the SGK-1 and pSGK-1 expressions decreased for squamous cell carcinoma specimens compared with benign keratosis or dysplastic specimens. Collectively, in this cross-sectional study, immunostaining patterns for proteins of interest do not seemingly differentiate epithelial dysplasia from squamous cell carcinoma. However, subcellular localization and expression profiles for GILZ, SGK-1, and pSGK-1 are suggestive of differential functional roles in dysplastic or malignant oral lesions compared with benign keratosis.
Collapse
Affiliation(s)
- Mahmood S Mozaffari
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, Augusta, Georgia
| | - Rafik Abdelsayed
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
5
|
Kuan LL, Dennison AR, Garcea G. Outcomes of peri-operative glucocorticosteroid use in major pancreatic resections: a systematic review. HPB (Oxford) 2021; 23:1789-1798. [PMID: 34593313 DOI: 10.1016/j.hpb.2021.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 07/12/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND There is increasing evidence that peri-operative glucocorticosteroid can ameliorate the systemic response following major surgery. Preliminary evidence suggests peri-operative usage of glucocorticosteroid may decrease post-operative complications. These positive associations have been observed in a range of different operations including intra-abdominal, thoracic, cardiac, and orthopaedic surgery. This review aims to investigate the impact of peri-operative glucocorticosteroid in major pancreatic resections. METHODS A systematic review based on a search in Medline and Embase databases was performed. PRISMA guidelines for systematic reviews were followed. RESULTS A total of five studies were analysed; three randomised controlled trials and two retrospective cohort studies. The total patient population was 1042. The glucocorticosteroids used were intravenous hydrocortisone or dexamethasone. Three studies reported significantly lower morbidity in the peri-operative glucocorticosteroid group. The number needed to treat to prevent one major complication with hydrocortisone is four patients. Two studies demonstrated that dexamethasone was associated with a statistically significantly improved median overall survival in pancreatic cancer. CONCLUSION This is the first systematic review conducted to investigate the significance of peri-operative glucocorticosteroid in patients undergoing pancreatic resection. This review shows a correlation of positive outcomes with the administration of glucocorticosteroid in the peri-operative setting following a major pancreatic resection.. More randomised clinical trials are required to confirm if this is a true effect, as it would have significant implications.
Collapse
Affiliation(s)
- Li Lian Kuan
- Department of Hepatobiliary and Pancreatic Surgery, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom; Discipline of Surgery, The University of Adelaide, The Queen Elizabeth Hospital, Adelaide, South Australia, Australia.
| | - Ashley R Dennison
- Department of Hepatobiliary and Pancreatic Surgery, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Giuseppe Garcea
- Department of Hepatobiliary and Pancreatic Surgery, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| |
Collapse
|
6
|
Cai X, Tao W, Li L. Glioma cell-derived FGF20 suppresses macrophage function by activating β-catenin. Cell Signal 2021; 89:110181. [PMID: 34757019 DOI: 10.1016/j.cellsig.2021.110181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 10/23/2021] [Accepted: 10/26/2021] [Indexed: 01/19/2023]
Abstract
Macrophages, which are the main regulators of the tumor-associated microenvironment, play a crucial role in the progression of various tumors. The anti-inflammatory role of β-catenin in macrophages has been extensively studied in recent years. However, the association between macrophages and β-catenin with regards to the development of glioma has not yet been investigated, at least to the best of our knowledge. The present study found that fibroblast growth factor 20 (FGF20), as a paracrine cytokine, was secreted by glioma cells and acted on macrophages. FGF20 treated macrophages exhibited a decreased pro-inflammatory phenotype upon LPS and IFN-γ stimulation, characterized by the decreased the level of M1 macrophage markers and the reduced production of pro-inflammatory cytokines. Mechanistic analysis revealed that FGF20 interacted with FGF receptor 1 isoform of macrophages, and subsequently increased the stability of β-catenin via phosphorylating GSK3β, which suppressed macrophage polarization to the M1-phenotype. Finally, it was found that FGF20 of glioma cells expression was upregulated by the glucocorticoids (GCs) treatment, and decreased FGF20 expression of glioma cells markedly blocked the effects of GCs on the polarization of macrophages. On the whole, the present study demonstrates that FGF20, secreted from glioma cells, participates the GCs regulated macrophage function and exerts anti-inflammatory effects during the treatment of glioma by GCs. Moreover, a molecular link was identified between glioma cells and macrophages, demonstrating that FGF20 modulates the GCs-induced dysfunction of macrophages during glioma development.
Collapse
Affiliation(s)
- Xue Cai
- Department of Emergency, ShengJing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China.
| | - Weichen Tao
- Department of Emergency, ShengJing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China
| | - Lei Li
- Department of Emergency, ShengJing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China.
| |
Collapse
|
7
|
Sridharan K, Rathore B, Yousuf M, Reddy Rachamalla HK, Jinka S, Jaggarapu MMCS, Banerjee R. Self-Assembling Derivative of Hydrocortisone as Glucocorticoid Receptor-Targeted Nanotherapeutics for Synergistic, Combination Therapy against Colorectal Tumor. Mol Pharm 2020; 18:1208-1228. [PMID: 33371687 DOI: 10.1021/acs.molpharmaceut.0c01091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Hydrocortisone, a natural glucocorticoid secreted by adrenal and extra-adrenal tissues, locally governs the transcription of genes involved in inflammation, immune response, metabolism, and energy homeostasis via binding to its cognate glucocorticoid receptor (GR). In this study, we show that modified hydrocortisone (HC16), a cancer-selective cytotoxic molecule, showed synergism in combination with drugs like Doxorubicin and docetaxel, self-assembled into vesicles, entrapped docetaxel and complexed with anti-cancer plasmid DNA for enhanced killing of cancer cells. These vesicles exhibited GR-mediated nuclear localization, delivery of the p53 gene, and also inhibited cell viability selectively in RKO, HCT15, and CT26 colon cancer cells but not in normal cells like CHO and HEK293T. Apart from exerting its own anti-cancer activity, the self-assembled HC16 vesicles loaded with docetaxel sensitized the cancer cells to its drug cargo by downregulating the drug metabolizing CYP3A4 gene. This indirectly reduces the risk of nonspecific adverse effects in normal cells, as the viability of sensitized cancer cells could be significantly reduced even in low doses of cytotoxic docetaxel. The near infrared (NIR)-dye-associated self-assemblies accumulated in a colon tumor with higher orders of NIR intensity compared to those in a colon of healthy mice. Thereafter, the treatment of HC16-docetaxel-p53 vesicle/DNA complex led to significant tumor regression, which resulted in a cecum/body weight ratio in tumor-bearing mice similar to that of healthy mice measured at 24 h postcompletion of treatment. There was an up to 2.5-fold enhancement in the overall survivability of colon-tumor-bearing mice treated with HC16-docetaxel-p53 vesicle/DNA complexes when compared against the pristine docetaxel-treated groups. Further, the HC16-docetaxel-p53 vesicle/DNA complex-treated group showed reduced nuclear accumulation of cell proliferation marker Ki67, reduced protein levels of prosurvival and mesenchymal proteins like Bcl-2, PARP, vimentin, and N-cadherin, and increased the levels of pro-apoptotic activated caspases as compared to the pristine docetaxel-treated groups. The therapeutic package described herein is expected to find future use as a rational, multifaceted, GR-targeted approach for inhibiting colon tumor progression.
Collapse
Affiliation(s)
- Kathyayani Sridharan
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| | - Bhowmira Rathore
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - Md Yousuf
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.,Department of Chemistry, Ramnagar College, Purba Medinipur, West Bengal 721 453, India
| | - Hari Krishna Reddy Rachamalla
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| | - Sudhakar Jinka
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| | - Madhan Mohan Chandra Sekhar Jaggarapu
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| | - Rajkumar Banerjee
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| |
Collapse
|
8
|
Suzuki S, Okada M, Sanomachi T, Togashi K, Seino S, Sato A, Yamamoto M, Kitanaka C. Therapeutic targeting of pancreatic cancer stem cells by dexamethasone modulation of the MKP-1-JNK axis. J Biol Chem 2020; 295:18328-18342. [PMID: 33115754 PMCID: PMC7939393 DOI: 10.1074/jbc.ra120.015223] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/16/2020] [Indexed: 12/24/2022] Open
Abstract
Postoperative recurrence from microscopic residual disease must be prevented to cure intractable cancers, including pancreatic cancer. Key to this goal is the elimination of cancer stem cells (CSCs) endowed with tumor-initiating capacity and drug resistance. However, current therapeutic strategies capable of accomplishing this are insufficient. Using in vitro models of CSCs and in vivo models of tumor initiation in which CSCs give rise to xenograft tumors, we show that dexamethasone induces expression of MKP-1, a MAPK phosphatase, via glucocorticoid receptor activation, thereby inactivating JNK, which is required for self-renewal and tumor initiation by pancreatic CSCs as well as for their expression of survivin, an anti-apoptotic protein implicated in multidrug resistance. We also demonstrate that systemic administration of clinically relevant doses of dexamethasone together with gemcitabine prevents tumor formation by CSCs in a pancreatic cancer xenograft model. Our study thus provides preclinical evidence for the efficacy of dexamethasone as an adjuvant therapy to prevent postoperative recurrence in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Shuhei Suzuki
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan; Department of Clinical Oncology, Yamagata University School of Medicine, Yamagata, Japan
| | - Masashi Okada
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan.
| | - Tomomi Sanomachi
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan; Department of Clinical Oncology, Yamagata University School of Medicine, Yamagata, Japan
| | - Keita Togashi
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan; Department of Ophthalmology and Visual Sciences, Yamagata University School of Medicine, Yamagata, Japan
| | - Shizuka Seino
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan
| | - Atsushi Sato
- Department of Neurosurgery, Yamagata University School of Medicine, Yamagata, Japan
| | - Masahiro Yamamoto
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan
| | - Chifumi Kitanaka
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan; Research Institute for Promotion of Medical Sciences, Faculty of Medicine, Yamagata University, Yamagata, Japan.
| |
Collapse
|
9
|
Conway ME, McDaniel JM, Graham JM, Guillen KP, Oliver PG, Parker SL, Yue P, Turkson J, Buchsbaum DJ, Welm BE, Myers RM, Varley KE. STAT3 and GR Cooperate to Drive Gene Expression and Growth of Basal-Like Triple-Negative Breast Cancer. Cancer Res 2020; 80:4355-4370. [PMID: 32816914 DOI: 10.1158/0008-5472.can-20-1379] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/17/2020] [Accepted: 08/13/2020] [Indexed: 11/16/2022]
Abstract
Breast cancers are divided into subtypes with different prognoses and treatment responses based on global differences in gene expression. Luminal breast cancer gene expression and proliferation are driven by estrogen receptor alpha, and targeting this transcription factor is the most effective therapy for this subtype. By contrast, it remains unclear which transcription factors drive the gene expression signature that defines basal-like triple-negative breast cancer, and there are no targeted therapies approved to treat this aggressive subtype. In this study, we utilized integrated genomic analysis of DNA methylation, chromatin accessibility, transcription factor binding, and gene expression in large collections of breast cancer cell lines and patient tumors to identify transcription factors responsible for the basal-like gene expression program. Glucocorticoid receptor (GR) and STAT3 bind to the same genomic regulatory regions, which were specifically open and unmethylated in basal-like breast cancer. These transcription factors cooperated to regulate expression of hundreds of genes in the basal-like gene expression signature, which were associated with poor prognosis. Combination treatment with small-molecule inhibitors of both transcription factors resulted in synergistic decreases in cell growth in cell lines and patient-derived organoid models. This study demonstrates that GR and STAT3 cooperate to regulate the basal-like breast cancer gene expression program and provides the basis for improved therapy for basal-like triple-negative breast cancer through rational combination of STAT3 and GR inhibitors. SIGNIFICANCE: This study demonstrates that GR and STAT3 cooperate to activate the canonical gene expression signature of basal-like triple-negative breast cancer and that combination treatment with STAT3 and GR inhibitors could provide synergistic therapeutic efficacy.
Collapse
Affiliation(s)
- Megan E Conway
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Joy M McDaniel
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama
| | - James M Graham
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Katrin P Guillen
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Patsy G Oliver
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Peibin Yue
- Department of Medicine and Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - James Turkson
- Department of Medicine and Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Donald J Buchsbaum
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bryan E Welm
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama
| | - Katherine E Varley
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
| |
Collapse
|
10
|
High-Dose Dexamethasone Manipulates the Tumor Microenvironment and Internal Metabolic Pathways in Anti-Tumor Progression. Int J Mol Sci 2020; 21:ijms21051846. [PMID: 32156004 PMCID: PMC7084511 DOI: 10.3390/ijms21051846] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/13/2022] Open
Abstract
High-dose dexamethasone (DEX) is used to treat chemotherapy-induced nausea and vomiting or to control immunotherapy-related autoimmune diseases in clinical practice. However, the underlying mechanisms of high-dose DEX in tumor progression remain unaddressed. Therefore, we explored the effects of high-dose DEX on tumor progression and the potential mechanisms of its anti-tumor function using immunohistochemistry, histological examination, real-time quantitative PCR (qPCR), and Western blotting. Tumor volume, blood vessel invasion, and levels of the cell proliferation markers Ki67 and c-Myc and the anti-apoptotic marker Bcl2 decreased in response to high-dose DEX. However, the cell apoptosis marker cleaved caspase 3 increased significantly in mice treated with 50 mg/kg DEX compared with controls. Some genes associated with immune responses were significantly downregulated following treatment with 50 mg/kg DEX e.g., Cxcl9, Cxcl10, Cd3e, Gzmb, Ifng, Foxp3, S100a9, Arg1, and Mrc1. In contrast, the M1-like tumor-associated macrophages (TAMs) activation marker Nos2 was shown to be increased. Moreover, the expression of peroxisome proliferator-activated receptors α and γ (Pparα and Pparg, respectively) was shown to be significantly upregulated in livers or tumors treated with DEX. However, high-dose DEX treatment decreased the expression of glucose and lipid metabolic pathway-related genes such as glycolysis-associated genes (Glut1, Hk2, Pgk1, Idh3a), triglyceride (TG) synthesis genes (Gpam, Agpat2, Dgat1), exogenous free fatty acid (FFA) uptake-related genes (Fabp1, Slc27a4, and CD36), and fatty acid oxidation (FAO) genes (Acadm, Acaa1, Cpt1a, Pnpla2). In addition, increased serum glucose and decreased serum TG and non-esterified fatty acid (NEFA) were observed in DEX treated-xenografted tumor mice. These findings indicate that high-dose DEX-inhibited tumor progression is a complicated process, not only activated by M1-like TAMs, but also decreased by the uptake and consumption of glucose and lipids that block the raw material and energy supply of cancer cells. Activated M1-like TAMs and inefficient glucose and lipid metabolism delayed tumor cell growth and promoted apoptosis. These findings have important implications for the application of DEX combined with drugs that target key metabolism pathways for tumor therapy in clinical practice.
Collapse
|
11
|
Aston WJ, Hope DE, Cook AM, Boon L, Dick I, Nowak AK, Lake RA, Lesterhuis WJ. Dexamethasone differentially depletes tumour and peripheral blood lymphocytes and can impact the efficacy of chemotherapy/checkpoint blockade combination treatment. Oncoimmunology 2019; 8:e1641390. [PMID: 31646089 PMCID: PMC6791454 DOI: 10.1080/2162402x.2019.1641390] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 07/03/2019] [Accepted: 07/04/2019] [Indexed: 10/26/2022] Open
Abstract
Dexamethasone is a synthetic glucocorticoid commonly used for the prevention and management of side effects in cancer patients undergoing chemotherapy. While it is effective as an anti-emetic and in preventing hypersensitivity reactions, dexamethasone depletes peripheral blood lymphocytes and impacts immune responses. The effect of dexamethasone on the number and quality of tumour-infiltrating leukocytes has not been reported. To address this, we calibrated the dose in two different strains of mice to achieve the same extent of peripheral blood lymphocyte depletion observed in patients with cancer. Doses that caused analogous depletion of T and B lymphocytes and NK cells from the peripheral blood, elicited no change in these populations within the tumour. The expression of immune checkpoint molecules PD-1, OX40, GITR and TIM3 on tumour-infiltrating lymphocytes was not altered. We found that dexamethasone had a small but significant deleterious impact on weakly efficacious chemoimmunotherapy but had no effect when the protocol was highly efficacious. Based on these results, we predict that dexamethasone will have a modest negative influence on the overall effectiveness of chemoimmunotherapy treatment.
Collapse
Affiliation(s)
- Wayne J. Aston
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- Medical School, The University of Western Australia, Nedlands, WA, Australia
| | - Danika E. Hope
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Alistair M. Cook
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- Medical School, The University of Western Australia, Nedlands, WA, Australia
| | | | - Ian Dick
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Anna K. Nowak
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- Medical School, The University of Western Australia, Nedlands, WA, Australia
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Richard A. Lake
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - W. Joost Lesterhuis
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
12
|
Mitre-Aguilar IB, Barrios-Garcia T, Ruiz-Lopez VM, Cabrera-Quintero AJ, Mejia-Dominguez NR, Ventura-Gallegos JL, Moreno-Mitre D, Aranda-Gutierrez A, Mejia-Rangel J, Escalona-Guzman AR, Chavarri-Guerra Y, Leon-Del-Rio A, Zentella-Dehesa A. Glucocorticoid-dependent expression of IAP participates in the protection against TNF-mediated cytotoxicity in MCF7 cells. BMC Cancer 2019; 19:356. [PMID: 30987626 PMCID: PMC6466787 DOI: 10.1186/s12885-019-5563-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 03/31/2019] [Indexed: 12/11/2022] Open
Abstract
Background Glucocorticoid receptor (GR) activation has been associated with breast cancer cell survival in vitro. Glucocorticoid (GC)-dependent protection against tumor necrosis factor (TNF)-induced cell death has been well characterized in MCF7 luminal A breast cancer cells. The GR activates a variety of protective mechanisms, such as inhibitors of apoptosis proteins (IAPs). However, the relative contribution of the GR-dependent expression of IAPs in the protection of cell death has not, to our knowledge, been evaluated. Methods MCF7 cells were used for all experiments. GR was activated with cortisol (CORT) or dexamethasone (DEX) and inhibited with mifepristone (RU486). Cell viability was determined in real-time with the xCELLigence™ RTCA System and at specific endpoints using crystal violet stain. The mRNA levels of the eight members of the IAP family were measured by qRT-PCR. The protein levels of GR, PR, ERα, HER2, PARP1, c-IAP1 and XIAP were evaluated by Western blot analysis. The knockdown of c-IAP1 and XIAP was accomplished via transient transfection with specific siRNAs. GR activation was verified by a gene reporter assay. Via the cBioportal interphase we queried the mRNA levels of GR and IAPs in breast cancer tumors. Results RU486 significantly inhibited the anti-cytotoxic effect of both GCs. PARP1 processing was diminished in the presence of both GCs. The combined treatments of GCs + TNF increased the relative mRNA levels of Survivin>c-IAP1 > NAIP>Apollon>XIAP>Ts-IAP > ML-IAP > c-IAP2. Additionally, GR mRNA content increased with the combined treatments of GCs + TNF. Sustained levels of the proteins c-IAP1 and XIAP were observed after 48 h of the combined treatments with GCs + TNF. With c-IAP1 and XIAP gene silencing, the GC-mediated protection was diminished. In the breast tumor samples, the GR mRNA was coexpressed with Apollon and XIAP with a Pearson coefficient greater than 0.3. Conclusions The effect of GCs against TNF-mediated cytotoxicity involves increased mRNA expression and sustained protein levels of c-IAP1 and XIAP. The antagonist effects of RU486 and the qRT-PCR results also suggest the role of the GR in this process. This finding may have clinical implications because the GR and IAPs are expressed in breast tumor samples. Electronic supplementary material The online version of this article (10.1186/s12885-019-5563-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Irma B Mitre-Aguilar
- Departamento de Medicina Genomica y Toxicologia Ambiental, Instituto de Investigaciones Biomedicas (IIBO), Universidad Nacional Autonoma de Mexico (UNAM), 04510 Ciudad de Mexico (CDMX), Mexico, Mexico.,Unidad de Bioquimica, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran (INCMNSZ), 14080, Mexico, CDMX, Mexico
| | - Tonatiuh Barrios-Garcia
- Programa de Investigacion en Cancer de Mama, IIBO, UNAM, 04510, Mexico, CDMX, Mexico.,Departamento de Biologia Molecular y Biotecnologia, IIBO, UNAM, 04510, Mexico, CDMX, Mexico
| | - Victor M Ruiz-Lopez
- Departamento de Biologia Molecular, Instituto Nacional de Enfermedades Respiratorias (INER), 14080, Mexico, CDMX, Mexico
| | - Alberto J Cabrera-Quintero
- Departamento de Medicina Genomica y Toxicologia Ambiental, Instituto de Investigaciones Biomedicas (IIBO), Universidad Nacional Autonoma de Mexico (UNAM), 04510 Ciudad de Mexico (CDMX), Mexico, Mexico.,Unidad de Bioquimica, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran (INCMNSZ), 14080, Mexico, CDMX, Mexico
| | - Nancy R Mejia-Dominguez
- Red de Apoyo a la Investigacion-Coordinacion de la Investigacion Cientifica (RAI-CIC), UNAM, 14080, Mexico, CDMX, Mexico
| | - Jose L Ventura-Gallegos
- Departamento de Medicina Genomica y Toxicologia Ambiental, Instituto de Investigaciones Biomedicas (IIBO), Universidad Nacional Autonoma de Mexico (UNAM), 04510 Ciudad de Mexico (CDMX), Mexico, Mexico.,Unidad de Bioquimica, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran (INCMNSZ), 14080, Mexico, CDMX, Mexico
| | - Daniel Moreno-Mitre
- Unidad de Bioquimica, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran (INCMNSZ), 14080, Mexico, CDMX, Mexico
| | - Alejandro Aranda-Gutierrez
- Unidad de Bioquimica, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran (INCMNSZ), 14080, Mexico, CDMX, Mexico
| | - Janini Mejia-Rangel
- Departamento de Medicina Genomica y Toxicologia Ambiental, Instituto de Investigaciones Biomedicas (IIBO), Universidad Nacional Autonoma de Mexico (UNAM), 04510 Ciudad de Mexico (CDMX), Mexico, Mexico.,Unidad de Bioquimica, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran (INCMNSZ), 14080, Mexico, CDMX, Mexico
| | - Alma R Escalona-Guzman
- Departamento de Medicina Genomica y Toxicologia Ambiental, Instituto de Investigaciones Biomedicas (IIBO), Universidad Nacional Autonoma de Mexico (UNAM), 04510 Ciudad de Mexico (CDMX), Mexico, Mexico.,Unidad de Bioquimica, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran (INCMNSZ), 14080, Mexico, CDMX, Mexico
| | - Yanin Chavarri-Guerra
- Departamento de Hemato-Oncologia, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, 14080, Mexico, CDMX, Mexico
| | - Alfonso Leon-Del-Rio
- Programa de Investigacion en Cancer de Mama, IIBO, UNAM, 04510, Mexico, CDMX, Mexico.,Departamento de Biologia Molecular y Biotecnologia, IIBO, UNAM, 04510, Mexico, CDMX, Mexico
| | - Alejandro Zentella-Dehesa
- Departamento de Medicina Genomica y Toxicologia Ambiental, Instituto de Investigaciones Biomedicas (IIBO), Universidad Nacional Autonoma de Mexico (UNAM), 04510 Ciudad de Mexico (CDMX), Mexico, Mexico. .,Programa de Investigacion en Cancer de Mama, IIBO, UNAM, 04510, Mexico, CDMX, Mexico. .,Unidad de Bioquimica, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran (INCMNSZ), 14080, Mexico, CDMX, Mexico. .,Centro de Cancer, Centro Medico ABC, 01120, Mexico, CDMX, Mexico.
| |
Collapse
|
13
|
Silva CO, Pinho JO, Lopes JM, Almeida AJ, Gaspar MM, Reis C. Current Trends in Cancer Nanotheranostics: Metallic, Polymeric, and Lipid-Based Systems. Pharmaceutics 2019; 11:E22. [PMID: 30625999 PMCID: PMC6359642 DOI: 10.3390/pharmaceutics11010022] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 12/28/2018] [Accepted: 01/01/2019] [Indexed: 02/07/2023] Open
Abstract
Theranostics has emerged in recent years to provide an efficient and safer alternative in cancer management. This review presents an updated description of nanotheranostic formulations under development for skin cancer (including melanoma), head and neck, thyroid, breast, gynecologic, prostate, and colon cancers, brain-related cancer, and hepatocellular carcinoma. With this focus, we appraised the clinical advantages and drawbacks of metallic, polymeric, and lipid-based nanosystems, such as low invasiveness, low toxicity to the surrounding healthy tissues, high precision, deeper tissue penetration, and dosage adjustment in a real-time setting. Particularly recognizing the increased complexity and multimodality in this area, multifunctional hybrid nanoparticles, comprising different nanomaterials and functionalized with targeting moieties and/or anticancer drugs, present the best characteristics for theranostics. Several examples, focusing on their design, composition, imaging and treatment modalities, and in vitro and in vivo characterization, are detailed herein. Briefly, all studies followed a common trend in the design of these theranostics modalities, such as the use of materials and/or drugs that share both inherent imaging (e.g., contrast agents) and therapeutic properties (e.g., heating or production reactive oxygen species). This rationale allows one to apparently overcome the heterogeneity, complexity, and harsh conditions of tumor microenvironments, leading to the development of successful targeted therapies.
Collapse
Affiliation(s)
- Catarina Oliveira Silva
- iMedUlisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Jacinta Oliveira Pinho
- iMedUlisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Joana Margarida Lopes
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - António J Almeida
- iMedUlisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Maria Manuela Gaspar
- iMedUlisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Catarina Reis
- iMedUlisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
- IBEB, Faculty of Sciences, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
| |
Collapse
|