1
|
Caragine CM, Le VT, Mustafa M, Diaz BJ, Morris JA, Müller S, Mendez-Mancilla A, Geller E, Liscovitch-Brauer N, Sanjana NE. Comprehensive dissection of cis-regulatory elements in a 2.8 Mb topologically associated domain in six human cancers. Nat Commun 2025; 16:1611. [PMID: 39948336 PMCID: PMC11825950 DOI: 10.1038/s41467-025-56568-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Cis-regulatory elements (CREs), such as enhancers and promoters, are fundamental regulators of gene expression and, across different cell types, the MYC locus utilizes a diverse regulatory architecture driven by multiple CREs. To better understand differences in CRE function, we perform pooled CRISPR inhibition (CRISPRi) screens to comprehensively probe the 2.8 Mb topologically-associated domain containing MYC in 6 human cancer cell lines with nucleotide resolution. We map 32 CREs where inhibition leads to changes in cell growth, including 8 that overlap previously identified enhancers. Targeting specific CREs decreases MYC expression by as much as 60%, and cell growth by as much as 50%. Using 3-D enhancer contact mapping, we find that these CREs almost always contact MYC but less than 10% of total MYC contacts impact growth when silenced, highlighting the utility of our approach to identify phenotypically-relevant CREs. We also detect an enrichment of lineage-specific transcription factors (TFs) at MYC CREs and, for some of these TFs, find a strong, tumor-specific correlation between TF and MYC expression not found in normal tissue. Taken together, these CREs represent systematically identified, functional regulatory regions and demonstrate how the same region of the human genome can give rise to complex, tissue-specific gene regulation.
Collapse
Affiliation(s)
- Christina M Caragine
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Victoria T Le
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Meer Mustafa
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Bianca Jay Diaz
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - John A Morris
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Simon Müller
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Alejandro Mendez-Mancilla
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Evan Geller
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Noa Liscovitch-Brauer
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Neville E Sanjana
- New York Genome Center, New York, NY, USA.
- Department of Biology, New York University, New York, NY, USA.
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA.
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
2
|
Rossi T, Iorio E, Chirico M, Pisanu ME, Amodio N, Cantafio MEG, Perrotta I, Colciaghi F, Fiorillo M, Gianferrari A, Puccio N, Neri A, Ciarrocchi A, Pistoni M. BET inhibitors (BETi) influence oxidative phosphorylation metabolism by affecting mitochondrial dynamics leading to alterations in apoptotic pathways in triple-negative breast cancer (TNBC) cells. Cell Prolif 2024; 57:e13730. [PMID: 39223828 PMCID: PMC11628750 DOI: 10.1111/cpr.13730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 07/04/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Repressing BET proteins' function using bromodomain inhibitors (BETi) has been shown to elicit antitumor effects by regulating the transcription of genes downstream of BRD4. We previously showed that BETi promoted cell death of triple-negative breast cancer (TNBC) cells. Here, we proved that BETi induce altered mitochondrial dynamics fitness in TNBC cells falling in cell death. We demonstrated that BETi treatment downregulated the expression of BCL-2, and proteins involved in mitochondrial fission and increased fused mitochondria. Impaired mitochondrial fission affected oxidative phosphorylation (OXPHOS) inducing the expression of OXPHOS-related genes, SDHa and ATP5a, and increased cell death. Consistently, the amount of mitochondrial DNA and mitochondrial membrane potential (∆Ψm) increased in BETi-treated cells compared to control cells. Lastly, BETi in combination with Metformin reduced cell growth. Our results indicate that mitochondrial dynamics and OXPHOS metabolism support breast cancer proliferation and represent novel BETi downstream targets in TNBC cells.
Collapse
Affiliation(s)
- Teresa Rossi
- Laboratory of Translational ResearchAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| | - Egidio Iorio
- High Resolution NMR UnitCore Facilities, Istituto Superiore di SanitàRomeItaly
| | - Mattea Chirico
- High Resolution NMR UnitCore Facilities, Istituto Superiore di SanitàRomeItaly
| | - Maria Elena Pisanu
- High Resolution NMR UnitCore Facilities, Istituto Superiore di SanitàRomeItaly
| | - Nicola Amodio
- Department of Experimental and Clinical MedicineUniversity Magna Graecia of CatanzaroCatanzaroItaly
| | | | - Ida Perrotta
- Department of Biology, Ecology and Earth SciencesCentre for Microscopy and Microanalysis (CM2), University of CalabriaCosenzaItaly
| | | | - Marco Fiorillo
- Department of Pharmacy, Health and Nutritional SciencesUniversity of CalabriaRendeItaly
| | - Alessia Gianferrari
- Laboratory of Translational ResearchAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| | - Noemi Puccio
- Laboratory of Translational ResearchAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| | - Antonino Neri
- Scientific DirectorateAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| | - Alessia Ciarrocchi
- Laboratory of Translational ResearchAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| | - Mariaelena Pistoni
- Laboratory of Translational ResearchAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| |
Collapse
|
3
|
Kwantwi LB, Tandoh T. Focal adhesion kinase-mediated interaction between tumor and immune cells in the tumor microenvironment: implications for cancer-associated therapies and tumor progression. Clin Transl Oncol 2024:10.1007/s12094-024-03723-x. [PMID: 39269597 DOI: 10.1007/s12094-024-03723-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024]
Abstract
Focal adhesion kinase (FAK) expression has been linked to tumor growth, immunosuppression, metastasis, angiogenesis, and therapeutic resistance through kinase-dependent and kinase scaffolding functions in the nucleus and cytoplasm. Hence, targeting FAK alone or with other agents has gained attention as a potential therapeutic strategy. Moreover, mounting evidence shows that FAK activity can influence the tumor immune microenvironment crosstalk to support tumor progression. Recently, tumor immune microenvironment interaction orchestrators have shown to be promising therapeutic agents for cancer immunotherapies. Therefore, this review highlights how FAK regulates the tumor immune microenvironment interplay to promote tumor immune evasive mechanisms and their potential for combination therapies with standard cancer treatments.
Collapse
Affiliation(s)
- Louis Boafo Kwantwi
- Department of Anatomy and Neurobiology, College of Medicine, Northeast Ohio Medical University, Rootstown, OH, 44272, USA.
| | - Theophilus Tandoh
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA, USA
| |
Collapse
|
4
|
González-Rodriguez MA, Troutman S, Bayle S, Lester DK, Grove M, Duckett D, Kareta MS, Kissil JL. Synergistic effects of combined BET and FAK inhibition against Vestibular Schwannomas in NF2-related Schwannomatosis. Oncogene 2024; 43:2995-3002. [PMID: 39209965 DOI: 10.1038/s41388-024-03144-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/14/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Neurofibromatosis type 2 (NF2) is a rare disorder that causes vestibular schwannomas (VS), meningiomas and ependymomas. To date, there is no FDA approved drug-based treatment for NF2. We have previously identified that BET inhibition can selectively reduce growth of the NF2-null schwannoma and Schwann cells in vitro and tumorigenesis in vivo and, separately, reported that inhibition of Focal Adhesion Kinase 1 (FAK1) via crizotinib has antiproliferative effects in NF2-null Schwann cells. The current study was aimed at determining whether combined BET and FAK inhibition can synergize and to identify the mechanisms of action. A panel of normal and NF2-null Schwann and schwannoma cell lines were used to characterize the effects of combined BET and FAK inhibition in vitro and in vivo using pharmacological and genetic approaches. The mechanism of action was explored by chromatin immunoprecipitation, ChIP-PCR, western blotting, and functional approaches. We find that combined BET and FAK inhibition are synergistic and inhibit the proliferation of NF2-null schwannoma and Schwann cell lines in vitro and in vivo, by arresting cells in the G1/S and G2/M phases of the cell cycle. Further, we identify the mechanism of action through the downregulation of FAK1 transcription by BET inhibition, which potentiates inhibition of FAK by 100-fold. Our findings suggest that combined targeting of BET and FAK1 may offer a potential therapeutic option for the treatment of NF2-related schwannomas.
Collapse
Affiliation(s)
| | - Scott Troutman
- Department of Molecular Oncology and H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Simon Bayle
- Drug Discovery, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Daniel K Lester
- Department of Molecular Oncology and H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Matthew Grove
- Department of Molecular Oncology and H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Derek Duckett
- Drug Discovery, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Michael S Kareta
- Genetics and Genomics Group, Sanford Research, Sioux Falls, SD, USA
| | - Joseph L Kissil
- Department of Molecular Oncology and H. Lee Moffitt Cancer Center, Tampa, FL, USA.
| |
Collapse
|
5
|
Kong Y, Lan T, Wang L, Gong C, Lv W, Zhang H, Zhou C, Sun X, Liu W, Huang H, Weng X, Cai C, Peng W, Zhang M, Jiang D, Yang C, Liu X, Rao Y, Chen C. BRD4-specific PROTAC inhibits basal-like breast cancer partially through downregulating KLF5 expression. Oncogene 2024; 43:2914-2926. [PMID: 39164524 PMCID: PMC11420083 DOI: 10.1038/s41388-024-03121-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 07/24/2024] [Accepted: 07/31/2024] [Indexed: 08/22/2024]
Abstract
Interest in the use of proteolysis-targeting chimeras (PROTACs) in cancer therapy has increased in recent years. Targeting bromodomain and extra terminal domain (BET) proteins, especially bromodomain-containing protein 4 (BRD4), has shown inhibitory effects on basal-like breast cancer (BLBC). However, the bioavailability of BRD4 PROTACs is restricted by their non-selective biodegradability and low tumor-targeting ability. We demonstrated that 6b (BRD4 PROTAC) suppresses BLBC cell growth by targeting BRD4, but not BRD2 and BRD3, for cereblon (CRBN)-mediated ubiquitination and proteasomal degradation. Compound 6b also inhibited expression of Krüppel-like factor 5 (KLF5) transcription factor, a key oncoprotein in BLBC, controlled by BRD4-mediated super-enhancers. Moreover, 6b inhibited HCC1806 tumor growth in a xenograft mouse model. The combination of 6b and KLF5 inhibitors showed additive effects on BLBC. These results suggest that BRD4-specific PROTAC can effectively inhibit BLBC by downregulating KLF5, and that 6b has potential as a novel therapeutic drug for BLBC.
Collapse
Grants
- This study was supported in part by grants National Key Research and Development Program of China (2020YFA0112300 to Chen, C. 2020YFE0202200, 2021YFA1300200 to Rao, Y.), The National Nature Science Foundation of China (U2102203 and 81830087 to Chen, C., 81402206, 82273216 and 81773149 to Kong, Y., 82125034 to Rao, Y.), Biomedical Projects of Yunnan Key Science and Technology Program (202302AA310046),Yunnan Fundamental Research Projects (202101AS070050), and Yunnan Revitalization Talent Support Program (Yunling Shcolar Project to CC). Yunnan (Kunming) Academician Expert Workstation (grant No. YSZJGZZ-2020025 to CC). Shenzhen Science and Technology program (RCYX20221008092911040 to Kong, Y.), Shenzhen Municipal Government of China (JCYJ20210324103603011 to Kong, Y.), Shenzhen Planned Projects for Post-doctors Stand out Research Funds to Kong, Y., Guangdong Basic and Applied Basic Research Foundation (2022A1515220051 to Kong, Y.).
Collapse
Affiliation(s)
- Yanjie Kong
- Pathology Department, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Tianlong Lan
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, China
| | - Luzhen Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Chen Gong
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wenxin Lv
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, China
| | - Hailin Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Chengang Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Xiuyun Sun
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, China
| | - Wenjing Liu
- The Third Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Haihui Huang
- Pathology Department, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Xin Weng
- Pathology Department, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Chang Cai
- Pathology Department, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Wenfeng Peng
- Pathology Department, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Meng Zhang
- Pathology Department, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Dewei Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Chuanyu Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Xia Liu
- Pathology Department, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China.
| | - Yu Rao
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, China.
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
- The Third Affiliated Hospital, Kunming Medical University, Kunming, China.
- Academy of Biomedical Engineering, Kunming Medical University, Kunming, China.
| |
Collapse
|
6
|
Vlahopoulos SA. Divergent Processing of Cell Stress Signals as the Basis of Cancer Progression: Licensing NFκB on Chromatin. Int J Mol Sci 2024; 25:8621. [PMID: 39201306 PMCID: PMC11354898 DOI: 10.3390/ijms25168621] [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: 07/10/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Inflammation is activated by diverse triggers that induce the expression of cytokines and adhesion molecules, which permit a succession of molecules and cells to deliver stimuli and functions that help the immune system clear the primary cause of tissue damage, whether this is an infection, a tumor, or a trauma. During inflammation, short-term changes in the expression and secretion of strong mediators of inflammation occur, while long-term changes occur to specific groups of cells. Long-term changes include cellular transdifferentiation for some types of cells that need to regenerate damaged tissue, as well as death for specific immune cells that can be detrimental to tissue integrity if they remain active beyond the boundaries of essential function. The transcriptional regulator NFκB enables some of the fundamental gene expression changes during inflammation, as well as during tissue development. During recurrence of malignant disease, cell stress-induced alterations enable the growth of cancer cell clones that are substantially resistant to therapeutic intervention and to the immune system. A number of those alterations occur due to significant defects in feedback signal cascades that control the activity of NFκB. Specifically, cell stress contributes to feedback defects as it overrides modules that otherwise control inflammation to protect host tissue. NFκB is involved in both the suppression and promotion of cancer, and the key distinctive feature that determines its net effect remains unclear. This paper aims to provide a clear answer to at least one aspect of this question, namely the mechanism that enables a divergent response of cancer cells to critical inflammatory stimuli and to cell stress in general.
Collapse
|
7
|
Das D, Leung JY, Balamurugan S, Tergaonkar V, Loh AHP, Chiang CM, Taneja R. BRD4 isoforms have distinct roles in tumour progression and metastasis in rhabdomyosarcoma. EMBO Rep 2024; 25:832-852. [PMID: 38191874 PMCID: PMC10897194 DOI: 10.1038/s44319-023-00033-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 01/10/2024] Open
Abstract
BRD4, a bromodomain and extraterminal (BET) protein, is deregulated in multiple cancers and has emerged as a promising drug target. However, the function of the two main BRD4 isoforms (BRD4-L and BRD4-S) has not been analysed in parallel in most cancers. This complicates determining therapeutic efficacy of pan-BET inhibitors. In this study, using functional and transcriptomic analysis, we show that BRD-L and BRD4-S isoforms play distinct roles in fusion negative embryonal rhabdomyosarcoma. BRD4-L has an oncogenic role and inhibits myogenic differentiation, at least in part, by activating myostatin expression. Depletion of BRD4-L in vivo impairs tumour progression but does not impact metastasis. On the other hand, depletion of BRD4-S has no significant impact on tumour growth, but strikingly promotes metastasis in vivo. Interestingly, BRD4-S loss results in the enrichment of BRD4-L and RNA Polymerase II at integrin gene promoters resulting in their activation. In fusion positive alveolar rhabdomyosarcoma, BRD4-L is unrestricted in its oncogenic role, with no evident involvement of BRD4-S. Our work unveils isoform-specific functions of BRD4 in rhabdomyosarcoma.
Collapse
Affiliation(s)
- Dipanwita Das
- Department of Physiology, Healthy Longevity and NUS Center for Cancer Research Translation Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore
| | - Jia Yu Leung
- Department of Physiology, Healthy Longevity and NUS Center for Cancer Research Translation Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
| | - Shivaranjani Balamurugan
- Department of Physiology, Healthy Longevity and NUS Center for Cancer Research Translation Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore
| | - Vinay Tergaonkar
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Amos Hong Pheng Loh
- VIVA-KKH Paediatric Brain and Solid Tumour Programme, KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Cheng-Ming Chiang
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Reshma Taneja
- Department of Physiology, Healthy Longevity and NUS Center for Cancer Research Translation Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore.
| |
Collapse
|
8
|
Pratelli G, Carlisi D, Di Liberto D, Notaro A, Giuliano M, D'Anneo A, Lauricella M, Emanuele S, Calvaruso G, De Blasio A. MCL1 Inhibition Overcomes the Aggressiveness Features of Triple-Negative Breast Cancer MDA-MB-231 Cells. Int J Mol Sci 2023; 24:11149. [PMID: 37446326 DOI: 10.3390/ijms241311149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/30/2023] [Accepted: 07/02/2023] [Indexed: 07/15/2023] Open
Abstract
Triple-Negative Breast Cancer (TNBC) is a particularly aggressive subtype among breast cancers (BCs), characterized by anoikis resistance, high invasiveness, and metastatic potential as well as Epithelial-Mesenchymal Transition (EMT) and stemness features. In the last few years, our research focused on the function of MCL1, an antiapoptotic protein frequently deregulated in TNBC. Here, we demonstrate that MCL1 inhibition by A-1210477, a specific BH3-mimetic, promotes anoikis/apoptosis in the MDA-MB-231 cell line, as shown via an increase in proapoptotic markers and caspase activation. Our evidence also shows A-1210477 effects on Focal Adhesions (FAs) impairing the integrin trim and survival signaling pathways, such as FAK, AKT, ERK, NF-κB, and GSK3β-inducing anoikis, thus suggesting a putative role of MCL1 in regulation of FA dynamics. Interestingly, in accordance with these results, we observed a reduction in migratory and invasiveness capabilities as confirmed by a decrease in metalloproteinases (MMPs) levels following A-1210477 treatment. Moreover, MCL1 inhibition promotes a reduction in EMT characteristics as demonstrated by the downregulation of Vimentin, MUC1, DNMT1, and a surprising re-expression of E-Cadherin, suggesting a possible mesenchymal-like phenotype reversion. In addition, we also observed the downregulation of stemness makers such as OCT3/4, SOX2, NANOG, as well as CD133, EpCAM, and CD49f. Our findings support the idea that MCL1 inhibition in MDA-MB-231 could be crucial to reduce anoikis resistance, aggressiveness, and metastatic potential and to minimize EMT and stemness features that distinguish TNBC.
Collapse
Affiliation(s)
- Giovanni Pratelli
- Department of Physics and Chemistry (DiFC)-Emilio Segrè, University of Palermo, 90128 Palermo, Italy
| | - Daniela Carlisi
- Section of Biochemistry, Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy
| | - Diana Di Liberto
- Section of Biochemistry, Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy
| | - Antonietta Notaro
- Laboratory of Biochemistry, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90127 Palermo, Italy
| | - Michela Giuliano
- Laboratory of Biochemistry, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90127 Palermo, Italy
| | - Antonella D'Anneo
- Laboratory of Biochemistry, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90127 Palermo, Italy
| | - Marianna Lauricella
- Section of Biochemistry, Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy
| | - Sonia Emanuele
- Section of Biochemistry, Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy
| | - Giuseppe Calvaruso
- Laboratory of Biochemistry, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90127 Palermo, Italy
| | - Anna De Blasio
- Laboratory of Biochemistry, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
9
|
Li L, Gao L, Zhou H, Shi C, Zhang X, Zhang D, Liu H. High Expression Level of BRD4 Is Associated with a Poor Prognosis and Immune Infiltration in Esophageal Squamous Cell Carcinoma. Dig Dis Sci 2023:10.1007/s10620-023-07907-3. [PMID: 36933111 DOI: 10.1007/s10620-023-07907-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/28/2023] [Indexed: 03/19/2023]
Abstract
BACKGROUND Bromodomain-containing protein 4 (BRD4) is a reader of histone acetylation and is associated with a variety of diseases. AIM To investigate the expression level of BRD4 in esophageal squamous cell carcinoma (ESCC), its prognostic value and its relationship with immune infiltration. METHODS The study included 94 ESCC patients from The Cancer Genome Atlas (TCGA) database and 179 ESCC patients from Affiliated Hospital 2 of Nantong University. The expression levels of proteins in tissue microarray were detected by immunohistochemistry. The prognostic factors were analyzed by Kaplan-Meier curve and univariate and multivariate cox regression. The ESTIMATE website was used to calculate the stromal, immune and ESTIMATE score. CIBERSORT was used to calculate the abundance of immune infiltrates. Spearman and Phi coefficient were used for correlation analysis. The TIDE algorithm was used to predict treatment response to immune checkpoint blockade. RESULTS BRD4 is up-regulated in ESCC, and high BRD4 expression level is associated with poor prognosis and adverse clinicopathological features. In addition, the monocyte count, systemic inflammatory-immunologic index, platelet-lymphocyte ratio, and monocyte-lymphocyte ratio in the BRD4 high expression level group were higher than in the low expression level group. Finally, we found that BRD4 expression level correlated with immune infiltration and that it was inversely correlated with infiltration of CD8 + T cells. Higher TIDE scores in the BRD4 high expression group than in the low expression group. CONCLUSION BRD4 is associated with poor prognosis and immune infiltration in ESCC, and may be a potential biomarker for prognosis and immunotherapy application.
Collapse
Affiliation(s)
- Li Li
- Department of Pathology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Shengli Road No. 666, Nantong, 226001, Jiangsu, People's Republic of China
| | - Lin Gao
- Medical Research Center, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, People's Republic of China
| | - Hong Zhou
- Department of Pathology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Shengli Road No. 666, Nantong, 226001, Jiangsu, People's Republic of China
| | - Chao Shi
- Department of Pathology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Shengli Road No. 666, Nantong, 226001, Jiangsu, People's Republic of China
| | - Xiaojuan Zhang
- Department of Pathology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Shengli Road No. 666, Nantong, 226001, Jiangsu, People's Republic of China
| | - Dongmei Zhang
- Medical Research Center, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, People's Republic of China
| | - Hongbin Liu
- Department of Pathology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Shengli Road No. 666, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
10
|
Lei JH, Zhang L, Wang Z, Peltier R, Xie Y, Chen G, Lin S, Miao K, Deng CX, Sun H. FGFR2-BRD4 Axis Regulates Transcriptional Networks of Histone 3 Modification and Synergy Between Its Inhibitors and PD-1/PD-L1 in a TNBC Mouse Model. Front Immunol 2022; 13:861221. [PMID: 35547739 PMCID: PMC9084888 DOI: 10.3389/fimmu.2022.861221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Epigenetic reprogramming is an independent mode of gene expression that often involves changes in the transcription and chromatin structure due to tumor initiation and development. In this study, we developed a specifically modified peptide array and searched for a recognized epigenetic reader. Our results demonstrated that BRD4 is not only an acetylation reader but of propionylation as well. We also studied the quantitative binding affinities between modified peptides and epigenetic regulators by isothermal titration calorimetry (ITC). Furthermore, we introduced the Fgfr2-S252W transgenic mouse model to confirm that this acetylation is associated with the activation of c-Myc and drives tumor formation. Targeted disruption of BRD4 in Fgfr2-S252W mouse tumor cells also confirmed that BRD4 is a key regulator of histone 3 acetylation. Finally, we developed a tumor slice culture system and demonstrated the synergy between immune checkpoint blockade and targeted therapy in triple-negative breast cancer (TNBC). These data extend our understanding of epigenetic reprogramming and epigenetics-based therapies.
Collapse
Affiliation(s)
- Josh Haipeng Lei
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau, Macau SAR, China.,Ministry of Education (MOE) Frontier Science Centre for Precision Oncology, University of Macau, Taipa, Macau SAR, China.,Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Lei Zhang
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau, Macau SAR, China.,Department of Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zhenyi Wang
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Heifei, China
| | - Raoul Peltier
- Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Yusheng Xie
- Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong SAR, China.,Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Ganchao Chen
- Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Shiqi Lin
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau, Macau SAR, China.,Ministry of Education (MOE) Frontier Science Centre for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Kai Miao
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau, Macau SAR, China.,Ministry of Education (MOE) Frontier Science Centre for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Chu-Xia Deng
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau, Macau SAR, China.,Ministry of Education (MOE) Frontier Science Centre for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Hongyan Sun
- Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong SAR, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, City University of Hong Kong, Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
11
|
Liao M, Qin R, Huang W, Zhu HP, Peng F, Han B, Liu B. Targeting regulated cell death (RCD) with small-molecule compounds in triple-negative breast cancer: a revisited perspective from molecular mechanisms to targeted therapies. J Hematol Oncol 2022; 15:44. [PMID: 35414025 PMCID: PMC9006445 DOI: 10.1186/s13045-022-01260-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/28/2022] [Indexed: 02/08/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of human breast cancer with one of the worst prognoses, with no targeted therapeutic strategies currently available. Regulated cell death (RCD), also known as programmed cell death (PCD), has been widely reported to have numerous links to the progression and therapy of many types of human cancer. Of note, RCD can be divided into numerous different subroutines, including autophagy-dependent cell death, apoptosis, mitotic catastrophe, necroptosis, ferroptosis, pyroptosis and anoikis. More recently, targeting the subroutines of RCD with small-molecule compounds has been emerging as a promising therapeutic strategy, which has rapidly progressed in the treatment of TNBC. Therefore, in this review, we focus on summarizing the molecular mechanisms of the above-mentioned seven major RCD subroutines related to TNBC and the latest progress of small-molecule compounds targeting different RCD subroutines. Moreover, we further discuss the combined strategies of one drug (e.g., narciclasine) or more drugs (e.g., torin-1 combined with chloroquine) to achieve the therapeutic potential on TNBC by regulating RCD subroutines. More importantly, we demonstrate several small-molecule compounds (e.g., ONC201 and NCT03733119) by targeting the subroutines of RCD in TNBC clinical trials. Taken together, these findings will provide a clue on illuminating more actionable low-hanging-fruit druggable targets and candidate small-molecule drugs for potential RCD-related TNBC therapies.
Collapse
Affiliation(s)
- Minru Liao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Rui Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hong-Ping Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.,Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Fu Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
12
|
Marqués M, Sorolla MA, Urdanibia I, Parisi E, Hidalgo I, Morales S, Salud A, Sorolla A. Are Transcription Factors Plausible Oncotargets for Triple Negative Breast Cancers? Cancers (Basel) 2022; 14:cancers14051101. [PMID: 35267409 PMCID: PMC8909618 DOI: 10.3390/cancers14051101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Triple negative breast cancer is a type of breast cancer that does not have a selective and effective therapy. It is known that this cancer possesses high abundance of certain proteins called transcription factors, which are essential for their growth. However, inhibiting transcription factors is very difficult with common therapeutics due to their inaccessibility inside the cell and their molecular structure. In this work, we identified the most important transcription factors for the growth of triple negative breast cancers, and that can predict worse clinical outcome. Moreover, we described different strategies that have been utilised to inhibit them. A successful inhibition of these transcription factors could reduce the mortality and convalescence associated with triple negative breast cancers. Abstract Breast cancer (BC) is the most diagnosed cancer worldwide and one of the main causes of cancer deaths. BC is a heterogeneous disease composed of different BC intrinsic subtypes such as triple-negative BC (TNBC), which is one of the most aggressive subtypes and which lacks a targeted therapy. Recent comprehensive analyses across cell types and cancer types have outlined a vast network of protein–protein associations between transcription factors (TFs). Not surprisingly, protein–protein networks central to oncogenesis and disease progression are highly altered during TNBC pathogenesis and are responsible for the activation of oncogenic programs, such as uncontrollable proliferation, epithelial-to-mesenchymal transition (EMT) and stemness. From the therapeutic viewpoint, inhibiting the interactions between TFs represents a very significant challenge, as the contact surfaces of TFs are relatively large and featureless. However, promising tools have emerged to offer a solution to the targeting problem. At the clinical level, some TF possess diagnostic and prognostic value in TNBC. In this review, we outline the recent advances in TFs relevant to TNBC growth and progression. Moreover, we highlight different targeting approaches to inhibit these TFs. Furthermore, the validity of such TFs as clinical biomarkers has been explored. Finally, we discuss how research is likely to evolve in the field.
Collapse
Affiliation(s)
- Marta Marqués
- Research Group of Cancer Biomarkers, Lleida Institute for Biomedical Research Dr. Pifarré Foundation (IRBLleida), Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain; (M.M.); (M.A.S.); (I.U.); (E.P.); (I.H.); (S.M.); (A.S.)
- Department of Medicine, University of Lleida, Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain
| | - Maria Alba Sorolla
- Research Group of Cancer Biomarkers, Lleida Institute for Biomedical Research Dr. Pifarré Foundation (IRBLleida), Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain; (M.M.); (M.A.S.); (I.U.); (E.P.); (I.H.); (S.M.); (A.S.)
| | - Izaskun Urdanibia
- Research Group of Cancer Biomarkers, Lleida Institute for Biomedical Research Dr. Pifarré Foundation (IRBLleida), Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain; (M.M.); (M.A.S.); (I.U.); (E.P.); (I.H.); (S.M.); (A.S.)
| | - Eva Parisi
- Research Group of Cancer Biomarkers, Lleida Institute for Biomedical Research Dr. Pifarré Foundation (IRBLleida), Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain; (M.M.); (M.A.S.); (I.U.); (E.P.); (I.H.); (S.M.); (A.S.)
| | - Iván Hidalgo
- Research Group of Cancer Biomarkers, Lleida Institute for Biomedical Research Dr. Pifarré Foundation (IRBLleida), Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain; (M.M.); (M.A.S.); (I.U.); (E.P.); (I.H.); (S.M.); (A.S.)
- Department of Medicine, University of Lleida, Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain
| | - Serafín Morales
- Research Group of Cancer Biomarkers, Lleida Institute for Biomedical Research Dr. Pifarré Foundation (IRBLleida), Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain; (M.M.); (M.A.S.); (I.U.); (E.P.); (I.H.); (S.M.); (A.S.)
- Department of Medical Oncology, Arnau de Vilanova University Hospital (HUAV), Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain
| | - Antonieta Salud
- Research Group of Cancer Biomarkers, Lleida Institute for Biomedical Research Dr. Pifarré Foundation (IRBLleida), Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain; (M.M.); (M.A.S.); (I.U.); (E.P.); (I.H.); (S.M.); (A.S.)
- Department of Medicine, University of Lleida, Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain
- Department of Medical Oncology, Arnau de Vilanova University Hospital (HUAV), Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain
| | - Anabel Sorolla
- Research Group of Cancer Biomarkers, Lleida Institute for Biomedical Research Dr. Pifarré Foundation (IRBLleida), Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain; (M.M.); (M.A.S.); (I.U.); (E.P.); (I.H.); (S.M.); (A.S.)
- Correspondence:
| |
Collapse
|
13
|
Dzobo K. Integrins Within the Tumor Microenvironment: Biological Functions, Importance for Molecular Targeting, and Cancer Therapeutics Innovation. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:417-430. [PMID: 34191612 DOI: 10.1089/omi.2021.0069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Many cellular functions important for solid tumor initiation and progression are mediated by members of the integrin family, a diverse family of cell attachment receptors. With recent studies emphasizing the role of the tumor microenvironment (TME) in tumor initiation and progression, it is not surprising that considerable attention is being paid to integrins. Several integrin antagonists are under clinical trials, with many demonstrating promising activity in patients with different cancers. A deeper knowledge of the functions of integrins within the TME is still required and might lead to better inhibitors being discovered. Integrin expression is commonly dysregulated in many tumors with integrins playing key roles in signaling as well as promotion of tumor cell invasion and migration. Integrins also play a major role in adhesion of circulating tumor cells to new sites and the resulting formation of secondary tumors. Furthermore, integrins have demonstrated the ability to promoting stem cell-like properties in tumor cells as well as drug resistance. Anti-integrin therapies rely heavily on the doses or concentrations used as these determine whether the drugs act as antagonists or as integrin agonists. This expert review offers the latest synthesis in terms of the current knowledge of integrins functions within the TME and as potential molecular targets for cancer therapeutics innovation.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa.,Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
14
|
Expression of Phosphorylated BRD4 Is Markedly Associated with the Activation Status of the PP2A Pathway and Shows a Strong Prognostic Value in Triple Negative Breast Cancer Patients. Cancers (Basel) 2021; 13:cancers13061246. [PMID: 33809005 PMCID: PMC7999847 DOI: 10.3390/cancers13061246] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary The use of BRD4 inhibitors has emerged as a novel therapeutic approach in a wide variety of tumors including the triple negative breast cancer. Moreover, PP2A has been proposed as the phosphatase involved in regulating BRD4 phosphorylation and stabilization. Our aim was to evaluate for the first time the clinical impact of BRD4 phosphorylation in triple negative breast cancer patients and as well as its potential linking with the PP2A activation status in this disease. Our findings are special relevant since they suggest the prognostic value of BRD4 phosphorylation levels, and the potential clinical usefulness of PP2A inhibition markers to anticipate response to BRD4 inhibitors. Abstract The bromodomain-containing protein 4 (BRD4), a member of the bromodomain and extra-terminal domain (BET) protein family, has emerged in the last years as a promising molecular target in many tumors including breast cancer. The triple negative breast cancer (TNBC) represents the molecular subtype with the worst prognosis and a current therapeutic challenge, and TNBC cells have been reported to show a preferential sensitivity to BET inhibitors. Interestingly, BRD4 phosphorylation (pBRD4) was found as an alteration that confers resistance to BET inhibition and PP2A proposed as the phosphatase responsible to regulate pBRD4 levels. However, the potential clinical significance of pBRD4, as well as its potential correlation with the PP2A pathway in TNBC, remains to be investigated. Here, we evaluated the expression levels of pBRD4 in a series of 132 TNBC patients. We found high pBRD4 levels in 34.1% of cases (45/132), and this alteration was found to be associated with the development of patient recurrences (p = 0.007). Interestingly, BRD4 hyperphosphorylation predicted significantly shorter overall (p < 0.001) and event-free survival (p < 0.001). Moreover, multivariate analyses were performed to confirm its independent prognostic impact in our cohort. In conclusion, our findings show that BRD4 hyperphosphorylation is an alteration associated with PP2A inhibition that defines a subgroup of TNBC patients with unfavorable prognosis, suggesting the potential clinical and therapeutic usefulness of the PP2A/BRD4 axis as a novel molecular target to overcome resistance to treatments based on BRD4 inhibition.
Collapse
|