1
|
Kong Y, Jiang R, Zhou H, Ge M, Lin H, Wang Y, Yao R, Wang Q, Liang X, Li J, Zhou X. PHF12 regulates HDAC1 to promote tumorigenesis via EGFR/AKT signaling pathway in non-small cell lung cancer. J Transl Med 2024; 22:689. [PMID: 39075515 PMCID: PMC11287983 DOI: 10.1186/s12967-024-05488-x] [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: 09/24/2023] [Accepted: 07/03/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Lung cancer stands as the second most prevalent malignant neoplasm worldwide. Addressing the underlying mechanisms propelling the progression of non-small cell lung cancer is of paramount importance. In this study, we have elucidated the pivotal role of PHF12 in this context. MATERIALS AND METHODS We harnessed clinical lung cancer tissue samples and non-small cell lung cancer cell lines to discern the expression pattern of PHF12. In vitro assays probing cell proliferation were conducted to substantiate the functional impact of PHF12. Furthermore, an in vivo Xenograft model was employed to dissect the role of PHF12. Employing ChIP assays and qRT-PCR, we delved into the intricate binding dynamics between PHF12 and HDAC1. Mechanistic insights into the PHF12-HDAC1 axis in lung cancer progression were pursued via RNA-seq and GSEA analyses. RESULTS Notably, PHF12 exhibited a substantial upregulation within tumor tissue, concomitant with its correlation to HDAC1. The trilogy of cell proliferation assays, transwell assays, and the Xenograft model collectively underscored the promoting influence of PHF12 on lung cancer proliferation, both in vitro and in vivo. The ChIP assay unveiled the transcriptional regulatory role of PHF12 in governing HDAC1 expression. This correlation extended to both mRNA and protein levels. PHF12 promotes NSCLC progression through regulating HDCA1 expression. Intriguingly, the rescue of function within NSCLC cell lines post PHF12 knockdown was achievable through HDAC1 overexpression. Additionally, our findings unveiled the capacity of the PHF12-HDAC1 axis to activate the EGFR/AKT signaling pathway, thereby further corroborating its significance in lung cancer progression. CONCLUSION Our study identified PHF12 as an oncogenic role in lung cancer proliferation and migration for the first time. PHF12 transcriptionally regulate HDAC1 and activate EGFR/AKT signaling pathway in NSCLC progression. PHF12 may serve as an important target in lung cancer therapy.
Collapse
Affiliation(s)
- Yiru Kong
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Rongrong Jiang
- Department of Cardiothoracic Surgery, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Hui Zhou
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Mengxi Ge
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Hao Lin
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Yu Wang
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Rongrong Yao
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Qing Wang
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Xiaohua Liang
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Jing Li
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China.
| | - Xinli Zhou
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China.
| |
Collapse
|
2
|
Kim U, Debnath R, Maiz JE, Rico J, Sinha S, Blanco MA, Chakrabarti R. ΔNp63 regulates MDSC survival and metabolism in triple-negative breast cancer. iScience 2024; 27:109366. [PMID: 38510127 PMCID: PMC10951988 DOI: 10.1016/j.isci.2024.109366] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/20/2023] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
Triple-negative breast cancer (TNBC) contributes greatly to mortality of breast cancer, demanding new targetable options. We have shown that TNBC patients have high ΔNp63 expression in tumors. However, the function of ΔNp63 in established TNBC is yet to be explored. In current studies, targeting ΔNp63 with inducible CRISPR knockout and Histone deacetylase inhibitor Quisinostat showed that ΔNp63 is important for tumor progression and metastasis in established tumors by promoting myeloid-derived suppressor cell (MDSC) survival through tumor necrosis factor alpha. Decreasing ΔNp63 levels are associated with decreased CD4+ and FOXP3+ T-cells but increased CD8+ T-cells. RNA sequencing analysis indicates that loss of ΔNp63 alters multiple MDSC properties such as lipid metabolism, chemotaxis, migration, and neutrophil degranulation besides survival. We further demonstrated that targeting ΔNp63 sensitizes chemotherapy. Overall, we showed that ΔNp63 reprograms the MDSC-mediated immunosuppressive functions in TNBC, highlighting the benefit of targeting ΔNp63 in chemotherapy-resistant TNBC.
Collapse
Affiliation(s)
- Ukjin Kim
- Department of Surgery, Sylvester Comprehensive Cancer, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Rahul Debnath
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Javier E. Maiz
- Department of Surgery, Sylvester Comprehensive Cancer, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joshua Rico
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Satrajit Sinha
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Mario Andrés Blanco
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rumela Chakrabarti
- Department of Surgery, Sylvester Comprehensive Cancer, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
3
|
Napoli M, Deshpande AA, Chakravarti D, Rajapakshe K, Gunaratne PH, Coarfa C, Flores ER. Genome-wide p63-Target Gene Analyses Reveal TAp63/NRF2-Dependent Oxidative Stress Responses. CANCER RESEARCH COMMUNICATIONS 2024; 4:264-278. [PMID: 38165157 PMCID: PMC10832605 DOI: 10.1158/2767-9764.crc-23-0358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/14/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
The p53 family member TP63 encodes two sets of N-terminal isoforms, TAp63 and ΔNp63 isoforms. They each regulate diverse biological functions in epidermal morphogenesis and in cancer. In the skin, where their activities have been extensively characterized, TAp63 prevents premature aging by regulating the quiescence and genomic stability of stem cells required for wound healing and hair regeneration, while ΔNp63 controls maintenance and terminal differentiation of epidermal basal cells. This functional diversity is surprising given that these isoforms share a high degree of similarity, including an identical sequence for a DNA-binding domain. To understand the mechanisms of the transcriptional programs regulated by each p63 isoform and leading to diverse biological functions, we performed genome-wide analyses using p63 isoform-specific chromatin immunoprecipitation, RNA sequencing, and metabolomics of TAp63-/- and ΔNp63-/- mouse epidermal cells. Our data indicate that TAp63 and ΔNp63 physically and functionally interact with distinct transcription factors for the downstream regulation of their target genes, thus ultimately leading to the regulation of unique transcriptional programs and biological processes. Our findings unveil novel transcriptomes regulated by the p63 isoforms to control diverse biological functions, including the cooperation between TAp63 and NRF2 in the modulation of metabolic pathways and response to oxidative stress providing a mechanistic explanation for the TAp63 knock out phenotypes. SIGNIFICANCE The p63 isoforms, TAp63 and ΔNp63, control epithelial morphogenesis and tumorigenesis through the interaction with distinct transcription factors and the subsequent regulation of unique transcriptional programs.
Collapse
Affiliation(s)
- Marco Napoli
- Department of Molecular Oncology, Division of Basic Science, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Avani A. Deshpande
- Department of Molecular Oncology, Division of Basic Science, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | | | - Kimal Rajapakshe
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | | | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Elsa R. Flores
- Department of Molecular Oncology, Division of Basic Science, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
4
|
Wan X, Guo W, Zhan Z, Bai O. Dysregulation of FBW7 in malignant lymphoproliferative disorders. Front Oncol 2022; 12:988138. [PMID: 36457505 PMCID: PMC9707496 DOI: 10.3389/fonc.2022.988138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/18/2022] [Indexed: 11/10/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) is involved in various aspects of cell processes, including cell proliferation, differentiation, and cell cycle progression. F-box and WD repeat domain-containing protein 7 (FBW7), as a key component of UPS proteins and a critical tumor suppressor in human cancers, controls proteasome-mediated degradation by ubiquitinating oncoproteins such as c-Myc, Mcl-1, cyclin E, and Notch. It also plays a role in the development of various cancers, including solid and hematological malignancies, such as T-cell acute lymphoblastic leukemia, diffuse large B-cell lymphoma, and multiple myeloma. This comprehensive review emphasizes the functions, substrates, and expression of FBW7 in malignant lymphoproliferative disorders.
Collapse
Affiliation(s)
| | | | | | - Ou Bai
- Department of Hematology, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
5
|
Pokorna Z, Vyslouzil J, Vojtesek B, Coates PJ. Identifying pathways regulating the oncogenic p53 family member ΔNp63 provides therapeutic avenues for squamous cell carcinoma. Cell Mol Biol Lett 2022; 27:18. [PMID: 35196980 PMCID: PMC8903560 DOI: 10.1186/s11658-022-00323-x] [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: 12/01/2021] [Accepted: 02/15/2022] [Indexed: 12/17/2022] Open
Abstract
Background ΔNp63 overexpression is a common event in squamous cell carcinoma (SCC) that contributes to tumorigenesis, making ΔNp63 a potential target for therapy. Methods We created inducible TP63-shRNA cells to study the effects of p63-depletion in SCC cell lines and non-malignant HaCaT keratinocytes. DNA damaging agents, growth factors, signaling pathway inhibitors, histone deacetylase inhibitors, and metabolism-modifying drugs were also investigated for their ability to influence ΔNp63 protein and mRNA levels. Results HaCaT keratinocytes, FaDu and SCC-25 cells express high levels of ΔNp63. HaCaT and FaDu inducible TP63-shRNA cells showed reduced proliferation after p63 depletion, with greater effects on FaDu than HaCaT cells, compatible with oncogene addiction in SCC. Genotoxic insults and histone deacetylase inhibitors variably reduced ΔNp63 levels in keratinocytes and SCC cells. Growth factors that regulate proliferation/survival of squamous cells (IGF-1, EGF, amphiregulin, KGF, and HGF) and PI3K, mTOR, MAPK/ERK or EGFR inhibitors showed lesser and inconsistent effects, with dual inhibition of PI3K and mTOR or EGFR inhibition selectively reducing ΔNp63 levels in HaCaT cells. In contrast, the antihyperlipidemic drug lovastatin selectively increased ΔNp63 in HaCaT cells. Conclusions These data confirm that ΔNp63-positive SCC cells require p63 for continued growth and provide proof of concept that p63 reduction is a therapeutic option for these tumors. Investigations of ΔNp63 regulation identified agent-specific and cell-specific pathways. In particular, dual inhibition of the PI3K and mTOR pathways reduced ΔNp63 more effectively than single pathway inhibition, and broad-spectrum histone deacetylase inhibitors showed a time-dependent biphasic response, with high level downregulation at the transcriptional level within 24 h. In addition to furthering our understanding of ΔNp63 regulation in squamous cells, these data identify novel drug combinations that may be useful for p63-based therapy of SCC. Supplementary Information The online version contains supplementary material available at 10.1186/s11658-022-00323-x.
Collapse
Affiliation(s)
- Zuzana Pokorna
- Research Center of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53, Brno, Czech Republic
| | - Jan Vyslouzil
- Research Center of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53, Brno, Czech Republic
| | - Borivoj Vojtesek
- Research Center of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53, Brno, Czech Republic
| | - Philip J Coates
- Research Center of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53, Brno, Czech Republic.
| |
Collapse
|
6
|
ΔNp63 regulates a common landscape of enhancer associated genes in non-small cell lung cancer. Nat Commun 2022; 13:614. [PMID: 35105868 PMCID: PMC8807845 DOI: 10.1038/s41467-022-28202-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Distinct lung stem cells give rise to lung adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC). ΔNp63, the p53 family member and p63 isoform, guides the maturation of these stem cells through the regulation of their self-renewal and terminal differentiation; however, the underlying mechanistic role regulated by ∆Np63 in lung cancer development has remained elusive. By utilizing a ΔNp63-specific conditional knockout mouse model and xenograft models of LUAD and LUSC, we found that ∆Np63 promotes non-small cell lung cancer by maintaining the lung stem cells necessary for lung cancer cell initiation and progression in quiescence. ChIP-seq analysis of lung basal cells, alveolar type 2 (AT2) cells, and LUAD reveals robust ∆Np63 regulation of a common landscape of enhancers of cell identity genes. Importantly, one of these genes, BCL9L, is among the enhancer associated genes regulated by ∆Np63 in Kras-driven LUAD and mediates the oncogenic effects of ∆Np63 in both LUAD and LUSC. Accordingly, high BCL9L levels correlate with poor prognosis in LUAD patients. Taken together, our findings provide a unifying oncogenic role for ∆Np63 in both LUAD and LUSC through the regulation of a common landscape of enhancer associated genes. The mechanistic role regulated by the oncogene ∆Np63 in lung cancer development is currently unclear. Here, the authors show that ΔNp63 is pro-tumorigenic in lung adenocarcinoma as well as squamous cell carcinoma, and maintains lung cancer progenitor cells via regulation of super-enhancer-associated genes, including BCL9L
Collapse
|
7
|
Napoli M, Deshpande AA, Flores ER. The Landmark Discovery That Paved the Way to a Mechanistic Understanding of P53 Gain of Function and Personalized Medicine. Cancer Res 2021; 81:4394-4396. [PMID: 34470782 DOI: 10.1158/0008-5472.can-21-2382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022]
Abstract
In 1990, Baker and colleagues reported their seminal findings in Cancer Research focusing on the transition from adenoma to carcinoma of the colon. By sequencing the TP53 locus in 58 colorectal tumors (25 adenomas and 33 carcinomas) and measuring its allelic deletions, they discovered that this transition requires the loss of one TP53 allele and the mutation of the other one. Here, we discuss how this landmark discovery shed a new light on p53 mutations, prompting the generation of novel mouse models that definitively proved the mutant p53 gain-of-function hypothesis suggested by these results. Finally, we evaluate the implications that the Vogelstein model of cancer progression had on numerous aspects of cancer biology and cancer care, including the characterization of tumor evolution and the response to therapy, and how it ultimately contributed to the wider adoption of early detection screenings and personalized medicine.See related article by Baker and colleagues, Cancer Res 1990;50:7717-22.
Collapse
Affiliation(s)
- Marco Napoli
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Avani A Deshpande
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Elsa R Flores
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida. .,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
8
|
Wen N, Lv Q, Du ZG. MicroRNAs involved in drug resistance of breast cancer by regulating autophagy. J Zhejiang Univ Sci B 2021; 21:690-702. [PMID: 32893526 DOI: 10.1631/jzus.b2000076] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Autophagy is a conserved catabolic process characterized by degradation and recycling of cytosolic components or organelles through a lysosome-dependent pathway. It has a complex and close relationship to drug resistance in breast cancer. MicroRNAs (miRNAs) are small noncoding molecules that can influence numerous cellular processes including autophagy, through the posttranscriptional regulation of gene expression. Autophagy is regulated by many proteins and pathways, some of which in turn have been found to be regulated by miRNAs. These miRNAs may affect the drug resistance of breast cancer. Drug resistance is the main cause of distant recurrence, metastasis and death in breast cancer patients. In this review, we summarize the causative relationship between autophagy and drug resistance of breast cancer. The roles of autophagy-related proteins and pathways and their associated miRNAs in drug resistance of breast cancer are also discussed.
Collapse
Affiliation(s)
- Nan Wen
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qing Lv
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zheng-Gui Du
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
9
|
Nie X, Jia W, Li X, Pan X, Yin R, Liu N, Su Z. FBXW7 induces apoptosis in glioblastoma cells by regulating HDAC7. Cell Biol Int 2021; 45:2150-2158. [PMID: 34288252 DOI: 10.1002/cbin.11668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 01/01/2023]
Abstract
Glioblastoma is an aggressive type of brain cancer with an extremely poor prognosis. Additionally, the F-box WD repeat-containing protein 7 (FBXW7) is a component of the ubiquitin-proteasome system that has been widely implicated in human cancers. In this study, we investigated the role and mechanism of FBXW7 in glioblastoma. FBXW7 expression was analyzed in normal and glioblastoma tissue samples using The Cancer Genome Atlas Glioblastoma Multiforme (TCGA-GBM) database. Then, quantitative reverse transcription-polymerase chain reaction (RT-PCR) was used to examine mRNA expression, whereas, western blot analysis was conducted to determine protein levels of the samples. Furthermore, cell apoptosis was assessed using the Annexin V staining method, followed by flow cytometry analysis. Immunoprecipitation (IP) assay was conducted as well to test protein-protein interactions. Lastly, protein expression in tissues was examined by conducting immunohistochemistry (IHC). Results showed that the glioblastoma tissue samples displayed an FBXW7 downregulation compared with normal tissues. In vitro, the overexpression of FBXW7 in glioblastoma cells induced apoptosis, whereas, its knockdown displayed the opposite effect. Mechanistically, FBXW7 interacted with HDAC7 to promote HDAC7 ubiquitination, however, the overexpression of HDAC7 in glioblastoma cells blocked FBXW7-induced apoptosis. Finally, FBXW7 and HDAC7 displayed an inverse correlation in glioblastoma tissues in vivo. Therefore, our data demonstrated an important function of FBXW7 in promoting glioblastoma apoptosis by interacting with HDAC7 and promoting HDAC7 ubiquitination.
Collapse
Affiliation(s)
- Xiaohu Nie
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Cent Hospital HuZhou University, Huzhou, China
| | - Weiqiang Jia
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaobin Li
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Cent Hospital HuZhou University, Huzhou, China
| | - Xuyan Pan
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Cent Hospital HuZhou University, Huzhou, China
| | - Rui Yin
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Cent Hospital HuZhou University, Huzhou, China
| | - Ning Liu
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Cent Hospital HuZhou University, Huzhou, China
| | - Zhongzhou Su
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Cent Hospital HuZhou University, Huzhou, China
| |
Collapse
|
10
|
Non-canonical function of DGCR8 in DNA double-strand break repair signaling and tumor radioresistance. Nat Commun 2021; 12:4033. [PMID: 34188037 PMCID: PMC8242032 DOI: 10.1038/s41467-021-24298-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 06/11/2021] [Indexed: 12/21/2022] Open
Abstract
In response to DNA double-strand breaks (DSBs), repair proteins are recruited to the damaged sites. Ubiquitin signaling plays a critical role in coordinating protein recruitment during the DNA damage response. Here, we find that the microRNA biogenesis factor DGCR8 promotes tumor resistance to X-ray radiation independently of its Drosha-binding ability. Upon radiation, the kinase ATM and the deubiquitinase USP51 mediate the activation and stabilization of DGCR8 through phosphorylation and deubiquitination. Specifically, radiation-induced ATM-dependent phosphorylation of DGCR8 at serine 677 facilitates USP51 to bind, deubiquitinate, and stabilize DGCR8, which leads to the recruitment of DGCR8 and DGCR8’s binding partner RNF168 to MDC1 and RNF8 at DSBs. This, in turn, promotes ubiquitination of histone H2A, repair of DSBs, and radioresistance. Altogether, these findings reveal the non-canonical function of DGCR8 in DSB repair and suggest that radiation treatment may result in therapy-induced tumor radioresistance through ATM- and USP51-mediated activation and upregulation of DGCR8. The molecular mechanisms underlying cancer cell radioresistance need to be elucidated. In this study, the authors show that the microRNA biogenesis factor DGCR8 is stabilized by USP51 and ATM upon irradiation and by consequence it promotes the repair of DNA double-strand breaks and radioresistance by recruiting RNF168 to sites of damage.
Collapse
|
11
|
Pokorná Z, Vysloužil J, Hrabal V, Vojtěšek B, Coates PJ. The foggy world(s) of p63 isoform regulation in normal cells and cancer. J Pathol 2021; 254:454-473. [PMID: 33638205 DOI: 10.1002/path.5656] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/10/2021] [Accepted: 02/24/2021] [Indexed: 12/19/2022]
Abstract
The p53 family member p63 exists as two major protein variants (TAp63 and ΔNp63) with distinct expression patterns and functional properties. Whilst downstream target genes of p63 have been studied intensively, how p63 variants are themselves controlled has been relatively neglected. Here, we review advances in understanding ΔNp63 and TAp63 regulation, highlighting their distinct pathways. TAp63 has roles in senescence and metabolism, and in germ cell genome maintenance, where it is activated post-transcriptionally by phosphorylation cascades after DNA damage. The function and regulation of TAp63 in mesenchymal and haematopoietic cells is less clear but may involve epigenetic control through DNA methylation. ΔNp63 functions to maintain stem/progenitor cells in various epithelia and is overexpressed in squamous and certain other cancers. ΔNp63 is transcriptionally regulated through multiple enhancers in concert with chromatin modifying proteins. Many signalling pathways including growth factors, morphogens, inflammation, and the extracellular matrix influence ΔNp63 levels, with inconsistent results reported. There is also evidence for reciprocal regulation, including ΔNp63 activating its own transcription. ΔNp63 is downregulated during cell differentiation through transcriptional regulation, while post-transcriptional events cause proteasomal degradation. Throughout the review, we identify knowledge gaps and highlight discordances, providing potential explanations including cell-context and cell-matrix interactions. Identifying individual p63 variants has roles in differential diagnosis and prognosis, and understanding their regulation suggests clinically approved agents for targeting p63 that may be useful combination therapies for selected cancer patients. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Zuzana Pokorná
- Research Centre of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Jan Vysloužil
- Research Centre of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Václav Hrabal
- Research Centre of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Borˇivoj Vojtěšek
- Research Centre of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Philip J Coates
- Research Centre of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Brno, Czech Republic
| |
Collapse
|
12
|
Jin Q, Hu H, Yan S, Jin L, Pan Y, Li X, Peng Y, Cao P. lncRNA MIR22HG-Derived miR-22-5p Enhances the Radiosensitivity of Hepatocellular Carcinoma by Increasing Histone Acetylation Through the Inhibition of HDAC2 Activity. Front Oncol 2021; 11:572585. [PMID: 33718133 PMCID: PMC7943860 DOI: 10.3389/fonc.2021.572585] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 01/05/2021] [Indexed: 12/30/2022] Open
Abstract
Background With the development of radiotherapy technology, radiotherapy has been increasingly used to treat primary hepatocellular carcinoma (HCC). However, due to radioresistance and the intolerance of the adjacent organs to radiation, the effects of radiotherapy are often unsatisfactory. Therefore, it is necessary to study radiosensitization in HCC. Method A microarray was used to analyze the genes that were significantly associated with radiosensitivity. HCC cells, HepG2 and MHCC97H, were subjected to radiation in vitro. Real-time PCR was performed to determine MIR22HG (microRNA22 host gene) and miR-22-5p expression levels. Western blotting was performed to determine histone expression levels. A histone deacetylase (HDAC) whole cell assay was used to determine the activity of HDAC2. MTT, colony formation, 5-ethynyl-2′-deoxyuridine, and wound healing assays were performed to examine the function of MIR22HG and miR-22-5p in cellular radiosensitivity. Chromatin immunoprecipitation-PCR was used to confirm that HDAC2 affects the acetylation level of the MIR22HG promoter region. Finally, animal experiments were performed to demonstrate the in vivo effect of MIR22HG on the radiosensitivity of hepatoma. Results Irradiation can up-regulate MIR22HG expression and down-regulate HDAC2 expression. Inhibition of HDAC2 expression promotes histone acetylation in the MIR22HG promoter region and up-regulates MIR22HG expression. MIR22HG can increase radiosensitivity via miR-22-5p in HCC. Conclusion Inhibition of HDAC2 expression promotes histone acetylation in the MIR22HG promoter region, thereby up-regulating the expression of MIR22HG and promoting the production of miR-22-5p, and ultimately increasing the sensitivity of liver cancer radiotherapy.
Collapse
Affiliation(s)
- Qiao Jin
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Hao Hu
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Siqi Yan
- Department of Oncological Radiotherapy, Hunan Academy of Traditional Chinese Medicine Affiliated Hospital, Changsha, China
| | - Long Jin
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Yuliang Pan
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiangjun Li
- Department of Oncology, The Second People's Hospital of Hunan Province, Changsha, China
| | - Yayi Peng
- Department of Oncology, The Second People's Hospital of Hunan Province, Changsha, China
| | - Peiguo Cao
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
13
|
Napoli M, Li X, Ackerman HD, Deshpande AA, Barannikov I, Pisegna MA, Bedrosian I, Mitsch J, Quinlan P, Thompson A, Rajapakshe K, Coarfa C, Gunaratne PH, Marchion DC, Magliocco AM, Tsai KY, Flores ER. Pan-cancer analysis reveals TAp63-regulated oncogenic lncRNAs that promote cancer progression through AKT activation. Nat Commun 2020; 11:5156. [PMID: 33056990 PMCID: PMC7561725 DOI: 10.1038/s41467-020-18973-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 09/24/2020] [Indexed: 12/16/2022] Open
Abstract
The most frequent genetic alterations across multiple human cancers are mutations in TP53 and the activation of the PI3K/AKT pathway, two events crucial for cancer progression. Mutations in TP53 lead to the inhibition of the tumour and metastasis suppressor TAp63, a p53 family member. By performing a mouse-human cross species analysis between the TAp63 metastatic mammary adenocarcinoma mouse model and models of human breast cancer progression, we identified two TAp63-regulated oncogenic lncRNAs, TROLL-2 and TROLL-3. Further, using a pan-cancer analysis of human cancers and multiple mouse models of tumour progression, we revealed that these two lncRNAs induce the activation of AKT to promote cancer progression by regulating the nuclear to cytoplasmic translocation of their effector, WDR26, via the shuttling protein NOLC1. Our data provide preclinical rationale for the implementation of these lncRNAs and WDR26 as therapeutic targets for the treatment of human tumours dependent upon mutant TP53 and/or the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Marco Napoli
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Xiaobo Li
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Hayley D Ackerman
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Avani A Deshpande
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Ivan Barannikov
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Marlese A Pisegna
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Isabelle Bedrosian
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jürgen Mitsch
- Advanced Data Analysis Centre, Nottingham, NG7 2RD, UK.,School of Computer Sciences University of Nottingham, Nottingham, NG7 2RD, UK
| | - Philip Quinlan
- Advanced Data Analysis Centre, Nottingham, NG7 2RD, UK.,School of Computer Sciences University of Nottingham, Nottingham, NG7 2RD, UK
| | - Alastair Thompson
- Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Preethi H Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77004, USA
| | - Douglas C Marchion
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Anthony M Magliocco
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Kenneth Y Tsai
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.,Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.,Department of Tumour Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Elsa R Flores
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA. .,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
14
|
Jiang YY, Jiang Y, Li CQ, Zhang Y, Dakle P, Kaur H, Deng JW, Lin RYT, Han L, Xie JJ, Yan Y, Doan N, Zheng Y, Mayakonda A, Hazawa M, Xu L, Li Y, Aswad L, Jeitany M, Kanojia D, Guan XY, Said JW, Yang W, Fullwood MJ, Lin DC, Koeffler HP. TP63, SOX2, and KLF5 Establish a Core Regulatory Circuitry That Controls Epigenetic and Transcription Patterns in Esophageal Squamous Cell Carcinoma Cell Lines. Gastroenterology 2020; 159:1311-1327.e19. [PMID: 32619460 DOI: 10.1053/j.gastro.2020.06.050] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 06/12/2020] [Accepted: 06/21/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS We investigated the transcriptome of esophageal squamous cell carcinoma (ESCC) cells, activity of gene regulatory (enhancer and promoter regions), and the effects of blocking epigenetic regulatory proteins. METHODS We performed chromatin immunoprecipitation sequencing with antibodies against H3K4me1, H3K4me3, and H3K27ac and an assay for transposase-accessible chromatin to map the enhancer regions and accessible chromatin in 8 ESCC cell lines. We used the CRC_Mapper algorithm to identify core regulatory circuitry transcription factors in ESCC cell lines, and determined genome occupancy profiles for 3 of these factors. In ESCC cell lines, expression of transcription factors was knocked down with small hairpin RNAs, promoter and enhancer regions were disrupted by CRISPR/Cas9 genome editing, or bromodomains and extraterminal (BET) family proteins and histone deacetylases (HDACs) were inhibited with ARV-771 and romidepsin, respectively. ESCC cell lines were then analyzed by whole-transcriptome sequencing, immunoprecipitation, immunoblots, immunohistochemistry, and viability assays. Interactions between distal enhancers and promoters were identified and verified with circular chromosome conformation capture sequencing. NOD-SCID mice were given injections of modified ESCC cells, some mice where given injections of HDAC or BET inhibitors, and growth of xenograft tumors was measured. RESULTS We identified super-enhancer-regulated circuits and transcription factors TP63, SOX2, and KLF5 as core regulatory factors in ESCC cells. Super-enhancer regulation of ALDH3A1 mediated by core regulatory factors was required for ESCC viability. We observed direct interactions between the promoter region of TP63 and functional enhancers, mediated by the core regulatory circuitry transcription factors. Deletion of enhancer regions from ESCC cells decreased expression of the core regulatory circuitry transcription factors and reduced cell viability; these same results were observed with knockdown of each core regulatory circuitry transcription factor. Incubation of ESCC cells with BET and HDAC disrupted the core regulatory circuitry program and the epigenetic modifications observed in these cells; mice given injections of HDAC or BET inhibitors developed smaller xenograft tumors from the ESCC cell lines. Xenograft tumors grew more slowly in mice given the combination of ARV-771 and romidepsin than mice given either agent alone. CONCLUSIONS In epigenetic and transcriptional analyses of ESCC cell lines, we found the transcription factors TP63, SOX2, and KLF5 to be part of a core regulatory network that determines chromatin accessibility, epigenetic modifications, and gene expression patterns in these cells. A combination of epigenetic inhibitors slowed growth of xenograft tumors derived from ESCC cells in mice.
Collapse
Affiliation(s)
- Yan-Yi Jiang
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California; Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Yuan Jiang
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California; Cancer Science Institute of Singapore, National University of Singapore, Singapore.
| | - Chun-Quan Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Ying Zhang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Pushkar Dakle
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Harvinder Kaur
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Jian-Wen Deng
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Ruby Yu-Tong Lin
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Lin Han
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Jian-Jun Xie
- Department of Biochemistry and Molecular Biology, Medical College of Shantou University, Shantou, China
| | - Yiwu Yan
- Cedars-Sinai Medical Center, Departments of Surgery and Biomedical Sciences, Los Angeles, California
| | - Ngan Doan
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Yueyuan Zheng
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Anand Mayakonda
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Masaharu Hazawa
- Cell-Bionomics Research Unit, Innovative Integrated Bio-Research Core, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Liang Xu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - YanYu Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Luay Aswad
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore
| | - Maya Jeitany
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Deepika Kanojia
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Xin-Yuan Guan
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jonathan W Said
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Wei Yang
- Cedars-Sinai Medical Center, Departments of Surgery and Biomedical Sciences, Los Angeles, California
| | - Melissa J Fullwood
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore.
| | - De-Chen Lin
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California.
| | - H Phillip Koeffler
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California; Cancer Science Institute of Singapore, National University of Singapore, Singapore; National University Cancer Institute, National University Hospital Singapore, Singapore
| |
Collapse
|
15
|
Cao D, Zhu H, Zhao Q, Huang J, Zhou C, He J, Liang Y. MiR-128 suppresses metastatic capacity by targeting metadherin in breast cancer cells. Biol Res 2020; 53:43. [PMID: 32993809 PMCID: PMC7526227 DOI: 10.1186/s40659-020-00311-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/17/2020] [Indexed: 01/17/2023] Open
Abstract
Background Breast cancer, the most common cancer in women worldwide, causes the vast majority of cancer-related deaths. Undoubtedly, tumor metastasis and recurrence are responsible for more than 90 percent of these deaths. MicroRNAs are endogenous noncoding RNAs that have been integrated into almost all the physiological and pathological processes, including metastasis. In the present study, the role of miR-128 in breast cancer was investigated. Results Compared to the corresponding adjacent normal tissue, the expression of miR-128 was significantly suppressed in human breast cancer specimens. More importantly, its expression level was reversely correlated to histological grade of the cancer. Ectopic expression of miR-128 in the aggressive breast cancer cell line MDA-MB-231 could inhibit cell motility and invasive capacity remarkably. Afterwards, Metadherin (MTDH), also known as AEG-1 (Astrocyte Elevated Gene 1) and Lyric that implicated in various aspects of cancer progression and metastasis, was further identified as a direct target gene of miR-128 and its expression level was up-regulated in clinical samples as expected. Moreover, knockdown of MTDH in MDA-MB-231 cells obviously impaired the migration and invasion capabilities, whereas re-expression of MTDH abrogated the suppressive effect caused by miR-128. Conclusions Overall, these findings demonstrate that miR-128 could serve as a novel biomarker for breast cancer metastasis and a potent target for treatment in the future.
Collapse
Affiliation(s)
- Danxia Cao
- Comprehensive Breast Health Center, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Rui-Jin Er Road, Shanghai, 200025, China
| | - Han Zhu
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, No. 2800, Gong-Wei Road, Shanghai, 201399, China
| | - Qian Zhao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, No. 280, Chong-Qing South Road, Shanghai, 200025, China
| | - Jianming Huang
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, No. 2800, Gong-Wei Road, Shanghai, 201399, China
| | - Cixiang Zhou
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, No. 280, Chong-Qing South Road, Shanghai, 200025, China
| | - Jianrong He
- Comprehensive Breast Health Center, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Rui-Jin Er Road, Shanghai, 200025, China.
| | - Yongjun Liang
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, No. 2800, Gong-Wei Road, Shanghai, 201399, China.
| |
Collapse
|
16
|
Gatti V, Bernassola F, Talora C, Melino G, Peschiaroli A. The Impact of the Ubiquitin System in the Pathogenesis of Squamous Cell Carcinomas. Cancers (Basel) 2020; 12:1595. [PMID: 32560247 PMCID: PMC7352818 DOI: 10.3390/cancers12061595] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/11/2020] [Accepted: 06/13/2020] [Indexed: 02/07/2023] Open
Abstract
The ubiquitin system is a dynamic regulatory pathway controlling the activity, subcellular localization and stability of a myriad of cellular proteins, which in turn affects cellular homeostasis through the regulation of a variety of signaling cascades. Aberrant activity of key components of the ubiquitin system has been functionally linked with numerous human diseases including the initiation and progression of human tumors. In this review, we will contextualize the importance of the two main components of the ubiquitin system, the E3 ubiquitin ligases (E3s) and deubiquitinating enzymes (DUBs), in the etiology of squamous cell carcinomas (SCCs). We will discuss the signaling pathways regulated by these enzymes, emphasizing the genetic and molecular determinants underlying their deregulation in SCCs.
Collapse
Affiliation(s)
- Veronica Gatti
- National Research Council of Italy, Institute of Translational Pharmacology, 00133 Rome, Italy;
| | - Francesca Bernassola
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy; (F.B.); (G.M.)
| | - Claudio Talora
- Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy;
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy; (F.B.); (G.M.)
| | - Angelo Peschiaroli
- National Research Council of Italy, Institute of Translational Pharmacology, 00133 Rome, Italy;
| |
Collapse
|
17
|
Bui NHB, Napoli M, Davis AJ, Abbas HA, Rajapakshe K, Coarfa C, Flores ER. Spatiotemporal Regulation of ΔNp63 by TGFβ-Regulated miRNAs Is Essential for Cancer Metastasis. Cancer Res 2020; 80:2833-2847. [PMID: 32312834 DOI: 10.1158/0008-5472.can-19-2733] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 03/18/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023]
Abstract
ΔNp63 is a transcription factor of the p53 family and has crucial functions in normal development and disease. The expression pattern of ΔNp63 in human cancer suggests dynamic regulation of this isoform during cancer progression and metastasis. Many primary and metastatic tumors express high levels of ΔNp63, while ΔNp63 loss is crucial for tumor dissemination, indicating an oscillatory expression of ΔNp63 during cancer progression. Here, we use genetically engineered orthotopic mouse models of breast cancer to show that while depletion of ΔNp63 inhibits primary mammary adenocarcinoma development, oscillatory expression of ΔNp63 in established tumors is crucial for metastatic dissemination in breast cancer. A TGFβ-regulated miRNA network acted as upstream regulators of this oscillatory expression of ΔNp63 during cancer progression. This work sheds light on the pleiotropic roles of ΔNp63 in cancer and unveils critical functions of TGFβ in the metastatic process. SIGNIFICANCE: This study unveils TGFβ signaling and a network of four miRNAs as upstream regulators of ΔNp63, providing key information for the development of therapeutic strategies to treat cancers that commonly overexpress ΔNp63.
Collapse
Affiliation(s)
- Ngoc H B Bui
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Marco Napoli
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Andrew John Davis
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Hussein A Abbas
- Hematology/Oncology Fellowship Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Elsa R Flores
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida. .,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
18
|
Patel A, Garcia LF, Mannella V, Gammon L, Borg TM, Maffucci T, Scatolini M, Chiorino G, Vergani E, Rodolfo M, Maurichi A, Posch C, Matin RN, Harwood CA, Bergamaschi D. Targeting p63 Upregulation Abrogates Resistance to MAPK Inhibitors in Melanoma. Cancer Res 2020; 80:2676-2688. [PMID: 32291316 DOI: 10.1158/0008-5472.can-19-3230] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/04/2020] [Accepted: 04/09/2020] [Indexed: 11/16/2022]
Abstract
Targeting the MAPK pathway by combined inhibition of BRAF and MEK has increased overall survival in advanced BRAF-mutant melanoma in both therapeutic and adjuvant clinical settings. However, a significant proportion of tumors develop acquired resistance, leading to treatment failure. We have previously shown p63 to be an important inhibitor of p53-induced apoptosis in melanoma following genotoxic drug exposure. Here, we investigated the role of p63 in acquired resistance to MAPK inhibition and show that p63 isoforms are upregulated in melanoma cell lines chronically exposed to BRAF and MEK inhibition, with consequent increased resistance to apoptosis. This p63 upregulation was the result of its reduced degradation by the E3 ubiquitin ligase FBXW7. FBXW7 was itself regulated by MDM2, and in therapy-resistant melanoma cell lines, nuclear accumulation of MDM2 caused downregulation of FBXW7 and consequent upregulation of p63. Consistent with this, both FBXW7-inactivating mutations and MDM2 upregulation were found in melanoma clinical samples. Treatment of MAPK inhibitor-resistant melanoma cells with MDM2 inhibitor Nutlin-3A restored FBXW7 expression and p63 degradation in a dose-dependent manner and sensitized these cells to apoptosis. Collectively, these data provide a compelling rationale for future investigation of Nutlin-3A as an approach to abrogate acquired resistance of melanoma to MAPK inhibitor targeted therapy. SIGNIFICANCE: Upregulation of p63, an unreported mechanism of MAPK inhibitor resistance in melanoma, can be abrogated by treatment with the MDM2 inhibitor Nutlin-3A, which may serve as a strategy to overcome resistance.
Collapse
Affiliation(s)
- Ankit Patel
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Lucia Fraile Garcia
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Viviana Mannella
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Luke Gammon
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Tiffanie-Marie Borg
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Tania Maffucci
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Maria Scatolini
- Molecular Oncology Lab, Fondazione Edo ed Elvo Tempia, Biella, Italy
| | | | - Elisabetta Vergani
- Department of Experimental Oncology and Molecular Medicine, Immunotherapy Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Monica Rodolfo
- Department of Experimental Oncology and Molecular Medicine, Immunotherapy Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Andrea Maurichi
- Department of Surgery, Melanoma and Sarcoma Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Christian Posch
- Department of Dermatology, The Rudolfstiftung Hospital, Vienna, Austria.,Faculty of Medicine, Sigmund Freud University, Vienna, Austria.,Department of Dermatology, Technical University of Munich, Munich, Germany
| | - Rubeta N Matin
- Department of Dermatology, Oxford University Hospitals NHS Foundation Trust, Headington, Oxford, United Kingdom
| | - Catherine A Harwood
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Daniele Bergamaschi
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.
| |
Collapse
|
19
|
Citro S, Bellini A, Medda A, Sabatini ME, Tagliabue M, Chu F, Chiocca S. Human Papilloma Virus Increases ΔNp63α Expression in Head and Neck Squamous Cell Carcinoma. Front Cell Infect Microbiol 2020; 10:143. [PMID: 32322564 PMCID: PMC7156594 DOI: 10.3389/fcimb.2020.00143] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 03/18/2020] [Indexed: 12/13/2022] Open
Abstract
P63, and in particular the most expressed ΔNp63α isoform, seems to have a critical role in the outcome of head and neck cancer. Many studies have been conducted to assess the possible use of p63 as a prognostic marker in squamous cell carcinoma cancers, but the results are still not well-defined. Moreover, a clear relationship between the expression of ΔNp63α and the presence of high-risk HPV E6 and E7 oncoproteins has been delineated. Here we describe how ΔNp63α is mostly expressed in HPV-positive compared to HPV-negative head and neck cancer cell lines, with a very good correlation between ΔNp63α mRNA and protein levels.
Collapse
Affiliation(s)
- Simona Citro
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Alice Bellini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Alessandro Medda
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Maria Elisa Sabatini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Marta Tagliabue
- Division of Otolaryngology Head & Neck Surgery, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Francesco Chu
- Division of Otolaryngology Head & Neck Surgery, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Susanna Chiocca
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
20
|
xCT Inhibition Increases Sensitivity to Vorinostat in a ROS-Dependent Manner. Cancers (Basel) 2020; 12:cancers12040827. [PMID: 32235498 PMCID: PMC7226257 DOI: 10.3390/cancers12040827] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/20/2020] [Accepted: 03/25/2020] [Indexed: 12/30/2022] Open
Abstract
As histone deacetylase inhibitors (HDACIs) have limited efficacy against solid tumors, we investigated whether and how oxidative stress is involved in sensitivity to HDACIs to develop a novel therapeutic option of HDACIs treatment. We first tested whether a reduction of the antioxidant glutathione (GSH) by glutamine deprivation affects sensitivity to a commercially available HDACI vorinostat and reactive oxygen species (ROS) accumulation. Next we investigated the relationship between a glutamate-cystine transporter xCT and the efficacy of vorinostat using siRNA of xCT and bioinformatic analyses. Finally, we verified the combinatory effects of vorinostat and the xCT inhibitor salazosulfapyridine (SASP) on ROS accumulation, cell death induction, and colony formation. Glutamine deprivation increased vorinostat-mediated cell death with ROS accumulation. Genetic ablation of xCT improved the efficacy of vorinostat, consistent with the results of public data analyses demonstrating that xCT expressions positively correlate with insensitivity to HDACIs in many types of cancer cell lines. Vorinostat caused ROS accumulation when combined with SASP, possibly resulting in synergistic ferroptosis. Our study provides a novel mechanistic insight into the mechanism underlying sensitivity to HDACIs involving xCT, suggesting xCT to be a promising predictive marker of HDACIs and rationalizing combinatory therapy of HDACIs with xCT inhibitors to induce ferroptosis.
Collapse
|
21
|
Davis AJ, Tsinkevich M, Rodencal J, Abbas HA, Su XH, Gi YJ, Fang B, Rajapakshe K, Coarfa C, Gunaratne PH, Koomen JM, Tsai KY, Flores ER. TAp63-Regulated miRNAs Suppress Cutaneous Squamous Cell Carcinoma through Inhibition of a Network of Cell-Cycle Genes. Cancer Res 2020; 80:2484-2497. [PMID: 32156775 DOI: 10.1158/0008-5472.can-19-1892] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 01/18/2020] [Accepted: 03/05/2020] [Indexed: 12/26/2022]
Abstract
TAp63 is a p53 family member and potent tumor and metastasis suppressor. Here, we show that TAp63-/- mice exhibit an increased susceptibility to ultraviolet radiation-induced cutaneous squamous cell carcinoma (cuSCC). A human-to-mouse comparison of cuSCC tumors identified miR-30c-2* and miR-497 as underexpressed in TAp63-deficient cuSCC. Reintroduction of these miRNAs significantly inhibited the growth of cuSCC cell lines and tumors. Proteomic profiling of cells expressing either miRNA showed downregulation of cell-cycle progression and mitosis-associated proteins. A mouse to human and cross-platform comparison of RNA-sequencing and proteomics data identified a 7-gene signature, including AURKA, KIF18B, PKMYT1, and ORC1, which were overexpressed in cuSCC. Knockdown of these factors in cuSCC cell lines suppressed tumor cell proliferation and induced apoptosis. In addition, selective inhibition of AURKA suppressed cuSCC cell proliferation, induced apoptosis, and showed antitumor effects in vivo. Finally, treatment with miR-30c-2* or miR-497 miRNA mimics was highly effective in suppressing cuSCC growth in vivo. Our data establish TAp63 as an essential regulator of novel miRNAs that can be therapeutically targeted for potent suppression of cuSCC. SIGNIFICANCE: This study provides preclinical evidence for the use of miR-30c-2*/miR-497 delivery and AURKA inhibition in the treatment of cuSCC, which currently has no FDA-approved targeted therapies.See related commentary by Parrales and Iwakuma, p. 2439.
Collapse
Affiliation(s)
- Andrew John Davis
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Maksym Tsinkevich
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jason Rodencal
- Department of Biology, Stanford University School of Medicine, Stanford, California
| | - Hussein A Abbas
- Hematology/Oncology Fellowship Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiao-Hua Su
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Young-Jin Gi
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bin Fang
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Preethi H Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - John M Koomen
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kenneth Y Tsai
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Elsa R Flores
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
22
|
Napoli M, Flores ER. The p53 family reaches the final frontier: the variegated regulation of the dark matter of the genome by the p53 family in cancer. RNA Biol 2020; 17:1636-1647. [PMID: 31910062 PMCID: PMC7567494 DOI: 10.1080/15476286.2019.1710054] [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] [Indexed: 02/08/2023] Open
Abstract
The tumour suppressor p53 and its paralogues, p63 and p73, are essential to maintain cellular homoeostasis and the integrity of the cell's genetic material, thus meriting the title of 'guardians of the genome'. The p53 family members are transcription factors and fulfill their activities by controlling the expression of protein-coding and non-coding genes. Here, we review how the latter group transcended from the 'dark matter' of the transcriptome, providing unexpected and intriguing anti-cancer therapeutic strategies.
Collapse
Affiliation(s)
- Marco Napoli
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute , Tampa, FL, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute , Tampa, FL, USA
| | - Elsa R Flores
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute , Tampa, FL, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute , Tampa, FL, USA
| |
Collapse
|
23
|
He Y, Tai S, Deng M, Fan Z, Ping F, He L, Zhang C, Huang Y, Cheng B, Xia J. Metformin and 4SC-202 synergistically promote intrinsic cell apoptosis by accelerating ΔNp63 ubiquitination and degradation in oral squamous cell carcinoma. Cancer Med 2019; 8:3479-3490. [PMID: 31025540 PMCID: PMC6601594 DOI: 10.1002/cam4.2206] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/03/2019] [Accepted: 04/11/2019] [Indexed: 02/06/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common and aggressive epithelial tumor in the head and neck region with a rising incidence. Despite the advances in basic science and clinical research, the overall survival rate of OSCC remains low. Thus finding novel effective therapeutic agents for OSCC is necessary. In this study, we investigated the effects and mechanisms of combined metformin and 4SC-202 in OSCC. Our results showed that metformin and 4SC-202 synergistically suppressed the proliferation and promoted the intrinsic apoptosis of OSCC cells in vitro and in vivo. Importantly, the proteasome inhibitor MG132 impeded the ΔNp63-decreasing effects after metformin and 4SC-202 treatment, indicating that metformin and 4SC-202 could promote the degradation of ΔNp63 protein. Moreover, ubiquitination level of ΔNp63 increased after metformin or/and 4SC-202 administration. Furthermore, we revealed that ΔNp63 mediated anticancer effects of metformin and 4SC-202, as overexpression or suppression of ΔNp63 could attenuate or facilitate the apoptosis rate of OSCC under metformin or/and 4SC-202 treatment. Collectively, metformin and 4SC-202 synergistically promote intrinsic apoptosis through accelerating ubiquitin-mediated degradation of ΔNp63 in OSCC, and this co-treatment can serve as a potential therapeutic scheme for OSCC.
Collapse
Affiliation(s)
- Yuan He
- Department of Oral MedicineGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
- Guangdong Provincial Key Laboratory of StomatologyGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
| | - Shanshan Tai
- Department of Oral MedicineGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
- Guangdong Provincial Key Laboratory of StomatologyGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
| | - Miao Deng
- Department of Oral MedicineGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
- Guangdong Provincial Key Laboratory of StomatologyGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
| | - Zhaona Fan
- Department of Oral MedicineGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
- Guangdong Provincial Key Laboratory of StomatologyGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
| | - Fan Ping
- Department of Oral MedicineGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
- Guangdong Provincial Key Laboratory of StomatologyGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
| | - Lihong He
- Department of Oral MedicineGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
- Guangdong Provincial Key Laboratory of StomatologyGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
| | - Chi Zhang
- Department of Oral MedicineGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
- Guangdong Provincial Key Laboratory of StomatologyGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
| | - Yulei Huang
- Department of Oral MedicineGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
- Guangdong Provincial Key Laboratory of StomatologyGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
| | - Bin Cheng
- Department of Oral MedicineGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
- Guangdong Provincial Key Laboratory of StomatologyGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
| | - Juan Xia
- Department of Oral MedicineGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
- Guangdong Provincial Key Laboratory of StomatologyGuanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhouP.R. China
| |
Collapse
|
24
|
Alsaegh MA, Altaie AM, Zhu S. p63 Expression and its Relation to Epithelial Cells Proliferation in Dentigerous Cyst, Odontogenic Keratocyst, and Ameloblastoma. Pathol Oncol Res 2019; 26:1175-1182. [PMID: 31187467 DOI: 10.1007/s12253-019-00680-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/30/2019] [Indexed: 10/26/2022]
Abstract
The current controversy about the classification of odontogenic keratocyst reflects the shortage in the understanding of the odontogenic cysts and tumors. The aim of the present study was to investigate p63 immunoexpression and its relation to the proliferation of the epithelial lining in dentigerous cyst (DC), odontogenic keratocyst (OKC), and follicular type of ameloblastoma (AB). The study involved 36 samples, which are DC (n = 12), OKC (n = 9), and AB (n = 15). p63 protein expression was evaluated by immunohistochemistry. The results on Ki-67 expression were obtained from our previous studies and correlated with p63 expressions. p63 was expressed differently in the studied lesions with various distribution in different study samples. Statistical analysis using Kruskal-Wallis test showed a significant difference in the expression of p63 protein among DC, OKC, and AB (p = 0.048). Subsequently, Mann-Whitney U test revealed the expression of p63 protein was significantly higher in OKC than DC (p = 0.018). Interestingly, Spearman's correlation analysis showed a positive correlation between the expression of p63 and Ki-67 in the odontogenic epithelium of DC (σ = 0.757, P = 0.004) and OKC (σ = 0.741, P = 0.022). While no such a positive correlation was found between the two studied markers in AB group (σ = 0.006, P = 0.983). In conclusion, the present results indicated various expression and correlation of p63 with the proliferation of odontogenic epithelial cells in DC, OKC, and AB. This diversity could reflect a different role and pathways of ΔNp63 in odontogenic tumor than that in odontogenic cyst. These together will help in better understanding the pathogenesis and biological behavior of odontogenic cysts and tumors.
Collapse
Affiliation(s)
- Mohammed Amjed Alsaegh
- Department of Oral and Maxillofacial Surgery, College of Dentistry, University of Science and Technology of Fujairah, Al-Hulifat, Fujairah, 2202, United Arab Emirates. .,Department of Oral and Maxillofacial Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China.
| | - Alaa Muayad Altaie
- Sharjah Medical Research Institute, Medical College, Sharjah University, Sharjah, United Arab Emirates
| | - Shengrong Zhu
- Department of Oral and Maxillofacial Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| |
Collapse
|
25
|
Synergistic antitumour activity of HDAC inhibitor SAHA and EGFR inhibitor gefitinib in head and neck cancer: a key role for ΔNp63α. Br J Cancer 2019; 120:658-667. [PMID: 30765872 PMCID: PMC6461861 DOI: 10.1038/s41416-019-0394-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 01/15/2019] [Accepted: 01/18/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) overexpression is associated with the development of head and neck cancer (HNC) and represents one of the main therapeutic targets for this disease. The use of EGFR inhibitors has limited efficacy due to their primary and acquired resistance, partially because of increased epithelial to mesenchymal transition (EMT). The HDAC inhibitor SAHA has been shown to revert EMT in different tumours, including HNC. In this study, we investigated the cooperative role of SAHA and the EGFR tyrosine kinase inhibitor gefitinib in both HPV-positive and HPV-negative HNC cell lines. METHODS A panel of 12 HPV-positive and HPV-negative HNC cell lines were screened for cell viability upon treatment with SAHA, gefitinib and the combination of the two. Epithelial/mesenchymal marker expression, as well as activation of signalling pathway, were assessed upon SAHA treatment. ΔNp63α silencing with shRNA lentiviral particles was used to determine its role in cell proliferation, migration and TGFβ pathway activation. RESULTS We found that both SAHA and gefitinib have antitumour activity in both HPV-positive and HPV-negative HNC cell lines and that their combination has a synergistic effect in inhibiting cell growth. SAHA treatment reverts EMT and inhibits the expression of the transcription factor ΔNp63α. Suppression of ΔNp63α reduces EGFR protein levels and decreases cell proliferation and TGFβ-dependent migration in both HPV-positive and HPV-negative HNC cell lines. CONCLUSIONS Our results, by giving a clear molecular mechanism at the basis of the antitumour activity of SAHA in HNC cell lines, provide a rationale for the clinical evaluation of SAHA in combination with gefitinib in both HPV-positive and HPV-negative HNC patients. Further knowledge is key to devising additional lines of combinatorial treatment strategies for this disease.
Collapse
|
26
|
Chen DQ, Yu C, Zhang XF, Liu ZF, Wang R, Jiang M, Chen H, Yan F, Tao M, Chen LB, Zhu H, Feng JF. HDAC3-mediated silencing of miR-451 decreases chemosensitivity of patients with metastatic castration-resistant prostate cancer by targeting NEDD9. Ther Adv Med Oncol 2018; 10:1758835918783132. [PMID: 30034549 PMCID: PMC6048672 DOI: 10.1177/1758835918783132] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 05/23/2018] [Indexed: 01/09/2023] Open
Abstract
Background: Treatment of metastatic castration-resistant prostate cancer (mCRPC) with
docetaxel often fails due to the emergence of chemoresistance. Thus,
restoring chemosensitivity to docetaxel-based therapies remains a challenge
in mCRPC treatment. Methods: microRNA (miR)-451 expression was measured in docetaxel-treated prostate
cancer cells and tumor tissues by quantitative reverse-transcription
polymerase chain reaction . Cell-counting kit 8 assay was performed to
determine docetaxel chemoresistance. Neural-precursor-cell-expressed
developmentally downregulated protein 9 (NEDD9) was identified as a novel
target of miR-451 by dual-luciferase reporter system. Chromatin
immunoprecipitation and co-immunoprecipitation assay were performed to
confirm that histone deacetylase 3 (HDAC3)/Sp1 (a highly evolutionarily
conserved transcription factor) interacted with the Sp1 binding sites in
miR-451 promoter. Results: miR-451 was found to be silenced in docetaxel-treated prostate cancer cells
and mCRPC tissues. Low miR-451 expression was closely associated with a high
Gleason score, high Eastern Cooperative Oncology Group performance status
score, visceral metastasis and poor prognosis. Low expression of miR-451 was
significantly correlated with short progression-free survival (PFS) and
overall survival (OS) according to Kaplan–Meier analysis, and miR-451 was
determined to be an independent poor prognostic factor for PFS and OS in
mCRPC patients by univariate and multivariate Cox regression analyses. NEDD9
was identified as a new and functional target of miR-451. Restoration of
NEDD9 partially reversed the effects of miR-451 on enhancing
chemosensitivity of prostate cancer cells. HDAC3 was confirmed to be
involved in silencing of miR-451 expression in prostate cancer cells. Conclusions: The current data revealed a new HDAC3/Sp1/miR-451/NEDD9 signaling axis that
regulates the chemosensitivity of prostate cancer cells and represents a
novel therapeutic target for chemosensitizing mCRPC.
Collapse
Affiliation(s)
- Dong-Qin Chen
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Chen Yu
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Xue-Feng Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, USA
| | - Zhong-Fang Liu
- Department of Medical Oncology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Rui Wang
- Department of Medical Oncology, Nanjing University, Nanjing, China
| | - Min Jiang
- Department of Medical Oncology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hao Chen
- Department of Urology, the First Hospital of Jiaxing, Jiaxing, China
| | - Feng Yan
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Min Tao
- Department of Medical Oncology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Long-Bang Chen
- Department of Medical Oncology, Nanjing University, Nanjing, China
| | - Hong Zhu
- The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, China
| | - Ji-Feng Feng
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting Road, Nanjing 210009, China
| |
Collapse
|
27
|
Novel Insights Into E3 Ubiquitin Ligase in Cancer Chemoresistance. Am J Med Sci 2017; 355:368-376. [PMID: 29661351 DOI: 10.1016/j.amjms.2017.12.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 12/20/2017] [Accepted: 12/23/2017] [Indexed: 12/18/2022]
Abstract
Drug resistance can obstruct successful cancer chemotherapy. The ubiquitin-proteasome pathway has emerged as a crucial player that controls steady-state protein levels regulating multiple biological processes, such as cell cycle, cellular proliferation, apoptosis, and DNA damage response, which are involved in oncogenesis, cancer development, prognosis, and drug resistance. E3 ligases perform the final step in the ubiquitination cascade, and determine which protein becomes ubiquitylated by specifically binding the substrate protein. They are promising drug targets thanks to their ability to regulate protein stability and functions. Although patient survival has increased in recent years with the availability of novel agents, chemoresistance remains a major problem in cancer management. E3 ligases attract increasing attention with advances in chemoresistance knowledge. To explore the role of E3 ligase in cancer chemotherapy resistance and the underlying mechanism, we summarize the growing number of E3 ligases and their substrate proteins, which have emerged as crucial players in cancer chemoresistance and targeted therapies.
Collapse
|
28
|
Wu Z, Jing S, Li Y, Gao Y, Yu S, Li Z, Zhao Y, Piao J, Ma S, Chen X. The effects of SAHA on radiosensitivity in pancreatic cancer cells by inducing apoptosis and targeting RAD51. Biomed Pharmacother 2017; 89:705-710. [PMID: 28267674 DOI: 10.1016/j.biopha.2017.02.067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 01/23/2017] [Accepted: 02/15/2017] [Indexed: 12/20/2022] Open
Abstract
Suberoyl anilide hydroxamic acid (SAHA) is one of the most promising Histone deacetylases(HDAC) inhibitors which has shown significant anti-tumor activity for many malignancies. We explored the potential mechanism of the radiosensitivity effect of SAHA in Panc-1 cells and attempted to develop SAHA as a systemic treatment strategy for pancreatic cancer. Growth inhibition was detected by CCK-8 assay. Radiosensitizing enhancement ratio was determined by clonogenic assay. The cell cycle and apoptosis assay was detected using flow cytometry and annexin-V/PI. The level of Bax, Bcl-2, Ku70, Ku86, RAD51, RAD54 protein expression were detected using Western blot analysis. Gene silencing was processed by lentiviral vector and qRT-PCR was performed to detect mRNA expression. The results revealed that SAHA inhibited the proliferation of Panc-1 cells. SAHA enhanced the radiosensitivity with a sensitization enhancement ratio(SER) of 1.10 of the Panc-1 cells. SAHA induced G2-M phase arrest and apoptosis of Panc-1 cells with radiation. SAHA upregulated Bax and downregulated Bcl-2, Ku70, Ku86, RAD51, RAD54 protein expression of irradiated Panc-1 cells. SAHA enhanced the radiosensitivity of Panc-1 cells by modulating RAD51 expression. SAHA enhanced radiosensitivity to pancreatic carcinoma Panc-1 cells. It was associated with the G2-M phase arrest and apoptosis via modulation of Bax and Bcl-2 expression. Downregulation of Ku70, Ku86, RAD51 and RAD54 expression caused suppression of HR-mediated DNA repair. SAHA is a good radiosensitizer for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Zhibing Wu
- Center of Hyperthermia Oncology, Nanjing Medical University Affiliated Hangzhou Hospital (Hangzhou First People's Hospital), Hangzhou, Zhejiang 310006, China; Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, China; Key Laboratory of molecular oncology of Chinese medicine and Western medicine, Hangzhou, Zhejiang 310006, China.
| | - Saisai Jing
- Department of Oncology, Cixi People's Hospital, Cixi, Zhejiang 315300, China
| | - Yanhong Li
- Department of Oncology, Cixi People's Hospital, Cixi, Zhejiang 315300, China
| | - Yabo Gao
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, China
| | - Shuhuan Yu
- Center of Hyperthermia Oncology, Nanjing Medical University Affiliated Hangzhou Hospital (Hangzhou First People's Hospital), Hangzhou, Zhejiang 310006, China
| | - Zhitian Li
- Center of Hyperthermia Oncology, Nanjing Medical University Affiliated Hangzhou Hospital (Hangzhou First People's Hospital), Hangzhou, Zhejiang 310006, China
| | - Yanyan Zhao
- Key Laboratory of molecular oncology of Chinese medicine and Western medicine, Hangzhou, Zhejiang 310006, China
| | - Jigang Piao
- Key Laboratory of molecular oncology of Chinese medicine and Western medicine, Hangzhou, Zhejiang 310006, China
| | - Shenglin Ma
- Center of Hyperthermia Oncology, Nanjing Medical University Affiliated Hangzhou Hospital (Hangzhou First People's Hospital), Hangzhou, Zhejiang 310006, China; Key Laboratory of molecular oncology of Chinese medicine and Western medicine, Hangzhou, Zhejiang 310006, China
| | - Xufeng Chen
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
29
|
Sun S, Wang Y, Zhou R, Deng Z, Han Y, Han X, Tao W, Yang Z, Shi C, Hong D, Li J, Shi D, Zhang Z. Targeting and Regulating of an Oncogene via Nanovector Delivery of MicroRNA using Patient-Derived Xenografts. Am J Cancer Res 2017; 7:677-693. [PMID: 28255359 PMCID: PMC5327642 DOI: 10.7150/thno.16357] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 12/18/2016] [Indexed: 12/11/2022] Open
Abstract
In precision cancer nanomedicine, the key is to identify the oncogenes that are responsible for tumorigenesis, based on which these genetic drivers can be each specifically regulated by a nanovector-directed, oncogene-targeted microRNA (miRNA) for tumor suppression. Fibroblast Growth Factor Receptor 3 (FGFR3) is such an oncogene. The molecular tumor-subtype harboring FGFR3 genomic alteration has been identified via genomic sequencing and referred to as the FGFR3-driven tumors. This genomics-based tumor classification provides further rationale for the development of the FGFR3-targeted miRNA replacement therapy in treating patients with FGFR3 gene abnormity. However, successful miRNA therapy has been hampered by lacking of an efficient delivery vehicle. In this study, a nanovector is developed for microRNA-100 (miR-100) -mediated FGFR3 regulation. The nanovector is composed of the mesoporous magnetic clusters that are conjugated with ternary polymers for efficient miRNA in-vivo delivery. The miRNA-loading capacity of the nanovector is found to be high due to the polycation polymer functionalized mesoporous structure, showing excellent tumor cell transfection and pH-sensitive miRNA release. Delivery of miR-100 to cancer cells effectively down-regulates the expression of FGFR3, inhibits cell proliferation, and induces cell apoptosis in vitro. Patient-derived xenografts (PDXs) are used to evaluate the efficacy of miRNA delivery in the FGFR3-driven tumors. Notably, sharp contrasts are observed between the FGFR3-driven tumors and those without FGFR3 genomic alteration. Only the FGFR3-driven PDXs are significantly inhibited via miR-100 delivery while the non-FGFR3-driven PDXs are not affected, showing promise of precision cancer nanomedicine.
Collapse
|
30
|
Flores ER. Commentary on “Apoptosis, p53, and Tumor Cell Sensitivity to Anticancer Agents”. Cancer Res 2016; 76:6763-6764. [DOI: 10.1158/0008-5472.can-16-2997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/02/2016] [Indexed: 11/16/2022]
|
31
|
The p53 family orchestrates the regulation of metabolism: physiological regulation and implications for cancer therapy. Br J Cancer 2016; 116:149-155. [PMID: 27884017 PMCID: PMC5243983 DOI: 10.1038/bjc.2016.384] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/18/2016] [Accepted: 10/23/2016] [Indexed: 12/20/2022] Open
Abstract
The p53 family of transcription factors is essential to counteract tumour formation and progression. Although previously this was exclusively associated with the ability of the p53 family to induce cell cycle arrest and apoptosis, an increasing number of reports have now indisputably demonstrated that the tumour suppressive functions of the p53 family members also rely on their ability to control and regulate cellular metabolism and maintain cellular oxidative homeostasis. Here, we review how each p53 family member, including p63 and p73, controls metabolic pathways in physiological conditions, and how these mechanisms could be exploited to provide anticancer therapeutic opportunities.
Collapse
|
32
|
Carbone C, Di Gennaro E, Piro G, Milone MR, Pucci B, Caraglia M, Budillon A. Tissue transglutaminase (TG2) is involved in the resistance of cancer cells to the histone deacetylase (HDAC) inhibitor vorinostat. Amino Acids 2016; 49:517-528. [PMID: 27761756 DOI: 10.1007/s00726-016-2338-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 09/20/2016] [Indexed: 12/22/2022]
Abstract
Vorinostat demonstrated preclinical and clinical efficacy in human cancers and is the first histone deacetylase inhibitor (HDACi) approved for cancer treatment. Tissue transglutaminase (TG2) is a multifunctional enzyme that catalyzes a Ca2+ dependent transamidating reaction resulting in covalent cross-links between proteins. TG2 acts also as G-protein in trans-membrane signaling and as a cell surface adhesion mediator. TG2 up-regulation has been demonstrated in several cancers and its expression levels correlate with resistance to chemotherapy and metastatic potential. We demonstrated that the anti-proliferative effect of the HDACi vorinostat is paralleled by the induction of TG2 mRNA and protein expression in cancer cells but not in ex vivo treated peripheral blood lymphocytes. This effect was also shared by other pan-HDACi and resulted in increased TG2 transamidating activity. Notably, high TG2 basal levels in a panel of cancer cell lines correlated with lower vorinostat antiproliferative activity. Notably, in TG2-knockdown cancer cells vorinostat anti-proliferative and pro-apoptotic effects were enhanced, whereas in TG2-full-length transfected cells were impaired, suggesting that TG2 could represent a mechanism of intrinsic or acquired resistance to vorinostat. In fact, co-treatment of tumor cells with inhibitors of TG2 transamidating activity potentiated the antitumor effect of vorinostat. Moreover, vorinostat-resistant MCF7 cells selected by stepwise increasing concentrations of the drug, significantly overexpressed TG2 protein compared to parental cells, and co-treatment of these cells with TG2 inhibitors reversed vorinostat-resistance. Taken together, our data demonstrated that TG2 is involved in the resistance of cancer cells to vorinostat, as well as to other HDACi.
Collapse
Affiliation(s)
- Carmine Carbone
- Experimental Pharmacology Unit, Istituto Nazionale Tumori Fondazione G. Pascale-IRCCS, 80131, Naples, Italy.,Digestive Molecular Clinical Oncology Research Unit, Department of Medicine, University of Verona, Verona, Italy
| | - Elena Di Gennaro
- Experimental Pharmacology Unit, Istituto Nazionale Tumori Fondazione G. Pascale-IRCCS, 80131, Naples, Italy
| | - Geny Piro
- Centro Ricerche Oncologiche Mercogliano, Istituto Nazionale Tumori Fondazione G. Pascale-IRCCS, Naples, Italy.,Laboratory of Oncology and Molecular Therapy, Department of Medicine, University of Verona, Verona, Italy
| | - Maria Rita Milone
- Centro Ricerche Oncologiche Mercogliano, Istituto Nazionale Tumori Fondazione G. Pascale-IRCCS, Naples, Italy
| | - Biagio Pucci
- Centro Ricerche Oncologiche Mercogliano, Istituto Nazionale Tumori Fondazione G. Pascale-IRCCS, Naples, Italy
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Alfredo Budillon
- Experimental Pharmacology Unit, Istituto Nazionale Tumori Fondazione G. Pascale-IRCCS, 80131, Naples, Italy. .,Centro Ricerche Oncologiche Mercogliano, Istituto Nazionale Tumori Fondazione G. Pascale-IRCCS, Naples, Italy.
| |
Collapse
|