1
|
Liang Y, Liu J, Ge J, Shi Q, Zhang G, Wan A, Luo T, Tian H, Fan L, Wang S, Chen L, Tang P, Zhu K, Jiang J, Bian X, Zhang Y, Qi X. Safety and efficacy of anlotinib combined with taxane and lobaplatin in neoadjuvant treatment of clinical stage II/III triple-negative breast cancer in China (the neoALTAL trial): a single-arm, phase 2 trial. EClinicalMedicine 2024; 71:102585. [PMID: 38638401 PMCID: PMC11024570 DOI: 10.1016/j.eclinm.2024.102585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 04/20/2024] Open
Abstract
Background Anlotinib is a new type of tyrosine kinase inhibitor that targets vascular endothelial growth factor receptors 1/2/3, platelet-derived growth factor receptors α/β, and fibroblast growth factor receptors 1-4 and c-Kit, with a broad spectrum of inhibitory effects on tumor angiogenesis and growth. It has been proven effective in HER2-negative metastatic breast cancer, but its efficacy in early-stage triple-negative breast cancer (TNBC) is unknown. This phase 2 study aims to evaluate the efficacy and safety of adding anlotinib to neoadjuvant chemotherapy in patients with TNBC. Methods Patients with clinical stage II/III TNBC were treated with 5 cycles of anlotinib (12 mg, d1-14, q3w) plus 6 cycles of taxanes (docetaxel 75 mg/m2 ,d1, q3w or nab-paclitaxel 125 mg/m2, d1 and d8, q3w) and lobaplatin (30 mg/m2, d1, q3w), followed by surgery. The primary endpoint was pathological complete response (pCR; ypT0/is ypN0) and the secondary endpoints include breast pCR (bpCR), axillary pCR (apCR), residual cancer burden (RCB), objective response rate (ORR), survival, and safety. Exploratory endpoints were efficacy biomarkers based on Fudan University Shanghai Cancer Center Immunohistochemical (FUSCC IHC) classification for TNBC and next-generation sequencing (NGS) of DNA from tumor tissue and blood samples of patients with 425-gene panel. This trial is registered with www.chictr.org.cn (ChiCTR2100043027). Findings From Jan 2021 to Aug 2022, 48 patients were assessed and 45 were enrolled. All patients received at least one dose of study treatment and underwent surgery. The median age was 48.5 years (SD: 8.7), 71% were nodal involved, and 20% had stage III. In the intention-to-treat population, 26 out of 45 patients achieved pCR (57.8%; 90% CI, 44.5%-70.3%), and 39 achieved residual cancer burden class 0-I (86.7%; 95% CI, 73.2%-94.9%). The bpCR and apCR rate were 64.4% (29/45) and 71.9% (23/32), respectively. No recurrence or metastasis occurred during the short-term follow-up. Based on the FUSCC IHC-based subtypes, the pCR rates were 68.8% (11/16) for immunomodulatory subtype, 58.3% (7/12) for basal-like immune-suppressed subtype and 33.3% (4/12) for luminal androgen receptor subtype, respectively. NGS revealed that the pCR were 77% (10/13) and 50% (14/28) in MYC-amplified and wild-type patients, respectively, and 78% (7/9) and 53% (17/32) in gBRCA1/2-mutated and wild-type patients, respectively. The median follow-up time of the study was 14.9 months (95% CI: 13.5-16.3 months). There was no disease progression or death during neoadjuvant therapy. No deaths occurred during postoperative follow-up. In the safety population (N = 45), Grade 3 or 4 treatment emergent adverse events occurred in 29 patients (64%), and the most common events were neutropenia (38%), leukopenia (27%), thrombocytopenia (25%), anemia (13%), and hypertension (13%), respectively. Interpretation The addition of anlotinib to neoadjuvant chemotherapy showed manageable toxicity and encouraging antitumor activity for patients with clinical stage II/III TNBC. Funding Chongqing Talents Project, Chongqing Key Project of Technology Innovation and Application Development and Chongqing Outstanding Youth Natural Science Foundation.
Collapse
Affiliation(s)
- Yan Liang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, No. 30 Gaoyantan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Jing Liu
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, No. 30 Gaoyantan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Jia Ge
- Department of Pathology, Southwest Hospital, Army Medical University, No. 30 Gaoyantan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Qiyun Shi
- The Eighth Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Guozhi Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, No. 30 Gaoyantan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Andi Wan
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, No. 30 Gaoyantan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Tao Luo
- Department of Pathology, Southwest Hospital, Army Medical University, No. 30 Gaoyantan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Hao Tian
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, No. 30 Gaoyantan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Linjun Fan
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, No. 30 Gaoyantan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Shushu Wang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, No. 30 Gaoyantan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Li Chen
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, No. 30 Gaoyantan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Peng Tang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, No. 30 Gaoyantan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Kai Zhu
- Central Medical Center, Chia Tai Tianqing Pharmaceutical Group Co., Ltd. L., No.1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, People's Republic of China
| | - Jun Jiang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, No. 30 Gaoyantan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Xiuwu Bian
- Department of Pathology, Southwest Hospital, Army Medical University, No. 30 Gaoyantan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Yi Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, No. 30 Gaoyantan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, No. 30 Gaoyantan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| |
Collapse
|
2
|
Jari M, Abdoli S, Bazi Z, Shamsabadi FT, Roshanmehr F, Shahbazi M. Enhancing protein production and growth in chinese hamster ovary cells through miR-107 overexpression. AMB Express 2024; 14:16. [PMID: 38302631 PMCID: PMC10834913 DOI: 10.1186/s13568-024-01670-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 01/16/2024] [Indexed: 02/03/2024] Open
Abstract
Chinese Hamster Ovary (CHO) cells are widely employed as host cells for biopharmaceutical production. The manufacturing of biopharmaceuticals poses several challenges, including restricted growth potential and inadequate productivity of the host cells. MicroRNAs play a crucial role in regulating gene expression and are considered highly promising tools for cell engineering to enhance protein production. Our study aimed to evaluate the effects of miR-107, which is recognized as an onco-miR, on erythropoietin-producing CHO cells (CHO-hEPO). To assess the impact of miR-107 on CHO cells, a DNA plasmid containing miR-107 was introduced to CHO-hEPO cells through transfection. Cell proliferation and viability were assessed using the trypan blue dye exclusion method. Cell cycle analysis was conducted by utilizing propidium iodide (PI) staining. The quantification of EPO was determined using an immunoassay test. Moreover, the impact of miR-107 on the expression of downstream target genes was evaluated using qRT-PCR. Our findings highlight and underscore the substantial impact of transient miR-107 overexpression, which led to a remarkable 2.7-fold increase in EPO titers and a significant 1.6-fold increase in the specific productivity of CHO cells (p < 0.01). Furthermore, this intervention resulted in significant enhancements in cell viability and growth rate (p < 0.05). Intriguingly, the overexpression of miR‑107 was linked to the downregulation of LATS2, PTEN, and TSC1 genes while concurrently driving upregulation in transcript levels of MYC, YAP, mTOR, and S6K genes within transgenic CHO cells. In conclusion, this study collectively underscores the feasibility of utilizing cancer-associated miRNAs as a powerful tool for CHO cell engineering. However, more in-depth exploration is warranted to unravel the precise molecular intricacies of miR-107's effects in the context of CHO cells.
Collapse
Affiliation(s)
- Maryam Jari
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Shastkola Road, Falsafi Complex, Gorgan, Zip code: 4934174611, Iran
- Department of Medical Biotechnology School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Shahriyar Abdoli
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Shastkola Road, Falsafi Complex, Gorgan, Zip code: 4934174611, Iran
- Department of Medical Biotechnology School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Zahra Bazi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Shastkola Road, Falsafi Complex, Gorgan, Zip code: 4934174611, Iran
- Department of Medical Biotechnology School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Fatemeh Tash Shamsabadi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Shastkola Road, Falsafi Complex, Gorgan, Zip code: 4934174611, Iran
- Department of Medical Biotechnology School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Farnaz Roshanmehr
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Shastkola Road, Falsafi Complex, Gorgan, Zip code: 4934174611, Iran
- Department of Medical Biotechnology School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Shahbazi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Shastkola Road, Falsafi Complex, Gorgan, Zip code: 4934174611, Iran.
- AryaTina Gene (ATG) Biopharmaceutical Company Gorgan, Gorgan, Iran.
| |
Collapse
|
3
|
Ido M, Fujii K, Mishima H, Kubo A, Saito M, Banno H, Ito Y, Goto M, Ando T, Mouri Y, Kousaka J, Imai T, Nakano S. Comprehensive genomic evaluation of advanced and recurrent breast cancer patients for tailored precision treatments. BMC Cancer 2024; 24:85. [PMID: 38229073 DOI: 10.1186/s12885-023-11442-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 09/25/2023] [Indexed: 01/18/2024] Open
Abstract
AIM The aim of this study was to investigate genetic alterations within breast cancer in the setting of recurrent or de novo stage IV disease. PATIENTS AND METHODS This study included 22 patients with recurrent breast cancer (n = 19) and inoperable de novo stage IV breast cancer (n = 3). For next generation sequencing, FoundationOneCDx (F1CDx) (Foundation Medicine Inc., Cambridge, MA, USA) was performed in 21 patients and FoundationOneLiquid CDx was performed in 1 patient. RESULTS Median age was 62.9 years (range, 33.4-82.1). Pathological diagnoses of specimens included invasive ductal carcinoma (n = 19), invasive lobular carcinoma (n = 2), and invasive micropapillary carcinoma (n = 1). F1CDx detected a median of 4.5 variants (range, 1-11). The most commonly altered gene were PIK3CA (n = 9), followed by TP53 (n = 7), MYC (n = 4), PTEN (n = 3), and CDH1 (n = 3). For hormone receptor-positive patients with PIK3CA mutations, hormonal treatment plus a phosphoinositide 3-kinase inhibitor was recommended as the treatment of choice. Patients in the hormone receptor-negative and no human epidermal growth factor receptor 2 expression group had significantly higher tumor mutational burden than patients in the hormone receptor-positive group. A BRCA2 reversion mutation was revealed by F1CDx in a patient with a deleterious germline BRCA2 mutation during poly ADP ribose polymerase inhibitor treatment. CONCLUSION Guidance on tailored precision therapy with consideration of genomic mutations was possible for some patients with information provided by F1CDx. Clinicians should consider using F1CDx at turning points in the course of the disease.
Collapse
Affiliation(s)
- Mirai Ido
- Department of Surgery, Division of Breast and Endocrine Surgery, Aichi Medical University Hospital, 1-1 Yazakokarimata, Nagakute city, 480-1195, Japan
| | - Kimihito Fujii
- Department of Surgery, Division of Breast and Endocrine Surgery, Aichi Medical University Hospital, 1-1 Yazakokarimata, Nagakute city, 480-1195, Japan.
| | - Hideyuki Mishima
- Cancer Center, Aichi Medical University Hospital, Nagakute city, Japan
| | - Akihito Kubo
- Cancer Center, Aichi Medical University Hospital, Nagakute city, Japan
| | - Masayuki Saito
- Department of Surgery, Division of Breast and Endocrine Surgery, Aichi Medical University Hospital, 1-1 Yazakokarimata, Nagakute city, 480-1195, Japan
| | - Hirona Banno
- Department of Surgery, Division of Breast and Endocrine Surgery, Aichi Medical University Hospital, 1-1 Yazakokarimata, Nagakute city, 480-1195, Japan
| | - Yukie Ito
- Department of Surgery, Division of Breast and Endocrine Surgery, Aichi Medical University Hospital, 1-1 Yazakokarimata, Nagakute city, 480-1195, Japan
| | - Manami Goto
- Department of Surgery, Division of Breast and Endocrine Surgery, Aichi Medical University Hospital, 1-1 Yazakokarimata, Nagakute city, 480-1195, Japan
| | - Takahito Ando
- Department of Surgery, Division of Breast and Endocrine Surgery, Aichi Medical University Hospital, 1-1 Yazakokarimata, Nagakute city, 480-1195, Japan
| | - Yukako Mouri
- Department of Surgery, Division of Breast and Endocrine Surgery, Aichi Medical University Hospital, 1-1 Yazakokarimata, Nagakute city, 480-1195, Japan
| | - Junko Kousaka
- Department of Surgery, Division of Breast and Endocrine Surgery, Aichi Medical University Hospital, 1-1 Yazakokarimata, Nagakute city, 480-1195, Japan
| | - Tsuneo Imai
- Department of Surgery, Division of Breast and Endocrine Surgery, Aichi Medical University Hospital, 1-1 Yazakokarimata, Nagakute city, 480-1195, Japan
| | - Shogo Nakano
- Department of Surgery, Division of Breast and Endocrine Surgery, Aichi Medical University Hospital, 1-1 Yazakokarimata, Nagakute city, 480-1195, Japan
| |
Collapse
|
4
|
Hernandez YA, Gonzalez J, Garcia R, Aristizabal-Pachón A. The Expression of Hsa-Mir-1225-5p Limits the Aggressive Biological Behaviour of Luminal Breast Cancer Cell Lines. Microrna 2024; 13:124-131. [PMID: 38204280 PMCID: PMC11348466 DOI: 10.2174/0122115366268128231201054005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/24/2023] [Accepted: 10/16/2023] [Indexed: 01/12/2024]
Abstract
INTRODUCTION Numerous genetic and biological processes have been linked to the function of microRNAs (miRNAs), which regulate gene expression by targeting messenger RNA (mRNA). It is commonly acknowledged that miRNAs play a role in the development of disease and the embryology of mammals. METHOD To further understand its function in the oncogenic process, the expression of the miRNA profile in cancer has been investigated. Despite being referred to as a noteworthy miRNA in cancer, it is unknown whether hsa-miR-1225-5p plays a part in the in vitro progression of the luminal A and luminal B subtypes of breast cancer. We proposed that a synthetic hsa-miR-1225-5p molecule be expressed in breast cancer cell lines and its activity be evaluated with the aim of studying its function in the development of luminal breast cancer. In terms of the typical cancer progression stages, such as proliferation, survival, migration, and invasion, we investigated the role of hsa-miR-1225-5p in luminal A and B breast cancer cell lines. RESULTS Additionally, using bioinformatics databases, we thoroughly explored the target score-based prediction of miRNA-mRNA interaction. Our study showed that the expression of miR-1225-5p significantly inhibited the in vitro growth of luminal A and B breast cancer cell lines. CONCLUSION The results were supported by a bioinformatic analysis and a detailed gene network that boosts the activation of signaling pathways required for cancer progression.
Collapse
Affiliation(s)
- Y-Andrés Hernandez
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Janeth Gonzalez
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Reggie Garcia
- Departamento de Ciencias Fisiológicas, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Andrés Aristizabal-Pachón
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| |
Collapse
|
5
|
Ahmad RM, Ali BR, Al-Jasmi F, Sinnott RO, Al Dhaheri N, Mohamad MS. A review of genetic variant databases and machine learning tools for predicting the pathogenicity of breast cancer. Brief Bioinform 2023; 25:bbad479. [PMID: 38149678 PMCID: PMC10782903 DOI: 10.1093/bib/bbad479] [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: 06/22/2023] [Revised: 09/22/2023] [Accepted: 12/04/2023] [Indexed: 12/28/2023] Open
Abstract
Studies continue to uncover contributing risk factors for breast cancer (BC) development including genetic variants. Advances in machine learning and big data generated from genetic sequencing can now be used for predicting BC pathogenicity. However, it is unclear which tool developed for pathogenicity prediction is most suited for predicting the impact and pathogenicity of variant effects. A significant challenge is to determine the most suitable data source for each tool since different tools can yield different prediction results with different data inputs. To this end, this work reviews genetic variant databases and tools used specifically for the prediction of BC pathogenicity. We provide a description of existing genetic variants databases and, where appropriate, the diseases for which they have been established. Through example, we illustrate how they can be used for prediction of BC pathogenicity and discuss their associated advantages and disadvantages. We conclude that the tools that are specialized by training on multiple diverse datasets from different databases for the same disease have enhanced accuracy and specificity and are thereby more helpful to the clinicians in predicting and diagnosing BC as early as possible.
Collapse
Affiliation(s)
- Rahaf M Ahmad
- Health Data Science Lab, Department of Genetics and Genomics, College of Medical and Health Sciences, United Arab Emirates University, Tawam road, Al Maqam district, Al Ain, Abu Dhabi, United Arab Emirates
| | - Bassam R Ali
- Health Data Science Lab, Department of Genetics and Genomics, College of Medical and Health Sciences, United Arab Emirates University, Tawam road, Al Maqam district, Al Ain, Abu Dhabi, United Arab Emirates
| | - Fatma Al-Jasmi
- Health Data Science Lab, Department of Genetics and Genomics, College of Medical and Health Sciences, United Arab Emirates University, Tawam road, Al Maqam district, Al Ain, Abu Dhabi, United Arab Emirates
- Division of Metabolic Genetics, Department of Pediatrics, Tawam Hospital, Al Ain, United Arab Emirates
| | - Richard O Sinnott
- School of Computing and Information System, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Noura Al Dhaheri
- Health Data Science Lab, Department of Genetics and Genomics, College of Medical and Health Sciences, United Arab Emirates University, Tawam road, Al Maqam district, Al Ain, Abu Dhabi, United Arab Emirates
- Division of Metabolic Genetics, Department of Pediatrics, Tawam Hospital, Al Ain, United Arab Emirates
| | - Mohd Saberi Mohamad
- Health Data Science Lab, Department of Genetics and Genomics, College of Medical and Health Sciences, United Arab Emirates University, Tawam road, Al Maqam district, Al Ain, Abu Dhabi, United Arab Emirates
| |
Collapse
|
6
|
Liu D, Peng Y, Li X, Zhu Z, Mi Z, Zhang Z, Fan H. Comprehensive landscape of TGFβ-related signature in osteosarcoma for predicting prognosis, immune characteristics, and therapeutic response. J Bone Oncol 2023; 40:100484. [PMID: 37234254 PMCID: PMC10205544 DOI: 10.1016/j.jbo.2023.100484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
Osteosarcoma (OS) is a highly heterogeneous malignant bone tumor, and its tendency to metastasize leads to a poor prognosis. TGFβ is an important regulator in the tumor microenvironment and is closely associated with the progression of various types of cancer. However, the role of TGFβ-related genes in OS is still unclear. In this study, we identified 82 TGFβ DEGs based on RNA-seq data from the TARGET and GETx databases and classified OS patients into two TGFβ subtypes. The KM curve showed that the Cluster 2 patients had a substantially poorer prognosis than the Cluster 1 patients. Subsequently, a novel TGFβ prognostic signatures (MYC and BMP8B) were developed based on the results of univariate, LASSO, and multifactorial Cox analyses. These signatures showed robust and reliable predictive performance for the prognosis of OS in the training and validation cohorts. To predict the three-year and five-year survival rate of OS, a nomogram that integrated clinical features and risk scores was also developed. The GSEA analysis showed that the different subgroups analyzed had distinct functions, particularly, the low-risk group was associated with high immune activity and a high infiltration abundance of CD8 T cells. Moreover, our results indicated that low-risk cases had higher sensitivity to immunotherapy, while high-risk cases were more sensitive to sorafenib and axitinib. scRNA-Seq analysis further revealed that MYC and BMP8B were strongly expressed mainly in tumor stromal cells. Finally, in this study, we confirmed the expression of MYC and BMP8B by performing qPCR, WB, and IHC analyses. To conclude, we developed and validated a TGFβ-related signature to accurately predict the prognosis of OS. Our findings might contribute to personalized treatment and making better clinical decisions for OS patients.
Collapse
Affiliation(s)
- Dong Liu
- Department of Orthopaedic Surgery, Xi-jing Hospital, The Fourth Military Medical University, Xi’an 710032, China
| | - Ye Peng
- Department of Orthopaedics, Air Force Medical Center, PLA, Beijing 100142, China
| | - Xian Li
- Department of Orthopaedics, Shenzhen University General Hospital, Shenzhen, China
| | - Zhijie Zhu
- Department of Orthopaedic Surgery, Xi-jing Hospital, The Fourth Military Medical University, Xi’an 710032, China
| | - Zhenzhou Mi
- Department of Orthopaedic Surgery, Xi-jing Hospital, The Fourth Military Medical University, Xi’an 710032, China
| | - Zhao Zhang
- Department of Orthopaedic Surgery, Xi-jing Hospital, The Fourth Military Medical University, Xi’an 710032, China
| | - Hongbin Fan
- Department of Orthopaedic Surgery, Xi-jing Hospital, The Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
7
|
Bloom SMK, O’Hare N, Forbes NS. Bacterial delivery of therapeutic proteins to the nuclei of cancer cells. Biotechnol Bioeng 2023; 120:1437-1448. [PMID: 36710503 PMCID: PMC10101893 DOI: 10.1002/bit.28340] [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: 10/29/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023]
Abstract
Targeting nucleic targets with therapeutic proteins would enhance the treatment of hard-to-treat cancers. However, exogenous proteins are excluded from the nucleus by both the cellular and nuclear membranes. We have recently developed Salmonella that deliver active proteins into the cytoplasm of cancer cells. Here, we hypothesized that bacterially delivered proteins accumulate within nuclei, nuclear localization sequences (NLSs) increase delivery, and bacterially delivered proteins kill cancer cells. To test this hypothesis, we developed intranuclear delivering (IND) Salmonella and quantified the delivery of three model proteins. IND Salmonella delivered both ovalbumin and green fluorescent protein to nuclei of MCF7 cancer cells. The amount of protein in nuclei was linearly dependent on the amount delivered to the cytoplasm. The addition of a NLSs increased both the amount of protein in each nucleus and the number of nuclei that received protein. Delivery of Omomyc, a protein inhibitor of the nuclear transcript factor, Myc, altered cell physiology, and significantly induced cell death. These results show that IND Salmonella deliver functional proteins to the nucleus of cancerous cells. Extending this method to other transcription factors will increase the number of accessible targets for cancer therapy.
Collapse
Affiliation(s)
| | - Nicholas O’Hare
- Department of Chemical Engineering, University of Massachusetts, Amherst
| | - Neil S. Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst
- Institute for Applied Life Sciences, University of Massachusetts, Amherst
| |
Collapse
|
8
|
Wedam R, Greer YE, Wisniewski DJ, Weltz S, Kundu M, Voeller D, Lipkowitz S. Targeting Mitochondria with ClpP Agonists as a Novel Therapeutic Opportunity in Breast Cancer. Cancers (Basel) 2023; 15:cancers15071936. [PMID: 37046596 PMCID: PMC10093243 DOI: 10.3390/cancers15071936] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Breast cancer is the most frequently diagnosed malignancy worldwide and the leading cause of cancer mortality in women. Despite the recent development of new therapeutics including targeted therapies and immunotherapy, triple-negative breast cancer remains an aggressive form of breast cancer, and thus improved treatments are needed. In recent decades, it has become increasingly clear that breast cancers harbor metabolic plasticity that is controlled by mitochondria. A myriad of studies provide evidence that mitochondria are essential to breast cancer progression. Mitochondria in breast cancers are widely reprogrammed to enhance energy production and biosynthesis of macromolecules required for tumor growth. In this review, we will discuss the current understanding of mitochondrial roles in breast cancers and elucidate why mitochondria are a rational therapeutic target. We will then outline the status of the use of mitochondria-targeting drugs in breast cancers, and highlight ClpP agonists as emerging mitochondria-targeting drugs with a unique mechanism of action. We also illustrate possible drug combination strategies and challenges in the future breast cancer clinic.
Collapse
Affiliation(s)
- Rohan Wedam
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yoshimi Endo Greer
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David J Wisniewski
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sarah Weltz
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Manjari Kundu
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Donna Voeller
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stanley Lipkowitz
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
9
|
Toyokuni S, Kong Y, Motooka Y, Akatsuka S. Environmental impact on carcinogenesis under BRCA1 haploinsufficiency. Genes Environ 2023; 45:2. [PMID: 36639692 PMCID: PMC9837898 DOI: 10.1186/s41021-023-00258-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/06/2023] [Indexed: 01/15/2023] Open
Abstract
Cancer is the primary cause of human mortality in Japan since 1981. Although numerous novel therapies have been developed and applied in clinics, the number of deaths from cancer is still increasing worldwide. It is time to consider the strategy of cancer prevention more seriously. Here we propose a hypothesis that cancer can be side effects of long time-use of iron and oxygen and that carcinogenesis is an evolution-like cellular events to obtain "iron addiction with ferroptosis-resistance" where genes and environment interact each other. Among the recognized genetic risk factors for carcinogenesis, we here focus on BRCA1 tumor suppressor gene and how environmental factors, including daily life exposure and diets, may impact toward carcinogenesis under BRCA1 haploinsufficiency. Although mice models of BRCA1 mutants have not been successful for decades in generating phenotype mimicking the human counterparts, a rat model of BRCA1 mutant was recently established that reasonably mimics the human phenotype. Two distinct categories of oxidative stress, one by radiation and one by iron-catalyzed Fenton reaction, promoted carcinogenesis in Brca1 rat mutants. Furthermore, mitochondrial damage followed by alteration of iron metabolism finally resulted in ferroptosis-resistance of target cells in carcinogenesis. These suggest a possibility that cancer prevention by active pharmacological intervention may be possible for BRCA1 mutants to increase the quality of their life rather than preventive mastectomy and/or oophorectomy.
Collapse
Affiliation(s)
- Shinya Toyokuni
- grid.27476.300000 0001 0943 978XDepartment of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550 Japan ,grid.27476.300000 0001 0943 978XCenter for Low-Temperature Plasma Sciences, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, 464-8603 Japan
| | - Yingyi Kong
- grid.27476.300000 0001 0943 978XDepartment of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550 Japan
| | - Yashiro Motooka
- grid.27476.300000 0001 0943 978XDepartment of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550 Japan
| | - Shinya Akatsuka
- grid.27476.300000 0001 0943 978XDepartment of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550 Japan
| |
Collapse
|
10
|
Lau TY, Kwan HY. Fucoxanthin Is a Potential Therapeutic Agent for the Treatment of Breast Cancer. Mar Drugs 2022; 20:md20060370. [PMID: 35736173 PMCID: PMC9229252 DOI: 10.3390/md20060370] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/18/2022] [Accepted: 05/24/2022] [Indexed: 12/04/2022] Open
Abstract
Breast cancer (BC) is one of the most common cancers diagnosed and the leading cause of cancer-related death in women. Although there are first-line treatments for BC, drug resistances and adverse events have been reported. Given the incidence of BC keeps increasing, seeking novel therapeutics is urgently needed. Fucoxanthin (Fx) is a dietary carotenoid commonly found in seaweeds and diatoms. Both in vitro and in vivo studies show that Fx and its deacetylated metabolite fucoxanthinol (Fxol) inhibit and prevent BC growth. The NF-κB signaling pathway is considered the major pathway contributing to the anti-proliferation, anti-angiogenesis and pro-apoptotic effects of Fx and Fxol. Other signaling molecules such as MAPK, MMP2/9, CYP and ROS are also involved in the anti-cancer effects by regulating the tumor microenvironment, cancer metastasis, carcinogen metabolism and oxidation. Besides, Fx also possesses anti-obesity effects by regulating UCP1 levels and lipid metabolism, which may help to reduce BC risk. More importantly, mounting evidence demonstrates that Fx overcomes drug resistance. This review aims to give an updated summary of the anti-cancer effects of Fx and summarize the underlying mechanisms of action, which will provide novel strategies for the development of Fx as an anti-cancer therapeutic agent.
Collapse
|
11
|
Kong Y, Akatsuka S, Motooka Y, Zheng H, Cheng Z, Shiraki Y, Mashimo T, Imaoka T, Toyokuni S. BRCA1 haploinsufficiency promotes chromosomal amplification under fenton reaction-based carcinogenesis through ferroptosis-resistance. Redox Biol 2022; 54:102356. [PMID: 35667247 PMCID: PMC9168618 DOI: 10.1016/j.redox.2022.102356] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 05/25/2022] [Indexed: 12/16/2022] Open
Abstract
Germline-mutation in BRCA1 tumor suppressor gene is an established risk for carcinogenesis not only in females but also in males. Deficiency in the repair of DNA double-strand breaks is hypothesized as a responsible mechanism for carcinogenesis. However, supporting data is insufficient both in the mutation spectra of cancers in the patients with BRCA1 germline-mutation and in murine knockout/knock-in models of Brca1 haploinsufficiency. Furthermore, information on the driving force toward carcinogenesis in BRCA1 mutation carriers is lacking. Here we applied Fenton reaction-based renal carcinogenesis to a rat heterozygously knockout model of BRCA1 haploinsufficiency (mutant [MUT] model; L63X/+). Rat MUT model revealed significant promotion of renal cell carcinoma (RCC) induced by ferric nitrilotriacetate (Fe-NTA). Array-based comparative genome hybridization of the RCCs identified significant increase in chromosomal amplification, syntenic to those in breast cancers of BRCA1 mutation carriers, including c-Myc, in comparison to those in the wild-type. Subacute-phase analysis of the kidney after repeated Fe-NTA treatment in the MUT model revealed dysregulated iron metabolism with mitochondrial malfunction assessed by expression microarray and electron microscopy, leading to renal tubular proliferation with iron overload. In conclusion, we for the first time demonstrate that biallelic wild-type BRCA1 provides more robust protection for mitochondrial metabolism under iron-catalyzed oxidative stress, preventing the emergence of neoplastic cells with chromosomal amplification. Our results suggest that oxidative stress via excess iron is a major driving force for carcinogenesis in BRCA1 haploinsufficiency, which can be a target for cancer prevention and therapeutics. Rat BRCA1 haploinsufficiency promoted Fenton reaction-based renal carcinogenesis. BRCA1 haploinsufficiency allowed chromosomal amplification under excess iron. BRCA1 haploinsufficiency caused more mitochondrial damage with ferroptosis resistance.
Collapse
|
12
|
Gomarasca M, Lombardi G, Maroni P. SUMOylation and NEDDylation in Primary and Metastatic Cancers to Bone. Front Cell Dev Biol 2022; 10:889002. [PMID: 35465332 PMCID: PMC9020829 DOI: 10.3389/fcell.2022.889002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/25/2022] [Indexed: 12/22/2022] Open
Abstract
Post-translational modifications comprise series of enzymatically-driven chemical modifications, virtually involving the entire cell proteome, that affect the fate of a target protein and, in turn, cell activity. Different classes of modifications can be established ranging from phosphorylation, glycosylation, ubiquitination, acetylation, methylation, lipidation and their inverse reactions. Among these, SUMOylation and NEDDylation are ubiquitin-like multi-enzymatic processes that determine the bound of SUMOs and NEDD8 labels, respectively, on defined amino acidic residues of a specific protein and regulate protein function. As fate-determinants of several effectors and mediators, SUMOylation and NEDDylation play relevant roles in many aspects of tumor cell biology. Bone represents a preferential site of metastasis for solid tumors (e.g., breast and prostate cancers) and the primary site of primitive tumors (e.g., osteosarcoma, chondrosarcoma). Deregulation of SUMOylation and NEDDylation affects different aspects of neoplastic transformation and evolution such as epithelial-mesenchymal transition, adaptation to hypoxia, expression and action of tumor suppressors and oncogenic mediators, and drug resistance. Thereby, they represent potential therapeutic targets. This narrative review aims at describing the involvement and regulation of SUMOylation and NEDDylation in tumor biology, with a specific focus on primary and secondary bone tumors, and to summarize and highlight their potentiality in diagnostics and therapeutic strategies.
Collapse
Affiliation(s)
- Marta Gomarasca
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Polska
- *Correspondence: Giovanni Lombardi,
| | - Paola Maroni
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| |
Collapse
|
13
|
Hanasoge Somasundara AV, Moss MA, Feigman MJ, Chen C, Cyrill SL, Ciccone MF, Trousdell MC, Vollbrecht M, Li S, Kendall J, Beyaz S, Wilkinson JE, Dos Santos CO. Parity-induced changes to mammary epithelial cells control NKT cell expansion and mammary oncogenesis. Cell Rep 2021; 37:110099. [PMID: 34879282 PMCID: PMC8719356 DOI: 10.1016/j.celrep.2021.110099] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/25/2021] [Accepted: 11/15/2021] [Indexed: 12/19/2022] Open
Abstract
Pregnancy reprograms mammary epithelial cells (MECs) to control their responses to pregnancy hormone re-exposure and carcinoma progression. However, the influence of pregnancy on the mammary microenvironment is less clear. Here, we used single-cell RNA sequencing to profile the composition of epithelial and non-epithelial cells in mammary tissue from nulliparous and parous female mice. Our analysis indicates an expansion of γδ natural killer T-like immune cells (NKTs) following pregnancy and upregulation of immune signaling molecules in post-pregnancy MECs. We show that expansion of NKTs following pregnancy is due to elevated expression of the antigen-presenting molecule CD1d on MECs. Loss of CD1d expression on post-pregnancy MECs, or overall lack of activated NKTs, results in mammary oncogenesis. Collectively, our findings illustrate how pregnancy-induced changes modulate the communication between MECs and the immune microenvironment and establish a causal link between pregnancy, the immune microenvironment, and mammary oncogenesis.
Collapse
MESH Headings
- Animals
- Antigens, CD1d/metabolism
- Cell Communication
- Cell Proliferation
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Epithelial Cells/immunology
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Female
- Gene Expression Regulation, Neoplastic
- Genes, BRCA1
- Genes, myc
- Lymphocyte Activation
- Mammary Glands, Animal/immunology
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice, Inbred BALB C
- Mice, Inbred NOD
- Mice, SCID
- Mice, Transgenic
- Natural Killer T-Cells/immunology
- Natural Killer T-Cells/metabolism
- Parity
- Pregnancy
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Signal Transduction
- Tumor Microenvironment
- Mice
Collapse
Affiliation(s)
| | - Matthew A Moss
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Mary J Feigman
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Chen Chen
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | | | | | - Macy Vollbrecht
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Siran Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Jude Kendall
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Semir Beyaz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - John E Wilkinson
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
14
|
Wang S, Zhang H, Liu H, Guo X, Ma R, Zhu W, Gao P. ELK1-induced up-regulation of KIF26B promotes cell cycle progression in breast cancer. Med Oncol 2021; 39:15. [PMID: 34817735 DOI: 10.1007/s12032-021-01607-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 11/01/2021] [Indexed: 10/19/2022]
Abstract
KIF26B is a member of the kinesin superfamily that is up-regulated in various tumors, including breast cancer (BC), which can promote tumor progression. This study aimed to investigate the potential function of KIF26B in BC, and the underlying mechanisms, focusing mainly on cell proliferation. KIF26B expression was examined in BC tissue samples obtained from 99 patients. Then, we performed MTS, EdU and flow cytometry assays to detect cell proliferation, and western blotting to measure the expression of cell cycle-related proteins in MDA-MB-231 and MDA-MB-468 cells following KIF26B knockdown. Promoter analysis was used to study the upstream regulatory mechanism of KIF26B. KIF26B was upregulated in BC tissues. High expression of KIF26B was associated with clinicopathological parameters, such as positive lymph node metastasis, higher tumor grade, and higher proliferative index in BC. Furthermore, knockdown of KIF26B expression inhibited MDA-MB-231 and MDA-MB-468 cell proliferation, arresting cells in the G1 phase of the cell cycle in vitro. Similarly, KIF26B silencing decreased the expression levels of Wnt, β-catenin, and cell cycle-related proteins such as c-Myc, cyclin D1, and cyclin-dependent kinase 4, while increasing the expression of p27. Moreover, ELK1 could bind to the core promoter region of KIF26B and activate its transcription. KIF26B acts as an oncogene in BC by regulating multiple proteins involved in the cell cycle. ELK1 activates KIF26B transcription.
Collapse
Affiliation(s)
- SuXia Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan Wen Hua Xi Road 44, Jinan, 250012, Shandong, China.,Department of Pathology, Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China.,Department of Pathology, Qilu Hospital, Shandong University Jinan, Shandong, 250012, China
| | - Hui Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan Wen Hua Xi Road 44, Jinan, 250012, Shandong, China.,Department of Pathology, Qilu Hospital, Shandong University Jinan, Shandong, 250012, China
| | - HaiTing Liu
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan Wen Hua Xi Road 44, Jinan, 250012, Shandong, China.,Department of Pathology, Qilu Hospital, Shandong University Jinan, Shandong, 250012, China
| | - XiangYu Guo
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan Wen Hua Xi Road 44, Jinan, 250012, Shandong, China
| | - RanRan Ma
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan Wen Hua Xi Road 44, Jinan, 250012, Shandong, China.,Department of Pathology, Qilu Hospital, Shandong University Jinan, Shandong, 250012, China
| | - WenJie Zhu
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan Wen Hua Xi Road 44, Jinan, 250012, Shandong, China.
| | - P Gao
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan Wen Hua Xi Road 44, Jinan, 250012, Shandong, China. .,Department of Pathology, Qilu Hospital, Shandong University Jinan, Shandong, 250012, China.
| |
Collapse
|
15
|
Tibatan MA, Sarısaman M. Unitary structure of palindromes in DNA. Biosystems 2021; 211:104565. [PMID: 34740704 DOI: 10.1016/j.biosystems.2021.104565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/20/2021] [Accepted: 10/20/2021] [Indexed: 12/13/2022]
Abstract
We investigate the quantum behavior encountered in palindromes within DNA structure. In particular, we reveal the unitary structure of usual palindromic sequences found in genomic DNAs of all living organisms, using the Schwinger's approach. We clearly demonstrate the role played by palindromic configurations with special emphasis on physical symmetries, in particular subsymmetries of unitary structure. We unveil the prominence of unitary structure in palindromic sequences in the sense that vitally significant information endowed within DNA could be transformed unchangeably in the process of transcription. We introduce a new symmetry relation, namely purine-purine or pyrimidine-pyrimidine symmetries (p-symmetry) in addition to the already known symmetry relation of purine-pyrimidine symmetries (pp-symmetry) given by Chargaff's rule. Therefore, important vital functions of a living organisms are protected by means of these symmetric features. It is understood that higher order palindromic sequences could be generated in terms of the basis of the highest prime numbers that make up the palindrome sequence number. We propose that violation of this unitary structure of palindromic sequences by means of our proposed symmetries leads to a mutation in DNA, which could offer a new perspective in the scientific studies on the origin and cause of mutation.
Collapse
Affiliation(s)
- Mehmet Ali Tibatan
- Department of Biotechnology, Istanbul University, 34134, Vezneciler, Istanbul, Turkey.
| | - Mustafa Sarısaman
- Department of Physics, Istanbul University, 34134, Vezneciler, Istanbul, Turkey.
| |
Collapse
|
16
|
The genomic architecture of metastasis in breast cancer: focus on mechanistic aspects, signalling pathways and therapeutic strategies. Med Oncol 2021; 38:95. [PMID: 34268641 DOI: 10.1007/s12032-021-01547-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 07/03/2021] [Indexed: 12/13/2022]
Abstract
Breast cancer is a multifactorial, heterogeneous disease and the second most frequent cancer amongst women worldwide. Metastasis is one of the most leading causes of death in these patients. Early-stage or locally advanced breast cancer is limited to the breast or nearby lymph nodes. When breast cancer spreads to farther tissues/organs from its original site, it is referred to as metastatic or stage IV breast cancer. Normal breast development is regulated by specific genes and signalling pathways controlling cell proliferation, cell death, cell differentiation and cell motility. Dysregulation of genes involved in various signalling pathways not only leads to the formation of primary tumour but also to the metastasis as well. The metastatic cascade is represented by a multi-step process including invasion of the local tumour cell followed by its entry into the vasculature, exit of malignant cells from the circulation and ultimately their colonization at the distant sites. These stages are referred to as formation of primary tumour, angiogenesis, invasion, intravasation and extravasation, respectively. The major sites of metastasis of breast cancer are the lymph nodes, bone, brain and lung. Only about 28% five-year survival rate has been reported for stage IV breast cancer. Metastasis is a serious concern for breast cancer and therefore, various therapeutic strategies such as tyrosine kinase inhibitors have been developed to target specific dysregulated genes and various signalling pathways involved in different steps of metastasis. In addition, other therapies like hyperbaric oxygen therapy, RNA interference and CRISPR/Cas9 are also being explored as novel strategies to cure the stage IV/metastatic breast cancer. Therefore, the current review has been compiled with an aim to evaluate the genetic basis of stage IV breast cancer with a focus on the molecular mechanisms. In addition, the therapeutic strategies targeting these dysregulated genes involved in various signalling pathways have also been discussed. Genome editing technologies that can target specific genes in the affected areas by making knock-in and knock-out alternations and thereby bring significant treatment outcomes in breast cancer have also been summarized.
Collapse
|
17
|
Zhao C, Tang C, Li C, Ning W, Hu Z, Xin L, Zhou HB, Huang J. Novel hybrid conjugates with dual estrogen receptor α degradation and histone deacetylase inhibitory activities for breast cancer therapy. Bioorg Med Chem 2021; 40:116185. [PMID: 33965842 DOI: 10.1016/j.bmc.2021.116185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/17/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023]
Abstract
Hormone therapy targeting estrogen receptors is widely used clinically for the treatment of breast cancer, such as tamoxifen, but most of them are partial agonists, which can cause serious side effects after long-term use. The use of selective estrogen receptor down-regulators (SERDs) may be an effective alternative to breast cancer therapy by directly degrading ERα protein to shut down ERα signaling. However, the solely clinically used SERD fulvestrant, is low orally bioavailable and requires intravenous injection, which severely limits its clinical application. On the other hand, double- or multi-target conjugates, which are able to synergize antitumor activity by different pathways, thus may enhance therapeutic effect in comparison with single targeted therapy. In this study, we designed and synthesized a series of novel dual-functional conjugates targeting both ERα degradation and histone deacetylase inhibiton by combining a privileged SERD skeleton 7-oxabicyclo[2.2.1]heptane sulfonamide (OBHSA) with a histone deacetylase inhibitor side chain. We found that substituents on both the sulfonamide nitrogen and phenyl group of OBHSA unit had significant effect on biological activities. Among them, conjugate 16i with N-methyl and naphthyl groups exhibited potent antiproliferative activity against MCF-7 cells, and excellent ERα degradation activity and HDACs inhibitory ability. A further molecular docking study indicated the interaction patterns of these conjugates with ERα, which may provide guidance to design novel SERDs or PROTAC-like SERDs for breast cancer therapy.
Collapse
Affiliation(s)
- Chenxi Zhao
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Chu Tang
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Frontier Science Center for Immunology and Metabolism, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China
| | - Changhao Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Wentao Ning
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Frontier Science Center for Immunology and Metabolism, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China
| | - Zhiye Hu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Frontier Science Center for Immunology and Metabolism, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China
| | - Lilan Xin
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Frontier Science Center for Immunology and Metabolism, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China
| | - Hai-Bing Zhou
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Frontier Science Center for Immunology and Metabolism, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China.
| | - Jian Huang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
18
|
Ghozlan H, Showalter A, Lee E, Zhu X, Khaled AR. Chaperonin-Containing TCP1 Complex (CCT) Promotes Breast Cancer Growth Through Correlations With Key Cell Cycle Regulators. Front Oncol 2021; 11:663877. [PMID: 33996588 PMCID: PMC8121004 DOI: 10.3389/fonc.2021.663877] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Uncontrolled proliferation as a result of dysregulated cell cycling is one of the hallmarks of cancer. Therapeutically targeting pathways that control the cell cycle would improve patient outcomes. However, the development of drug resistance and a limited number of inhibitors that target multiple cell cycle modulators are challenges that impede stopping the deregulated growth that leads to malignancy. To advance the discovery of new druggable targets for cell cycle inhibition, we investigated the role of Chaperonin-Containing TCP1 (CCT or TRiC) in breast cancer cells. CCT, a type II chaperonin, is a multi-subunit protein-folding complex that interacts with many oncoproteins and mutant tumor suppressors. CCT subunits are highly expressed in a number of cancers, including breast cancer. We found that expression of one of the CCT subunits, CCT2, inversely correlates with breast cancer patient survival and is subject to copy number alterations through genomic amplification. To investigate a role for CCT2 in the regulation of the cell cycle, we expressed an exogenous CCT2-FLAG construct in T47D and MCF7 luminal A breast cancer cells and examined cell proliferation under conditions of two-dimensional (2D) monolayer and three-dimensional (3D) spheroid cultures. Exogenous CCT2 increased the proliferation of cancer cells, resulting in larger and multiple spheroids as compared to control cells. CCT2-expressing cells were also able to undergo spheroid growth reversal, re-attaching, and resuming growth in 2D cultures. Such cells gained anchorage-independent growth. CCT2 expression in cells correlated with increased expression of MYC, especially in spheroid cultures, and other cell cycle regulators like CCND1 and CDK2, indicative of a novel activity that could contribute to the increase in cell growth. Statistically significant correlations between CCT2, MYC, and CCND1 were shown. Since CCT2 is located on chromosome 12q15, an amplicon frequently found in soft tissue cancers as well as breast cancer, CCT2 may have the basic characteristics of an oncogene. Our findings suggest that CCT2 could be an essential driver of cell division that may be a node through which pathways involving MYC, cyclin D1 and other proliferative factors could converge. Hence the therapeutic inhibition of CCT2 may have the potential to achieve multi-target inhibition, overcoming the limitations associated with single agent inhibitors.
Collapse
Affiliation(s)
- Heba Ghozlan
- Division of Cancer Research, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Adrian Showalter
- Division of Cancer Research, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Eunkyung Lee
- Department of Health Sciences, College of Health Professions and Sciences, University of Central Florida, Orlando, FL, United States
| | - Xiang Zhu
- Division of Cancer Research, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Annette R Khaled
- Division of Cancer Research, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| |
Collapse
|
19
|
Actin-like protein 6A/MYC/CDK2 axis confers high proliferative activity in triple-negative breast cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:56. [PMID: 33541412 PMCID: PMC7863242 DOI: 10.1186/s13046-021-01856-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/25/2021] [Indexed: 12/31/2022]
Abstract
Background Triple negative breast cancer (TNBC) is an aggressive subtype of breast cancer with high proliferative activity. TNBC tumors exhibit elevated MYC expression and altered expression of MYC regulatory genes, which are associated with tumor progression and poor prognosis; however, the underlying mechanisms by which MYC retains its high expression and mediates TNBC tumorigenesis require further exploration. Methods ACTL6A regulation of MYC and its target gene, CDK2, was defined using Co-IP, mass spectrometry and ChIP assays. To study the role of ACTL6A in TNBC, we performed soft-agar, colony formation, flow cytometry and tumor formation in nude mice. CDK2 inhibitor and paclitaxel were used in testing combination therapy in vitro and in vivo. Results ACTL6A bound MYC to suppress glycogen synthase kinase 3 beta (GSK3β)-induced phosphorylation on MYC T58, which inhibited ubiquitination of MYC and stabilized it. Moreover, ACTL6A promoted the recruitment of MYC and histone acetyltransferase KAT5 on CDK2 promoters, leading to hyperactivation of CDK2 transcription. ACTL6A overexpression promoted, while silencing ACTL6A suppressed cell proliferation and tumor growth in TNBC cells in vitro and in vivo, which was dependent on MYC signaling. Furthermore, co-therapy with paclitaxel and CDK2 inhibitor showed synergistic effects in tumor suppression. Notably, ACTL6A/MYC/CDK2 axis was specifically up-regulated in TNBC and high expression of ACTL6A was correlated to shorter survival in patients with TNBC. Conclusions These findings reveal a novel mechanism by which ACTL6A prolongs the retention of MYC in TNBC and suggest that pharmacological targeting ACTL6A/MYC/CDK2 axis might have therapeutic potential in patients with TNBC. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01856-3.
Collapse
|
20
|
Dass SA, Tan KL, Selva Rajan R, Mokhtar NF, Mohd Adzmi ER, Wan Abdul Rahman WF, Tengku Din TADAA, Balakrishnan V. Triple Negative Breast Cancer: A Review of Present and Future Diagnostic Modalities. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:62. [PMID: 33445543 PMCID: PMC7826673 DOI: 10.3390/medicina57010062] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast type of cancer with no expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2). It is a highly metastasized, heterogeneous disease that accounts for 10-15% of total breast cancer cases with a poor prognosis and high relapse rate within five years after treatment compared to non-TNBC cases. The diagnostic and subtyping of TNBC tumors are essential to determine the treatment alternatives and establish personalized, targeted medications for every TNBC individual. Currently, TNBC is diagnosed via a two-step procedure of imaging and immunohistochemistry (IHC), which are operator-dependent and potentially time-consuming. Therefore, there is a crucial need for the development of rapid and advanced technologies to enhance the diagnostic efficiency of TNBC. This review discusses the overview of breast cancer with emphasis on TNBC subtypes and the current diagnostic approaches of TNBC along with its challenges. Most importantly, we have presented several promising strategies that can be utilized as future TNBC diagnostic modalities and simultaneously enhance the efficacy of TNBC diagnostic.
Collapse
Affiliation(s)
- Sylvia Annabel Dass
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, USM, Penang 11800, Malaysia; (S.A.D.); (K.L.T.); (R.S.R.)
| | - Kim Liu Tan
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, USM, Penang 11800, Malaysia; (S.A.D.); (K.L.T.); (R.S.R.)
| | - Rehasri Selva Rajan
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, USM, Penang 11800, Malaysia; (S.A.D.); (K.L.T.); (R.S.R.)
| | - Noor Fatmawati Mokhtar
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia; (N.F.M.); (E.R.M.A.)
| | - Elis Rosliza Mohd Adzmi
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia; (N.F.M.); (E.R.M.A.)
| | - Wan Faiziah Wan Abdul Rahman
- Department of Pathology, School of Medical Sciences, Health Campus, Kubang Kerian, Kelantan 16150, Malaysia;
- Breast Cancer Awareness & Research Unit, Hospital Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia;
| | - Tengku Ahmad Damitri Al-Astani Tengku Din
- Breast Cancer Awareness & Research Unit, Hospital Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia;
- Chemical Pathology Department, School of Medical Sciences, Health Campus, Kubang Kerian, Kelantan 16150, Malaysia
| | - Venugopal Balakrishnan
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, USM, Penang 11800, Malaysia; (S.A.D.); (K.L.T.); (R.S.R.)
| |
Collapse
|
21
|
Oliphant MUJ, Kong D, Zhou H, Lewis MT, Ford HL. Two Sides of the Same Coin: The Role of Developmental pathways and pluripotency factors in normal mammary stem cells and breast cancer metastasis. J Mammary Gland Biol Neoplasia 2020; 25:85-102. [PMID: 32323111 PMCID: PMC7395869 DOI: 10.1007/s10911-020-09449-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
Breast cancer initiation and progression are often observed as the result of dysregulation of normal developmental processes and pathways. Studies focused on normal mammary stem/progenitor cell activity have led to an understanding of how breast cancer cells acquire stemness-associated properties including tumor initiation, survival and multi-lineage differentiation into heterogeneous tumors that become difficult to target therapeutically. Importantly, more recent investigations have provided valuable insight into how key developmental regulators can impact multiple phases of metastasis, where they are repurposed to not only promote metastatic phenotypes such as migration, invasion and EMT at the primary site, but also to regulate the survival, initiation and maintenance of metastatic lesions at secondary organs. Herein, we discuss findings that have led to a better understanding of how embryonic and pluripotency factors contribute not only to normal mammary development, but also to metastatic progression. We further examine the therapeutic potential of targeting these developmental pathways, and discuss how a better understanding of compensatory mechanisms, crosstalk between pathways, and novel experimental models could provide critical insight into how we might exploit embryonic and pluripotency regulators to inhibit tumor progression and metastasis.
Collapse
Affiliation(s)
- M U J Oliphant
- Integrated Physiology Program, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
- Department of Cell Biology and Ludwig Center at Harvard, Harvard Medical School, 240 Longwood Avenue, Building C1, Room 513B, Boston, MA, 02115, USA
| | - Deguang Kong
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
| | - Hengbo Zhou
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
| | - M T Lewis
- Departments of Molecular and Cellular Biology and Radiology. Lester and Sue Smith Breast Center, Baylor College of Medicine. One Baylor Plaza BCM600, Room N1210, Houston, TX, 77030, USA
| | - H L Ford
- Integrated Physiology Program, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA.
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA.
- Cancer Biology Program, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA.
| |
Collapse
|
22
|
Feigman MJ, Moss MA, Chen C, Cyrill SL, Ciccone MF, Trousdell MC, Yang ST, Frey WD, Wilkinson JE, Dos Santos CO. Pregnancy reprograms the epigenome of mammary epithelial cells and blocks the development of premalignant lesions. Nat Commun 2020; 11:2649. [PMID: 32461571 PMCID: PMC7253414 DOI: 10.1038/s41467-020-16479-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 05/05/2020] [Indexed: 02/06/2023] Open
Abstract
Pregnancy causes a series of cellular and molecular changes in mammary epithelial cells (MECs) of female adults. In addition, pregnancy can also modify the predisposition of rodent and human MECs to initiate oncogenesis. Here, we investigate how pregnancy reprograms enhancer chromatin in the mammary epithelium of mice and influences the transcriptional output of the oncogenic transcription factor cMYC. We find that pregnancy induces an expansion of the active cis-regulatory landscape of MECs, which influences the activation of pregnancy-related programs during re-exposure to pregnancy hormones in vivo and in vitro. Using inducible cMYC overexpression, we demonstrate that post-pregnancy MECs are resistant to the downstream molecular programs induced by cMYC, a response that blunts carcinoma initiation, but does not perturb the normal pregnancy-induced epigenomic landscape. cMYC overexpression drives post-pregnancy MECs into a senescence-like state, and perturbations of this state increase malignant phenotypic changes. Taken together, our findings provide further insight into the cell-autonomous signals in post-pregnancy MECs that underpin the regulation of gene expression, cellular activation, and resistance to malignant development. Mammary epithelial cells are epigenetically modified during pregnancy, these changes can influence the pre-disposition to cancer. Here, the authors examine the epigenetic landscape of mammary epithelial cells pre and post pregnancy and identify changes to the epigenetic landscape, which can protect mice from Myc induced cancer.
Collapse
Affiliation(s)
- Mary J Feigman
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | - Matthew A Moss
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA
| | - Chen Chen
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | - Samantha L Cyrill
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | - Michael F Ciccone
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | | | - Shih-Ting Yang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | - Wesley D Frey
- School of Medicine, Tulane University, New Orleans, LA, 70118, USA
| | - John E Wilkinson
- Department of Comparative Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Camila O Dos Santos
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA.
| |
Collapse
|
23
|
Lu W, Yang C, He H, Liu H. The CARM1-p300-c-Myc-Max (CPCM) transcriptional complex regulates the expression of CUL4A/4B and affects the stability of CRL4 E3 ligases in colorectal cancer. Int J Biol Sci 2020; 16:1071-1085. [PMID: 32140074 PMCID: PMC7053342 DOI: 10.7150/ijbs.41230] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 01/02/2020] [Indexed: 02/06/2023] Open
Abstract
The transcription factor c-Myc and two cullin family members CUL4A/4B function as oncogenes in colorectal cancer. Our recent publication reveals that c-Myc specifically activates the expression of CUL4A/4B through binding to their promoters. However, the underlying mechanism of how c-Myc actions in this process is still unknown. Using mass spectrometry and immunoprecipitation assays, we identified c-Myc formed a transcriptional complex with its partner Max (Myc-associated factor X), a histone acetyltransferase p300 and a coactivator associated arginine methyltransferase 1 (CARM1) in the present study. Knockdown or overexpression of the components of CARM1-p300-c-Myc-Max (CPCM) complex resulted in a decrease or increase of CUL4A/4B levels, respectively. Individual knockdown or inhibition of CPCM components decreased cell proliferation, colony formation, and cell invasion. Biochemically, knockdown or inhibition of CPCM components decreased their occupancies on the promoters of CUL4A/4B and resulted in their downregulation. Importantly, inhibition of CPCM components also caused a decrease of CRL4 E3 ligase activities and eventually led to an accumulation of ST7 (suppression of tumorigenicity 7), the specific substrate of CRL4 E3 ligases in colorectal cancer. Moreover, the in vivo tumor formation results indicated that knockdown or inhibition of CPCM components significantly decreased the tumor volumes. Together, our results suggest that the CPCM complex mediates explicitly the expression of CUL4A/4B, and thus affects the stability of CRL4 E3 ligases and the ubiquitination of ST7. These results provide more options by targeting the CPCM components to inhibit tumor growth in the therapy of colorectal cancer.
Collapse
Affiliation(s)
- Wenzhu Lu
- Department of Integrated Traditional and Western Medicine, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Chunmei Yang
- Department of Integrated Traditional and Western Medicine, Chengdu Shangjinnanfu Hospital/West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Hongbo He
- Department of Integrated Traditional and Western Medicine, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Hong Liu
- Department of Integrated Traditional and Western Medicine, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
24
|
Zhang Y, Xu B, Shi J, Li J, Lu X, Xu L, Yang H, Hamad N, Wang C, Napier D, He S, Liu C, Liu Z, Qian H, Chen L, Wei X, Zheng X, Huang JA, Thibault O, Craven R, Wei D, Pan Y, Zhou BP, Wu Y, Yang XH. BRD4 modulates vulnerability of triple-negative breast cancer to targeting of integrin-dependent signaling pathways. Cell Oncol (Dordr) 2020; 43:1049-1066. [PMID: 33006750 PMCID: PMC7716866 DOI: 10.1007/s13402-020-00537-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2020] [Indexed: 12/30/2022] Open
Abstract
PURPOSE Stemming from a myriad of genetic and epigenetic alterations, triple-negative breast cancer (TNBC) is tied to poor clinical outcomes and aspires for individualized therapies. Here we investigated the therapeutic potential of co-inhibiting integrin-dependent signaling pathway and BRD4, a transcriptional and epigenetic mediator, for TNBC. METHODS Two independent patient cohorts were subjected to bioinformatic and IHC examination for clinical association of candidate cancer drivers. The efficacy and biological bases for co-targeting these drivers were interrogated using cancer cell lines, a protein kinase array, chemical inhibitors, RNAi/CRISPR/Cas9 approaches, and a 4 T1-Balb/c xenograft model. RESULTS We found that amplification of the chromosome 8q24 region occurred in nearly 20% of TNBC tumors, and that it coincided with co-upregulation or amplification of c-Myc and FAK, a key effector of integrin-dependent signaling. This co-upregulation at the mRNA or protein level correlated with a poor patient survival (p < 0.0109 or p < 0.0402, respectively). Furthermore, we found that 14 TNBC cell lines exhibited high vulnerabilities to the combination of JQ1 and VS-6063, potent pharmacological antagonists of the BRD4/c-Myc and integrin/FAK-dependent pathways, respectively. We also observed a cooperative inhibitory effect of JQ1 and VS-6063 on tumor growth and infiltration of Ly6G+ myeloid-derived suppressor cells in vivo. Finally, we found that JQ1 and VS-6063 cooperatively induced apoptotic cell death by altering XIAP, Bcl2/Bcl-xl and Bim levels, impairing c-Src/p130Cas-, PI3K/Akt- and RelA-associated signaling, and were linked to EMT-inducing transcription factor Snail- and Slug-dependent regulation. CONCLUSION Based on our results, we conclude that the BRD4/c-Myc- and integrin/FAK-dependent pathways act in concert to promote breast cancer cell survival and poor clinical outcomes. As such, they represent promising targets for a synthetic lethal-type of therapy against TNBC.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, and Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Respiratory Medicine, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Bingwei Xu
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, and Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Junfeng Shi
- Department of Oncology, Nanjing Medical University, Nanjing, Jiangsu Province, People's Republic of China
| | - Jieming Li
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, and Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Center of Drug Discovery, China Pharmaceutical University, Nanjing, Jiangsu Province, People's Republic of China
| | - Xinlan Lu
- Department of Medical Oncology, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shanxi Province, People's Republic of China
| | - Li Xu
- Department of Statistics, University of Kentucky, Lexington, KY, USA
| | - Helen Yang
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, and Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Nevean Hamad
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, and Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Chi Wang
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, and Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Dana Napier
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, and Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Shuixiang He
- Department of Medical Oncology, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shanxi Province, People's Republic of China
| | - Chunming Liu
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, and Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Zeyi Liu
- Department of Respiratory Medicine, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Hai Qian
- Center of Drug Discovery, China Pharmaceutical University, Nanjing, Jiangsu Province, People's Republic of China
| | - Li Chen
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, and Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Xiaowei Wei
- Department of Oncology, Nanjing Medical University, Nanjing, Jiangsu Province, People's Republic of China
| | - Xucai Zheng
- The First Affiliated Hospital of University of Science & Technology of China and Provincial Hospital, Hefei, Anhui Province, People's Republic of China
| | - Jian-An Huang
- Department of Respiratory Medicine, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Olivier Thibault
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, and Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Rolf Craven
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, and Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Dongping Wei
- Department of Oncology, Nanjing Medical University, Nanjing, Jiangsu Province, People's Republic of China.
| | - Yueyin Pan
- The First Affiliated Hospital of University of Science & Technology of China and Provincial Hospital, Hefei, Anhui Province, People's Republic of China.
| | - Binhua P Zhou
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, and Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, USA.
| | - Yadi Wu
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, and Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, USA.
| | - Xiuwei H Yang
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, and Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
25
|
High MYC mRNA Expression Is More Clinically Relevant than MYC DNA Amplification in Triple-Negative Breast Cancer. Int J Mol Sci 2019; 21:ijms21010217. [PMID: 31905596 PMCID: PMC6981812 DOI: 10.3390/ijms21010217] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/21/2019] [Accepted: 12/25/2019] [Indexed: 12/13/2022] Open
Abstract
DNA abnormalities are used in inclusion criteria of clinical trials for treatments with specific targeted molecules. MYC is one of the most powerful oncogenes and is known to be associated with triple-negative breast cancer (TNBC). Its DNA amplification is often part of the targeted DNA-sequencing panels under the assumption of reflecting upregulated signaling. However, it remains unclear if MYC DNA amplification is a surrogate of its upregulated signaling. Thus, we investigated the difference between MYC DNA amplification and mRNA high expression in TNBCs utilizing publicly available cohorts. MYC DNA amplified tumors were found to have various mRNA expression levels, suggesting that MYC DNA amplification does not always result in elevated MYC mRNA expression. Compared to other subtypes, both MYC DNA amplification and mRNA high expression were more frequent in the TNBCs. MYC mRNA high expression, but not DNA amplification, was significantly associated with worse overall survival in the TNBCs. The TNBCs with MYC mRNA high expression enriched MYC target genes, cell cycle related genes, and WNT/β-catenin gene sets, whereas none of them were enriched in MYC DNA amplified TNBCs. In conclusion, MYC mRNA high expression, but not DNA amplification, reflects not only its upregulated signaling pathway, but also clinical significance in TNBCs.
Collapse
|
26
|
Liu Z, Mi M, Li X, Zheng X, Wu G, Zhang L. lncRNA OSTN-AS1 May Represent a Novel Immune-Related Prognostic Marker for Triple-Negative Breast Cancer Based on Integrated Analysis of a ceRNA Network. Front Genet 2019; 10:850. [PMID: 31572452 PMCID: PMC6753250 DOI: 10.3389/fgene.2019.00850] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/14/2019] [Indexed: 12/24/2022] Open
Abstract
The competing endogenous RNA (ceRNA) networks are an effective method for investigating cancer; however, construction of ceRNA networks among different subtypes of breast cancer has not been previously performed. Based on analysis of differentially expressed RNAs between 150 triple-negative breast cancer (TNBC) tissues and 823 non–triple-negative breast cancer (nTNBC) tissues downloaded from TCGA database, a ceRNA network was constructed based on database comparisons using Cytoscape. Survival analysis and receiver operating characteristic curve data were combined to screen out prognostic candidate genes, which were subsequently analyzed using co-expressed functionally related analysis, Gene Set Variation Analysis (GSVA) pathway-related analysis, and immune infiltration and tumor mutational burden immune-related analysis. A total of 190 differentially expressed lncRNAs (DElncRNAs), 48 differentially expressed mRNAs (DEmRNAs), and 13 differentially expressed miRNAs (DEmiRNAs) were included in the ceRNA network between TNBC and nTNBC subtypes. Gene ontology analysis of mRNAs coexpressed with prognostic candidate lncRNAs (AC104472.1, PSORS1C3, DSCR9, OSTN-AS1, AC012074.1, AC005035.1, SIAH2-AS1, and ERVMER61-1) were utilized for functional prediction. Consequently, OSTN-AS1 was primarily related to immunologic function, for instance, immune cell infiltration and immune-related markers coexpression. The GSVA deviation degree was increased with OSTN increased expression. In addition, many important immune molecules, such as PDCD1 and CTLA-4, were strongly correlated in terms of their quantitative expression. Competing endogenous RNA networks may identify candidate therapeutic targets and potential prognostic biomarkers in breast cancer. In particular, OSTN-AS1 serves as a novel immune-related molecule and could be involved in immunotherapy efforts in the future.
Collapse
Affiliation(s)
- Zijian Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mi Mi
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqian Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Zheng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liling Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Sengupta P, Banerjee N, Roychowdhury T, Dutta A, Chattopadhyay S, Chatterjee S. Site-specific amino acid substitution in dodecameric peptides determines the stability and unfolding of c-MYC quadruplex promoting apoptosis in cancer cells. Nucleic Acids Res 2019; 46:9932-9950. [PMID: 30239898 PMCID: PMC6212778 DOI: 10.1093/nar/gky824] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 09/11/2018] [Indexed: 12/11/2022] Open
Abstract
c-MYC proto-oncogene harbours a transcription-inhibitory quadruplex-forming scaffold (Pu27) upstream P1 promoter providing anti-neoplastic therapeutic target. Previous reports showed the binding profile of human Cathelicidin peptide (LL37) and telomeric G-quadruplex. Here, we truncated the quadruplex-binding domain of LL37 to prepare a small library of peptides through site-specific amino acid substitution. We investigated the intracellular selectivity of peptides for Pu27 over other oncogenic quadruplexes and their role in c-MYC promoter repression by dual-luciferase assays. We analysed their thermodynamics of binding reactions with c-MYC quadruplex isomers (Pu27, Myc22, Pu19) by Isothermal Titration Calorimetry. We discussed how amino acid substitutions and peptide helicity enhanced/weakened their affinities for c-MYC quadruplexes and characterized specific non-covalent inter-residual interactions determining their selectivity. Solution NMR structure indicated that KR12C, the best peptide candidate, selectively stabilized the 5′-propeller loop of c-MYC quadruplex by arginine-driven electrostatic-interactions at the sugar-phosphate backbone while KR12A peptide destabilized the quadruplex inducing a single-stranded hairpin-like conformation. Chromatin immunoprecipitations envisaged that KR12C and KR12A depleted and enriched Sp1 and NM23-H2 (Nucleoside diphosphate kinase) occupancy at Pu27 respectively supporting their regulation in stabilizing and unfolding c-MYC quadruplex in MCF-7 cells. We deciphered that selective arresting of c-MYC transcription by KR12C triggered apoptotic-signalling pathway via VEGF-A-BCL-2 axis.
Collapse
Affiliation(s)
- Pallabi Sengupta
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata 700054, India
| | - Nilanjan Banerjee
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata 700054, India
| | - Tanaya Roychowdhury
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata 700032, India
| | - Anindya Dutta
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata 700054, India
| | - Samit Chattopadhyay
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata 700032, India
| | - Subhrangsu Chatterjee
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata 700054, India
| |
Collapse
|
28
|
Lourenco C, Kalkat M, Houlahan KE, De Melo J, Longo J, Done SJ, Boutros PC, Penn LZ. Modelling the MYC-driven normal-to-tumour switch in breast cancer. Dis Model Mech 2019; 12:12/7/dmm038083. [PMID: 31350286 PMCID: PMC6679384 DOI: 10.1242/dmm.038083] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 06/03/2019] [Indexed: 12/13/2022] Open
Abstract
The potent MYC oncoprotein is deregulated in many human cancers, including breast carcinoma, and is associated with aggressive disease. To understand the mechanisms and vulnerabilities of MYC-driven breast cancer, we have generated an in vivo model that mimics human disease in response to MYC deregulation. MCF10A cells ectopically expressing a common breast cancer mutation in the phosphoinositide 3 kinase pathway (PIK3CAH1047R) led to the development of organised acinar structures in mice. Expressing both PIK3CAH1047R and deregulated MYC led to the development of invasive ductal carcinoma. Therefore, the deregulation of MYC expression in this setting creates a MYC-dependent normal-to-tumour switch that can be measured in vivo. These MYC-driven tumours exhibit classic hallmarks of human breast cancer at both the pathological and molecular level. Moreover, tumour growth is dependent upon sustained deregulated MYC expression, further demonstrating addiction to this potent oncogene and regulator of gene transcription. We therefore provide a MYC-dependent model of breast cancer, which can be used to assay invivo tumour signalling pathways, proliferation and transformation from normal breast acini to invasive breast carcinoma. We anticipate that this novel MYC-driven transformation model will be a useful research tool to better understand the oncogenic function of MYC and for the identification of therapeutic vulnerabilities. Summary: We present a MYC-driven transformation model of breast cancer that recapitulates the disease in vivo and which can be used to identify MYC-dependent cancer vulnerabilities.
Collapse
Affiliation(s)
- Corey Lourenco
- Princess Margaret Cancer Centre, University Health Network, 101 College St, Toronto, ON M5G 0A3, Canada.,Department of Medical Biophysics, University of Toronto, 101 College Street Suite 15-701, Toronto, ON M5G 1L7, Canada
| | - Manpreet Kalkat
- Princess Margaret Cancer Centre, University Health Network, 101 College St, Toronto, ON M5G 0A3, Canada.,Department of Medical Biophysics, University of Toronto, 101 College Street Suite 15-701, Toronto, ON M5G 1L7, Canada
| | - Kathleen E Houlahan
- Department of Medical Biophysics, University of Toronto, 101 College Street Suite 15-701, Toronto, ON M5G 1L7, Canada.,Ontario Institute for Cancer Research, 661 University Ave, Suite 510, Toronto, ON M5G 0A3, Canada
| | - Jason De Melo
- Princess Margaret Cancer Centre, University Health Network, 101 College St, Toronto, ON M5G 0A3, Canada
| | - Joseph Longo
- Princess Margaret Cancer Centre, University Health Network, 101 College St, Toronto, ON M5G 0A3, Canada.,Department of Medical Biophysics, University of Toronto, 101 College Street Suite 15-701, Toronto, ON M5G 1L7, Canada
| | - Susan J Done
- Princess Margaret Cancer Centre, University Health Network, 101 College St, Toronto, ON M5G 0A3, Canada.,Department of Medical Biophysics, University of Toronto, 101 College Street Suite 15-701, Toronto, ON M5G 1L7, Canada
| | - Paul C Boutros
- Department of Medical Biophysics, University of Toronto, 101 College Street Suite 15-701, Toronto, ON M5G 1L7, Canada.,Ontario Institute for Cancer Research, 661 University Ave, Suite 510, Toronto, ON M5G 0A3, Canada
| | - Linda Z Penn
- Princess Margaret Cancer Centre, University Health Network, 101 College St, Toronto, ON M5G 0A3, Canada .,Department of Medical Biophysics, University of Toronto, 101 College Street Suite 15-701, Toronto, ON M5G 1L7, Canada
| |
Collapse
|
29
|
Savva C, De Souza K, Ali R, Rakha EA, Green AR, Madhusudan S. Clinicopathological significance of ataxia telangiectasia-mutated (ATM) kinase and ataxia telangiectasia-mutated and Rad3-related (ATR) kinase in MYC overexpressed breast cancers. Breast Cancer Res Treat 2019; 175:105-115. [PMID: 30746633 PMCID: PMC6491658 DOI: 10.1007/s10549-018-05113-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 12/18/2018] [Indexed: 12/20/2022]
Abstract
PURPOSE MYC transcription factor has critical roles in cell growth, proliferation, metabolism, differentiation, transformation and angiogenesis. MYC overexpression is seen in about 15% of breast cancers and linked to aggressive phenotypes. MYC overexpression also induces oxidative stress and replication stress in cells. ATM signalling and ATR-mediated signalling are critical for MYC-induced DNA damage response. Whether ATM and ATR expressions influence clinical outcomes in MYC overexpressed breast cancers is unknown. METHODS We investigated ATM, ATR and MYC at the transcriptional level [Molecular Taxonomy of Breast Cancer International Consortium cohort (n = 1950)] and at the protein level in the Nottingham series comprising 1650 breast tumours. We correlated ATM, ATR and MYC expressions to clinicopathological features and survival outcomes. RESULTS In MYC over expressed tumours, high ATR or low ATM levels were associated with aggressive breast cancer features such as higher tumour grade, de-differentiation, pleomorphism, high mitotic index, high-risk Nottingham Prognostic Index, triple negative and basal-like breast cancers (all adjusted p values < 0.05). Tumours with low ATM or high ATR levels in conjunction with MYC overexpression also have worse overall breast cancer-specific survival (BCSS) (p value < 0.05). CONCLUSIONS We conclude that ATR/ATM-directed stratification and personalisation of therapy may be feasible in MYC overexpressed breast cancer.
Collapse
Affiliation(s)
- Constantinos Savva
- Department of Oncology, Nottingham University Hospitals, Nottingham, NG5 1PB, UK
| | - Karen De Souza
- Department of Oncology, Nottingham University Hospitals, Nottingham, NG5 1PB, UK
| | - Reem Ali
- Translational Oncology, Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, Academic Unit of Oncology, School of Medicine, University of Nottingham, Nottingham, NG51 PB, UK
| | - Emad A Rakha
- Department of Pathology, Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, NG5 1PB, UK
| | - Andrew R Green
- Department of Pathology, Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, NG5 1PB, UK.
| | - Srinivasan Madhusudan
- Department of Oncology, Nottingham University Hospitals, Nottingham, NG5 1PB, UK.
- Translational Oncology, Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, Academic Unit of Oncology, School of Medicine, University of Nottingham, Nottingham, NG51 PB, UK.
| |
Collapse
|
30
|
Heart myxoma develops oncogenic and metastatic phenotype. J Cancer Res Clin Oncol 2019; 145:1283-1295. [PMID: 30900156 DOI: 10.1007/s00432-019-02897-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 03/13/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Heart myxomas have been frequently considered as benign lesions associated with Carney's complex. However, after surgical removal, myxomas re-emerge causing dysfunctional heart. METHODS To identify whether cardiac myxomas may develop a metastatic phenotype as occurs in malignant cancers, a profile of several proteins involved in malignancy such as oncogenes (c-MYC, K-RAS and H-RAS), cancer-associated metabolic transcriptional factors (HIF-1α, p53 and PPAR-γ) and epithelial-mesenchymal transition proteins (fibronectin, vimentin, β-catenin, SNAIL and MMP-9) were evaluated in seven samples from a cohort of patients with atrial and ventricular myxomas. The analysis was also performed in: (1) cardiac tissue surrounding the area where myxoma was removed; (2) non-cancer heart tissue (NCHT); and (3) malignant triple negative breast cancer biopsies for comparative purposes. RESULTS Statistical analysis applying univariate (Kruskal-Wallis and Dunn's tests) and multivariate analyses (PCA, principal component analysis) revealed that heart myxomas (7-15 times) and myxoma surrounding tissue (22-99 times) vs. NCHT showed high content of c-MYC, p53, vimentin, and HIF-1α, indicating that both myxoma and its surrounding area express oncogenes and malignancy-related proteins as occurs in triple negative breast cancer. CONCLUSIONS Based on ROC (receiver operating characteristics) statistical analysis, c-MYC, HIF-1α, p53, and vimentin may be considered potential biomarkers for malignancy detection in myxoma.
Collapse
|
31
|
Qian XL, Pan YH, Huang QY, Shi YB, Huang QY, Hu ZZ, Xiong LX. Caveolin-1: a multifaceted driver of breast cancer progression and its application in clinical treatment. Onco Targets Ther 2019; 12:1539-1552. [PMID: 30881011 PMCID: PMC6398418 DOI: 10.2147/ott.s191317] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Human breast cancer is one of the most frequent cancer diseases and causes of death among female population worldwide. It appears at a high incidence and has a high malignancy, mortality, recurrence rate and poor prognosis. Caveolin-1 (Cav1) is the main component of caveolae and participates in various biological events. More and more experimental studies have shown that Cav1 plays a critical role in the progression of breast cancer including cell proliferation, apoptosis, autophagy, invasion, migration and breast cancer metastasis. Besides, Cav1 has been found to be involved in chemotherapeutics and radiotherapy resistance, which are still the principal problems encountered in clinical breast cancer treatment. In addition, stromal Cav1 may be a potential indicator for breast cancer patients' prognosis. In the current review, we cover the state-of-the-art study, development and progress on Cav1 and breast cancer, altogether describing the role of Cav1 in breast cancer progression and application in clinical treatment, in the hope of providing a basis for further research and promoting CAV1 gene as a potential target to diagnose and treat aggressive breast cancers.
Collapse
Affiliation(s)
- Xian-Ling Qian
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China, ;
- First Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Yi-Hang Pan
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China, ;
- First Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Qi-Yuan Huang
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China, ;
- Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Yu-Bo Shi
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China, ;
| | - Qing-Yun Huang
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China, ;
| | - Zhen-Zhen Hu
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China, ;
- Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, Nanchang 330006, China, ;
| | - Li-Xia Xiong
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China, ;
- Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, Nanchang 330006, China, ;
| |
Collapse
|
32
|
Li M, Li A, Zhou S, Lv H, Yang W. SPAG5 upregulation contributes to enhanced c-MYC transcriptional activity via interaction with c-MYC binding protein in triple-negative breast cancer. J Hematol Oncol 2019; 12:14. [PMID: 30736840 PMCID: PMC6367803 DOI: 10.1186/s13045-019-0700-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/22/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype that lacks effective therapeutic targets. Sperm-associated antigen 5 (SPAG5) is a mitotic spindle-associated protein that is involved in various biological processes in cervical cancer and bladder urothelial carcinoma. However, the role of SPAG5 in TNBC remains undefined. METHODS The expression of SPAG5 was examined in TNBC patients via quantitative real-time polymerase chain reaction (qRT-PCR), western blotting, and immunohistochemistry (IHC). The biological functions of SPAG5 in TNBC and the underlying mechanisms were investigated in vitro and in vivo. RESULTS SPAG5 expression was significantly upregulated in TNBC tissues compared with that in paired adjacent noncancerous tissues (ANTs). High SPAG5 expression was associated with increased lymph node metastasis and high risk of local recurrence. SPAG5 protein expression was significantly associated with poor disease-free survival in TNBC. Gene set enrichment analysis of TNBC data from The Cancer Genome Atlas (TCGA) indicated that high SPAG5 expression was significantly associated with cell cycle and the ATR-BRCA pathway. Functional assays demonstrated that SPAG5 expression promoted tumor growth in vitro and in vivo. In addition, SPAG5-silenced cells were more sensitive to the PARP inhibitor (PARPi) olaparib. Mechanistically, SPAG5 interacted with c-MYC binding protein (MYCBP), thereby increasing MYCBP protein levels and leading to increased c-MYC transcriptional activity, which promoted the expression of the c-MYC target genes: CDC20, CDC25C, BRCA1, BRCA2, and RAD51.Knockdown of MYCBP or c-MYC abolished the SPAG5-induced cell-cycle progression and cell proliferation of TNBC. CONCLUSIONS Collectively, our results indict that SPAG5 is an efficient prognostic factor in TNBC, and that SPAG5 knockdown increases the sensitivity of TNBC to the PARPi olaparib. SPAG5 promotes tumor growth and DNA repair by increasing c-MYC transcriptional activity via interaction with MYCBP. The SPAG5/MYCBP/c-MYC axis may represent a potential therapeutic target for TNBC treatment.
Collapse
Affiliation(s)
- Ming Li
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Anqi Li
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Shuling Zhou
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Hong Lv
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Wentao Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China. .,Institute of Pathology, Fudan University, Shanghai, China.
| |
Collapse
|
33
|
Jiang D, Song Y, Cao W, Wang X, Jiang D, Lv Z, Yang Z, Li F. p53-independent role of MYC mutant T58A in the proliferation and apoptosis of breast cancer cells. Oncol Lett 2019; 17:1071-1079. [PMID: 30655867 PMCID: PMC6312996 DOI: 10.3892/ol.2018.9688] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 09/24/2018] [Indexed: 12/31/2022] Open
Abstract
Myc proto-oncogene (MYC) is an oncoprotein that promotes proliferation and apoptosis. MYC mutations frequently disrupt the apoptotic processes during tumorigenesis. In the present study, the effects of the MYC point mutation T58A on the progression of a cellular tumor antigen p53 (p53)-/- human breast cancer cell line was analyzed, and the mechanism of p53-independent MYC-induced apoptosis was investigated. HCC1937 cells were transfected with mutant (T58A) or wild-type (WT) MYC using lentiviral vectors. The proliferation of transfected cells was evaluated by colony formation and MTT assays, and apoptosis was analyzed by flow cytometry and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assays. WT MYC was transfected into HCC1937 cells exhibiting p14/p21 silencing through lentivirus-mediated RNA interference. The expression levels of Bim were detected by reverse transcription-quantitative polymerase chain reaction and western blot analyses. Mutant MYC proteins retained the ability to stimulate the proliferation of HCC1937 cells, although they were defective at promoting apoptosis due to a failure to induce the Bcl-2 homology 3 domain-only protein Bim. When p14 was silenced, the effects of mutant MYC on proliferation and apoptosis were weakened. When p21 was silenced, the effects of mutant MYC were strengthened. Breast cancer-derived T58A MYC mutations are unable to activate Bim due to their failure to regulate p14/p21. It was concluded that mutant MYC was more effective compared with WT MYC at promoting the progression of breast cancer.
Collapse
Affiliation(s)
- Dandan Jiang
- Breast Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yuhua Song
- Breast Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Weihong Cao
- Breast Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Xingang Wang
- Breast Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Danni Jiang
- Imaging Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Zhidong Lv
- Breast Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Zhaochuan Yang
- Breast Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Funian Li
- Breast Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
34
|
Archer TC, Ehrenberger T, Mundt F, Gold MP, Krug K, Mah CK, Mahoney EL, Daniel CJ, LeNail A, Ramamoorthy D, Mertins P, Mani DR, Zhang H, Gillette MA, Clauser K, Noble M, Tang LC, Pierre-François J, Silterra J, Jensen J, Tamayo P, Korshunov A, Pfister SM, Kool M, Northcott PA, Sears RC, Lipton JO, Carr SA, Mesirov JP, Pomeroy SL, Fraenkel E. Proteomics, Post-translational Modifications, and Integrative Analyses Reveal Molecular Heterogeneity within Medulloblastoma Subgroups. Cancer Cell 2018; 34:396-410.e8. [PMID: 30205044 PMCID: PMC6372116 DOI: 10.1016/j.ccell.2018.08.004] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 06/28/2018] [Accepted: 08/03/2018] [Indexed: 12/18/2022]
Abstract
There is a pressing need to identify therapeutic targets in tumors with low mutation rates such as the malignant pediatric brain tumor medulloblastoma. To address this challenge, we quantitatively profiled global proteomes and phospho-proteomes of 45 medulloblastoma samples. Integrated analyses revealed that tumors with similar RNA expression vary extensively at the post-transcriptional and post-translational levels. We identified distinct pathways associated with two subsets of SHH tumors, and found post-translational modifications of MYC that are associated with poor outcomes in group 3 tumors. We found kinases associated with subtypes and showed that inhibiting PRKDC sensitizes MYC-driven cells to radiation. Our study shows that proteomics enables a more comprehensive, functional readout, providing a foundation for future therapeutic strategies.
Collapse
Affiliation(s)
- Tenley C Archer
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA; Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Tobias Ehrenberger
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Filip Mundt
- Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Maxwell P Gold
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Karsten Krug
- Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Clarence K Mah
- Department of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Elizabeth L Mahoney
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Colin J Daniel
- Department of Molecular and Medical Genetics, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Alexander LeNail
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Divya Ramamoorthy
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Philipp Mertins
- Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - D R Mani
- Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hailei Zhang
- Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael A Gillette
- Harvard Medical School, Boston, MA, USA; Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital (MGH), Boston, MA, USA
| | - Karl Clauser
- Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael Noble
- Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lauren C Tang
- Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jessica Pierre-François
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Jacob Silterra
- Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - James Jensen
- Department of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Pablo Tamayo
- Department of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Andrey Korshunov
- CCU Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neuropathology, Heidelberg University, Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children's Cancer Center at the NCT Heidelberg (KiTZ), Heidelberg, Germany; Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany; Department of Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marcel Kool
- Hopp Children's Cancer Center at the NCT Heidelberg (KiTZ), Heidelberg, Germany; Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Paul A Northcott
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Rosalie C Sears
- Department of Molecular and Medical Genetics, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Jonathan O Lipton
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Steven A Carr
- Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Jill P Mesirov
- Department of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California San Diego (UCSD), La Jolla, CA, USA.
| | - Scott L Pomeroy
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Ernest Fraenkel
- Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA.
| |
Collapse
|
35
|
Pourteimoor V, Paryan M, Mohammadi‐Yeganeh S. microRNA as a systemic intervention in the specific breast cancer subtypes with C‐MYC impacts; introducing subtype‐based appraisal tool. J Cell Physiol 2018; 233:5655-5669. [DOI: 10.1002/jcp.26399] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 12/11/2017] [Indexed: 12/18/2022]
Affiliation(s)
| | - Mahdi Paryan
- Department of Research and Development, Production and Research ComplexPasteur Institute of IranTehranIran
| | - Samira Mohammadi‐Yeganeh
- Cellular and Molecular Biology Research CenterShahid Beheshti University of Medical SciencesTehranIran
- Department of Biotechnology, School of Advanced Technologies in MedicineShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
36
|
Penkert J, Ripperger T, Schieck M, Schlegelberger B, Steinemann D, Illig T. On metabolic reprogramming and tumor biology: A comprehensive survey of metabolism in breast cancer. Oncotarget 2018; 7:67626-67649. [PMID: 27590516 PMCID: PMC5341901 DOI: 10.18632/oncotarget.11759] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/25/2016] [Indexed: 12/20/2022] Open
Abstract
Altered metabolism in tumor cells has been a focus of cancer research for as long as a century but has remained controversial and vague due to an inhomogeneous overall picture. Accumulating genomic, metabolomic, and lastly panomic data as well as bioenergetics studies of the past few years enable a more comprehensive, systems-biologic approach promoting deeper insight into tumor biology and challenging hitherto existing models of cancer bioenergetics. Presenting a compendium on breast cancer-specific metabolome analyses performed thus far, we review and compile currently known aspects of breast cancer biology into a comprehensive network, elucidating previously dissonant issues of cancer metabolism. As such, some of the aspects critically discussed in this review include the dynamic interplay or metabolic coupling between cancer (stem) cells and cancer-associated fibroblasts, the intratumoral and intertumoral heterogeneity and plasticity of cancer cell metabolism, the existence of distinct metabolic tumor compartments in need of separate yet simultaneous therapeutic targeting, the reliance of cancer cells on oxidative metabolism and mitochondrial power, and the role of pro-inflammatory, pro-tumorigenic stromal conditioning. Comprising complex breast cancer signaling networks as well as combined metabolomic and genomic data, we address metabolic consequences of mutations in tumor suppressor genes and evaluate their contribution to breast cancer predisposition in a germline setting, reasoning for distinct personalized preventive and therapeutic measures. The review closes with a discussion on central root mechanisms of tumor cell metabolism and rate-limiting steps thereof, introducing essential strategies for therapeutic targeting.
Collapse
Affiliation(s)
- Judith Penkert
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Tim Ripperger
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | | | | | - Doris Steinemann
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Thomas Illig
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany.,Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| |
Collapse
|
37
|
Woo SH, Seo SK, Park Y, Kim EK, Seong MK, Kim HA, Song JY, Hwang SG, Lee JK, Noh WC, Park IC. Dichloroacetate potentiates tamoxifen-induced cell death in breast cancer cells via downregulation of the epidermal growth factor receptor. Oncotarget 2018; 7:59809-59819. [PMID: 27494858 PMCID: PMC5312350 DOI: 10.18632/oncotarget.10999] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/22/2016] [Indexed: 01/01/2023] Open
Abstract
Metabolic reprogramming in cancer cells has recently been recognized as an essential hallmark of neoplasia. In this context, metabolic alterations represent an attractive therapeutic target, and encouraging results with drugs targeting various metabolic processes have been obtained in preclinical studies. Recently, several studies have suggested that dichloroacetate (DCA), a specific pyruvate dehydrogenase kinase inhibitor, may be a potential anticancer drug in a large number of diverse tumors. However, the precise mechanism is not fully understood, which is important for the use of DCA in cancer treatment. In the present study, we found that DCA sensitized MCF7 breast cancer cells to tamoxifen-induced cell death by decreasing epidermal growth factor receptor (EGFR) expression. The downregulation of EGFR was caused by degradation of the protein. Furthermore, p38 mitogen-activated protein kinase played an important role in DCA/tamoxifen-induced EGFR degradation. Finally, DCA also promoted comparable tamoxifen-induced cell death in tamoxifen-resistant MCF7 cells, which were established by long-term treatment with tamoxifen. In summary, our results suggest that DCA is an attractive potential drug that sensitizes cells to tamoxifen-induced cell death and overcome tamoxifen resistance via downregulation of EGFR expression in breast cancer cells.
Collapse
Affiliation(s)
- Sang Hyeok Woo
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Sung-Keum Seo
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Yoonhwa Park
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea.,School of Life Science and Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Eun-Kyu Kim
- Department of Surgery, Breast Cancer Center, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Bundang-gu, Seongnam, 13620, Republic of Korea
| | - Min-Ki Seong
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Hyun-Ah Kim
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Jie-Young Song
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Sang-Gu Hwang
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Jin Kyung Lee
- KIRAMS Radiation Biobank, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Woo Chul Noh
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| | - In-Chul Park
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| |
Collapse
|
38
|
The ERβ4 variant induces transformation of the normal breast mammary epithelial cell line MCF-10A; the ERβ variants ERβ2 and ERβ5 increase aggressiveness of TNBC by regulation of hypoxic signaling. Oncotarget 2018; 9:12201-12211. [PMID: 29552303 PMCID: PMC5844739 DOI: 10.18632/oncotarget.24134] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 11/05/2017] [Indexed: 01/03/2023] Open
Abstract
Triple negative breast cancer (TNBC) still remains a challenge to treat in the clinic due to a lack of good targets for treatment. Although TNBC lacks expression of ERα, the expression of ERβ and its variants are detected quite frequently in this cancer type and can represent an avenue for treatment. We show that two of the variants of ERβ, namely ERβ2 and ERβ5, control aggressiveness of TNBC by regulating hypoxic signaling through stabilization of HIF-1α. RNA-seq of patient derived xenografts (PDX) from TNBC shows expression of ERβ2, ERβ4 and ERβ5 variants in more than half of the samples. Furthermore, expression of ERβ4 in the immortalized, normal mammary epithelial cell line MCF-10A that is resistant to tumorsphere formation caused transformation and development of tumorspheres. By contrast, ERβ1, ERβ2 or ERβ5 were unable to support tumorsphere formation. We have previously shown that all variants except ERβ1 stabilize HIF-1α but only ERβ4 appears to have the ability to transform normal mammary epithelial cells, pointing towards a unique property of ERβ4. We propose that ERβ variants may be good diagnostic tools and also serve as novel targets for treatment of breast cancer.
Collapse
|
39
|
Zheng W, Li J, Wang X, Yuan Y, Zhang J, Xiu Z. Effects of Antarctic krill docosahexaenoic acid on MCF-7 cell migration and invasion induced by the interaction of CD95 with caveolin-1. Life Sci 2018; 192:270-277. [DOI: 10.1016/j.lfs.2017.11.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/09/2017] [Accepted: 11/07/2017] [Indexed: 12/31/2022]
|
40
|
Sun YS, Zhao Z, Yang ZN, Xu F, Lu HJ, Zhu ZY, Shi W, Jiang J, Yao PP, Zhu HP. Risk Factors and Preventions of Breast Cancer. Int J Biol Sci 2017; 13:1387-1397. [PMID: 29209143 PMCID: PMC5715522 DOI: 10.7150/ijbs.21635] [Citation(s) in RCA: 720] [Impact Index Per Article: 102.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/28/2017] [Indexed: 12/28/2022] Open
Abstract
Breast cancer is the second leading cause of cancer deaths among women. The development of breast cancer is a multi-step process involving multiple cell types, and its prevention remains challenging in the world. Early diagnosis of breast cancer is one of the best approaches to prevent this disease. In some developed countries, the 5-year relative survival rate of breast cancer patients is above 80% due to early prevention. In the recent decade, great progress has been made in the understanding of breast cancer as well as in the development of preventative methods. The pathogenesis and tumor drug-resistant mechanisms are revealed by discovering breast cancer stem cells, and many genes are found related to breast cancer. Currently, people have more drug options for the chemoprevention of breast cancer, while biological prevention has been recently developed to improve patients' quality of life. In this review, we will summarize key studies of pathogenesis, related genes, risk factors and preventative methods on breast cancer over the past years. These findings represent a small step in the long fight against breast cancer.
Collapse
Affiliation(s)
- Yi-Sheng Sun
- Key Lab of Vaccine against Hemorrhagic Fever with Renal Syndrome, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Zhao Zhao
- Centre of Laboratory Medicine, Zhejiang Provincial People's Hospital
| | - Zhang-Nv Yang
- Key Lab of Vaccine against Hemorrhagic Fever with Renal Syndrome, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Fang Xu
- Key Lab of Vaccine against Hemorrhagic Fever with Renal Syndrome, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Hang-Jing Lu
- Key Lab of Vaccine against Hemorrhagic Fever with Renal Syndrome, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Zhi-Yong Zhu
- Key Lab of Vaccine against Hemorrhagic Fever with Renal Syndrome, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Wen Shi
- Key Lab of Vaccine against Hemorrhagic Fever with Renal Syndrome, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Jianmin Jiang
- Key Lab of Vaccine against Hemorrhagic Fever with Renal Syndrome, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Ping-Ping Yao
- Key Lab of Vaccine against Hemorrhagic Fever with Renal Syndrome, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Han-Ping Zhu
- Key Lab of Vaccine against Hemorrhagic Fever with Renal Syndrome, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| |
Collapse
|
41
|
Jeong Y, Hoe H, Cho H, Park K, Kim D, Kim C, Magae J, Kang DW, Lee S, Chang Y. Suppression of c‐Myc enhances p21
WAF1/CIP1
‐mediated G1 cell cycle arrest through the modulation of ERK phosphorylation by ascochlorin. J Cell Biochem 2017; 119:2036-2047. [PMID: 28833404 DOI: 10.1002/jcb.26366] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 08/17/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Yun‐Jeong Jeong
- Research Institute of Biomedical Engineering and Department of MedicineCatholic University of Daegu School of MedicineDaeguRepublic of Korea
| | - Hyang‐Sook Hoe
- Department of Neural Development and DiseaseKorea Brain Research Institute (KBRI)DaeguRepublic of Korea
| | - Hyun‐Ji Cho
- Research Institute of Biomedical Engineering and Department of MedicineCatholic University of Daegu School of MedicineDaeguRepublic of Korea
- Department of Neural Development and DiseaseKorea Brain Research Institute (KBRI)DaeguRepublic of Korea
| | - Kwan‐Kyu Park
- Research Institute of Biomedical Engineering and Department of MedicineCatholic University of Daegu School of MedicineDaeguRepublic of Korea
| | - Dae‐Dong Kim
- Research Institute of Biomedical Engineering and Department of MedicineCatholic University of Daegu School of MedicineDaeguRepublic of Korea
| | - Cheorl‐Ho Kim
- Department of Biological ScienceSungkyunkwan UniversitySuwonKyunggi‐DoRepublic of Korea
| | | | - Dong Wook Kang
- Department of Pharmaceutical Science and TechnologyDaegu Catholic UniversityGyeongsan‐siGyeongbukRepublic of Korea
| | - Sang‐Rae Lee
- National Primate Research Center (NPRC)Korea Research Institute of Bioscience and Biotechnology (KRIBB), OchangChungbukRepublic of Korea
| | - Young‐Chae Chang
- Research Institute of Biomedical Engineering and Department of MedicineCatholic University of Daegu School of MedicineDaeguRepublic of Korea
| |
Collapse
|
42
|
Batistatou A, Kotoula V, Bobos M, Kouvatseas G, Zagouri F, Tsolaki E, Gogas H, Koutras A, Pentheroudakis G, Timotheadou E, Pervana S, Goussia A, Petraki K, Sotiropoulou M, Koletsa T, Razis E, Kosmidis P, Aravantinos G, Papadimitriou C, Pectasides D, Fountzilas G. Correlation of MYC Gene and Protein Status With Breast Cancer Subtypes and Outcome of Patients Treated With Anthracycline-Based Adjuvant Chemotherapy. Pooled Analysis of 2 Hellenic Cooperative Group Phase III Trials. Clin Breast Cancer 2017; 18:53-62.e3. [PMID: 28870680 DOI: 10.1016/j.clbc.2017.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/29/2017] [Accepted: 07/07/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND The prognostic/predictive value of aberrant MYC gene copies and protein expression is not clear in breast cancer. PATIENTS AND METHODS Early breast cancer patients were treated with anthracycline-containing chemotherapy within 2 randomized adjuvant trials. MYC gene and centromere-8 status, as well as Myc protein expression were investigated on 1060 paraffin tumors with fluorescence in situ hybridization and immunohistochemistry, respectively. RESULTS MYC amplification was present in 45% and polysomy-8 in 23% of the tumors. Cytoplasmic staining was observed in 60% and nuclear staining in 26% of the tumors, strongly correlating with each other but not with MYC gene status. MYC gene amplification in the absence of polysomy-8 was associated with adverse disease-free survival (DFS) and overall survival (OS), and remained as an independent unfavorable prognostic factor in multivariate analysis (Wald P = .022 for DFS; P = .032 for OS), whereas patients with MYC amplification and polysomy-8, with polysomy-8 only, and with normal MYC without polysomy-8 performed significantly better compared with those with MYC gene amplification only. Nuclear Myc protein expression benefitted patients treated with paclitaxel (interaction P = .052 for DFS; P = .049 for OS). This interaction remained independently significant in multivariate analysis for OS (overall P = .028). CONCLUSION The effect of MYC gene status on breast cancer patient outcome seems to depend on the underlying chromosomal instability and appears unfavorable for tumors with MYC amplification without polysomy. Nuclear Myc protein expression seems predictive for benefit from adjuvant paclitaxel. These data might aid in the interpretation of relevant findings from large clinical trials.
Collapse
Affiliation(s)
- Anna Batistatou
- Department of Pathology, Ioannina University Hospital, Ioannina, Greece.
| | - Vassiliki Kotoula
- Department of Pathology, Aristotle University of Thessaloniki, School of Health Sciences, Faculty of Medicine, Thessaloniki, Greece; Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Mattheos Bobos
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Flora Zagouri
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Eleftheria Tsolaki
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Helen Gogas
- First Department of Medicine, Laiko General Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Angelos Koutras
- Division of Oncology, Department of Medicine, University Hospital, University of Patras Medical School, Patras, Greece
| | | | - Eleni Timotheadou
- Department of Medical Oncology, Papageorgiou Hospital, Aristotle University of Thessaloniki, School of Health Sciences, Faculty of Medicine, Thessaloniki, Greece
| | - Stavroula Pervana
- Department of Pathology, Papageorgiou Hospital, Thessaloniki, Greece
| | - Anna Goussia
- Department of Pathology, Ioannina University Hospital, Ioannina, Greece
| | | | | | | | - Evangelia Razis
- Third Department of Medical Oncology, Hygeia Hospital, Athens, Greece
| | - Paris Kosmidis
- Second Department of Medical Oncology, Hygeia Hospital, Athens, Greece
| | - Gerasimos Aravantinos
- Second Department of Medical Oncology, Agii Anargiri Cancer Hospital, Athens, Greece
| | - Christos Papadimitriou
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Dimitrios Pectasides
- Oncology Section, Second Department of Internal Medicine, Hippokration Hospital, Athens, Greece
| | - George Fountzilas
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, Greece; Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
43
|
Frey WD, Chaudhry A, Slepicka PF, Ouellette AM, Kirberger SE, Pomerantz WCK, Hannon GJ, Dos Santos CO. BPTF Maintains Chromatin Accessibility and the Self-Renewal Capacity of Mammary Gland Stem Cells. Stem Cell Reports 2017; 9:23-31. [PMID: 28579392 PMCID: PMC5783326 DOI: 10.1016/j.stemcr.2017.04.031] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 04/26/2017] [Accepted: 04/27/2017] [Indexed: 11/16/2022] Open
Abstract
Chromatin remodeling is a key requirement for transcriptional control of cellular differentiation. However, the factors that alter chromatin architecture in mammary stem cells (MaSCs) are poorly understood. Here, we show that BPTF, the largest subunit of the NURF chromatin remodeling complex, is essential for MaSC self-renewal and differentiation of mammary epithelial cells (MECs). BPTF depletion arrests cells at a previously undefined stage of epithelial differentiation that is associated with an incapacity to achieve the luminal cell fate. Moreover, genome-wide analysis of DNA accessibility following genetic or chemical inhibition, suggests a role for BPTF in maintaining the open chromatin landscape at enhancers regions in MECs. Collectively, our study implicates BPTF in maintaining the unique epigenetic state of MaSCs.
Collapse
Affiliation(s)
- Wesley D Frey
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Anisha Chaudhry
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Priscila F Slepicka
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Adam M Ouellette
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Steven E Kirberger
- Department of Chemistry, University of Minnesota, 207 Pleasant Street Southeast, Minneapolis, MN 55455, USA
| | - William C K Pomerantz
- Department of Chemistry, University of Minnesota, 207 Pleasant Street Southeast, Minneapolis, MN 55455, USA
| | - Gregory J Hannon
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge CB20RE, UK.
| | - Camila O Dos Santos
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
44
|
Kourie HR, El Rassy E, Clatot F, de Azambuja E, Lambertini M. Emerging treatments for HER2-positive early-stage breast cancer: focus on neratinib. Onco Targets Ther 2017; 10:3363-3372. [PMID: 28744140 PMCID: PMC5513878 DOI: 10.2147/ott.s122397] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Over the last decades, a better understanding of breast cancer heterogeneity provided tools for a biologically based personalization of anticancer treatments. In particular, the overexpression of the human epidermal growth factor receptor 2 (HER2) by tumor cells provided a specific target in these HER2-positive tumors. The development of the monoclonal antibody trastuzumab, and its approval in 1998 for the treatment of patients with metastatic disease, radically changed the natural history of this aggressive subtype of breast cancer. These findings provided strong support for the continuous research in targeting the HER2 pathway and implementing the development of new anti-HER2 targeted agents. Besides trastuzumab, a series of other anti-HER2 agents have been developed and are currently being explored for the treatment of breast cancer patients, including those diagnosed with early-stage disease. Among these agents, neratinib, an oral tyrosine kinase inhibitor that irreversibly inhibits HER1, HER2, and HER4 at the intracellular level, has shown promising results, including when administered to patients previously exposed to trastuzumab-based treatment. This article aims to review the available data on the role of the HER2 pathway in breast cancer and on the different targeted agents that have been studied or are currently under development for the treatment of patients with early-stage HER2-positive disease with a particular focus on neratinib.
Collapse
Affiliation(s)
- Hampig Raphael Kourie
- Department of Oncology, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Elie El Rassy
- Department of Oncology, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Florian Clatot
- Department of Medical Oncology and IRON/U1245, Centre Henri Becquerel, Rouen, France.,Breast Cancer Translational Research Laboratory
| | - Evandro de Azambuja
- Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Matteo Lambertini
- Breast Cancer Translational Research Laboratory.,Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
45
|
Stucky A, Sedghizadeh PP, Mahabady S, Chen X, Zhang C, Zhang G, Zhang X, Zhong JF. Single-cell genomic analysis of head and neck squamous cell carcinoma. Oncotarget 2017; 8:73208-73218. [PMID: 29069864 PMCID: PMC5641207 DOI: 10.18632/oncotarget.18021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 05/10/2017] [Indexed: 01/16/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) incidence or rates have increased dramatically recently with little improvement in patient outcomes. There is an unmet need in HNSCC to develop reliable molecular markers capable of evaluating patient risks and advising treatments. This review focuses on recent developments in single-cell molecular analysis of cancer, and its applications for HNSCC diagnosis and treatments. For proof of concept, we examined gene expression levels of 62 patients with HNSCC, and correlate the gene expression profiles to single-cell gene expression profiles obtained from a pilot single-cell study of CCR5-positive breast carcinoma cells. The single-cell molecular analyses complemented the lysate data and reveals heterogeneity of oncogenesis pathways with the cancer cell population. Our single-cell molecular analysis indicated that molecular heterogeneity exists in HNSCC and should be addressed in treatment strategy of HNSCC. Single-cell molecular technology can have significant impact on diagnosis, therapeutic decision making, and prognosis of HNSCC.
Collapse
Affiliation(s)
- Andres Stucky
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene, and Division of Biomedical Sciences, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Parish P Sedghizadeh
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene, and Division of Biomedical Sciences, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Susan Mahabady
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene, and Division of Biomedical Sciences, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Xuelian Chen
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene, and Division of Biomedical Sciences, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Cheng Zhang
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene, and Division of Biomedical Sciences, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA.,Department of Hematology and Blood Transfusion, Xinqiao Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Gang Zhang
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene, and Division of Biomedical Sciences, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA.,Department of Oral and Maxillofacial Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Xi Zhang
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene, and Division of Biomedical Sciences, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA.,Department of Hematology and Blood Transfusion, Xinqiao Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Jiang F Zhong
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene, and Division of Biomedical Sciences, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
46
|
Sarrafzadeh S, Geranpayeh L, Tasharrofi B, Soudyab M, Nikpayam E, Iranpour M, Mirfakhraie R, Gharesouran J, Ghafouri-Fard S, Ghafouri-Fard S. Expression Study and Clinical Correlations of MYC and CCAT2 in Breast Cancer Patients. IRANIAN BIOMEDICAL JOURNAL 2017; 21:303-11. [PMID: 28480695 PMCID: PMC5548962 DOI: 10.18869/acadpub.ibj.21.5.303] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background: Colon cancer-associated transcript 2 (CCAT2) is a newly recognized lncRNA transcribed from the 8q24 genomic region. It functions as an oncogene in various types of cancers including breast cancer, in which it affects Wnt/β-catenin pathway. Previous studies have shown a putative interaction between this lncRNA and MYC proto-oncogene. Methods: In the current study, we evaluated the expression of CCAT2 in breast cancer tissues with regards to the expression of its target MYC. In addition, we assessed the relationship between CCAT2 and MYC expression levels in tumor tissues and the clinical prognostic characteristics of breast cancer patients. Results: MYC expression levels were significantly up-regulated in tumor tissues compared with adjacent non-cancerous tissues (ANCTs), while such analysis showed no statistically significant difference between these two tissue types in CCAT2 expression. Starkly increased CCAT2 gene expression levels were found in 12/48 (25%) of cancer tissue samples compared with their corresponding ANCTs. Furthermore, significant inverse correlations were found between CCAT2 expression and stage, as well as lymph node involvement. Besides, a significant inverse correlation was found between the relative MYC expression in tumor tissues compared with their corresponding ANCTs and disease stage. Conclusions: These results highlight the significance of MYC and CCAT2 expressions in the early stages of breast cancer development and suggest a potentially significant role for CCAT2 in a subset of breast cancer patients, which could be applied as a potential therapeutic target in these patients.
Collapse
Affiliation(s)
- Shaghayegh Sarrafzadeh
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Lobat Geranpayeh
- Department of Surgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnoosh Tasharrofi
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Soudyab
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elahe Nikpayam
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Iranpour
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Mirfakhraie
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jalal Gharesouran
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayyeh Ghafouri-Fard
- Department of Community and Preventive Medicine, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
47
|
Li XX, Shi L, Zhou XJ, Wu J, Xia TS, Zhou WB, Sun X, Zhu L, Wei JF, Ding Q. The role of c-Myc-RBM38 loop in the growth suppression in breast cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:49. [PMID: 28399911 PMCID: PMC5387383 DOI: 10.1186/s13046-017-0521-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/31/2017] [Indexed: 01/01/2023]
Abstract
Background RNA-binding protein 38 (RBM38) is a member of the RNA recognition motif (RRM) family of RNA-binding proteins (RBPs). RBM38 often exerts its function by forming regulatory loops with relevant genes. c-Myc is an oncogenic transcription factor that is upregulated in one-third of breast cancers and involved in many cellular processes in this malignancy. In our previous study, RBM38 was identified as a tumor suppressor in breast cancer. In the present study, we investigated the mechanisms underlying the regulation of this tumor suppressor. Methods Lentivirus transfections, Western blotting analysis, qRT-PCR and immunohistochemistry were employed to study the expression of c-Myc and RBM38. Chromatin immunoprecipitation and dual-luciferase reporter assays were performed to investigate the direct relationship between c-Myc protein and the RBM38 gene. RNA immunoprecipitation combined with dual-luciferase reporter assays was conducted to confirm the direct relationship between the RBM38 protein and the c-Myc transcript. Results Knockdown of c-Myc increased RBM38 expression by binding directly to specific DNA sequences (5′-CACGTG-3′), known as the E-box motif, in the promoter region of RBM38 gene. Additionally, RBM38 destabilized the c-Myc transcript by directly targeting AU-rich elements (AREs) in the 3′-untranslated region (3′-UTR) of c-Myc mRNA to suppress c-Myc expression. Moreover, specific inhibitors of c-Myc transcriptional activity inhibited RBM38-induced suppression of growth, implying that RBM38 acts as a tumor suppressor via a mechanism that depends, at least partially, on the reduction of c-Myc expression in breast cancer. Conclusions RBM38 and c-Myc form a unique mutually antagonistic RBM38-c-Myc loop in breast cancer. Electronic supplementary material The online version of this article (doi:10.1186/s13046-017-0521-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiao-Xia Li
- Jiangsu Breast Disease Center, the First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Liang Shi
- Jiangsu Breast Disease Center, the First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Xu-Jie Zhou
- Jiangsu Breast Disease Center, the First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Jing Wu
- Jiangsu Breast Disease Center, the First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Tian-Song Xia
- Jiangsu Breast Disease Center, the First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Wen-Bin Zhou
- Jiangsu Breast Disease Center, the First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Xi Sun
- Jiangsu Breast Disease Center, the First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Lei Zhu
- Jiangsu Breast Disease Center, the First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Ji-Fu Wei
- Research Division of Clinical Pharmacology, the First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China.
| | - Qiang Ding
- Jiangsu Breast Disease Center, the First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China.
| |
Collapse
|
48
|
Mahas A, Potluri K, Kent MN, Naik S, Markey M. Copy number variation in archival melanoma biopsies versus benign melanocytic lesions. Cancer Biomark 2017; 16:575-97. [PMID: 27002761 DOI: 10.3233/cbm-160600] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Skin melanocytes can give rise to benign and malignant neoplasms. Discrimination of an early melanoma from an unusual/atypical benign nevus can represent a significant challenge. However, previous studies have shown that in contrast to benign nevi, melanoma demonstrates pervasive chromosomal aberrations. OBJECTIVE This substantial difference between melanoma and benign nevi can be exploited to discriminate between melanoma and benign nevi. METHODS Array-comparative genomic hybridization (aCGH) is an approach that can be used on DNA extracted from formalin-fixed paraffin-embedded (FFPE) tissues to assess the entire genome for the presence of changes in DNA copy number. In this study, high resolution, genome-wide single-nucleotide polymorphism (SNP) arrays were utilized to perform comprehensive and detailed analyses of recurrent copy number aberrations in 41 melanoma samples in comparison with 21 benign nevi. RESULTS We found statistically significant copy number gains and losses within melanoma samples. Some of the identified aberrations are previously implicated in melanoma. Moreover, novel regions of copy number alterations were identified, revealing new candidate genes potentially involved in melanoma pathogenesis. CONCLUSIONS Taken together, these findings can help improve melanoma diagnosis and introduce novel melanoma therapeutic targets.
Collapse
Affiliation(s)
- Ahmed Mahas
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH, USA
| | - Keerti Potluri
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH, USA
| | - Michael N Kent
- Department of Dermatology, Wright State University Boonshoft School of Medicine, Dayton, OH, USA.,Dermatopathology Laboratory of Central States, Dayton, OH, USA
| | - Sameep Naik
- Dermatopathology Laboratory of Central States, Dayton, OH, USA
| | - Michael Markey
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH, USA
| |
Collapse
|
49
|
Myc suppresses tumor invasion and cell migration by inhibiting JNK signaling. Oncogene 2017; 36:3159-3167. [PMID: 28068320 DOI: 10.1038/onc.2016.463] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 10/24/2016] [Accepted: 11/07/2016] [Indexed: 01/01/2023]
Abstract
Tumor metastasis, but not primary overgrowth, is the leading cause of mortality for cancer patients. During the past decade, Drosophila melanogaster has been well-accepted as an excellent model to address the intrinsic mechanism of different aspects of cancer progression, ranging from tumor initiation to metastasis. In a genetic screen performed in Drosophila, aiming to find novel modulators of tumor invasion, we identified the oncoprotein Myc as a negative regulator. While expression of Myc dramatically blocks tumor invasion and cell migration, loss of Myc promotes cell migration in vivo. The activity of Myc is further enhanced by the co-expression of its transcription partner Max. Mechanistically, we found Myc/Max directly upregulates the transcription of puc, which encodes an inhibitor of JNK signaling crucial for tumor invasion and cell migration. Furthermore, we demonstrated that human cMyc potently suppresses JNK-dependent cell invasion and migration in both Drosophila and lung adenocarcinoma cell lines. These findings provide novel molecular insights into Myc-mediated cancer progression and raise the noteworthy problem in therapeutic strategies as inhibiting Myc might conversely accelerate tumor metastasis.
Collapse
|
50
|
Bryson BL, Junk DJ, Cipriano R, Jackson MW. STAT3-mediated SMAD3 activation underlies Oncostatin M-induced Senescence. Cell Cycle 2016; 16:319-334. [PMID: 27892764 DOI: 10.1080/15384101.2016.1259037] [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] [Indexed: 12/16/2022] Open
Abstract
Cytokines in the developing tumor microenvironment (TME) can drive transformation and subsequent progression toward metastasis. Elevated levels of the Interleukin-6 (IL-6) family cytokine Oncostatin M (OSM) in the breast TME correlate with aggressive, metastatic cancers, increased tumor recurrence, and poor patient prognosis. Paradoxically, OSM engages a tumor-suppressive, Signal Transducer and Activator of Transcription 3 (STAT3)-dependent senescence response in normal and non-transformed human mammary epithelial cells (HMEC). Here, we identify a novel link between OSM-activated STAT3 signaling and the Transforming Growth Factor-β (TGF-β) signaling pathway that engages senescence in HMEC. Inhibition of functional TGF-β/SMAD signaling by expressing a dominant-negative TGF-β receptor, treating with a TGF-β receptor inhibitor, or suppressing SMAD3 expression using a SMAD3-shRNA prevented OSM-induced senescence. OSM promoted a protein complex involving activated-STAT3 and SMAD3, induced the nuclear localization of SMAD3, and enhanced SMAD3-mediated transcription responsible for senescence. In contrast, expression of MYC (c-MYC) from a constitutive promoter abrogated senescence and strikingly, cooperated with OSM to promote a transformed phenotype, epithelial-mesenchymal transition (EMT), and invasiveness. Our findings suggest that a novel STAT3/SMAD3-signaling axis is required for OSM-mediated senescence that is coopted during the transformation process to confer aggressive cancer cell properties. Understanding how developing cancer cells bypass OSM/STAT3/SMAD3-mediated senescence may help identify novel targets for future "pro-senescence" therapies aiming to reengage this hidden tumor-suppressive response.
Collapse
Affiliation(s)
- Benjamin L Bryson
- a Department of Pathology , School of Medicine, Case Western Reserve University , Cleveland , OH , USA
| | - Damian J Junk
- a Department of Pathology , School of Medicine, Case Western Reserve University , Cleveland , OH , USA
| | - Rocky Cipriano
- a Department of Pathology , School of Medicine, Case Western Reserve University , Cleveland , OH , USA
| | - Mark W Jackson
- a Department of Pathology , School of Medicine, Case Western Reserve University , Cleveland , OH , USA.,b Case Comprehensive Cancer Center , Case Western Reserve University , Cleveland , OH , USA
| |
Collapse
|