1
|
Shi Y, Zhang Y, Yu S, Luo J, Pan Z, Wang X, Tian J. Activation of kappa opioid receptor (KOR) inhibits estrogen receptor (ER)-positive breast cancer through the KOR-ER-XBP1 pathway. Biomed Pharmacother 2023; 167:115462. [PMID: 37708692 DOI: 10.1016/j.biopha.2023.115462] [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: 03/02/2023] [Revised: 08/25/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023] Open
Abstract
Opioids are commonly used in patients with breast cancer (BC), both for perioperative analgesia and for the relief of chronic cancer pain. Studies have suggested a potential association of opioid receptors (ORs) with the prognosis of BC patients. However, the exact roles of different ORs remain poorly understood. In this study, we found that κ opioid receptor (KOR) was the only OR (among the four types of ORs) that was significantly decreased in BC tumor tissues compared with peritumoral normal tissues. In addition, decreased expression of KOR correlated with poor clinical outcomes in patients with estrogen receptor (ER)-positive BC. In vitro studies confirmed the anti-tumor effects of KOR agonists in ER-positive MCF-7 and T47D cells by showing that activation of KOR significantly inhibited cellular proliferation and promoted apoptosis. Using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and protein-protein interaction network (PPI) analysis, we found that KOR-ER-XBP1 was the potential downstream signaling pathway mediating the anti-tumor effects of KOR agonist. Finally, the role of XBP1 was confirmed as KOR activation-induced increase in the proliferative and monoclonal formation abilities of ER-positive BC cells were both significantly abolished after silencing of XBP1. These findings provide us a better understanding of the roles of different ORs in BC, identifying KOR agonists as better opioids than traditional μ opioid receptor (MOR) agonists for providing analgesia in ER-positive BC patients owing to their association with better prognosis.
Collapse
Affiliation(s)
- Yumiao Shi
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Yiqi Zhang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Suyang Yu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Jiamei Luo
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Zhiying Pan
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Xiaoqiang Wang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China.
| | - Jie Tian
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
2
|
X-Box Binding Protein 1 (XBP1): A Potential Role in Chemotherapy Response, Clinical Pathologic Features, Non-Inflamed Tumour Microenvironment for Breast Cancer. Biosci Rep 2022; 42:231292. [PMID: 35543228 PMCID: PMC9202509 DOI: 10.1042/bsr20220225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/17/2022] [Accepted: 04/04/2022] [Indexed: 12/09/2022] Open
Abstract
X-box binding protein 1 (XBP1) is mainly expressed in breast cancer (BC) in human cancers. Its tumorigenesis and favourable prognosis are contradictory, and its essential role in chemotherapeutic response and immunosuppression is unknown in BC. The study firstly identified XBP1 who received neoadjuvant chemotherapy (NAC) from GSE25055 and GSE24460. Associations between XBP1 expression and clinicopathological characteristics was investigated using Oncomine, TCGA, UALCAN and bc-GenExMiner. The prognostic value of XBP1 was assessed using the Kaplan–Meier Plotter, bc-GenExMiner, GSE25055, and GSE25056. Furthermore, we systematically correlated XBP1 and immunological characteristics in the BC tumour microenvironment (TME) using TISIDB, TIMER, GSE25055, GSE25056 and TCGA dataset. Finally, an essential role of XBP1 in chemotherapy response was evaluated based on GSE25055, GSE25065, GSE24460, GSE5846, ROC Plotter and CELL databases. Furthermore, XBP1 mRNA expression levels were obviously highest in BC among human cancers and were significantly related to a good prognosis. In addition, XBP1 mRNA and protein levels were higher in the luminal subtype than in normal tissues and basal-like subtype, which might be attributed to membrane transport-related processes. Apart from BC, negative immunological correlations of XBP1 were not observed in other malignancies. XBP1 might shape the non-inflamed TME in BC. Finally, XBP1 expression was higher in chemo-resistive than chemo-sensitive cases, it had a predictive value and could independently predict chemotherapy response in BC patients receiving NAC. Our study suggests that the essential role of XBP1 in clinical pathologic features, non-inflamed TME, chemotherapy response in BC.
Collapse
|
3
|
SOX2OT Long Noncoding RNA Is Regulated by the UPR in Oestrogen Receptor-Positive Breast Cancer. SCI 2021. [DOI: 10.3390/sci3020026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Endoplasmic reticulum (ENR) stress perturbs cell homeostasis and induces the unfolded protein response (UPR). In breast cancer, this process is activated by oestrogen deprivation and is associated with tamoxifen resistance. We present evidence that the transcription factor SOX2 and the long noncoding RNA SOX2 overlapping transcript (SOX2OT) are upregulated in oestrogen receptor-positive (ER+) breast cancer and in response to oestrogen deprivation. We examined the effect of the UPR on SOX2 and SOX2OT expression and the effect of SOX2OT on UPR pathways in breast cancer cell lines. The induction of the UPR by thapsigargin or glucose deprivation upregulates SOX2OT expression. This upregulation is also shown with the anti-oestrogen 4OH-tamoxifen and mTOR inhibitor everolimus in ER + breast cancer cells that are sensitive to oestrogen deprivation or everolimus treatment. SOX2OT overexpression decreased BiP and PERK expression. This effect of SOX2OT overexpression was confirmed on BiP and PERK pathway by q-PCR. Our results show that a long noncoding RNA regulates the UPR and evince a new function of SOX2OT as a participant of ENR stress reprogramming of breast cancer cells.
Collapse
|
4
|
Huang Y, Jiang L, Wei G. Circ_0006168 Promotes the Migration, Invasion and Proliferation of Esophageal Squamous Cell Carcinoma Cells via miR-516b-5p-Dependent Regulation of XBP1. Onco Targets Ther 2021; 14:2475-2488. [PMID: 33883902 PMCID: PMC8053607 DOI: 10.2147/ott.s293180] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/22/2021] [Indexed: 12/18/2022] Open
Abstract
Background Circular RNAs (circRNAs) exert important roles in carcinogenesis. Here, we aimed to uncover the working mechanism of circ_0006168 in esophageal squamous cell carcinoma (ESCC) development. Methods Western blot assay and real-time quantitative polymerase chain reaction (RT-qPCR) were used to determine protein and RNA expression, respectively. Wound healing assay and transwell migration assay were performed to assess cell migration ability, whereas cell invasion ability was evaluated by transwell invasion assay. 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and colony formation assay were utilized to analyze cell proliferation ability. Xenograft tumor model was utilized to assess the role of X-box binding protein 1 (XBP1) in xenograft tumor growth in vivo. Dual-luciferase reporter assay, RNA immunoprecipitation (RIP) assay and RNA pull down assay were used to verify intermolecular interactions. Results XBP1 silencing suppressed the migration, invasion and proliferation of ESCC cells in vitro and restrained the xenograft tumor growth in vivo. MicroRNA-516b-5p (miR-516b-5p) interacted with the 3ʹ untranslated region (3ʹUTR) of XBP1 in ESCC cells. MiR-516b-5p overexpression inhibited the proliferation and motility of ESCC cells. MiR-516b-5p was a molecular target of circ_0006168 in ESCC cells. The interference of circ_0006168 restrained the motility and proliferation of ESCC cells. Circ_0006168 acted as miR-516b-5p sponge to up-regulate XBP1 expression in ESCC cells. MiR-516b-5p silencing or the accumulation of XBP1 largely rescued the proliferation ability and motility in circ_0006168-silenced ESCC cells. Conclusion In conclusion, circ_0006168 contributed to ESCC development through promoting the proliferation and motility of ESCC cells via mediating miR-516b-5p/XBP1 axis.
Collapse
Affiliation(s)
- Yunhe Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Lei Jiang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Guangxia Wei
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| |
Collapse
|
5
|
Wang Q, Zhao Y, Zheng H, Wang Q, Wang W, Liu B, Han H, Zhang L, Chen K. CCDC170 affects breast cancer apoptosis through IRE1 pathway. Aging (Albany NY) 2020; 13:1332-1356. [PMID: 33291081 PMCID: PMC7835043 DOI: 10.18632/aging.202315] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/03/2020] [Indexed: 01/29/2023]
Abstract
Genome-wide association studies have revealed that multiple single-nucleotide polymorphisms in the intergenic region between estrogen receptor 1 and coiled-coil domain containing 170 (CCDC170) are associated with breast cancer risk. We performed microarray and bioinformatics analyses to identify genes that were induced upon CCDC170 overexpression, and confirmed our findings by evaluating paraffin-embedded breast cancer tissues and conducting cellular assays. In CCDC170-overexpressing MCF7 breast cancer cells, microarray analyses revealed that inositol-requiring enzyme 1 (IRE1) was the most elevated gene in enriched pathways. In breast cancer tissues, IRE1 expression correlated positively with CCDC170 and X-box binding protein 1 expression at both the mRNA and protein levels. In a survival analysis, patients with higher CCDC170 levels exhibited better disease-free survival. Western blotting indicated that overexpressing CCDC170 in MCF7 cells increased protein levels of IRE1α, estrogen receptor α and X-box binding protein 1, while silencing CCDC170 reduced them. CCDC170 overexpression promoted apoptosis in MCF7 cells, and this effect was more obvious under endoplasmic reticulum stress. MCF7 cells overexpressing CCDC170 were more sensitive to paclitaxel. Our study showed that higher CCDC170 expression is associated with a better prognosis in breast cancer patients and that CCDC170 may promote apoptosis through the IRE1α pathway.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China.,Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Molecular Cancer Epidemiology, Tianjin 300060, P.R. China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, P.R. China
| | - Yanrui Zhao
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China.,Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Molecular Cancer Epidemiology, Tianjin 300060, P.R. China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, P.R. China
| | - Hong Zheng
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China.,Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Molecular Cancer Epidemiology, Tianjin 300060, P.R. China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, P.R. China
| | - Qinghua Wang
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China.,Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Molecular Cancer Epidemiology, Tianjin 300060, P.R. China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, P.R. China
| | - Wei Wang
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China.,Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Molecular Cancer Epidemiology, Tianjin 300060, P.R. China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, P.R. China
| | - Ben Liu
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China.,Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Molecular Cancer Epidemiology, Tianjin 300060, P.R. China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, P.R. China
| | - Hongwei Han
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China.,Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Molecular Cancer Epidemiology, Tianjin 300060, P.R. China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, P.R. China
| | - Lina Zhang
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China.,Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Molecular Cancer Epidemiology, Tianjin 300060, P.R. China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, P.R. China.,Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China.,Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Molecular Cancer Epidemiology, Tianjin 300060, P.R. China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, P.R. China
| |
Collapse
|
6
|
Lyu L, Wang M, Zheng Y, Tian T, Deng Y, Xu P, Lin S, Yang S, Zhou L, Hao Q, Wu Y, Dai Z, Kang H. Overexpression of FAM234B Predicts Poor Prognosis in Patients with Luminal Breast Cancer. Cancer Manag Res 2020; 12:12457-12471. [PMID: 33299353 PMCID: PMC7721111 DOI: 10.2147/cmar.s280009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/12/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Family with sequence similarity 234 member B (FAM234B), a protein-coding gene, is mainly expressed in brain tissues. Its clinical significance and biological function in tumors, especially in breast cancer (BC), have not been elucidated. METHODS We firstly investigated the expression pattern of FAM234B at the mRNA and protein levels using Oncomine, TCGA portal, GEPIA, TIMER, HPA, and UALCAN databases, then applied bc-GenExMiner to assess the associations between expression level of FAM234B and clinicopathological features of BC. Besides, we also verified the expression of FAM234B expression in clinical BC samples using qRT-PCR. Subsequently, GEPIA, bc-GenExMiner, and TIMER databases were used to analyze the prognostic significance of FAM234B in all BC and different molecular subtypes. Finally, we conducted co-expression analysis and gene set enrichment analysis (GSEA). Additionally, we explored the regulatory mechanism of FAM234B in BC. RESULTS Both bioinformatics analysis and experimental verification confirmed that the FAM234B expression was significantly higher at the mRNA and protein levels in luminal BC tissues than in adjacent normal tissues. High FAM234B expression was significantly correlated with older age, estrogen receptor-positive, progesterone receptor-positive, human epidermal growth factor receptor 2-negative, wild-type p53, low Nottingham prognostic index, low Scarff-Bloom-Richardson grade, lymph node metastasis positivity, and high tumor stage. Moreover, survival analysis indicated that high FAM234B expression was significantly related to a worse prognosis in patients with luminal BC. GSEA indicated that FAM234B was positively related to membrane transport process and negatively associated with immune response function. Besides, mechanism exploration indicated that pseudogene HTR7P1 might act as endogenous RNA to compete with has-miR-1271-5p or has-miR-381-3p for binding to FAM234B, thereby upregulating the expression of FAM234B in luminal BC. CONCLUSION Our results suggest that FAM234B may be a candidate therapeutic target or prognostic marker for luminal breast cancer.
Collapse
Affiliation(s)
- Lijuan Lyu
- Department of Oncology, The 2nd Affiliated Hospital of Xi’an Jiaotong University, Xi’an710004, People’s Republic of China
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, People’s Republic of China
| | - Meng Wang
- Department of Oncology, The 2nd Affiliated Hospital of Xi’an Jiaotong University, Xi’an710004, People’s Republic of China
| | - Yi Zheng
- Department of Oncology, The 2nd Affiliated Hospital of Xi’an Jiaotong University, Xi’an710004, People’s Republic of China
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, People’s Republic of China
| | - Tian Tian
- Department of Oncology, The 2nd Affiliated Hospital of Xi’an Jiaotong University, Xi’an710004, People’s Republic of China
| | - Yujiao Deng
- Department of Oncology, The 2nd Affiliated Hospital of Xi’an Jiaotong University, Xi’an710004, People’s Republic of China
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, People’s Republic of China
| | - Peng Xu
- Department of Oncology, The 2nd Affiliated Hospital of Xi’an Jiaotong University, Xi’an710004, People’s Republic of China
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, People’s Republic of China
| | - Shuai Lin
- Department of Oncology, The 2nd Affiliated Hospital of Xi’an Jiaotong University, Xi’an710004, People’s Republic of China
| | - Si Yang
- Department of Oncology, The 2nd Affiliated Hospital of Xi’an Jiaotong University, Xi’an710004, People’s Republic of China
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, People’s Republic of China
| | - Linghui Zhou
- Department of Oncology, The 2nd Affiliated Hospital of Xi’an Jiaotong University, Xi’an710004, People’s Republic of China
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, People’s Republic of China
| | - Qian Hao
- Department of Oncology, The 2nd Affiliated Hospital of Xi’an Jiaotong University, Xi’an710004, People’s Republic of China
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, People’s Republic of China
| | - Ying Wu
- Department of Oncology, The 2nd Affiliated Hospital of Xi’an Jiaotong University, Xi’an710004, People’s Republic of China
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, People’s Republic of China
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, People’s Republic of China
| | - Huafeng Kang
- Department of Oncology, The 2nd Affiliated Hospital of Xi’an Jiaotong University, Xi’an710004, People’s Republic of China
| |
Collapse
|
7
|
Liu R, Wu Z, Zhang Y, Miao X, Zou Q, Yuan Y, Li D, Yang Z. Prognostic and Clinicopathological Significance of X-Box-Binding Protein 1 and N-Acetyltransferase 1 in Gallbladder Cancer. Front Oncol 2020; 10:1124. [PMID: 32793479 PMCID: PMC7393250 DOI: 10.3389/fonc.2020.01124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 06/04/2020] [Indexed: 12/29/2022] Open
Abstract
Background: X-box-binding protein 1 (XBP1) and N-acetyltransferase 1 (NAT1) are involved in oncogenesis and progression of many human cancer types. However, the roles of XBP1 and NAT1 in gallbladder cancer (GBC) are never reported. Methods: We examined XBP1 and NAT1 expression in GBC and matched adjacent non-tumor tissues via Western blotting. Then, we assayed XBP1 and NAT1 expression in 215 GBCs, including 69 squamous cell/adenosquamous carcinomas (SC/ASCs) and 146 adenocarcinomas (ACs) with immunohistochemistry. Their prognostic and clinicopathological significance was further evaluated using the χ2 test or Fisher's exact test, Kaplan–Meier univariate survival analysis, and log-rank tests. Results: XBP1 expression was upregulated, and NAT1 expression was downregulated in GBC. Immunohistochemical results showed that XBP1 expression was negatively associated with NAT1 expression in GBC, including SC/ASC and AC. The rate of patients with an age of more than 45 years, positivity of lymph node metastasis, and invasion were significantly higher in SC/ASC than those in AC (all P < 0.05). The percentage of XBP1-positive and NAT1-negative expression was significantly higher in the cases with poor differentiation, advanced tumor, nodes, and metastases (TNM) stage, lymph node metastasis, invasion, and only receiving biopsy in GBC, SC/ASC, and AC (all P < 0.05). XBP1-positive and NAT1-negative expression was positively related to larger tumor size (>3 cm) in GBC and AC. There was a negative association between XBP1 and NAT1 expression in GBC, SC/ASC, and AC (all P < 0.05). Positive XBP1 and negative NAT1 expression was closely associated with decreased overall survival in GBC, SC/ASC, and AC patients (all P < 0.05). The multivariate Cox regression analysis showed that positive XBP1 or negative NAT1 expression was an independent factor for poor prognosis in gallbladder SC/ASC and AC patients. Conclusions: This study indicates that positive XBP1 and negative NAT1 expression are closely associated with the clinicopathological and biological behaviors and poor prognosis in GBC.
Collapse
Affiliation(s)
- Rushi Liu
- Department of Medical Laboratory, Immunodiagnostic Reagents Engineering Research Center of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Zhengchun Wu
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research, Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yuanfang Zhang
- Department of Medical Laboratory, Immunodiagnostic Reagents Engineering Research Center of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Xiongying Miao
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research, Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiong Zou
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Yuan
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Daiqiang Li
- Department of Pathology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhulin Yang
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research, Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
8
|
SOX2OT Long Noncoding RNA Is Regulated by the UPR in Oestrogen Receptor-Positive Breast Cancer. SCI 2020. [DOI: 10.3390/sci2020024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Endoplasmic reticulum (ER) stress perturbs cell homeostasis and induces the unfolded protein response (UPR). In breast cancer, this process is activated by oestrogen deprivation and is associated with tamoxifen resistance. We present evidence that the transcription factor SOX2 and the long noncoding RNA SOX2 overlapping transcript (SOX2OT) are up-regulated in oestrogen receptor-positive (ER+) breast cancer and in response to oestrogen deprivation. We examined the effect of the UPR on SOX2 and SOX2OT expression, and the effect of SOX2OT on UPR pathways in breast cancer cell lines. The induction of the UPR by thapsigargin or glucose deprivation up-regulates SOX2OT expression. This up-regulation is also shown with the anti-oestrogen 4OH-tamoxifen and mTOR inhibitor everolimus in ER + breast cancer cells that are sensitive to oestrogen deprivation or everolimus treatment. SOX2OT overexpression decreased BiP and PERK expression. This effect of SOX2OT overexpression was confirmed on BiP and PERK pathway by q-PCR. Our results show that a long noncoding RNA regulates the UPR and evince a new function of SOX2OT as a participant of ER stress reprogramming of breast cancer cells.
Collapse
|
9
|
Sereff SB, Daniels MW, Wittliff JL. Relationships of protein biomarkers of the urokinase plasminogen activator system with expression of their cognate genes in primary breast carcinomas. J Clin Lab Anal 2019; 33:e22982. [PMID: 31359505 PMCID: PMC6868412 DOI: 10.1002/jcla.22982] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 06/30/2019] [Accepted: 07/01/2019] [Indexed: 01/20/2023] Open
Abstract
Background uPA, its receptor uPAR, and inhibitors PAI‐1 and PAI‐2 play key roles in membrane remodeling/invasion and in predicting response to chemotherapy. We identified novel relationships of these biomarkers with ER/PR that indicate clinical utility for assessing breast carcinoma outcomes. Methods Retrospective studies were performed with de‐identified results of (a) uPA, uPAR, and PAI‐1; (b) estrogen (ER) and progestin receptor (PR); and (c) clinical outcomes. Relative expression of 22 000 genes from microarray of RNA from LCM‐procured breast cancer cells was used with R Studio version 3.4.1. Results Primary ER/PR status was related to uPA, uPAR, or PAI‐1 levels. ER− or PR− cancers expressed elevated uPA, uPAR, and PAI2 mRNA compared to ER+ or PR+ cells. Inverse relationships between ER/PR protein and expression of uPA, uPAR, and PAI‐2 were observed, whereas HER2 status was unrelated. qPCR analyses showed RERG and NQO‐1 expressions were elevated in uPA− lesions, while CD34 and EDG‐1 were elevated in uPAR− cancers. ERBB4 was overexpressed in PAI‐1+ carcinomas. Cox regression analyses revealed relationships of ER/PR status and uPA system members with regard to clinical outcomes of breast cancer. Conclusions uPA, uPAR, PAI1, or PAI2 expression was increased in either ER− or PR− cancers similar to that of protein content in ER−/PR− carcinomas, suggesting sex hormones regulate the uPA system in breast cancer. Results revealed protein content of uPA system members was related to ER/PR status of primary lesions. Use of LCM‐procured carcinoma cells uncovered relationships between expression of known cancer−associated genes and protein content of uPA system members. Collectively, results indicate evaluation of ER and PR protein of primary breast cancers combined with analyses of uPA, uPAR, and PAI‐1 protein content improves assessment of clinical outcomes.
Collapse
Affiliation(s)
- Seth B Sereff
- Department of Biochemistry & Molecular Genetics, University of Louisville, Louisville, Kentucky.,Institute for Molecular Diversity & Drug Design, University of Louisville, Louisville, Kentucky
| | - Michael W Daniels
- Department of Biochemistry & Molecular Genetics, University of Louisville, Louisville, Kentucky.,Department of Biostatistics, University of Colorado at Aurora, Aurora, Colorado
| | - James L Wittliff
- Department of Biochemistry & Molecular Genetics, University of Louisville, Louisville, Kentucky.,Institute for Molecular Diversity & Drug Design, University of Louisville, Louisville, Kentucky
| |
Collapse
|
10
|
Musah S, Chen J, Schlueter C, Humphrey DM, Stocke K, Hoyle MI, Hoyle GW. Inhibition of chlorine-induced airway fibrosis by budesonide. Toxicol Appl Pharmacol 2018; 363:11-21. [PMID: 30189237 DOI: 10.1016/j.taap.2018.08.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 12/15/2022]
Abstract
Chlorine is a chemical threat agent that can be harmful to humans. Acute inhalation of high levels of chlorine results in the death of airway epithelial cells and can lead to persistent adverse effects on respiratory health, including airway remodeling and hyperreactivity. We previously developed a mouse chlorine exposure model in which animals developed inflammation and fibrosis in large airways. In the present study, examination by laser capture microdissection of developing fibroproliferative lesions in FVB/NJ mice exposed to 240 ppm-h chlorine revealed upregulation of genes related to macrophage function. Treatment of chlorine-exposed mice with the corticosteroid drug budesonide daily for 7 days (30-90 μg/mouse i.m.) starting 1 h after exposure prevented the influx of M2 macrophages and the development of airway fibrosis and hyperreactivity. In chlorine-exposed, budesonide-treated mice 7 days after exposure, large airways lacking fibrosis contained extensive denuded areas indicative of a poorly repaired epithelium. Damaged or poorly repaired epithelium has been considered a trigger for fibrogenesis, but the results of this study suggest that inflammation is the ultimate driver of fibrosis in our model. Examination at later times following 7-day budesonide treatment showed continued absence of fibrosis after cessation of treatment and regrowth of a poorly differentiated airway epithelium by 14 days after exposure. Delay in the start of budesonide treatment for up to 2 days still resulted in inhibition of airway fibrosis. Our results show the therapeutic potential of budesonide as a countermeasure for inhibiting persistent effects of chlorine inhalation and shed light on mechanisms underlying the initial development of fibrosis following airway injury.
Collapse
Affiliation(s)
- Sadiatu Musah
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, United States
| | - Jing Chen
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, United States
| | - Connie Schlueter
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, United States
| | - David M Humphrey
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, United States
| | - Kendall Stocke
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, United States
| | - Mona I Hoyle
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, United States
| | - Gary W Hoyle
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, United States.
| |
Collapse
|
11
|
Wittliff JL, Sereff SB, Daniels MW. Expression of Genes for Methylxanthine Pathway-Associated Enzymes Accompanied by Sex Steroid Receptor Status Impacts Breast Carcinoma Progression. Discov Oncol 2017; 8:298-313. [PMID: 28971320 DOI: 10.1007/s12672-017-0309-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/11/2017] [Indexed: 01/10/2023] Open
Abstract
Consumption of methylxanthine alkaloids appears to induce activities by antagonizing adenosine receptors, implicated in breast cancer behavior in vitro. Our goal was to evaluate expression of genes for methylxanthine receptors and metabolizing enzymes to assess risk of breast carcinoma recurrence. Clinical outcomes, estrogen/progestin receptor results, and gene expression assays guided selection. RNA was isolated from laser capture microdissection-procured carcinoma cells for microarray using established protocols. Gene expression levels of eight methylxanthine receptors, eight metabolizing enzymes, and various phosphodiesterases were retrieved from microarray results. Univariable Cox regressions and Kaplan-Meier plots were determined for each gene with R software. Individually, lower expressions of PDE4A, CYP2A6, or CYP2E were related to decreased progression-free survival (PFS) and overall survival (OS). PDE1A over-expression predicted decreased PFS and OS. ADORA2B and RYR1 over-expressions predicted diminished OS. ER+ cancers exhibited lower ADORA1, ADORA2B, and RYR1 and elevated PDE4A, CYP2A6, and CYP2E expressions. Of PR+ carcinomas, diminished ADORA2B and RYR1 and elevated expressions of ADORA3, PDE4A, CYP2C8, and CYP2E were noted. Least absolute shrinkage and selection operator (LASSO) revealed that CYP2E, PDE1A, and PDE4A expressions collectively predicted PFS whereas ADORA1, CYP2E, PDE1A, PDE1B, and PDE4A expressions jointly predicted OS. Models were clinically significant when validated externally. LASSO also derived a six-gene model and five-gene model that predicted PFS of ER- or PR- carcinomas, respectively. Similarly, five-gene and four-gene models predicted OS in ER- or PR- carcinomas, respectively. Collectively, expression of genes involved in methylxanthine action and metabolism in single-cell types predicted clinical outcomes of breast carcinoma indicating promise for developing diagnostics and design of new therapeutics.
Collapse
Affiliation(s)
- James L Wittliff
- Department of Biochemistry & Molecular Genetics, University of Louisville, HSC Bldg. A, Room 512A, Louisville, KY, 40292, USA.
- Institute for Molecular Diversity & Drug Design, University of Louisville, Louisville, KY, 40292, USA.
| | - Seth B Sereff
- Department of Biochemistry & Molecular Genetics, University of Louisville, HSC Bldg. A, Room 512A, Louisville, KY, 40292, USA
- Institute for Molecular Diversity & Drug Design, University of Louisville, Louisville, KY, 40292, USA
| | - Michael W Daniels
- Institute for Molecular Diversity & Drug Design, University of Louisville, Louisville, KY, 40292, USA
- Department of Bioinformatics & Biostatistics, University of Louisville, Louisville, KY, 40292, USA
| |
Collapse
|
12
|
Jiang D, Turner B, Song J, Li R, Diehn M, Le QT, Khatri P, Koong AC. Comprehensive Analysis of the Unfolded Protein Response in Breast Cancer Subtypes. JCO Precis Oncol 2017; 2017. [PMID: 29888341 DOI: 10.1200/po.16.00073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Purpose Triple-negative breast cancers (TNBCs) are associated with a worse prognosis and patients with TNBC have fewer therapeutic options than patients with non-TNBC. Recently, the IRE1α-XBP1 branch of the unfolded protein response (UPR) was implicated in TNBC prognosis on the basis of a relatively small patient population, suggesting the diagnostic and therapeutic value of this pathway in TNBCs. In addition, the IRE1α-XBP1 and hypoxia-induced factor 1 α (HIF1α) pathways have been identified as interacting partners in TNBC, suggesting a novel mechanism of regulation. To comprehensively evaluate and validate these findings, we investigated the relative activities and relevance to patient survival of the UPR and HIF1α pathways in different breast cancer subtypes in large populations of patients. Materials and Methods We performed a comprehensive analysis of gene expression and survival data from large cohorts of patients with breast cancer. The patients were stratified based on the average expression of the UPR or HIF1α gene signatures. Results We identified a strong positive association between the XBP1 gene signature and estrogen receptor-positive status or the HIF1α gene signature, as well as the predictive value of the XBP1 gene signature for survival of patients who are estrogen receptor negative, or have TNBC or HER2+. In contrast, another important UPR branch, the ATF4/CHOP pathway, lacks prognostic value in breast cancer in general. Activity of the HIF1α pathway is correlated with patient survival in all the subtypes evaluated. Conclusion These findings clarify the relevance of the UPR pathways in different breast cancer subtypes and underscore the potential therapeutic importance of the IRE1α-XBP1 branch in breast cancer treatment.
Collapse
Affiliation(s)
- Dadi Jiang
- Stanford University School of Medicine, Stanford, CA
| | | | - Jie Song
- Stanford University School of Medicine, Stanford, CA
| | - Ruijiang Li
- Stanford University School of Medicine, Stanford, CA
| | | | - Quynh-Thu Le
- Stanford University School of Medicine, Stanford, CA
| | - Purvesh Khatri
- Stanford Center for Biomedical Informatics Research, Stanford University, Stanford, CA
| | | |
Collapse
|
13
|
Daniels MW, Brock GN, Wittliff JL. Clinical outcomes linked to expression of gene subsets for protein hormones and their cognate receptors from LCM-procured breast carcinoma cells. Breast Cancer Res Treat 2016; 161:245-258. [PMID: 27858316 DOI: 10.1007/s10549-016-4049-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 11/05/2016] [Indexed: 10/20/2022]
Abstract
PURPOSE Certain peptide hormones and/or their cognate receptors influencing normal cellular pathways also have been detected in breast cancers. The hypothesis is that gene subsets of these regulatory molecules predict risk of breast carcinoma recurrence in patients with primary disease. METHODS Gene expression levels of 61 hormones and 81 receptors were determined by microarray with LCM-procured carcinoma cells of 247 de-identified biopsies. Univariable and multivariable Cox regressions were determined using expression levels of each hormone/receptor gene, individually or as a pair. RESULTS Molecular signatures for ER+/PR+, ER-/PR-, and ER- carcinoma cells deciphered by LASSO were externally validated at HRs (CI) of 2.8 (1.84-4.4), 1.53 (1.01-2.3), and 1.72 (1.15-2.56), respectively. Using LCM-procured breast carcinoma cells, a 16-gene molecular signature was derived for ER+/PR+ biopsies, whereas a 10-gene signature was deciphered for ER-/PR- cancers. Four genes, POMC, CALCR, AVPR1A, and GH1, of this 10-gene signature were identified in a 6-gene molecular signature for ER- specimens. CONCLUSIONS Applying these signatures, Kaplan-Meier plots definitively identified a cohort of patients with either ER-/PR- or ER- carcinomas that exhibited low risk of recurrence. In contrast, the ER+/PR+ signature identified a cohort of patients with high risk of breast cancer recurrence. Each of the three molecular signatures predicted clinical outcomes of breast cancer patients with greater accuracy than observed with either single-gene analysis or by ER/PR protein content alone. Collectively, our results suggest that gene expression profiles of breast carcinomas with suspected poor prognosis (ER-/PR-) have identified a subset of patients with decreased risk of recurrence.
Collapse
Affiliation(s)
- Michael W Daniels
- Department of Biochemistry & Molecular Genetics, Institute for Molecular Diversity and Drug Design, University of Louisville, Louisville, KY, 40202, USA.,Department of Bioinformatics & Biostatistics, University of Louisville, Louisville, KY, 40202, USA
| | - Guy N Brock
- Department of Bioinformatics & Biostatistics, University of Louisville, Louisville, KY, 40202, USA
| | - James L Wittliff
- Department of Biochemistry & Molecular Genetics, Institute for Molecular Diversity and Drug Design, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
14
|
Davis KR, Giesy SL, Long Q, Krumm CS, Harvatine KJ, Boisclair YR. XBP1 Regulates the Biosynthetic Capacity of the Mammary Gland During Lactation by Controlling Epithelial Expansion and Endoplasmic Reticulum Formation. Endocrinology 2016; 157:417-28. [PMID: 26562262 DOI: 10.1210/en.2015-1676] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cells composing the mammary secretory compartment have evolved a high capacity to secrete not only proteins but also triglycerides and carbohydrates. This feature is illustrated by the mouse, which can secrete nearly twice its own weight in milk proteins, triglycerides and lactose over a short 20-day lactation. The coordination of synthesis and export of products in other secretory cells is orchestrated in part by the transcription factor X-box binding protein 1 (XBP1). To assess the role of XBP1 in mammary epithelial cells (MEC), we studied floxed XBP1 female mice lacking (wild type; WT) or expressing the Cre recombinase under the control of the ovine β-lactoglobulin promoter (ΔXBP1(MEC)). Pregnant ΔXBP1(MEC) females had morphologically normal mammary development and gave birth to the same number of pups as WT mice. Their litters, however, suffered a weight gain deficit by lactation day 3 (L3)3 that grew to 80% by L14. ΔXBP1(MEC) dams had only modest changes in milk composition (-21% protein, +24% triglyceride) and in the expression of associated genes in isolated MEC. By L5, WT glands were fully occupied by dilated alveoli, whereas ΔXBP1(MEC) glands contained fewer, mostly unfilled alveoli and retained a prominent adipocyte population. The smaller epithelial compartment in ΔXBP1(MEC) glands was explained by lower MEC proliferation and increased apoptosis. Finally, endoplasmic reticulum ribbons were less abundant in ΔXBP1(MEC) at pregnancy day 18 and failed to increase in abundance by L5. Collectively, these results show that XBP1 is required for MEC population expansion during lactation and its ability to develop an elaborate endoplasmic reticulum compartment.
Collapse
Affiliation(s)
- Kristen R Davis
- Department of Animal Science (K.R.D., S.L.G., C.S.K., Y.R.B.), Cornell University, Ithaca, New York 14853; Cam-Su Genome Resources and Laboratory Animal Research Center (Q.L.), Suzhou University, Suzhou 215123, Peoples Republic of China; and Department of Animal Science (K.J.H.), Penn State University, University Park, Pennsylvania 16802
| | - Sarah L Giesy
- Department of Animal Science (K.R.D., S.L.G., C.S.K., Y.R.B.), Cornell University, Ithaca, New York 14853; Cam-Su Genome Resources and Laboratory Animal Research Center (Q.L.), Suzhou University, Suzhou 215123, Peoples Republic of China; and Department of Animal Science (K.J.H.), Penn State University, University Park, Pennsylvania 16802
| | - Qiaoming Long
- Department of Animal Science (K.R.D., S.L.G., C.S.K., Y.R.B.), Cornell University, Ithaca, New York 14853; Cam-Su Genome Resources and Laboratory Animal Research Center (Q.L.), Suzhou University, Suzhou 215123, Peoples Republic of China; and Department of Animal Science (K.J.H.), Penn State University, University Park, Pennsylvania 16802
| | - Christopher S Krumm
- Department of Animal Science (K.R.D., S.L.G., C.S.K., Y.R.B.), Cornell University, Ithaca, New York 14853; Cam-Su Genome Resources and Laboratory Animal Research Center (Q.L.), Suzhou University, Suzhou 215123, Peoples Republic of China; and Department of Animal Science (K.J.H.), Penn State University, University Park, Pennsylvania 16802
| | - Kevin J Harvatine
- Department of Animal Science (K.R.D., S.L.G., C.S.K., Y.R.B.), Cornell University, Ithaca, New York 14853; Cam-Su Genome Resources and Laboratory Animal Research Center (Q.L.), Suzhou University, Suzhou 215123, Peoples Republic of China; and Department of Animal Science (K.J.H.), Penn State University, University Park, Pennsylvania 16802
| | - Yves R Boisclair
- Department of Animal Science (K.R.D., S.L.G., C.S.K., Y.R.B.), Cornell University, Ithaca, New York 14853; Cam-Su Genome Resources and Laboratory Animal Research Center (Q.L.), Suzhou University, Suzhou 215123, Peoples Republic of China; and Department of Animal Science (K.J.H.), Penn State University, University Park, Pennsylvania 16802
| |
Collapse
|
15
|
Castro MAA, de Santiago I, Campbell TM, Vaughn C, Hickey TE, Ross E, Tilley WD, Markowetz F, Ponder BAJ, Meyer KB. Regulators of genetic risk of breast cancer identified by integrative network analysis. Nat Genet 2016; 48:12-21. [PMID: 26618344 PMCID: PMC4697365 DOI: 10.1038/ng.3458] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 11/06/2015] [Indexed: 12/14/2022]
Abstract
Genetic risk for breast cancer is conferred by a combination of multiple variants of small effect. To better understand how risk loci might combine, we examined whether risk-associated genes share regulatory mechanisms. We created a breast cancer gene regulatory network comprising transcription factors and groups of putative target genes (regulons) and asked whether specific regulons are enriched for genes associated with risk loci via expression quantitative trait loci (eQTLs). We identified 36 overlapping regulons that were enriched for risk loci and formed a distinct cluster within the network, suggesting shared biology. The risk transcription factors driving these regulons are frequently mutated in cancer and lie in two opposing subgroups, which relate to estrogen receptor (ER)(+) luminal A or luminal B and ER(-) basal-like cancers and to different luminal epithelial cell populations in the adult mammary gland. Our network approach provides a foundation for determining the regulatory circuits governing breast cancer, to identify targets for intervention, and is transferable to other disease settings.
Collapse
Affiliation(s)
- Mauro A A Castro
- Bioinformatics and Systems Biology Laboratory, Federal University of Paraná (UFPR), Polytechnic Center, Curitiba, Brazil
| | - Ines de Santiago
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- Department of Oncology, University of Cambridge, Hutchison/Medical Research Council (MRC) Research Centre, Cambridge, UK
| | - Thomas M Campbell
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- Department of Oncology, University of Cambridge, Hutchison/Medical Research Council (MRC) Research Centre, Cambridge, UK
| | - Courtney Vaughn
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- Department of Oncology, University of Cambridge, Hutchison/Medical Research Council (MRC) Research Centre, Cambridge, UK
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories, School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Edith Ross
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories, School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Florian Markowetz
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Bruce A J Ponder
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- Department of Oncology, University of Cambridge, Hutchison/Medical Research Council (MRC) Research Centre, Cambridge, UK
| | - Kerstin B Meyer
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- Department of Oncology, University of Cambridge, Hutchison/Medical Research Council (MRC) Research Centre, Cambridge, UK
| |
Collapse
|
16
|
Clarke R, Tyson JJ, Dixon JM. Endocrine resistance in breast cancer--An overview and update. Mol Cell Endocrinol 2015; 418 Pt 3:220-34. [PMID: 26455641 PMCID: PMC4684757 DOI: 10.1016/j.mce.2015.09.035] [Citation(s) in RCA: 256] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 09/29/2015] [Accepted: 09/29/2015] [Indexed: 02/07/2023]
Abstract
Tumors that express detectable levels of the product of the ESR1 gene (estrogen receptor-α; ERα) represent the single largest molecular subtype of breast cancer. More women eventually die from ERα+ breast cancer than from either HER2+ disease (almost half of which also express ERα) and/or from triple negative breast cancer (ERα-negative, progesterone receptor-negative, and HER2-negative). Antiestrogens and aromatase inhibitors are largely indistinguishable from each other in their abilities to improve overall survival and almost 50% of ERα+ breast cancers will eventually fail one or more of these endocrine interventions. The precise reasons why these therapies fail in ERα+ breast cancer remain largely unknown. Pharmacogenetic explanations for Tamoxifen resistance are controversial. The role of ERα mutations in endocrine resistance remains unclear. Targeting the growth factors and oncogenes most strongly correlated with endocrine resistance has proven mostly disappointing in their abilities to improve overall survival substantially, particularly in the metastatic setting. Nonetheless, there are new concepts in endocrine resistance that integrate molecular signaling, cellular metabolism, and stress responses including endoplasmic reticulum stress and the unfolded protein response (UPR) that provide novel insights and suggest innovative therapeutic targets. Encouraging evidence that drug combinations with CDK4/CDK6 inhibitors can extend recurrence free survival may yet translate to improvements in overall survival. Whether the improvements seen with immunotherapy in other cancers can be achieved in breast cancer remains to be determined, particularly for ERα+ breast cancers. This review explores the basic mechanisms of resistance to endocrine therapies, concluding with some new insights from systems biology approaches further implicating autophagy and the UPR in detail, and a brief discussion of exciting new avenues and future prospects.
Collapse
Affiliation(s)
- Robert Clarke
- Department of Oncology, Georgetown University Medical Center, Washington DC 20057, USA.
| | - John J Tyson
- Department of Biological Sciences, Virginia Polytechnic and State University, Blacksburg, VA 24061, USA
| | - J Michael Dixon
- Edinburgh Breast Unit, Western General Hospital, Edinburgh, UK
| |
Collapse
|
17
|
Interaction between smoking history and gene expression levels impacts survival of breast cancer patients. Breast Cancer Res Treat 2015. [DOI: 10.1007/s10549-015-3507-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
18
|
Clarke R, Cook KL. Unfolding the Role of Stress Response Signaling in Endocrine Resistant Breast Cancers. Front Oncol 2015; 5:140. [PMID: 26157705 PMCID: PMC4475795 DOI: 10.3389/fonc.2015.00140] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 06/03/2015] [Indexed: 11/24/2022] Open
Abstract
The unfolded protein response (UPR) is an ancient stress response that enables a cell to manage the energetic stress that accompanies protein folding. There has been a significant recent increase in our understanding of the UPR, how it integrates physiological processes within cells, and how this integration can affect cancer cells and cell fate decisions. Recent publications have highlighted the role of UPR signaling components on mediating various cell survival pathways, cellular metabolism and bioenergenics, and autophagy. We address the role of UPR on mediating endocrine therapy resistance and estrogen receptor-positive breast cancer cell survival.
Collapse
Affiliation(s)
- Robert Clarke
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center , Washington, DC , USA
| | - Katherine L Cook
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center , Washington, DC , USA
| |
Collapse
|
19
|
Metzler MA, Venkatesh SG, Lakshmanan J, Carenbauer AL, Perez SM, Andres SA, Appana S, Brock GN, Wittliff JL, Darling DS. A systems biology approach identifies a regulatory network in parotid acinar cell terminal differentiation. PLoS One 2015; 10:e0125153. [PMID: 25928148 PMCID: PMC4416001 DOI: 10.1371/journal.pone.0125153] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/16/2015] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE The transcription factor networks that drive parotid salivary gland progenitor cells to terminally differentiate, remain largely unknown and are vital to understanding the regeneration process. METHODOLOGY A systems biology approach was taken to measure mRNA and microRNA expression in vivo across acinar cell terminal differentiation in the rat parotid salivary gland. Laser capture microdissection (LCM) was used to specifically isolate acinar cell RNA at times spanning the month-long period of parotid differentiation. RESULTS Clustering of microarray measurements suggests that expression occurs in four stages. mRNA expression patterns suggest a novel role for Pparg which is transiently increased during mid postnatal differentiation in concert with several target gene mRNAs. 79 microRNAs are significantly differentially expressed across time. Profiles of statistically significant changes of mRNA expression, combined with reciprocal correlations of microRNAs and their target mRNAs, suggest a putative network involving Klf4, a differentiation inhibiting transcription factor, which decreases as several targeting microRNAs increase late in differentiation. The network suggests a molecular switch (involving Prdm1, Sox11, Pax5, miR-200a, and miR-30a) progressively decreases repression of Xbp1 gene transcription, in concert with decreased translational repression by miR-214. The transcription factor Xbp1 mRNA is initially low, increases progressively, and may be maintained by a positive feedback loop with Atf6. Transfection studies show that Xbp1 activates the Mist1 promoter [corrected]. In addition, Xbp1 and Mist1 each activate the parotid secretory protein (Psp) gene, which encodes an abundant salivary protein, and is a marker of terminal differentiation. CONCLUSION This study identifies novel expression patterns of Pparg, Klf4, and Sox11 during parotid acinar cell differentiation, as well as numerous differentially expressed microRNAs. Network analysis identifies a novel stemness arm, a genetic switch involving transcription factors and microRNAs, and transition to an Xbp1 driven differentiation network. This proposed network suggests key regulatory interactions in parotid gland terminal differentiation.
Collapse
Affiliation(s)
- Melissa A. Metzler
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, Kentucky, United States of America
- Department of Biochemistry & Molecular Biology, University of Louisville, Louisville, Kentucky, United States of America
- Institute for Molecular Diversity and Drug Design, University of Louisville, Louisville, Kentucky, United States of America and
- * E-mail:
| | - Srirangapatnam G. Venkatesh
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, Kentucky, United States of America
- Institute for Molecular Diversity and Drug Design, University of Louisville, Louisville, Kentucky, United States of America and
| | - Jaganathan Lakshmanan
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, Kentucky, United States of America
| | - Anne L. Carenbauer
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, Kentucky, United States of America
| | - Sara M. Perez
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, Kentucky, United States of America
| | - Sarah A. Andres
- Department of Biochemistry & Molecular Biology, University of Louisville, Louisville, Kentucky, United States of America
- Institute for Molecular Diversity and Drug Design, University of Louisville, Louisville, Kentucky, United States of America and
| | - Savitri Appana
- Department of Bioinformatics and Biostatistics, University of Louisville, Louisville, Kentucky, United States of America
| | - Guy N. Brock
- Department of Bioinformatics and Biostatistics, University of Louisville, Louisville, Kentucky, United States of America
| | - James L. Wittliff
- Department of Biochemistry & Molecular Biology, University of Louisville, Louisville, Kentucky, United States of America
- Institute for Molecular Diversity and Drug Design, University of Louisville, Louisville, Kentucky, United States of America and
| | - Douglas S. Darling
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, Kentucky, United States of America
- Department of Biochemistry & Molecular Biology, University of Louisville, Louisville, Kentucky, United States of America
- Institute for Molecular Diversity and Drug Design, University of Louisville, Louisville, Kentucky, United States of America and
| |
Collapse
|
20
|
Watanabe T, Kato A, Terashima H, Matsubara K, Chen YJ, Adachi K, Mizuno H, Suzuki M. The PFA-AMeX method achieves a good balance between the morphology of tissues and the quality of RNA content in DNA microarray analysis with laser-capture microdissection samples. J Toxicol Pathol 2015; 28:43-9. [PMID: 26023261 PMCID: PMC4337499 DOI: 10.1293/tox.2014-0045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 11/14/2014] [Indexed: 01/18/2023] Open
Abstract
Recently, large-scale gene expression profiling is often performed using RNA extracted from unfixed frozen or formalin-fixed paraffin embedded (FFPE) samples. However, both types of samples have drawbacks in terms of the morphological preservation and RNA quality. In the present study, we investigated 30 human prostate tissues using the PFA-AMeX method (fixation using paraformaldehyde (PFA) followed by embedding in paraffin by AMeX) with a DNA microarray combined with laser-capture microdissection. Morphologically, in contrast to the case of atypical adenomatous hyperplasia, loss of basal cells in prostate adenocarcinomas was as obvious in PFA-AMeX samples as in FFPE samples. As for quality, the loss of rRNA peaks 18S and 28S on the capillary electropherograms from both FFPE and PFA-AMeX samples showed that the RNA was degraded equally during processing. However, qRT-PCR with 3' and 5' primer sets designed against human beta-actin revealed that, although RNA degradation occurred in both methods, it occurred more mildly in the PFA-AMeX samples. In conclusion, the PFA-AMeX method is good with respect to morphology and RNA quality, which makes it a promising tool for DNA microarrays combined with laser-capture microdissection, and if the appropriate RNA quality criteria are used, the capture of credible GeneChip data is well over 80% efficient, at least in human prostate specimens.
Collapse
Affiliation(s)
- Takeshi Watanabe
- Chugai Research Institute for Medical Science Inc., 1-135 Komakado, Gotemba-shi, Shizuoka 412-8513, Japan
| | - Atsuhiko Kato
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba-shi, Shizuoka 412-8513, Japan
| | - Hiromichi Terashima
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba-shi, Shizuoka 412-8513, Japan
| | - Koichi Matsubara
- PharmaLogicals Research, Pte. Ltd., 11 Biopolis Way, #05-08/09 Helios, 138667, Singapore
- Chugai Pharmabody Research Pte. Ltd., 3 Biopolis Drive, #04-11 to 17 Synapse, 138623, Singapore
| | - Yu Jau Chen
- PharmaLogicals Research, Pte. Ltd., 11 Biopolis Way, #05-08/09 Helios, 138667, Singapore
- Chugai Pharmabody Research Pte. Ltd., 3 Biopolis Drive, #04-11 to 17 Synapse, 138623, Singapore
| | - Kenji Adachi
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba-shi, Shizuoka 412-8513, Japan
| | - Hideaki Mizuno
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba-shi, Shizuoka 412-8513, Japan
| | - Masami Suzuki
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba-shi, Shizuoka 412-8513, Japan
| |
Collapse
|
21
|
Andres SA, Smolenkova IA, Wittliff JL. Gender-associated expression of tumor markers and a small gene set in breast carcinoma. Breast 2014; 23:226-33. [PMID: 24656773 DOI: 10.1016/j.breast.2014.02.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 02/06/2014] [Accepted: 02/22/2014] [Indexed: 11/28/2022] Open
Abstract
Breast carcinomas in both genders share pathological features, although differences in incidence, prognosis and survival are reported. Expression of 33 genes was investigated in male and female breast carcinomas in association with ER, PR, HER-2/neu and EGF-receptor. Among 98 male breast cancers, 82 were ER+ and 78 were PR+. ER and PR protein levels were greater in males compared to females, although no differences were observed in ESR1 and PGR expression. A difference was observed in binding affinities of PR but not ER between genders. No differences were observed in HER-2/neu, EGFR protein, or patient age. Expression of NAT1, TBC1D9, IL6ST, RABEP1, PLK1 and LRBA was elevated in carcinomas of males compared to those of females, in which ER status appeared to be related to expression. Over-expression of protein products of these genes represents novel molecular targets for development of gender-specific therapeutics and companion diagnostics.
Collapse
Affiliation(s)
- Sarah A Andres
- Department of Biochemistry & Molecular Biology, J. Graham Brown Cancer Center and Institute for Molecular Diversity & Drug Design, University of Louisville, Louisville, KY 40202, USA
| | - Irina A Smolenkova
- Department of Biochemistry & Molecular Biology, J. Graham Brown Cancer Center and Institute for Molecular Diversity & Drug Design, University of Louisville, Louisville, KY 40202, USA
| | - James L Wittliff
- Department of Biochemistry & Molecular Biology, J. Graham Brown Cancer Center and Institute for Molecular Diversity & Drug Design, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
22
|
Overley-Adamson B, Artlett CM, Stephens C, Sassi-Gaha S, Weis RD, Thacker JD. Targeting the unfolded protein response, XBP1, and the NLRP3 inflammasome in fibrosis and cancer. Cancer Biol Ther 2014; 15:452-62. [PMID: 24496016 PMCID: PMC3979823 DOI: 10.4161/cbt.27820] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Increasing health care costs in the US are due in a large part to the increasing prevalence of chronic diseases in an aging population. Current therapeutic strategies for treating chronic diseases alleviate symptoms allowing patients to live longer with these diseases, but they do little, however, to alter the underlying disease course. Recent advances in molecular biology are revealing new drug targets that may significantly alter the course of these diseases and, as a result, offer economic relief from burgeoning health care costs. Endoplasmic reticulum (ER) stress has been implicated as an underlying pathology in many chronic diseases, and, therefore, the development of therapies designed to ameliorate ER stress may yield novel, effective treatment strategies. Herein, we report that X-box binding protein 1 (XBP1) may be one of the earliest proteins engaged in response to ER stress. We show that a new signaling peptide derived from the ER-embedded transient receptor potential calcium channel protein 1 (TRPC1) engages XBP1 upstream of NLRP3 inflammasome-mediated maturation and secretion of IL-1β/IL-18. Moreover, we show that a synthetic homolog of this signaling peptide (Naclynamide™) administered intravenously twice weekly over a 4-week treatment course induced suppuration and evoked partial or complete resolution of lesions associated with a fibrotic granuloma, a lymphosarcoma, and a colo-rectal carcinoma in canine patients. The mode of action for Naclynamide™ as a first-in-class anti-cancer drug candidate is discussed.
Collapse
Affiliation(s)
| | - Carol M Artlett
- Drexel University College of Medicine; Department of Microbiology and Immunology; Philadelphia, PA USA
| | | | - Sihem Sassi-Gaha
- Drexel University College of Medicine; Department of Microbiology and Immunology; Philadelphia, PA USA
| | | | - James D Thacker
- Drexel University College of Medicine; Department of Microbiology and Immunology; Philadelphia, PA USA; TherimuneX Pharmaceuticals, Inc.; Doylestown, PA USA
| |
Collapse
|
23
|
Seelan RS, Warner DR, Mukhopadhyay PM, Andres SA, Smolenkova IA, Wittliff JL, Michele Pisano M, Greene RM. Epigenetic analysis of laser capture microdissected fetal epithelia. Anal Biochem 2013; 442:68-74. [PMID: 23911529 DOI: 10.1016/j.ab.2013.07.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/16/2013] [Accepted: 07/19/2013] [Indexed: 12/30/2022]
Abstract
Laser capture microdissection (LCM) is a superior method for nondestructive collection of specific cell populations from tissue sections. Although DNA, RNA, and protein have been analyzed from LCM-procured samples, epigenetic analyses, particularly of fetal, highly hydrated tissue, have not been attempted. A standardized protocol with quality assurance measures was established to procure cells by LCM of the medial edge epithelia (MEE) of the fetal palatal processes for isolation of intact microRNA for expression analyses and genomic DNA (gDNA) for CpG methylation analyses. MicroRNA preparations, obtained using the RNAqueous Micro kit (Life Technologies), exhibited better yields and higher quality than those obtained using the Arcturus PicoPure RNA Isolation kit (Life Technologies). The approach was validated using real-time polymerase chain reaction (PCR) to determine expression of selected microRNAs (miR-99a and miR-200b) and pyrosequencing to determine CpG methylation status of selected genes (Aph1a and Dkk4) in the MEE. These studies describe an optimized approach for employing LCM of epithelial cells from fresh frozen fetal tissue that enables quantitative analyses of microRNA expression levels and CpG methylation.
Collapse
Affiliation(s)
- Ratnam S Seelan
- Birth Defects Center, Department of Molecular, Cellular, and Craniofacial Biology, University of Louisville, Louisville, KY 40202, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Protein tyrosine phosphatase 4A2 expression predicts overall and disease-free survival of human breast cancer and is associated with estrogen and progestin receptor status. Discov Oncol 2013; 4:208-21. [PMID: 23568563 DOI: 10.1007/s12672-013-0141-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 03/19/2013] [Indexed: 12/21/2022] Open
Abstract
Expression of protein tyrosine phosphatase PTP4A2 (also known as PRL2) has been examined in a variety of human carcinomas, although its role in breast cancer remains inconclusive. Since the majority of previous breast cancer studies utilized tissue biopsies composed of heterogeneous cell populations, we hypothesized that an examination of PTP4A2 expression in carcinoma cells isolated by laser capture microdissection (LCM) would provide a more accurate means of assessing its predictive value. From investigations of 247 human breast cancer biopsies collected under standardized, stringent conditions, total RNA was extracted from LCM-procured carcinoma cells to perform microarray analyses to identify gene signatures associated with breast cancer behavior. Expression of PTP4A2 was corroborated by real-time quantitative polymerase chain reaction (qPCR) and referenced to estrogen and progesterone receptor levels. Patient outcomes for overall and disease-free survival were more favorable (p = 0.004 and p = 0.001, respectively) when the expression of PTP4A2 in breast carcinomas was increased compared to patients with biopsies with decreased PTP4A2 levels. PTP4A2 expression determined either by microarray or qPCR was elevated in either estrogen receptor (ER)-positive or progestin receptor (PR)-positive breast cancer biopsies compared to ER-negative or PR-negative biopsies. However, PTP4A2 expression was only correlated with overall survival in PR-positive breast carcinomas. These data suggest that PTP4A2 mRNA expression alone may serve as a biomarker for prediction of a breast cancer patient's risk of recurrence and overall survival.
Collapse
|