1
|
Wang S, Guo J, Xian X, Li M, Zhang A, Liu Y, Zhang Y, Chen S, Gu G, Zhang X, Yan D, An M, Pan L, Fu B. Distinct 5-methylcytosine profiles of LncRNA in breast cancer brain metastasis. BMC Cancer 2025; 25:557. [PMID: 40148799 PMCID: PMC11951547 DOI: 10.1186/s12885-025-13948-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Recent studies have identified a complex relationship between methylation patterns and the development of various cancers. Breast cancer (BC) is the second leading cause of cancer mortality among women. Approximately 5-20% of BC patients are at risk of BC brain metastases (BCBM). Although 5-methylcytosine (m5C) has been identified as an important regulatory modifier, its distribution in BCBM is not well understood. This study aimed to investigate the distribution of m5C in BCBM. MATERIALS AND METHODS Samples from BCBM (231-BR cells) and BC (MDA-MB-231 cells) groups were subjected to a comprehensive analysis of the m5C methylation in long non-coding RNA (lncRNA) using methylated RNA immunoprecipitation next-generation sequencing (MeRIP-seq). The expression levels of methylated genes in BC and adjacent tissues were verified through quantitative real-time polymerase chain reaction (RT-qPCR). Enrichment pathway analyses were through Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) to predict the potential functions of m5C in BCBM. RESULTS The MeRIP-seq analysis identified 23,934 m5C peaks in BCBM and 21,236 m5C in BC. A total of 9,480 annotated genes (BCBM) and 8,481 annotated genes (BC) were mapped. Notably, 1,819 methylation sites in lncRNA were upregulated in BCBM, whereas 2,415 methylation sites were upregulated in BC. Significant m5C hypermethylated lncRNAs included ENST00000477316, ENST00000478098 and uc002gtt.1, whereas hypomethylated lncRNAs included ENST00000600912, ENST00000493668, ENST00000544651 and ENST00000464989. These results were verified by qPCR and MeRIP-qPCR in BC and BCBM. Considering the strong association between m5C RNA methylation regulators and lncRNA, we examined the expression levels of 13 m5C RNA methylation regulators and observed significant differences between BC tissues and adjacent normal tissues. In addition, the interaction between regulators of altered expression and the differentially expressed genes in vitro was analyzed. The GO and KEGG pathways analyses revealed that genes significantly associated with m5C sites in lncRNA were linked to the BCBM signaling pathways. CONCLUSION This uncovered significant variations in the levels and distribution of m5C in BCBM compared to BC. The findings provide a new theoretical understanding of the mechanisms of BCBM.
Collapse
Affiliation(s)
- Song Wang
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China
| | - Jianran Guo
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China
| | - Xinmiao Xian
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China
| | - Min Li
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China
| | - Anqi Zhang
- Department of Central Laboratory, Liaocheng People's Hospital, 67 Dongchang west Road, Liaocheng, 252000, Shandong, P. R. China
| | - Yujiao Liu
- Department of Stomatology, Liaocheng People's Hospital, Liaocheng, P. R. China
| | - Yifei Zhang
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China
| | - Shen Chen
- Department of Breast and Thyroid Surgery, Liaocheng People's Hospital, Liaocheng, P. R. China
| | - Guohao Gu
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China
| | - Xuehua Zhang
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China
| | - Dong Yan
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China
| | - Meng An
- Department of Clinical Laboratory, Liaocheng People's Hospital, 67 Dongchang west Road, Liaocheng, Shandong, 252000, P. R. China.
| | - Li Pan
- Department of Central Laboratory, Liaocheng People's Hospital, 67 Dongchang west Road, Liaocheng, 252000, Shandong, P. R. China.
| | - Bo Fu
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China.
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China.
| |
Collapse
|
2
|
Li M, Wang X, Hong J, Mao J, Chen J, Chen X, Du Y, Song D. Transglutaminase 2 in breast cancer metastasis and drug resistance. Front Cell Dev Biol 2024; 12:1485258. [PMID: 39544364 PMCID: PMC11560871 DOI: 10.3389/fcell.2024.1485258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024] Open
Abstract
Transglutaminase 2 (TG2) is a widely distributed multifunctional protein with various enzymatic and non-enzymatic activities. It is becoming increasingly evident that high levels of TG2 in tumors induce the occurrence of epithelial to mesenchymal transition (EMT) and the acquisition of stem cell-like phenotypes, promoting tumor metastasis and drug resistance. By regulating intracellular and extracellular signaling pathways, TG2 promotes breast cancer metastasis to lung, brain, liver and bone, as well as resistance to various chemotherapy drugs including docetaxel, doxorubicin, platinum and neratinib. More importantly, recent studies described the involvement of TG2 in PD-1/PD-L1 inhibitors resistance. An in-depth understanding of the role that TG2 plays in the progression of metastasis and drug resistance will offer new therapeutic targets for breast cancer treatment. This review covers the extensive and rapidly growing field of the role of TG2 in breast cancer. Based on the role of TG2 in EMT, we summarize TG2-related signaling pathways in breast cancer metastasis and drug resistance and discuss TG2 as a therapeutic target.
Collapse
Affiliation(s)
- Mengxin Li
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Xuanzhong Wang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
| | - Jinghui Hong
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Juanjuan Mao
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jiasi Chen
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xuyang Chen
- School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
| | - Ye Du
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Dong Song
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
3
|
Sanni A, Bennett AI, Huang Y, Gidi I, Adeniyi M, Nwaiwu J, Kang MH, Keyel ME, Gao C, Reynolds CP, Haab B, Mechref Y. An Optimized Liquid Chromatography-Mass Spectrometry Method for Ganglioside Analysis in Cell Lines. Cells 2024; 13:1640. [PMID: 39404403 PMCID: PMC11476222 DOI: 10.3390/cells13191640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024] Open
Abstract
Gangliosides are glycosphingolipids composed of a sialylated glycan head group and a ceramide backbone. These anionic lipids form lipid rafts and play crucial roles in regulating various proteins involved in signal transduction, adhesion, and cell-cell recognition. Neuroblastoma, a pediatric cancer of the sympathetic nervous system, is treated with intensive chemotherapy, radiation, and an antibody targeting the GD2 ganglioside. Gangliosides are critical in neuroblastoma development and serve as therapeutic targets, making it essential to establish a reliable, rapid, and cost-effective method for profiling gangliosides, particularly one capable of isomeric separation of intact species. In this study, liquid chromatography-mass spectrometry (LC-MS) was optimized using standard gangliosides, followed by the optimization of sphingolipid extraction methods from cell lines by comparing Folch and absolute methanol extraction techniques. Percent recovery and the number of identified sphingolipids were used to evaluate the analytical merits of these methods. A standard gangliosides calibration curve demonstrated excellent linearity (R2 = 0.9961-0.9975). The ZIC-HILIC column provided the best separation of ganglioside GD1 isomers with a 25 min runtime. GD1a elutes before GD1b on the ZIC-HILIC column. Absolute methanol yielded better percent recovery (96 ± 7) and identified 121 different sphingolipids, the highest number between the two extraction methods. The optimized method was applied to profile gangliosides in neuroblastoma (COG-N-683), pancreatic cancer (PSN1), breast cancer (MDA-MB-231BR), and brain tumor (CRL-1620) cell lines. The ganglioside profile of the neuroblastoma cell line COG-N-683 showed an inverse relationship between GD1 and GD2. Ceramide, Hex1Cer, GM1, and GM3 were highly abundant in CRL-1620, PSN1, and MDA-MB-231BR, respectively. These results suggest that our method provides a sensitive, reliable, and high-throughput workflow for ganglioside profiling across different cell types.
Collapse
Affiliation(s)
- Akeem Sanni
- Chemistry and Biochemistry Department, Texas Tech University, Lubbock, TX 79409, USA; (A.S.); (A.I.B.); (Y.H.); (I.G.); (J.N.)
| | - Andrew I. Bennett
- Chemistry and Biochemistry Department, Texas Tech University, Lubbock, TX 79409, USA; (A.S.); (A.I.B.); (Y.H.); (I.G.); (J.N.)
| | - Yifan Huang
- Chemistry and Biochemistry Department, Texas Tech University, Lubbock, TX 79409, USA; (A.S.); (A.I.B.); (Y.H.); (I.G.); (J.N.)
| | - Isabella Gidi
- Chemistry and Biochemistry Department, Texas Tech University, Lubbock, TX 79409, USA; (A.S.); (A.I.B.); (Y.H.); (I.G.); (J.N.)
| | - Moyinoluwa Adeniyi
- Chemistry and Biochemistry Department, Texas Tech University, Lubbock, TX 79409, USA; (A.S.); (A.I.B.); (Y.H.); (I.G.); (J.N.)
| | - Judith Nwaiwu
- Chemistry and Biochemistry Department, Texas Tech University, Lubbock, TX 79409, USA; (A.S.); (A.I.B.); (Y.H.); (I.G.); (J.N.)
| | - Min H. Kang
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center (TTUHSC), Lubbock, TX 79416, USA; (M.H.K.); (C.P.R.)
| | - Michelle E. Keyel
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center (TTUHSC), Lubbock, TX 79416, USA; (M.H.K.); (C.P.R.)
| | - ChongFeng Gao
- Van Andel Institute, Grand Rapids, MI 49503, USA; (C.G.); (B.H.)
| | - C. Patrick Reynolds
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center (TTUHSC), Lubbock, TX 79416, USA; (M.H.K.); (C.P.R.)
| | - Brian Haab
- Van Andel Institute, Grand Rapids, MI 49503, USA; (C.G.); (B.H.)
| | - Yehia Mechref
- Chemistry and Biochemistry Department, Texas Tech University, Lubbock, TX 79409, USA; (A.S.); (A.I.B.); (Y.H.); (I.G.); (J.N.)
| |
Collapse
|
4
|
Cai P, Li J, An M, Li M, Guo J, Li J, Li X, Chen S, Zhang A, Li P, Liu Y, Zhang W, Fu B. Comprehensive analysis of RNA-5-methylcytosine modification in breast cancer brain metastasis. Future Oncol 2024; 20:2993-3008. [PMID: 39345093 PMCID: PMC11572191 DOI: 10.1080/14796694.2024.2405459] [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: 03/21/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
Aim: To delineate the RNA-5-methylcytosine (m5C) modification of breast cancer brain metastasis (BCBM).Methods: Methylated RNA immunoprecipitation next-generation sequencing (MeRIP-seq) was performed to obtain RNA-m5C patterns of BCBM.Results: 1048 hypermethylation and 1866 hypomethylation m5C peaks were identified in BCBM compared with those in breast cancer. The most significant m5C hypermethylated genes included ENG, SHANK1, IGFN1, EVL and MMP9, whereas the most significant m5C hypomethylated genes included AREG, SAA2, TP53I11, KRT7 and LCN2. MeRIP-qPCR data were concordant with the corresponding MeRIP-seq results in terms of the observed m5C levels. Conjoint analysis identified 190 hyper-up genes characterized by concurrent m5C hypermethylation and up-regulation, alongside 284 hypo-down genes exhibiting both m5C hypomethylation and down-regulation.Conclusion: This study presents the first comprehensive analysis of RNA-m5C modification in BCBM.
Collapse
Affiliation(s)
- Peiying Cai
- Department of Central Laboratory, Liaocheng People's Hospital, Liaocheng, P.R. China
| | - Jichao Li
- Department of Clinical Laboratory, Liaocheng Women & Children Hospital, Liaocheng, P.R. China
| | - Meng An
- Department of Clinical Laboratory, Liaocheng People's Hospital, Liaocheng, P.R. China
| | - Min Li
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital; Shandong Provincial Key Medical & Health Laboratory of Precision Medicine for Aging Intervention & Active Health, Liaocheng, P.R. China
| | - Jianran Guo
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital; Shandong Provincial Key Medical & Health Laboratory of Precision Medicine for Aging Intervention & Active Health, Liaocheng, P.R. China
| | - Jun Li
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital; Shandong Provincial Key Medical & Health Laboratory of Precision Medicine for Aging Intervention & Active Health, Liaocheng, P.R. China
| | - Xuan Li
- Department of Molecular Pharmacology Key Laboratory, Liaocheng People's Hospital, Liaocheng, P.R. China
| | - Shen Chen
- Department of Breast & Thyroid Surgery, Liaocheng People's Hospital, Liaocheng, P.R. China
| | - Anqi Zhang
- Department of Central Laboratory, Liaocheng People's Hospital, Liaocheng, P.R. China
| | - Peng Li
- Department of Clinical Laboratory, Liaocheng People's Hospital, Liaocheng, P.R. China
| | - Yan Liu
- Department of Clinical Laboratory, Liaocheng People's Hospital, Liaocheng, P.R. China
| | - Wei Zhang
- Department of Breast & Thyroid Surgery, Liaocheng People's Hospital, Liaocheng, P.R. China
| | - Bo Fu
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital; Shandong Provincial Key Medical & Health Laboratory of Precision Medicine for Aging Intervention & Active Health, Liaocheng, P.R. China
| |
Collapse
|
5
|
Chen Y, Zhu L, Wang Y, Hu J, Zhang H, Zhu J, Gong W, Liu X, Xiao F, Li X. Tumor-derived mesenchymal progenitor cell-related genes in the regulation of breast cancer proliferation. Gland Surg 2024; 13:325-339. [PMID: 38601284 PMCID: PMC11002474 DOI: 10.21037/gs-23-387] [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: 09/13/2023] [Accepted: 01/20/2024] [Indexed: 04/12/2024]
Abstract
Background Breast cancer (BC) is one of the most common malignancies worldwide, and its development is affected in various ways by the tumor microenvironment (TME). Tumor-derived mesenchymal progenitor cells (MPCs), as the most important components of the TME, participate in the proliferation and metastasis of BC in several ways. In this study, we aimed to characterize the genes associated with tumor-derived MPCs and determine their effects on BC cells. Methods Tumor-derived MPCs and normal breast tissue-derived mesenchymal stem cells (MSCs) were isolated from tissues specimens of patients with BC. We conducted culture and passage, phenotype identification, proliferation and migration detection, inflammatory factor release detection, and other experiments on isolated MPCs from tumors and MSCs from normal breast tissues. Three paired tumor-derived MPCs and normal breast tissue-derived MSCs were then subjected to transcriptome analysis to determine the expression profiles of the relevant genes, and quantitative real-time polymerase chain reaction (qRT-PCR) was used to further confirm gene expression. Subsequently, the overexpression plasmids were transfected into tumor-derived MPCs, and the expression of various inflammatory factors of tumor-derived MPCs and their proliferation were characterized with a cell viability test reagent (Cell Counting Kit 8). Subsequently, the transfected tumor-derived MPCs were cocultured with BC cells using a conditioned medium coculture method to clarify the role of tumor-derived MSCs in BC. Results Tumor-derived MPCs expressed stem cell characteristics including CD105, CD90, and CD73 and exhibited adipogenic and osteogenic differentiation in vitro. The proliferation of tumor-derived MPCs was significantly lower than that of normal breast tissue-derived MSCs, and the invasive metastatic ability was comparable; however, MPCs were found to release inflammatory factors such as interleukin 6 (IL-6) and transforming growth factor β (TGF-β). Transcriptome analysis showed that stomatin (STOM), collagen and calcium binding EGF domains 1 (CCBE1), and laminin subunit alpha 5 (LAMA5) were significantly upregulated in tumor-derived MPCs. Among them, STOM was highly expressed in tumor-derived MPCs, which mediated the slow proliferation of MPCs and promoted the proliferation of BC cells. Conclusions STOM, CCBE1, and LAMA5 were highly expressed in tumor-derived MPCs, with STOM being found to retard the proliferation of MPCs but promote the proliferation of BC cells. There findings present new possibilities in targeted microenvironmental therapy for BC.
Collapse
Affiliation(s)
- Yizhu Chen
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Li Zhu
- Department of General Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yiming Wang
- School of Nursing, Jilin University, Changchun, China
| | - Jia Hu
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Hao Zhang
- Medical Research Institute, Hebei Yanda Hospital, Langfang, China
| | - Jingjin Zhu
- Department of General Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Wenye Gong
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Xiaohan Liu
- Department of General Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Fengjun Xiao
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiru Li
- Department of General Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
6
|
Benjamin M, Malakar P, Sinha RA, Nasser MW, Batra SK, Siddiqui JA, Chakravarti B. Molecular signaling network and therapeutic developments in breast cancer brain metastasis. ADVANCES IN CANCER BIOLOGY - METASTASIS 2023; 7:100079. [PMID: 36536947 PMCID: PMC7613958 DOI: 10.1016/j.adcanc.2022.100079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Breast cancer (BC) is one of the most frequently diagnosed cancers in women worldwide. It has surpassed lung cancer as the leading cause of cancer-related death. Breast cancer brain metastasis (BCBM) is becoming a major clinical concern that is commonly associated with ER-ve and HER2+ve subtypes of BC patients. Metastatic lesions in the brain originate when the cancer cells detach from a primary breast tumor and establish metastatic lesions and infiltrate near and distant organs via systemic blood circulation by traversing the BBB. The colonization of BC cells in the brain involves a complex interplay in the tumor microenvironment (TME), metastatic cells, and brain cells like endothelial cells, microglia, and astrocytes. BCBM is a significant cause of morbidity and mortality and presents a challenge to developing successful cancer therapy. In this review, we discuss the molecular mechanism of BCBM and novel therapeutic strategies for patients with brain metastatic BC.
Collapse
Affiliation(s)
- Mercilena Benjamin
- Lab Oncology, Dr. B.R.A.I.R.C.H. All India Institute of Medical Sciences, New Delhi, India
| | - Pushkar Malakar
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational and Research Institute, Narendrapur, West Bengal, 700103, India
| | - Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Mohd Wasim Nasser
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Jawed Akhtar Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Bandana Chakravarti
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| |
Collapse
|
7
|
Burguin A, Roy J, Ouellette G, Maltais R, Bherer J, Diorio C, Poirier D, Durocher F. Aminosteroid RM-581 Decreases Cell Proliferation of All Breast Cancer Molecular Subtypes, Alone and in Combination with Breast Cancer Treatments. J Clin Med 2023; 12:4241. [PMID: 37445276 DOI: 10.3390/jcm12134241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/07/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Breast cancer (BC) is a heterogenous disease classified into four molecular subtypes (Luminal A, Luminal B, HER2 and triple-negative (TNBC)) depending on the expression of the estrogen receptor (ER), the progesterone receptor (PR) and the human epidermal receptor 2 (HER2). The development of effective treatments for BC, especially TNBC, remains a challenge. Aminosteroid derivative RM-581 has previously shown an antiproliferative effect in multiple cancers in vitro and in vivo. In this study, we evaluated its effect in BC cell lines representative of BC molecular subtypes, including metastatic TNBC. We found that RM-581 has an antiproliferative effect on all BC molecular subtypes, especially on Luminal A and TNBC, in 2D and 3D cultures. The combination of RM-581 and trastuzumab or trastuzumab-emtansine enhanced the anticancer effect of each drug for HER2-positive BC cell lines, and the combination of RM-581 and taxanes (docetaxel or paclitaxel) improved the antiproliferative effect of RM-581 in TNBC and metastatic TNBC cell lines. We also confirmed that RM-581 is an endoplasmic reticulum (EnR)-stress aggravator by inducing an increase in EnR-stress-induced apoptosis markers such as BIP/GRP78 and CHOP and disrupting lipid homeostasis. This study demonstrates that RM-581 could be effective for the treatment of BC, especially TNBC.
Collapse
Affiliation(s)
- Anna Burguin
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC GIV 0A6, Canada
- Cancer Research Centre, CHU de Québec-Research Centre, Québec, QC G1R 3S3, Canada
| | - Jenny Roy
- Cancer Research Centre, CHU de Québec-Research Centre, Québec, QC G1R 3S3, Canada
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec-Research Center, Québec, QC G1V 4G2, Canada
| | - Geneviève Ouellette
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC GIV 0A6, Canada
- Cancer Research Centre, CHU de Québec-Research Centre, Québec, QC G1R 3S3, Canada
| | - René Maltais
- Cancer Research Centre, CHU de Québec-Research Centre, Québec, QC G1R 3S3, Canada
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec-Research Center, Québec, QC G1V 4G2, Canada
| | - Juliette Bherer
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC GIV 0A6, Canada
- Cancer Research Centre, CHU de Québec-Research Centre, Québec, QC G1R 3S3, Canada
| | - Caroline Diorio
- Cancer Research Centre, CHU de Québec-Research Centre, Québec, QC G1R 3S3, Canada
- Department of Social and Preventive Medicine, Faculty of Medicine, Université Laval, Québec, QC GIV 0A6, Canada
| | - Donald Poirier
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC GIV 0A6, Canada
- Cancer Research Centre, CHU de Québec-Research Centre, Québec, QC G1R 3S3, Canada
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec-Research Center, Québec, QC G1V 4G2, Canada
| | - Francine Durocher
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC GIV 0A6, Canada
- Cancer Research Centre, CHU de Québec-Research Centre, Québec, QC G1R 3S3, Canada
| |
Collapse
|
8
|
Westerhof TM, Yang BA, Merill NM, Yates JA, Altemus M, Russell L, Miller AJ, Bao L, Wu Z, Ulintz PJ, Aguilar CA, Morikawa A, Castro MG, Merajver SD, Oliver CR. Blood-brain barrier remodeling in an organ-on-a-chip device shows Dkk1 to be a regulator of early metastasis. ADVANCED NANOBIOMED RESEARCH 2023; 3:2200036. [PMID: 37234365 PMCID: PMC10208594 DOI: 10.1002/anbr.202200036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024] Open
Abstract
Brain metastases are the most lethal progression event, in part because the biological processes underpinning brain metastases are poorly understood. There is a paucity of realistic models of metastasis, as current in vivo murine models are slow to manifest metastasis. We set out to delineate metabolic and secretory modulators of brain metastases by utilizing two models consisting of in vitro microfluidic devices: 1) a blood brain niche (BBN) chip that recapitulates the blood-brain-barrier and niche; and 2) a migration chip that assesses cell migration. We report secretory cues provided by the brain niche that attract metastatic cancer cells to colonize the brain niche region. Astrocytic Dkk-1 is increased in response to brain-seeking breast cancer cells and stimulates cancer cell migration. Brain-metastatic cancer cells under Dkk-1 stimulation increase gene expression of FGF-13 and PLCB1. Further, extracellular Dkk-1 modulates cancer cell migration upon entering the brain niche.
Collapse
Affiliation(s)
- Trisha M Westerhof
- Michigan Medicine, Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Benjamin A Yang
- School of Engineering, Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nathan M Merill
- Michigan Medicine, Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Joel A Yates
- Michigan Medicine, Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Megan Altemus
- Michigan Medicine, Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Liam Russell
- School of Engineering, Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anna J Miller
- School of Engineering, Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Liwei Bao
- Michigan Medicine, Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Zhifen Wu
- Michigan Medicine, Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Peter J Ulintz
- Michigan Medicine, Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Carlos A Aguilar
- School of Engineering, Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Aki Morikawa
- Michigan Medicine, Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria G Castro
- Michigan Medicine, Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA
- Michigan Medicine, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sofia D Merajver
- Michigan Medicine, Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Christopher R Oliver
- Michigan Medicine, Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
9
|
Germon ZP, Sillar JR, Mannan A, Duchatel RJ, Staudt D, Murray HC, Findlay IJ, Jackson ER, McEwen HP, Douglas AM, McLachlan T, Schjenken JE, Skerrett-Byrne DA, Huang H, Melo-Braga MN, Plank MW, Alvaro F, Chamberlain J, De Iuliis G, Aitken RJ, Nixon B, Wei AH, Enjeti AK, Huang Y, Lock RB, Larsen MR, Lee H, Vaghjiani V, Cain JE, de Bock CE, Verrills NM, Dun MD. Blockade of ROS production inhibits oncogenic signaling in acute myeloid leukemia and amplifies response to precision therapies. Sci Signal 2023; 16:eabp9586. [PMID: 36976863 DOI: 10.1126/scisignal.abp9586] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Mutations in the type III receptor tyrosine kinase FLT3 are frequent in patients with acute myeloid leukemia (AML) and are associated with a poor prognosis. AML is characterized by the overproduction of reactive oxygen species (ROS), which can induce cysteine oxidation in redox-sensitive signaling proteins. Here, we sought to characterize the specific pathways affected by ROS in AML by assessing oncogenic signaling in primary AML samples. The oxidation or phosphorylation of signaling proteins that mediate growth and proliferation was increased in samples from patient subtypes with FLT3 mutations. These samples also showed increases in the oxidation of proteins in the ROS-producing Rac/NADPH oxidase-2 (NOX2) complex. Inhibition of NOX2 increased the apoptosis of FLT3-mutant AML cells in response to FLT3 inhibitors. NOX2 inhibition also reduced the phosphorylation and cysteine oxidation of FLT3 in patient-derived xenograft mouse models, suggesting that decreased oxidative stress reduces the oncogenic signaling of FLT3. In mice grafted with FLT3 mutant AML cells, treatment with a NOX2 inhibitor reduced the number of circulating cancer cells, and combining FLT3 and NOX2 inhibitors increased survival to a greater extent than either treatment alone. Together, these data raise the possibility that combining NOX2 and FLT3 inhibitors could improve the treatment of FLT3 mutant AML.
Collapse
Affiliation(s)
- Zacary P Germon
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jonathan R Sillar
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Department of Haematology, Calvary Mater Hospital, Waratah, NSW, Australia
| | - Abdul Mannan
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Ryan J Duchatel
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Dilana Staudt
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Heather C Murray
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Izac J Findlay
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Evangeline R Jackson
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Holly P McEwen
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Alicia M Douglas
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Tabitha McLachlan
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - John E Schjenken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW, Australia
| | - David A Skerrett-Byrne
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW, Australia
| | - Honggang Huang
- Department of Molecular Biology and Biochemistry, Protein Research Group, University of Southern Denmark, Odense, Denmark
| | - Marcella N Melo-Braga
- Department of Molecular Biology and Biochemistry, Protein Research Group, University of Southern Denmark, Odense, Denmark
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maximilian W Plank
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- GlaxoSmithKline, Abbotsford, Victoria, Australia
| | - Frank Alvaro
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- John Hunter Children's Hospital, New Lambton Heights, NSW, Australia
| | - Janis Chamberlain
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- John Hunter Children's Hospital, New Lambton Heights, NSW, Australia
| | - Geoff De Iuliis
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW, Australia
| | - R John Aitken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW, Australia
| | - Andrew H Wei
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Anoop K Enjeti
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Department of Haematology, Calvary Mater Hospital, Waratah, NSW, Australia
- NSW Health Pathology, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Yizhou Huang
- Children's Cancer Institute, Lowy Cancer Centre, School of Women's and Children's Health, University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia
| | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Centre, School of Women's and Children's Health, University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia
| | - Martin R Larsen
- Department of Molecular Biology and Biochemistry, Protein Research Group, University of Southern Denmark, Odense, Denmark
| | - Heather Lee
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Vijesh Vaghjiani
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Jason E Cain
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Charles E de Bock
- Children's Cancer Institute, Lowy Cancer Centre, School of Women's and Children's Health, University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia
| | - Nicole M Verrills
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Matthew D Dun
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
10
|
Staudt DE, Murray HC, Skerrett-Byrne DA, Smith ND, Jamaluddin MFB, Kahl RGS, Duchatel RJ, Germon ZP, McLachlan T, Jackson ER, Findlay IJ, Kearney PS, Mannan A, McEwen HP, Douglas AM, Nixon B, Verrills NM, Dun MD. Phospho-heavy-labeled-spiketide FAIMS stepped-CV DDA (pHASED) provides real-time phosphoproteomics data to aid in cancer drug selection. Clin Proteomics 2022; 19:48. [PMID: 36536316 PMCID: PMC9762002 DOI: 10.1186/s12014-022-09385-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Global high-throughput phosphoproteomic profiling is increasingly being applied to cancer specimens to identify the oncogenic signaling cascades responsible for promoting disease initiation and disease progression; pathways that are often invisible to genomics analysis. Hence, phosphoproteomic profiling has enormous potential to inform and improve individualized anti-cancer treatment strategies. However, to achieve the adequate phosphoproteomic depth and coverage necessary to identify the activated, and hence, targetable kinases responsible for driving oncogenic signaling pathways, affinity phosphopeptide enrichment techniques are required and often coupled with offline high-pressure liquid chromatographic (HPLC) separation prior to nanoflow liquid chromatography-tandem mass spectrometry (nLC-MS/MS). These complex and time-consuming procedures, limit the utility of phosphoproteomics for the analysis of individual cancer patient specimens in real-time, and restrict phosphoproteomics to specialized laboratories often outside of the clinical setting. To address these limitations, here we have optimized a new protocol, phospho-heavy-labeled-spiketide FAIMS Stepped-CV DDA (pHASED), that employs online phosphoproteome deconvolution using high-field asymmetric waveform ion mobility spectrometry (FAIMS) and internal phosphopeptide standards to provide accurate label-free quantitation (LFQ) data in real-time. Compared with traditional single-shot LFQ phosphoproteomics workflows, pHASED provided increased phosphoproteomic depth and coverage (phosphopeptides = 4617 pHASED, 2789 LFQ), whilst eliminating the variability associated with offline prefractionation. pHASED was optimized using tyrosine kinase inhibitor (sorafenib) resistant isogenic FLT3-mutant acute myeloid leukemia (AML) cell line models. Bioinformatic analysis identified differential activation of the serine/threonine protein kinase ataxia-telangiectasia mutated (ATM) pathway, responsible for sensing and repairing DNA damage in sorafenib-resistant AML cell line models, thereby uncovering a potential therapeutic opportunity. Herein, we have optimized a rapid, reproducible, and flexible protocol for the characterization of complex cancer phosphoproteomes in real-time, a step towards the implementation of phosphoproteomics in the clinic to aid in the selection of anti-cancer therapies for patients.
Collapse
Affiliation(s)
- Dilana E. Staudt
- grid.266842.c0000 0000 8831 109XSchool of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308 Australia ,grid.413648.cPrecision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305 Australia
| | - Heather C. Murray
- grid.266842.c0000 0000 8831 109XSchool of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308 Australia ,grid.413648.cPrecision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305 Australia
| | - David A. Skerrett-Byrne
- grid.266842.c0000 0000 8831 109XSchool of Environmental and Life Sciences, College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW 2308 Australia ,grid.413648.cInfertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305 Australia
| | - Nathan D. Smith
- grid.266842.c0000 0000 8831 109XAnalytical and Biomolecular Research Facility (ABRF), Research Services, University of Newcastle, NSW, Callaghan, 2308 Australia
| | - M. Fairuz B. Jamaluddin
- grid.266842.c0000 0000 8831 109XSchool of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308 Australia
| | - Richard G. S. Kahl
- grid.266842.c0000 0000 8831 109XSchool of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308 Australia
| | - Ryan J. Duchatel
- grid.266842.c0000 0000 8831 109XSchool of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308 Australia ,grid.413648.cPrecision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305 Australia
| | - Zacary P. Germon
- grid.266842.c0000 0000 8831 109XSchool of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308 Australia ,grid.413648.cPrecision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305 Australia
| | - Tabitha McLachlan
- grid.266842.c0000 0000 8831 109XSchool of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308 Australia ,grid.413648.cPrecision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305 Australia
| | - Evangeline R. Jackson
- grid.266842.c0000 0000 8831 109XSchool of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308 Australia ,grid.413648.cPrecision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305 Australia
| | - Izac J. Findlay
- grid.266842.c0000 0000 8831 109XSchool of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308 Australia ,grid.413648.cPrecision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305 Australia
| | - Padraic S. Kearney
- grid.266842.c0000 0000 8831 109XSchool of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308 Australia ,grid.413648.cPrecision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305 Australia
| | - Abdul Mannan
- grid.266842.c0000 0000 8831 109XSchool of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308 Australia ,grid.413648.cPrecision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305 Australia
| | - Holly P. McEwen
- grid.266842.c0000 0000 8831 109XSchool of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308 Australia ,grid.413648.cPrecision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305 Australia
| | - Alicia M. Douglas
- grid.266842.c0000 0000 8831 109XSchool of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308 Australia
| | - Brett Nixon
- grid.266842.c0000 0000 8831 109XSchool of Environmental and Life Sciences, College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW 2308 Australia ,grid.413648.cInfertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305 Australia
| | - Nicole M. Verrills
- grid.266842.c0000 0000 8831 109XSchool of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308 Australia ,grid.413648.cPrecision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305 Australia
| | - Matthew D. Dun
- grid.266842.c0000 0000 8831 109XSchool of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308 Australia ,grid.413648.cPrecision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305 Australia
| |
Collapse
|
11
|
DeCastro AJL, Pranda MA, Gray KM, Merlo-Coyne J, Girma N, Hurwitz M, Zhang Y, Stroka KM. Morphological Phenotyping of Organotropic Brain- and Bone-Seeking Triple Negative Metastatic Breast Tumor Cells. Front Cell Dev Biol 2022; 10:790410. [PMID: 35252171 PMCID: PMC8891987 DOI: 10.3389/fcell.2022.790410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/31/2022] [Indexed: 11/22/2022] Open
Abstract
Triple negative breast cancer (TNBC) follows a non-random pattern of metastasis to the bone and brain tissue. Prior work has found that brain-seeking breast tumor cells display altered proteomic profiles, leading to alterations in pathways related to cell signaling, cell cycle, metabolism, and extracellular matrix remodeling. Given the unique microenvironmental characteristics of brain and bone tissue, we hypothesized that brain- or bone-seeking TNBC cells may have altered morphologic or migratory phenotypes from each other, or from the parental TNBC cells, as a function of the biochemical or mechanical microenvironment. In this study, we utilized TNBC cells (MDA-MB-231) that were conditioned to metastasize solely to brain (MDA-BR) or bone (MDA-BO) tissue. We quantified characteristics such as cell morphology, migration, and stiffness in response to cues that partially mimic their final metastatic niche. We have shown that MDA-BO cells have a distinct protrusive morphology not found in MDA-P or MDA-BR. Further, MDA-BO cells migrate over a larger area when on a collagen I (abundant in bone tissue) substrate when compared to fibronectin (abundant in brain tissue). However, migration in highly confined environments was similar across the cell types. Modest differences were found in the stiffness of MDA-BR and MDA-BO cells plated on collagen I vs. fibronectin-coated surfaces. Lastly, MDA-BO cells were found to have larger focal adhesion area and density in comparison with the other two cell types. These results initiate a quantitative profile of mechanobiological phenotypes in TNBC, with future impacts aiming to help predict metastatic propensities to organ-specific sites in a clinical setting.
Collapse
Affiliation(s)
- Ariana Joy L. DeCastro
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Marina A. Pranda
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Kelsey M. Gray
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - John Merlo-Coyne
- Department of Biology, University of Maryland, College Park, MD, United States
| | - Nathaniel Girma
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Madelyn Hurwitz
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Yuji Zhang
- Department of Epidemiology and Public Health, University of Maryland, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, United States
| | - Kimberly M. Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, United States
- Biophysics Program, University of Maryland, College Park, MD, United States
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland, Baltimore, MD, United States
- *Correspondence: Kimberly M. Stroka,
| |
Collapse
|
12
|
Wang L, Zeng D, Wang Q, Liu L, Lu T, Gao Y. Screening and Identification of Novel Potential Biomarkers for Breast Cancer Brain Metastases. Front Oncol 2022; 11:784096. [PMID: 35096583 PMCID: PMC8792448 DOI: 10.3389/fonc.2021.784096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/09/2021] [Indexed: 12/29/2022] Open
Abstract
Brain metastases represent a major cause of mortality among patients with breast cancer, and few effective targeted treatment options are currently available. Development of new biomarkers and therapeutic targets for breast cancer brain metastases (BCBM) is therefore urgently needed. In this study, we compared the gene expression profiles of the brain metastatic cell line MDA-MB-231-BR (231-BR) and its parental MDA-MB-231, and identified a total of 84 genes in the primary screening through a series of bioinformatic analyses, including construction of protein-protein interaction (PPI) networks by STRING database, identification of hub genes by applying of MCODE and Cytohubba algorithms, identification of leading-edge subsets of Gene Set Enrichment Analysis (GSEA), and identification of most up-regulated genes. Eight genes were identified as candidate genes due to their elevated expression in brain metastatic 231-BR cells and prognostic values in patients with BCBM. Then we knocked down the eight individual candidate genes in 231-BR cells and evaluated their impact on cell migration through a wound-healing assay, and four of them (KRT19, FKBP10, GSK3B and SPANXB1) were finally identified as key genes. Furthermore, the expression of individual key genes showed a correlation with the infiltration of major immune cells in the brain tumor microenvironment (TME) as analyzed by Tumor Immune Estimation Resource (TIMER) and Gene Expression Profiling Interactive Analysis (GEPIA), suggesting possible roles of them in regulation of the tumor immune response in TME. Therefore, the present work may provide new potential biomarkers for BCBM. Additionally, using GSEA, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) Enrichment Analysis, we determined the top enriched cellular functions or pathways in 231-BR cells, which may help better understand the biology governing the development and progression of BCBM.
Collapse
Affiliation(s)
- Lulu Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,Cancer Institute of Capital Medical University, Beijing, China
| | - Dan Zeng
- Department of Human Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Qi Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Li Liu
- Department of Experimental Center for Basic Medical Teaching, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Tao Lu
- Department of Human Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yan Gao
- Department of Human Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,Cancer Institute of Capital Medical University, Beijing, China
| |
Collapse
|
13
|
EphrinB2-EphB4 Signaling in Neurooncological Disease. Int J Mol Sci 2022; 23:ijms23031679. [PMID: 35163601 PMCID: PMC8836162 DOI: 10.3390/ijms23031679] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 02/06/2023] Open
Abstract
EphrinB2-EphB4 signaling is critical during embryogenesis for cardiovascular formation and neuronal guidance. Intriguingly, critical expression patterns have been discovered in cancer pathologies over the last two decades. Multiple connections to tumor migration, growth, angiogenesis, apoptosis, and metastasis have been identified in vitro and in vivo. However, the molecular signaling pathways are manifold and signaling of the EphB4 receptor or the ephrinB2 ligand is cancer type specific. Here we explore the impact of these signaling pathways in neurooncological disease, including glioma, brain metastasis, and spinal bone metastasis. We identify potential downstream pathways that mediate cancer suppression or progression and seek to understand it´s role in antiangiogenic therapy resistance in glioma. Despite the Janus-faced functions of ephrinB2-EphB4 signaling in cancer Eph signaling remains a promising clinical target.
Collapse
|
14
|
Duan J, Bao C, Xie Y, Guo H, Liu Y, Li J, Liu R, Li P, Bai J, Yan Y, Mu L, Li X, Wang G, Lu W. Targeted core-shell nanoparticles for precise CTCF gene insert in treatment of metastatic breast cancer. Bioact Mater 2021; 11:1-14. [PMID: 34938908 DOI: 10.1016/j.bioactmat.2021.10.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/27/2021] [Accepted: 10/01/2021] [Indexed: 12/13/2022] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR) technology emerges a remarkable potential for cure of refractory cancer like metastatic breast cancer. However, how to efficiently deliver the CRISPR system with non-viral carrier remains a major issue to be solved. Here, we report a kind of targeted core-shell nanoparticles (NPs) carrying dual plasmids (pHR-pCas9) for precise CCCTC-binding factor (CTCF) gene insert to circumvent metastatic breast cancer. The targeted core-shell NPs carrying pHR-pCas9 can accomplish γGTP-mediated cellular uptake and endosomal escape, facilitate the precise insert and stable expression of CTCF gene, inhibit the migration, metastasis, and colonization of metastatic breast cancer cells. Besides, the finding further reveals that the inhibitory mechanism of metastasis could be associated with up-regulating CTCF protein, followed by down-regulating stomatin (STOM) protein. The study offers a universal nanostrategy enabling the robust non-viral delivery of gene-editing system for treatment of severe illness.
Collapse
Affiliation(s)
- Jialun Duan
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, And School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Chunjie Bao
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, And School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Ying Xie
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, And School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Haitao Guo
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, And School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yixuan Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, And School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jianwei Li
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, And School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100875, China
| | - Rui Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, And School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Peishan Li
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, And School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jing Bai
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, And School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yan Yan
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, And School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Limin Mu
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, And School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xueqi Li
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, And School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Guiling Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, And School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Wanliang Lu
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, And School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| |
Collapse
|
15
|
Dynamic Landscape of Extracellular Vesicle-Associated Proteins Is Related to Treatment Response of Patients with Metastatic Breast Cancer. MEMBRANES 2021; 11:membranes11110880. [PMID: 34832109 PMCID: PMC8619728 DOI: 10.3390/membranes11110880] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 12/13/2022]
Abstract
Breast cancer is the leading cause of cancer death in women. The majority of these deaths are due to disease metastasis, in which cancer cells disseminate to multiple organs and disrupt vital physiological functions. It is widely accepted that breast cancer cells secrete extracellular vesicles (EVs), which contain dynamic molecular cargo that act as versatile mediators of intercellular communication. Therefore, Evs. secreted by breast cancer cells could be involved in the development of metastatic disease and resistance to treatment. Moreover, changes in EV cargo could reflect the effects of therapy on their parent tumor cells. The aim of this feasibility study was to quantitatively profile the proteomes of Evs. isolated from blood samples taken from treatment sensitive and resistant metastatic breast cancer patients to identify proteins associated with responses. Three serial blood samples were collected from three patients with metastatic breast cancer receiving systemic therapy including a responder, a non-responder, and a mixed-responder. Evs. were isolated from plasma using size exclusion chromatography and their protein cargo was prepared for tandem mass tag (TMT)-labelling and quantitative analyses using two-dimensional high-performance liquid chromatography followed by tandem mass spectrometry. After filtering, we quantitatively identified 286 proteins with high confidence using a q value of 0.05. Of these, 149 were classified as EV associated candidate proteins and 137 as classical, high abundant plasma proteins. After comparing EV protein abundance between the responder and non-responder, we identified 35 proteins with unique de-regulated abundance patterns that was conserved at multiple time points. We propose that this proof-of-concept approach can be used to identify proteins which have potential as predictors of metastatic breast cancer response to treatment.
Collapse
|
16
|
Márquez-Ortiz RA, Contreras-Zárate MJ, Tesic V, Alvarez-Eraso KLF, Kwak G, Littrell Z, Costello JC, Sreekanth V, Ormond DR, Karam SD, Kabos P, Cittelly DM. IL13Rα2 Promotes Proliferation and Outgrowth of Breast Cancer Brain Metastases. Clin Cancer Res 2021; 27:6209-6221. [PMID: 34544797 PMCID: PMC8595859 DOI: 10.1158/1078-0432.ccr-21-0361] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 08/05/2021] [Accepted: 09/16/2021] [Indexed: 01/09/2023]
Abstract
PURPOSE The survival of women with brain metastases (BM) from breast cancer remains very poor, with over 80% dying within a year of their diagnosis. Here, we define the function of IL13Rα2 in outgrowth of breast cancer brain metastases (BCBM) in vitro and in vivo, and postulate IL13Rα2 as a suitable therapeutic target for BM. EXPERIMENTAL DESIGN We performed IHC staining of IL13Rα2 in BCBM to define its prognostic value. Using inducible shRNAs in TNBC and HER2+ breast-brain metastatic models, we assessed IL13Rα2 function in vitro and in vivo. We performed RNAseq and functional studies to define the molecular mechanisms underlying IL13Rα2 function in BCBM. RESULTS High IL13Rα2 expression in BCBM predicted worse survival after BM diagnoses. IL13Rα2 was essential for cancer-cell survival, promoting proliferation while repressing invasion. IL13Rα2 KD resulted in FAK downregulation, repression of cell cycle and proliferation mediators, and upregulation of Ephrin B1 signaling. Ephrin-B1 (i) promoted invasion of BC cells in vitro, (ii) marked micrometastasis and invasive fronts in BCBM, and (iii) predicted shorter disease-free survival and BM-free survival (BMFS) in breast primary tumors known to metastasize to the brain. In experimental metastases models, which bypass early tumor invasion, downregulation of IL13Rα2 before or after tumor seeding and brain intravasation decreased BMs, suggesting that IL13Rα2 and the promotion of a proliferative phenotype is critical to BM progression. CONCLUSIONS Non-genomic phenotypic adaptations at metastatic sites are critical to BM progression and patients' prognosis. This study opens the road to use IL13Rα2 targeting as a therapeutic strategy for BM.
Collapse
Affiliation(s)
| | | | - Vesna Tesic
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | | | - Gina Kwak
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Zachary Littrell
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - James C Costello
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Varsha Sreekanth
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - D Ryan Ormond
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Peter Kabos
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Diana M Cittelly
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
17
|
Pharmaco-proteogenomic profiling of pediatric diffuse midline glioma to inform future treatment strategies. Oncogene 2021; 41:461-475. [PMID: 34759345 PMCID: PMC8782719 DOI: 10.1038/s41388-021-02102-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 12/13/2022]
Abstract
Diffuse midline glioma (DMG) is a deadly pediatric and adolescent central nervous system (CNS) tumor localized along the midline structures of the brain atop the spinal cord. With a median overall survival (OS) of just 9–11-months, DMG is characterized by global hypomethylation of histone H3 at lysine 27 (H3K27me3), driven by recurring somatic mutations in H3 genes including, HIST1H3B/C (H3.1K27M) or H3F3A (H3.3K27M), or through overexpression of EZHIP in patients harboring wildtype H3. The recent World Health Organization’s 5th Classification of CNS Tumors now designates DMG as, ‘H3 K27-altered’, suggesting that global H3K27me3 hypomethylation is a ubiquitous feature of DMG and drives devastating transcriptional programs for which there are no treatments. H3-alterations co-segregate with various other somatic driver mutations, highlighting the high-level of intertumoral heterogeneity of DMG. Furthermore, DMG is also characterized by very high-level intratumoral diversity with tumors harboring multiple subclones within each primary tumor. Each subclone contains their own combinations of driver and passenger lesions that continually evolve, making precision-based medicine challenging to successful execute. Whilst the intertumoral heterogeneity of DMG has been extensively investigated, this is yet to translate to an increase in patient survival. Conversely, our understanding of the non-genomic factors that drive the rapid growth and fatal nature of DMG, including endogenous and exogenous microenvironmental influences, neurological cues, and the posttranscriptional and posttranslational architecture of DMG remains enigmatic or at best, immature. However, these factors are likely to play a significant role in the complex biological sequelae that drives the disease. Here we summarize the heterogeneity of DMG and emphasize how analysis of the posttranslational architecture may improve treatment paradigms. We describe factors that contribute to treatment response and disease progression, as well as highlight the potential for pharmaco-proteogenomics (i.e., the integration of genomics, proteomics and pharmacology) in the management of this uniformly fatal cancer.
Collapse
|
18
|
Trigg NA, Skerrett-Byrne DA, Xavier MJ, Zhou W, Anderson AL, Stanger SJ, Katen AL, De Iuliis GN, Dun MD, Roman SD, Eamens AL, Nixon B. Acrylamide modulates the mouse epididymal proteome to drive alterations in the sperm small non-coding RNA profile and dysregulate embryo development. Cell Rep 2021; 37:109787. [PMID: 34610313 DOI: 10.1016/j.celrep.2021.109787] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 08/10/2021] [Accepted: 09/09/2021] [Indexed: 12/12/2022] Open
Abstract
Paternal exposure to environmental stressors elicits distinct changes to the sperm sncRNA profile, modifications that have significant post-fertilization consequences. Despite this knowledge, there remains limited mechanistic understanding of how paternal exposures modify the sperm sncRNA landscape. Here, we report the acute sensitivity of the sperm sncRNA profile to the reproductive toxicant acrylamide. Furthermore, we trace the differential accumulation of acrylamide-responsive sncRNAs to coincide with sperm transit of the proximal (caput) segment of the epididymis, wherein acrylamide exposure alters the abundance of several transcription factors implicated in the expression of acrylamide-sensitive sncRNAs. We also identify extracellular vesicles secreted from the caput epithelium in relaying altered sncRNA profiles to maturing spermatozoa and dysregulated gene expression during early embryonic development following fertilization by acrylamide-exposed spermatozoa. These data provide mechanistic links to account for how environmental insults can alter the sperm epigenome and compromise the transcriptomic profile of early embryos.
Collapse
Affiliation(s)
- Natalie A Trigg
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - David A Skerrett-Byrne
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Miguel J Xavier
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Wei Zhou
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia; Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC 3052, Australia; Gynaecology Research Centre, The Royal Women's Hospital, Parkville, VIC 3052, Australia
| | - Amanda L Anderson
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Simone J Stanger
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Aimee L Katen
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia; Priority Research Centre for Drug Development, School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Geoffry N De Iuliis
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Matthew D Dun
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Research Centre for Cancer Research Innovation and Translation, Hunter Medical Research Institute, Lambton, NSW 2305, Australia
| | - Shaun D Roman
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia; Priority Research Centre for Drug Development, School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Andrew L Eamens
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| |
Collapse
|
19
|
Ebrahim AS, Hailat Z, Bandyopadhyay S, Neill D, Kandouz M. The Value of EphB2 Receptor and Cognate Ephrin Ligands in Prognostic and Predictive Assessments of Human Breast Cancer. Int J Mol Sci 2021; 22:ijms22158098. [PMID: 34360867 PMCID: PMC8348398 DOI: 10.3390/ijms22158098] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/22/2021] [Accepted: 07/22/2021] [Indexed: 01/01/2023] Open
Abstract
Cell–cell communication proteins Eph and ephrin constitute the largest family of receptor tyrosine kinases (RTKs). They are distinguished by the fact that both receptors and ligands are membrane-bound, and both can drive intracellular signaling in their respective cells. Ever since these RTKs have been found to be involved in cancer development, strategies to target them therapeutically have been actively pursued. However, before this goal can be rationally achieved, the contributions of either Eph receptors or their ephrin ligands to cancer development and progression should be scrutinized in depth. To assess the clinical pertinence of this concern, we performed a systematic review and meta-analysis of the prognostic/predictive value of EphB2 and its multiple cognate ephrin ligands in breast cancer. We found that EphB2 has prognostic value, as indicated by the association of higher EphB2 expression levels with lower distant metastasis-free survival (DMFS), and the association of lower EphB2 expression levels with poorer relapse-free survival (RFS). We also found that higher EphB2 expression could be a prognostic factor for distant metastasis, specifically in the luminal subtypes of breast cancer. EFNB2 showed a marked correlation between higher expression levels and shorter DMFS. EFNA5 or EFNB1 overexpression is correlated with longer RFS. Increased EFNB1 expression is correlated with longer OS in lymph node (LN)-negative patients and the luminal B subtype. Higher levels of EFNB2 or EFNA5 are significantly correlated with shorter RFS, regardless of LN status. However, while this correlation with shorter RFS is true for EFNB2 in all subtypes except basal, it is also true for EFNA5 in all subtypes except HER2+. The analysis also points to possible predictive value for EphB2. In systemically treated patients who have undergone either endocrine therapy or chemotherapy, we found that higher expression of EphB2 is correlated with better rates of RFS. Bearing in mind the limitations inherent to any mRNA-based profiling method, we complemented our analysis with an immunohistochemical assessment of expression levels of both the EphB2 receptor and cognate ephrin ligands. We found that the latter are significantly more expressed in cancers than in normal tissues, and even more so in invasive and metastatic samples than in ductal carcinoma in situ (DCIS). Finally, in an in vitro cellular model of breast cancer progression, based on H-Ras-transformation of the MCF10A benign mammary cell line, we observed dramatic increases in the mRNA expression of EphB2 receptor and EFNB1 and EFNB2 ligands in transformed and invasive cells in comparison with their benign counterparts. Taken together, these data show the clinical validity of a model whereby EphB2, along with its cognate ephrin ligands, have dual anti- and pro-tumor progression effects. In so doing, they reinforce the necessity of further biological investigations into Ephs and ephrins, prior to using them in targeted therapies.
Collapse
Affiliation(s)
- Abdul Shukkur Ebrahim
- Department of Ophthalmology, Visual & Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| | - Zeyad Hailat
- Department of Computer Science, Wayne State University, Detroit, MI 48201, USA;
| | - Sudeshna Bandyopadhyay
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.B.); (D.N.)
| | - Daniel Neill
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.B.); (D.N.)
| | - Mustapha Kandouz
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.B.); (D.N.)
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
- Correspondence:
| |
Collapse
|
20
|
Skerrett-Byrne DA, Bromfield EG, Murray HC, Jamaluddin MFB, Jarnicki AG, Fricker M, Essilfie AT, Jones B, Haw TJ, Hampsey D, Anderson AL, Nixon B, Scott RJ, Wark PAB, Dun MD, Hansbro PM. Time-resolved proteomic profiling of cigarette smoke-induced experimental chronic obstructive pulmonary disease. Respirology 2021; 26:960-973. [PMID: 34224176 DOI: 10.1111/resp.14111] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 05/01/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVE Chronic obstructive pulmonary disease (COPD) is the third leading cause of illness and death worldwide. Current treatments aim to control symptoms with none able to reverse disease or stop its progression. We explored the major molecular changes in COPD pathogenesis. METHODS We employed quantitative label-based proteomics to map the changes in the lung tissue proteome of cigarette smoke-induced experimental COPD that is induced over 8 weeks and progresses over 12 weeks. RESULTS Quantification of 7324 proteins enabled the tracking of changes to the proteome. Alterations in protein expression profiles occurred in the induction phase, with 18 and 16 protein changes at 4- and 6-week time points, compared to age-matched controls, respectively. Strikingly, 269 proteins had altered expression after 8 weeks when the hallmark pathological features of human COPD emerge, but this dropped to 27 changes at 12 weeks with disease progression. Differentially expressed proteins were validated using other mouse and human COPD bronchial biopsy samples. Major changes in RNA biosynthesis (heterogeneous nuclear ribonucleoproteins C1/C2 [HNRNPC] and RNA-binding protein Musashi homologue 2 [MSI2]) and modulators of inflammatory responses (S100A1) were notable. Mitochondrial dysfunction and changes in oxidative stress proteins also occurred. CONCLUSION We provide a detailed proteomic profile, identifying proteins associated with the pathogenesis and disease progression of COPD establishing a platform to develop effective new treatment strategies.
Collapse
Affiliation(s)
- David A Skerrett-Byrne
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Elizabeth G Bromfield
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia.,Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Heather C Murray
- University of Newcastle, Callaghan, New South Wales, Australia.,Cancer Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - M Fairuz B Jamaluddin
- University of Newcastle, Callaghan, New South Wales, Australia.,Cancer Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Andrew G Jarnicki
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Michael Fricker
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Ama T Essilfie
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Queensland Institute of Medical Research, Herston, Queensland, Australia
| | - Bernadette Jones
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Tatt J Haw
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Daniel Hampsey
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Amanda L Anderson
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Brett Nixon
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Rodney J Scott
- University of Newcastle, Callaghan, New South Wales, Australia.,Cancer Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Matthew D Dun
- University of Newcastle, Callaghan, New South Wales, Australia.,Cancer Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia.,Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
21
|
An M, Zang X, Wang J, Kang J, Tan X, Fu B. Comprehensive analysis of differentially expressed long noncoding RNAs, miRNAs and mRNAs in breast cancer brain metastasis. Epigenomics 2021; 13:1113-1128. [PMID: 34148372 DOI: 10.2217/epi-2021-0152] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Aim: To delineate the transcriptomic landscape and potential molecular mechanisms of breast cancer brain metastasis (BCBM). Materials & methods: Whole-transcriptome sequencing was performed to identify long noncoding RNA (lncRNA), miRNA and mRNA expression profiles associated with BCBM. Results: A total of 739 differentially expressed lncRNAs, 115 differentially expressed miRNAs and 5749 differentially expressed mRNAs were identified in 231-BR cells compared with MDA-MB-231 cells. Real-time quantitative PCR results revealed the expression levels of candidate molecules were consistent with their correspondence RNA-seq data. Protein-protein interaction analysis identified some hub genes associated with BCBM, such as PTBP1, NUP98 and HYOU1. LncRNA-miRNA-mRNA network highlighted a potential mechanism of BCBM in which lncRNA FIRRE and RP11-169F17.1 sponging hsa-miR-501-5p to regulate the expression of MMS19, PTBP1 and NUP98. Conclusion: This study provides a framework for better understanding molecular mechanisms of BCBM.
Collapse
Affiliation(s)
- Meng An
- Department of Clinical Laboratory, Liaocheng People's Hospital, Liaocheng, PR China
| | - Xiaowen Zang
- Department of Neurology First Ward, Liaocheng Veterans Hospital, Liaocheng, PR China
| | - Jimin Wang
- Department of Clinical Laboratory, Liaocheng People's Hospital, Liaocheng, PR China
| | - Jie Kang
- Department of Stomatology, Liaocheng People's Hospital, Liaocheng, PR China
| | - Xiaoyu Tan
- Department of Clinical Laboratory, Liaocheng People's Hospital, Liaocheng, PR China
| | - Bo Fu
- Department of Central Laboratory, Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, PR China
| |
Collapse
|
22
|
Skerrett-Byrne DA, Trigg NA, Bromfield EG, Dun MD, Bernstein IR, Anderson AL, Stanger SJ, MacDougall LA, Lord T, Aitken RJ, Roman SD, Robertson SA, Nixon B, Schjenken JE. Proteomic Dissection of the Impact of Environmental Exposures on Mouse Seminal Vesicle Function. Mol Cell Proteomics 2021; 20:100107. [PMID: 34089863 PMCID: PMC8250459 DOI: 10.1016/j.mcpro.2021.100107] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/19/2021] [Accepted: 05/28/2021] [Indexed: 12/15/2022] Open
Abstract
Seminal vesicles are an integral part of the male reproductive accessory gland system. They produce a complex array of secretions containing bioactive constituents that support gamete function and promote reproductive success, with emerging evidence suggesting these secretions are influenced by our environment. Despite their significance, the biology of seminal vesicles remains poorly defined. Here, we complete the first proteomic assessment of mouse seminal vesicles and assess the impact of the reproductive toxicant acrylamide. Mice were administered acrylamide (25 mg/kg bw/day) or control daily for five consecutive days prior to collecting seminal vesicle tissue. A total of 5013 proteins were identified in the seminal vesicle proteome with bioinformatic analyses identifying cell proliferation, protein synthesis, cellular death, and survival pathways as prominent biological processes. Secreted proteins were among the most abundant, and several proteins are linked with seminal vesicle phenotypes. Analysis of the effect of acrylamide on the seminal vesicle proteome revealed 311 differentially regulated (FC ± 1.5, p ≤ 0.05, 205 up-regulated, 106 downregulated) proteins, orthogonally validated via immunoblotting and immunohistochemistry. Pathways that initiate protein synthesis to promote cellular survival were prominent among the dysregulated pathways, and rapamycin-insensitive companion of mTOR (RICTOR, p = 6.69E-07) was a top-ranked upstream driver. Oxidative stress was implicated as contributing to protein changes, with acrylamide causing an increase in 8-OHdG in seminal vesicle epithelial cells (fivefold increase, p = 0.016) and the surrounding smooth muscle layer (twofold increase, p = 0.043). Additionally, acrylamide treatment caused a reduction in seminal vesicle secretion weight (36% reduction, p = 0.009) and total protein content (25% reduction, p = 0.017). Together these findings support the interpretation that toxicant exposure influences male accessory gland physiology and highlights the need to consider the response of all male reproductive tract tissues when interpreting the impact of environmental stressors on male reproductive function.
Collapse
Affiliation(s)
- David A Skerrett-Byrne
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Natalie A Trigg
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Elizabeth G Bromfield
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Matthew D Dun
- Cancer Signalling Research Group, Faculty of Health and Medicine, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Priority Research Centre for Cancer Research Innovation and Translation, Hunter Medical Research Institute, Lambton, NSW, Australia
| | - Ilana R Bernstein
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Amanda L Anderson
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Simone J Stanger
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Lily A MacDougall
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Tessa Lord
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - R John Aitken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Shaun D Roman
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Sarah A Robertson
- The Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - John E Schjenken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| |
Collapse
|
23
|
Gao F, Griffin N, Faulkner S, Li X, King SJ, Jobling P, Denham JW, Jiang CC, Hondermarck H. The Membrane Protein Sortilin Can Be Targeted to Inhibit Pancreatic Cancer Cell Invasion. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1931-1942. [PMID: 32526166 DOI: 10.1016/j.ajpath.2020.05.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/03/2020] [Accepted: 05/26/2020] [Indexed: 12/16/2022]
Abstract
Pancreatic cancer has a dismal prognosis, and there is no targeted therapy against this malignancy. The neuronal membrane protein sortilin is emerging as a regulator of cancer cell development, but its expression and impact in pancreatic cancer are unknown. This study found that sortilin expression was higher in pancreatic cell lines versus normal pancreatic ductal epithelial cells, as shown by Western blot analysis and mass spectrometry. The increased sortilin level in pancreatic cancer cells was confirmed by immunohistochemistry in a series of 99 human pancreatic adenocarcinomas versus 48 normal pancreatic tissues (P = 0.0014). Sortilin inhibition by siRNA and the pharmacologic inhibitor AF38469 strongly reduced the adhesion and invasion of pancreatic cancer cells without affecting cell survival and viability. Sortilin inhibition also decreased the phosphorylation of the focal adhesion kinase in Tyr925. Together, these data show that sortilin contributes to pancreatic cancer invasion and could eventually be targeted in therapy.
Collapse
Affiliation(s)
- Fangfang Gao
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, Australia; Hunter Medical Research Institute, University of Newcastle, New Lambton, New South Wales, Australia
| | - Nathan Griffin
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, Australia; Hunter Medical Research Institute, University of Newcastle, New Lambton, New South Wales, Australia
| | - Sam Faulkner
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, Australia; Hunter Medical Research Institute, University of Newcastle, New Lambton, New South Wales, Australia
| | - Xiang Li
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, Australia; Hunter Medical Research Institute, University of Newcastle, New Lambton, New South Wales, Australia
| | - Simon J King
- Hunter Medical Research Institute, University of Newcastle, New Lambton, New South Wales, Australia
| | - Phillip Jobling
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, Australia; Hunter Medical Research Institute, University of Newcastle, New Lambton, New South Wales, Australia
| | - Jim W Denham
- Hunter Medical Research Institute, University of Newcastle, New Lambton, New South Wales, Australia
| | - Chen Chen Jiang
- School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, Australia; Hunter Medical Research Institute, University of Newcastle, New Lambton, New South Wales, Australia
| | - Hubert Hondermarck
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, Australia; Hunter Medical Research Institute, University of Newcastle, New Lambton, New South Wales, Australia.
| |
Collapse
|
24
|
Oliver CR, Westerhof TM, Castro MG, Merajver SD. Quantifying the Brain Metastatic Tumor Micro-Environment using an Organ-On-A Chip 3D Model, Machine Learning, and Confocal Tomography. J Vis Exp 2020. [PMID: 32865534 DOI: 10.3791/61654] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Brain metastases are the most lethal cancer lesions; 10-30% of all cancers metastasize to the brain, with a median survival of only ~5-20 months, depending on the cancer type. To reduce the brain metastatic tumor burden, gaps in basic and translational knowledge need to be addressed. Major challenges include a paucity of reproducible preclinical models and associated tools. Three-dimensional models of brain metastasis can yield the relevant molecular and phenotypic data used to address these needs when combined with dedicated analysis tools. Moreover, compared to murine models, organ-on-a-chip models of patient tumor cells traversing the blood brain barrier into the brain microenvironment generate results rapidly and are more interpretable with quantitative methods, thus amenable to high throughput testing. Here we describe and demonstrate the use of a novel 3D microfluidic blood brain niche (µmBBN) platform where multiple elements of the niche can be cultured for an extended period (several days), fluorescently imaged by confocal microscopy, and the images reconstructed using an innovative confocal tomography technique; all aimed to understand the development of micro-metastasis and changes to the tumor micro-environment (TME) in a repeatable and quantitative manner. We demonstrate how to fabricate, seed, image, and analyze the cancer cells and TME cellular and humoral components, using this platform. Moreover, we show how artificial intelligence (AI) is used to identify the intrinsic phenotypic differences of cancer cells that are capable of transit through a model µmBBN and to assign them an objective index of brain metastatic potential. The data sets generated by this method can be used to answer basic and translational questions about metastasis, the efficacy of therapeutic strategies, and the role of the TME in both.
Collapse
Affiliation(s)
- C Ryan Oliver
- Department of Internal Medicine, University of Michigan Ann Arbor; Rogel Cancer Center, University of Michigan Ann Arbor
| | - Trisha M Westerhof
- Department of Internal Medicine, University of Michigan Ann Arbor; Rogel Cancer Center, University of Michigan Ann Arbor
| | - Maria G Castro
- Rogel Cancer Center, University of Michigan Ann Arbor; Department of Neurosurgery, University of Michigan Ann Arbor; Department of Cell and Developmental Biology, University of Michigan Ann Arbor
| | - Sofia D Merajver
- Department of Internal Medicine, University of Michigan Ann Arbor; Rogel Cancer Center, University of Michigan Ann Arbor;
| |
Collapse
|
25
|
Wong AHH, Shin EM, Tergaonkar V, Chng WJ. Targeting NF-κB Signaling for Multiple Myeloma. Cancers (Basel) 2020; 12:cancers12082203. [PMID: 32781681 PMCID: PMC7463546 DOI: 10.3390/cancers12082203] [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: 07/12/2020] [Revised: 07/30/2020] [Accepted: 08/01/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) is the second most common hematologic malignancy in the world. Even though survival rates have significantly risen over the past years, MM remains incurable, and is also far from reaching the point of being managed as a chronic disease. This paper reviews the evolution of MM therapies, focusing on anti-MM drugs that target the molecular mechanisms of nuclear factor kappa B (NF-κB) signaling. We also provide our perspectives on contemporary research findings and insights for future drug development.
Collapse
Affiliation(s)
- Ada Hang-Heng Wong
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore; (E.M.S.); (V.T.)
- AW Medical Company Limited, Macau, China
- Correspondence: (A.H.-H.W.); (W.-J.C.); Tel.: +65-6586-9709 (A.H.-H.W.); +65-6772-4612 (W.-J.C.)
| | - Eun Myoung Shin
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore; (E.M.S.); (V.T.)
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore; (E.M.S.); (V.T.)
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
- Department of Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Wee-Joo Chng
- Cancer Science Institute of Singapore, Singapore 117599, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Department of Hematology-Oncology, National University Cancer Institute of Singapore, National University Health System, Singapore 119074, Singapore
- Correspondence: (A.H.-H.W.); (W.-J.C.); Tel.: +65-6586-9709 (A.H.-H.W.); +65-6772-4612 (W.-J.C.)
| |
Collapse
|
26
|
Schaffner-Reckinger E, Machado RAC. The actin-bundling protein L-plastin-A double-edged sword: Beneficial for the immune response, maleficent in cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 355:109-154. [PMID: 32859369 DOI: 10.1016/bs.ircmb.2020.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The dynamic organization of the actin cytoskeleton into bundles and networks is orchestrated by a large variety of actin-binding proteins. Among them, the actin-bundling protein L-plastin is normally expressed in hematopoietic cells, where it is involved in the immune response. However, L-plastin is also often ectopically expressed in malignant cancer cells of non-hematopoietic origin and is even considered as a marker for cancer progression. Post-translational modification modulates L-plastin activity. In particular, L-plastin Ser5 phosphorylation has been shown to be important for the immune response in leukocytes as well as for invasion and metastasis formation of carcinoma cells. This chapter discusses the physiological and pathological role of L-plastin with a special focus on the importance of L-plastin Ser5 phosphorylation for the protein functions. The potential use of Ser5 phosphorylated L-plastin as a biomarker and/or therapeutic target will be evoked.
Collapse
Affiliation(s)
- Elisabeth Schaffner-Reckinger
- Cancer Cell Biology and Drug Discovery Group, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| | - Raquel A C Machado
- Cancer Cell Biology and Drug Discovery Group, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
27
|
Afrin F, Chi M, Eamens AL, Duchatel RJ, Douglas AM, Schneider J, Gedye C, Woldu AS, Dun MD. Can Hemp Help? Low-THC Cannabis and Non-THC Cannabinoids for the Treatment of Cancer. Cancers (Basel) 2020; 12:cancers12041033. [PMID: 32340151 PMCID: PMC7226605 DOI: 10.3390/cancers12041033] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 04/20/2020] [Indexed: 12/21/2022] Open
Abstract
Cannabis has been used to relieve the symptoms of disease for thousands of years. However, social and political biases have limited effective interrogation of the potential benefits of cannabis and polarised public opinion. Further, the medicinal and clinical utility of cannabis is limited by the psychotropic side effects of ∆9-tetrahydrocannabinol (∆9-THC). Evidence is emerging for the therapeutic benefits of cannabis in the treatment of neurological and neurodegenerative diseases, with potential efficacy as an analgesic and antiemetic for the management of cancer-related pain and treatment-related nausea and vomiting, respectively. An increasing number of preclinical studies have established that ∆9-THC can inhibit the growth and proliferation of cancerous cells through the modulation of cannabinoid receptors (CB1R and CB2R), but clinical confirmation remains lacking. In parallel, the anti-cancer properties of non-THC cannabinoids, such as cannabidiol (CBD), are linked to the modulation of non-CB1R/CB2R G-protein-coupled receptors, neurotransmitter receptors, and ligand-regulated transcription factors, which together modulate oncogenic signalling and redox homeostasis. Additional evidence has also demonstrated the anti-inflammatory properties of cannabinoids, and this may prove relevant in the context of peritumoural oedema and the tumour immune microenvironment. This review aims to document the emerging mechanisms of anti-cancer actions of non-THC cannabinoids.
Collapse
Affiliation(s)
- Farjana Afrin
- Cancer Signalling Research Group, Medical Biochemistry, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; (F.A.); (M.C.); (R.J.D.); (A.M.D.); (C.G.)
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health and Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;
| | - Mengna Chi
- Cancer Signalling Research Group, Medical Biochemistry, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; (F.A.); (M.C.); (R.J.D.); (A.M.D.); (C.G.)
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health and Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;
| | - Andrew L. Eamens
- Centre for Plant Science, School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia;
| | - Ryan J. Duchatel
- Cancer Signalling Research Group, Medical Biochemistry, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; (F.A.); (M.C.); (R.J.D.); (A.M.D.); (C.G.)
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health and Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;
| | - Alicia M. Douglas
- Cancer Signalling Research Group, Medical Biochemistry, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; (F.A.); (M.C.); (R.J.D.); (A.M.D.); (C.G.)
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health and Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;
| | - Jennifer Schneider
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health and Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;
- Priority Research Centre for Chemical Biology and Clinical Pharmacology, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Craig Gedye
- Cancer Signalling Research Group, Medical Biochemistry, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; (F.A.); (M.C.); (R.J.D.); (A.M.D.); (C.G.)
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health and Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;
- Calvary Mater Newcastle, Waratah, NSW 2298, Australia
| | - Ameha S. Woldu
- Cancer Signalling Research Group, Medical Biochemistry, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; (F.A.); (M.C.); (R.J.D.); (A.M.D.); (C.G.)
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health and Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;
- Correspondence: (A.S.W.); (M.D.D.); Tel.: +61-02-4921-7807 (A.S.W.); +61-02-4921-5693 (M.D.D.)
| | - Matthew D. Dun
- Cancer Signalling Research Group, Medical Biochemistry, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; (F.A.); (M.C.); (R.J.D.); (A.M.D.); (C.G.)
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health and Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;
- Correspondence: (A.S.W.); (M.D.D.); Tel.: +61-02-4921-7807 (A.S.W.); +61-02-4921-5693 (M.D.D.)
| |
Collapse
|
28
|
Mousa H, Elgamal M, Marei RG, Souchelnytskyi N, Lin KW, Souchelnytskyi S. Acquisition of Invasiveness by Breast Adenocarcinoma Cells Engages Established Hallmarks and Novel Regulatory Mechanisms. Cancer Genomics Proteomics 2019; 16:505-518. [PMID: 31659104 PMCID: PMC6885374 DOI: 10.21873/cgp.20153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND/AIM Proteomics of invasiveness opens a window on the complexity of the metastasis-engaged mechanisms. The extend and types of this complexity require elucidation. MATERIALS AND METHODS Proteomics, immunohistochemistry, immunoblotting, network analysis and systems cancer biology were used to analyse acquisition of invasiveness by human breast adenocarcinoma cells. RESULTS We report here that invasiveness network highlighted the involvement of hallmarks such as cell proliferation, migration, cell death, genome stability, immune system regulation and metabolism. Identified involvement of cell-virus interaction and gene silencing are potentially novel cancer mechanisms. Identified 6,113 nodes with 11,055 edges affecting 1,085 biological processes show extensive re-arrangements in cell physiology. These high numbers are in line with a similar broadness of networks built with diagnostic signatures approved for clinical use. CONCLUSION Our data emphasize a broad systemic regulation of invasiveness, and describe the network of this regulation.
Collapse
Affiliation(s)
- Hanaa Mousa
- College of Medicine, Qatar University, Doha, Qatar
| | | | | | | | - Kah-Wai Lin
- College of Medicine, Qatar University, Doha, Qatar
- Neurocentrum, Karolinska University Hospital, Solna, Sweden
| | | |
Collapse
|
29
|
Pranda MA, Gray KM, DeCastro AJL, Dawson GM, Jung JW, Stroka KM. Tumor Cell Mechanosensing During Incorporation into the Brain Microvascular Endothelium. Cell Mol Bioeng 2019; 12:455-480. [PMID: 31719927 DOI: 10.1007/s12195-019-00591-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 08/17/2019] [Indexed: 12/11/2022] Open
Abstract
Introduction Tumor metastasis to the brain occurs in approximately 20% of all cancer cases and often occurs due to tumor cells crossing the blood-brain barrier (BBB). The brain microenvironment is comprised of a soft hyaluronic acid (HA)-rich extracellular matrix with an elastic modulus of 0.1-1 kPa, whose crosslinking is often altered in disease states. Methods To explore the effects of HA crosslinking on breast tumor cell migration, we developed a biomimetic model of the human brain endothelium, consisting of brain microvascular endothelial cell (HBMEC) monolayers on HA and gelatin (HA/gelatin) films with different degrees of crosslinking, as established by varying the concentration of the crosslinker Extralink. Results and Discussion Metastatic breast tumor cell migration speed, diffusion coefficient, spreading area, and aspect ratio increased with decreasing HA crosslinking, a mechanosensing trend that correlated with tumor cell actin organization but not CD44 expression. Meanwhile, breast tumor cell incorporation into endothelial monolayers was independent of HA crosslinking density, suggesting that alterations in HA crosslinking density affect tumor cells only after they exit the vasculature. Tumor cells appeared to exploit both the paracellular and transcellular routes of trans-endothelial migration. Quantitative phenotyping of HBMEC junctions via a novel Python software revealed a VEGF-dependent decrease in punctate VE-cadherin junctions and an increase in continuous and perpendicular junctions when HBMECs were treated with tumor cell-secreted factors. Conclusions Overall, our quantitative results suggest that a combination of biochemical and physical factors promote tumor cell migration through the BBB.
Collapse
Affiliation(s)
- Marina A Pranda
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA
| | - Kelsey M Gray
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA
| | - Ariana Joy L DeCastro
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA
| | - Gregory M Dawson
- Department of Biology, University of Maryland, College Park, College Park, MD 20742 USA
| | - Jae W Jung
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA
| | - Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA.,Biophysics Program, University of Maryland, College Park, College Park, MD 20742 USA.,Center for Stem Cell Biology and Regenerative Medicine, University of Maryland - Baltimore, Baltimore, MD 21201 USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland - Baltimore, Baltimore, MD 21201 USA.,Fischell Department of Bioengineering, University of Maryland, College Park, 3110 A. James Clark Hall, 8278 Paint Branch Drive, College Park, MD 20742 USA
| |
Collapse
|
30
|
Duchatel RJ, Jackson ER, Alvaro F, Nixon B, Hondermarck H, Dun MD. Signal Transduction in Diffuse Intrinsic Pontine Glioma. Proteomics 2019; 19:e1800479. [DOI: 10.1002/pmic.201800479] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 05/03/2019] [Indexed: 11/12/2022]
Affiliation(s)
- Ryan J. Duchatel
- Cancer Signalling Research Group School of Biomedical Sciences and Pharmacy Faculty of Health and Medicine University of Newcastle Callaghan NSW 2308 Australia
- Priority Research Centre for Cancer Research Innovation and Translation Hunter Medical Research Institute Lambton NSW 2305 Australia
| | - Evangeline R. Jackson
- Cancer Signalling Research Group School of Biomedical Sciences and Pharmacy Faculty of Health and Medicine University of Newcastle Callaghan NSW 2308 Australia
- Priority Research Centre for Cancer Research Innovation and Translation Hunter Medical Research Institute Lambton NSW 2305 Australia
| | - Frank Alvaro
- Priority Research Centre for Cancer Research Innovation and Translation Hunter Medical Research Institute Lambton NSW 2305 Australia
- John Hunter Children's Hospital Faculty of Health and Medicine University of Newcastle New Lambton Heights NSW 2305 Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science School of Environmental and Life Sciences University of Newcastle Callaghan NSW 2308 Australia
| | - Hubert Hondermarck
- Priority Research Centre for Cancer Research Innovation and Translation Hunter Medical Research Institute Lambton NSW 2305 Australia
- Cancer Neurobiology Group School of Biomedical Sciences and Pharmacy Faculty of Health and Medicine University of Newcastle Callaghan NSW 2308 Australia
| | - Matthew D. Dun
- Cancer Signalling Research Group School of Biomedical Sciences and Pharmacy Faculty of Health and Medicine University of Newcastle Callaghan NSW 2308 Australia
- Priority Research Centre for Cancer Research Innovation and Translation Hunter Medical Research Institute Lambton NSW 2305 Australia
| |
Collapse
|
31
|
Fu B, Zhang A, Li M, Pan L, Tang W, An M, Liu W, Zhang J. Circular RNA profile of breast cancer brain metastasis: identification of potential biomarkers and therapeutic targets. Epigenomics 2019; 10:1619-1630. [PMID: 30810051 DOI: 10.2217/epi-2018-0090] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
AIM To explore the circular RNA (circRNA) profile of breast cancer brain metastasis (BCBM). MATERIALS & METHODS RNA-seq was performed to identify the circRNA expression profile of brain metastatic breast cancer cell line 231-BR, in comparison with its parental nonspecific metastatic cell line MDA-MB-231. RESULTS A total of 215 upregulated and 191 downregulated circRNAs were identified. The expression levels of ten randomly selected majorly altered circRNAs were re-examined by real-time quantitative PCR and agarose gel electrophoresis, and the following alterations were verified: upregulation of hsa_circ_0001944, hsa_circ_0001481, hsa_circ_0000646, hsa_circ_0001006 and hsa_circ_0000732, and downregulation of hsa_circ_0001910, hsa_circ_0008285 and hsa_circ_0000002. CircRNA/miRNA analysis revealed that hsa_circ_0001944 may be involved in BCBM through sponging up miR-509 and interfering with its binding to the downstream targets. CONCLUSION This study provides a leading and fundamental circRNA profile of BCBM.
Collapse
Affiliation(s)
- Bo Fu
- Department of Central Laboratory, Liaocheng People's Hospital, Liaocheng, PR China.,School of Basic Medical Science, Shandong University, Jinan, PR China
| | - Anqi Zhang
- Department of Central Laboratory, Liaocheng People's Hospital, Liaocheng, PR China
| | - Mingli Li
- Department of Clinical Laboratory, Weifang Traditional Chinese Hospital, Weifang, PR China
| | - Li Pan
- Department of Central Laboratory, Liaocheng People's Hospital, Liaocheng, PR China
| | - Wenqiang Tang
- Department of Central Laboratory, Liaocheng People's Hospital, Liaocheng, PR China
| | - Meng An
- Department of Clinical Laboratory, Liaocheng People's Hospital, Liaocheng, PR China
| | - Wei Liu
- Department of Central Laboratory, Liaocheng People's Hospital, Liaocheng, PR China
| | - Jianqiong Zhang
- Jiangsu Key Laboratory of Molecule Imaging & Functional Imaging, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, PR China
| |
Collapse
|
32
|
Kalita-de Croft P, Straube J, Lim M, Al-Ejeh F, Lakhani SR, Saunus JM. Proteomic Analysis of the Breast Cancer Brain Metastasis Microenvironment. Int J Mol Sci 2019; 20:ijms20102524. [PMID: 31121957 PMCID: PMC6567270 DOI: 10.3390/ijms20102524] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/09/2019] [Accepted: 05/21/2019] [Indexed: 12/30/2022] Open
Abstract
Patients with brain-metastatic breast cancer face a bleak prognosis marked by morbidity and premature death. A deeper understanding of molecular interactions in the metastatic brain tumour microenvironment may inform the development of new therapeutic strategies. In this study, triple-negative MDA-MB-231 breast cancer cells or PBS (modelling traumatic brain injury) were stereotactically injected into the cerebral cortex of NOD/SCID mice to model metastatic colonization. Brain cells were isolated from five tumour-associated samples and five controls (pooled uninvolved and injured tissue) by immunoaffinity chromatography, and proteomic profiles were compared using the Sequential Window Acquisition of All Theoretical Mass Spectra (SWATH-MS) discovery platform. Ontology and cell type biomarker enrichment analysis of the 125 differentially abundant proteins (p < 0.05) showed the changes largely represent cellular components involved in metabolic reprogramming and cell migration (min q = 4.59 × 10-5), with high-throughput PubMed text mining indicating they have been most frequently studied in the contexts of mitochondrial dysfunction, oxidative stress and autophagy. Analysis of mouse brain cell type-specific biomarkers suggested the changes were paralleled by increased proportions of microglia, mural cells and interneurons. Finally, we orthogonally validated three of the proteins in an independent xenograft cohort, and investigated their expression in craniotomy specimens from triple-negative metastatic breast cancer patients, using a combination of standard and fluorescent multiplex immunohistochemistry. This included 3-Hydroxyisobutyryl-CoA Hydrolase (HIBCH), which is integral for gluconeogenic valine catabolism in the brain, and was strongly induced in both graft-associated brain tissue (13.5-fold by SWATH-MS; p = 7.2 × 10-4), and areas of tumour-associated, reactive gliosis in human clinical samples. HIBCH was also induced in the tumour compartment, with expression frequently localized to margins and haemorrhagic areas. These observations raise the possibility that catabolism of valine is an effective adaptation in metastatic cells able to access it, and that intermediates or products could be transferred from tumour-associated glia. Overall, our findings indicate that metabolic reprogramming dominates the proteomic landscape of graft-associated brain tissue in the intracranial MDA-MB-231 xenograft model. Brain-derived metabolic provisions could represent an exploitable dependency in breast cancer brain metastases.
Collapse
Affiliation(s)
- Priyakshi Kalita-de Croft
- Faculty of Medicine, the University of Queensland, Centre for Clinical Research, Herston 4029, QLD, Australia.
| | - Jasmin Straube
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia.
| | - Malcolm Lim
- Faculty of Medicine, the University of Queensland, Centre for Clinical Research, Herston 4029, QLD, Australia.
| | - Fares Al-Ejeh
- QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia.
| | - Sunil R Lakhani
- Faculty of Medicine, the University of Queensland, Centre for Clinical Research, Herston 4029, QLD, Australia.
- Pathology Queensland, The Royal Brisbane & Women's Hospital, Herston 4029, QLD, Australia.
| | - Jodi M Saunus
- Faculty of Medicine, the University of Queensland, Centre for Clinical Research, Herston 4029, QLD, Australia.
| |
Collapse
|
33
|
Tyran M, Carbuccia N, Garnier S, Guille A, Adelaïde J, Finetti P, Toulzian J, Viens P, Tallet A, Goncalves A, Metellus P, Birnbaum D, Chaffanet M, Bertucci F. A Comparison of DNA Mutation and Copy Number Profiles of Primary Breast Cancers and Paired Brain Metastases for Identifying Clinically Relevant Genetic Alterations in Brain Metastases. Cancers (Basel) 2019; 11:cancers11050665. [PMID: 31086113 PMCID: PMC6562582 DOI: 10.3390/cancers11050665] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/08/2019] [Accepted: 05/11/2019] [Indexed: 12/15/2022] Open
Abstract
Improving the systemic treatment of brain metastases (BM) in primary breast cancer (PBC) is impaired by the lack of genomic characterization of BM. To estimate the concordance of DNA copy-number-alterations (CNAs), mutations, and actionable genetic alterations (AGAs) between paired samples, we performed whole-genome array-comparative-genomic-hybridization, and targeted-next-generation-sequencing on 14 clinical PBC–BM pairs. We found more CNAs, more mutations, and higher tumor mutational burden, and more AGAs in BM than in PBC; 92% of the pairs harbored at least one AGA in the BM not observed in the paired PBC. This concerned various therapeutic classes, including tyrosine-kinase-receptor-inhibitors, phosphatidylinositol 3-kinase/AKT/ mammalian Target of Rapamycin (PI3K/AKT/MTOR)-inhibitors, poly ADP ribose polymerase (PARP)-inhibitors, or cyclin-dependent kinase (CDK)-inhibitors. With regards to the PARP-inhibitors, the homologous recombination defect score was positive in 79% of BM, compared to 43% of PBC, discordant in 7 out of 14 pairs, and positive in the BM in 5 out of 14 cases. CDK-inhibitors were associated with the largest percentage of discordant AGA appearing in the BM. When considering the AGA with the highest clinical-evidence level, for each sample, 50% of the pairs harbored an AGA in the BM not detected or not retained from the analysis of the paired PBC. Thus, the profiling of BM provided a more reliable opportunity, than that of PBC, for diagnostic decision-making based on genomic analysis. Patients with BM deserve an investigation of several targeted therapies.
Collapse
Affiliation(s)
- Marguerite Tyran
- Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Aix-Marseille Université, Institut Paoli-Calmettes, F-13009 Marseille, France.
- Département de Radiothérapie, Institut Paoli-Calmettes, 13009 Marseille, France.
| | - Nadine Carbuccia
- Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Aix-Marseille Université, Institut Paoli-Calmettes, F-13009 Marseille, France.
| | - Séverine Garnier
- Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Aix-Marseille Université, Institut Paoli-Calmettes, F-13009 Marseille, France.
| | - Arnaud Guille
- Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Aix-Marseille Université, Institut Paoli-Calmettes, F-13009 Marseille, France.
| | - José Adelaïde
- Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Aix-Marseille Université, Institut Paoli-Calmettes, F-13009 Marseille, France.
| | - Pascal Finetti
- Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Aix-Marseille Université, Institut Paoli-Calmettes, F-13009 Marseille, France.
| | - Julien Toulzian
- Département d'Anatomopathologie, Institut Paoli-Calmettes, 13009 Marseille, France.
| | - Patrice Viens
- Département d'Oncologie Médicale, Institut Paoli-Calmettes, 13009 Marseille, France.
- Faculté de Médecine, Aix-Marseille Université, 13005 Marseille, France.
| | - Agnès Tallet
- Département de Radiothérapie, Institut Paoli-Calmettes, 13009 Marseille, France.
| | - Anthony Goncalves
- Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Aix-Marseille Université, Institut Paoli-Calmettes, F-13009 Marseille, France.
- Département d'Oncologie Médicale, Institut Paoli-Calmettes, 13009 Marseille, France.
- Faculté de Médecine, Aix-Marseille Université, 13005 Marseille, France.
| | - Philippe Metellus
- Département de Neurochirurgie et de Neuro-oncologie, Hôpital Privé Clairval, Ramsay-Générale de Santé and Institut de Neurophysiopathologie Equipe 10, UMR0751, CNRS, 13009 Marseille, France.
| | - Daniel Birnbaum
- Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Aix-Marseille Université, Institut Paoli-Calmettes, F-13009 Marseille, France.
| | - Max Chaffanet
- Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Aix-Marseille Université, Institut Paoli-Calmettes, F-13009 Marseille, France.
| | - François Bertucci
- Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Aix-Marseille Université, Institut Paoli-Calmettes, F-13009 Marseille, France.
- Département d'Oncologie Médicale, Institut Paoli-Calmettes, 13009 Marseille, France.
- Faculté de Médecine, Aix-Marseille Université, 13005 Marseille, France.
| |
Collapse
|
34
|
Nixon B, De Iuliis GN, Hart HM, Zhou W, Mathe A, Bernstein IR, Anderson AL, Stanger SJ, Skerrett-Byrne DA, Jamaluddin MFB, Almazi JG, Bromfield EG, Larsen MR, Dun MD. Proteomic Profiling of Mouse Epididymosomes Reveals their Contributions to Post-testicular Sperm Maturation. Mol Cell Proteomics 2019; 18:S91-S108. [PMID: 30213844 PMCID: PMC6427233 DOI: 10.1074/mcp.ra118.000946] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/28/2018] [Indexed: 01/31/2023] Open
Abstract
The functional maturation of spermatozoa that is necessary to achieve fertilization occurs as these cells transit through the epididymis, a highly specialized region of the male reproductive tract. A defining feature of this maturation process is that it occurs in the complete absence of nuclear gene transcription or de novo, protein translation in the spermatozoa. Rather, it is driven by sequential interactions between spermatozoa and the complex external milieu in which they are bathed within lumen of the epididymal tubule. A feature of this dynamic microenvironment are epididymosomes, small membrane encapsulated vesicles that are secreted from the epididymal soma. Herein, we report comparative proteomic profiling of epididymosomes isolated from different segments of the mouse epididymis using multiplexed tandem mass tag (TMT) based quantification coupled with high resolution LC-MS/MS. A total of 1640 epididymosome proteins were identified and quantified via this proteomic method. Notably, this analysis revealed pronounced segment-to-segment variation in the encapsulated epididymosome proteome. Thus, 146 proteins were identified as being differentially accumulated between caput and corpus epididymosomes, and a further 344 were differentially accumulated between corpus and cauda epididymosomes (i.e., fold change of ≤ -1.5 or ≥ 1.5; p, < 0.05). Application of gene ontology annotation revealed a substantial portion of the epididymosome proteins mapped to the cellular component of extracellular exosome and to the biological processes of transport, oxidation-reduction, and metabolism. Additional annotation of the subset of epididymosome proteins that have not previously been identified in exosomes revealed enrichment of categories associated with the acquisition of sperm function (e.g., fertilization and binding to the zona pellucida). In tandem with our demonstration that epididymosomes are able to convey protein cargo to the head of maturing spermatozoa, these data emphasize the fundamental importance of epididymosomes as key elements of the epididymal microenvironment responsible for coordinating post-testicular sperm maturation.
Collapse
Affiliation(s)
- Brett Nixon
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - Geoffry N De Iuliis
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - Hanah M Hart
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - Wei Zhou
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - Andrea Mathe
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia;; School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Ilana R Bernstein
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - Amanda L Anderson
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - Simone J Stanger
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - David A Skerrett-Byrne
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - M Fairuz B Jamaluddin
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW 2308, Australia;; Hunter Medical Research Institute, Cancer Research Program, New Lambton Heights, NSW 2305, Australia
| | - Juhura G Almazi
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW 2308, Australia;; Hunter Medical Research Institute, Cancer Research Program, New Lambton Heights, NSW 2305, Australia
| | - Elizabeth G Bromfield
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Matthew D Dun
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW 2308, Australia;; Hunter Medical Research Institute, Cancer Research Program, New Lambton Heights, NSW 2305, Australia.
| |
Collapse
|
35
|
Nixon B, Johnston SD, Skerrett-Byrne DA, Anderson AL, Stanger SJ, Bromfield EG, Martin JH, Hansbro PM, Dun MD. Modification of Crocodile Spermatozoa Refutes the Tenet That Post-testicular Sperm Maturation Is Restricted To Mammals. Mol Cell Proteomics 2019; 18:S58-S76. [PMID: 30072580 PMCID: PMC6427239 DOI: 10.1074/mcp.ra118.000904] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/24/2018] [Indexed: 12/24/2022] Open
Abstract
Competition to achieve paternity has contributed to the development of a multitude of elaborate male reproductive strategies. In one of the most well-studied examples, the spermatozoa of all mammalian species must undergo a series of physiological changes, termed capacitation, in the female reproductive tract before realizing their potential to fertilize an ovum. However, the evolutionary origin and adaptive advantage afforded by capacitation remains obscure. Here, we report the use of comparative and quantitative proteomics to explore the biological significance of capacitation in an ancient reptilian species, the Australian saltwater crocodile (Crocodylus porosus,). Our data reveal that exposure of crocodile spermatozoa to capacitation stimuli elicits a cascade of physiological responses that are analogous to those implicated in the functional activation of their mammalian counterparts. Indeed, among a total of 1119 proteins identified in this study, we detected 126 that were differentially phosphorylated (± 1.2 fold-change) in capacitated versus, noncapacitated crocodile spermatozoa. Notably, this subset of phosphorylated proteins shared substantial evolutionary overlap with those documented in mammalian spermatozoa, and included key elements of signal transduction, metabolic and cellular remodeling pathways. Unlike mammalian sperm, however, we noted a distinct bias for differential phosphorylation of serine (as opposed to tyrosine) residues, with this amino acid featuring as the target for ∼80% of all changes detected in capacitated spermatozoa. Overall, these results indicate that the phenomenon of sperm capacitation is unlikely to be restricted to mammals and provide a framework for understanding the molecular changes in sperm physiology necessary for fertilization.
Collapse
Affiliation(s)
- Brett Nixon
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia;; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;.
| | - Stephen D Johnston
- School of Agriculture and Food Science, The University of Queensland, Gatton, QLD 4343, Australia
| | | | - Amanda L Anderson
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Simone J Stanger
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Elizabeth G Bromfield
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia;; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Jacinta H Martin
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia;; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Philip M Hansbro
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;; Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW 2308, Australia
| | - Matthew D Dun
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;; Priority Research Centre for Cancer Research, Innovation and Translation, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
36
|
Viewing the Eph receptors with a focus on breast cancer heterogeneity. Cancer Lett 2018; 434:160-171. [PMID: 30055288 DOI: 10.1016/j.canlet.2018.07.030] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/18/2018] [Accepted: 07/23/2018] [Indexed: 02/07/2023]
Abstract
Aberrant expression of different family members of the Eph/ephrin system, which comprises the Eph receptors (Ephs) and their ligands (ephrins), has been implicated in various malignancies including breast cancer. The latter presents as a heterogeneous disease with diverse molecular, morphologic and clinical behavior signatures. This review reflects the existing Eph/ephrin literature while focusing on breast cancer heterogeneity. Hormone positive, HER2 positive and triple negative breast cancer (TNBC) cell lines, xenografts/mutant animal models and patient samples are examined separately as, in humans, they represent entities with differences in prognosis and treatment. EphA2, EphB4 and EphB6 are the members most extensively studied in breast cancer. Existing research points to the potential use of various Eph/ephrin members as biomarkers for assessing prognosis and selecting the most suitable therapeutic strategies in variable clinical scenarios, also for overcoming drug resistance, in the era of breast cancer heterogeneity.
Collapse
|
37
|
Xiong Y, Li KX, Wei H, Jiao L, Yu SY, Zeng L. Eph/ephrin signalling serves a bidirectional role in lipopolysaccharide‑induced intestinal injury. Mol Med Rep 2018; 18:2171-2181. [PMID: 29901151 PMCID: PMC6072232 DOI: 10.3892/mmr.2018.9169] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 06/06/2018] [Indexed: 12/22/2022] Open
Abstract
A growing body of evidence has demonstrated that Eph/ephrin signalling may serve a central role in intestinal diseases. However, whether erythropoietin-producing hepatocellular (Eph)/ephrin signalling is associated with the development of post-infectious irritable bowel syndrome (PI-IBS) is still unknown. In the present study, the role of Eph/Ephrin signalling in lipopolysaccharide (LPS)-induced intestinal injury was evaluated in vivo and in vitro. LPS treatment significantly increased the levels of proinflammatory mediators [monocyte chemoattractant protein-1, tumour necrosis factor α, interleukin (IL)-1β, IL-6, intercellular adhesion molecule 1 and vascular cell adhesion molecule-1], activated the EphA2-Ephrin A1, protein kinase B (Akt)-nuclear factor (NF)-κB, Src-NF-κB and Wnt/β-catenin signalling pathways, and inhibited EphB1-Ephrin B3 signalling in colon tissues, and primary cultured enteric neuronal and glial cells. Notably, EphA2 monoclonal antibody (mAb) treatment or Ephrin B3 overexpression could partially alleviate the LPS-induced upregulation of proinflammatory mediators, and Akt-NF-κB, Src-NF-κB and Wnt/β-catenin signalling pathways. In addition, EphA2 mAb treatment could partially inhibit LPS-induced inactivation of EphB-Ephrin B3 signalling, while Ephrin B3 overexpression could abrogate LPS-induced activation of EphA2-Ephrin A1 signalling. EphB1/Ephrin B3 signalling may antagonise the EphA2/Ephrin A1-dependent pathway following LPS treatment. The results associated with the EphA2 signaling pathway, indicated that Eph/ephrin signalling may serve a bidirectional role in LPS-induced intestinal injury. Eph/ephrin signalling may be a novel therapeutic target for LPS-induced intestinal injury and potentially PI-IBS.
Collapse
Affiliation(s)
- Ying Xiong
- Department of Gastroenterology, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong 518110, P.R. China
| | - Kai-Xue Li
- Department of Gastroenterology, The Second People's Hospital of Shenzhen, Shenzhen, Guangdong 518035, P.R. China
| | - Hong Wei
- Department of Gastroenterology, The Second People's Hospital of Shenzhen, Shenzhen, Guangdong 518035, P.R. China
| | - Lu Jiao
- Department of Gastroenterology, The Second People's Hospital of Shenzhen, Shenzhen, Guangdong 518035, P.R. China
| | - Shao-Yong Yu
- Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, MD 21205‑2195, USA
| | - Li Zeng
- Department of Gastroenterology, The Second People's Hospital of Shenzhen, Shenzhen, Guangdong 518035, P.R. China
| |
Collapse
|
38
|
Brzozowski JS, Bond DR, Jankowski H, Goldie BJ, Burchell R, Naudin C, Smith ND, Scarlett CJ, Larsen MR, Dun MD, Skelding KA, Weidenhofer J. Extracellular vesicles with altered tetraspanin CD9 and CD151 levels confer increased prostate cell motility and invasion. Sci Rep 2018; 8:8822. [PMID: 29891991 PMCID: PMC5995928 DOI: 10.1038/s41598-018-27180-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 05/25/2018] [Indexed: 02/06/2023] Open
Abstract
To facilitate intercellular communication, cells release nano-sized, extracellular vesicles (EVs) to transfer biological cargo to both local and distant sites. EVs are enriched in tetraspanins, two of which (CD9 and CD151) have altered expression patterns in many solid tumours, including prostate cancer, as they advance toward metastasis. We aimed to determine whether EVs from prostate cells with altered CD9 and CD151 expression could influence cellular behaviour and increase the metastatic capabilities of non-tumourigenic prostate cells. EVs were isolated by ultrafiltration and characterised for their tetraspanin expression and size distribution. iTRAQ was used to identify differences between RWPE1 and tetraspanin-modified RWPE1 EV proteomes, showing an enrichment in protein degradation pathways. Addition of EVs from RWPE1 cells with reduced CD9 or increased CD151 abundance resulted in increased invasion of RWPE1 cells, and increased migration in the case of high CD151 abundance. We have been able to show that alteration of CD9 and CD151 on prostate cells alters the proteome of their resultant EVs, and that these EVs can enhance the migratory and invasive capabilities of a non-tumourigenic prostate cellular population. This work suggests that cellular tetraspanin levels can alter EVs, potentially acting as a driver of metastasis in prostate cancer.
Collapse
Affiliation(s)
- Joshua S Brzozowski
- Cancer Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
| | - Danielle R Bond
- Cancer Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.,School of Environmental and Life Sciences, The University of Newcastle, Ourimbah, NSW, Australia
| | - Helen Jankowski
- Cancer Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
| | - Belinda J Goldie
- Cancer Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.,Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Rachel Burchell
- Cancer Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
| | - Crystal Naudin
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Emory University, Atlanta, Georgia, USA
| | - Nathan D Smith
- ABRF, Research Services, University of Newcastle, Callaghan, NSW, Australia
| | - Christopher J Scarlett
- Cancer Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.,School of Environmental and Life Sciences, The University of Newcastle, Ourimbah, NSW, Australia
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Matthew D Dun
- Cancer Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
| | - Kathryn A Skelding
- Cancer Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
| | - Judith Weidenhofer
- Cancer Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia. .,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.
| |
Collapse
|
39
|
Li X, Dun MD, Faulkner S, Hondermarck H. Neuroproteins in Cancer: Assumed Bystanders Become Culprits. Proteomics 2018; 18:e1800049. [PMID: 29745056 DOI: 10.1002/pmic.201800049] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Indexed: 12/30/2022]
Abstract
Recent breakthrough discoveries have highlighted the stimulatory role of nerves in cancer initiation and progression, through the release of neurotransmitters and growth factors by nerve terminals in the tumor microenvironment. Intriguingly, neuroproteins such as neuronal membrane proteins, synaptic proteins, neurotransmitters, and neurotrophic growth factors as well as their corresponding receptors, to name only a few, are frequently found in proteomic analyses of cancer tissues external to the brain and central nervous system. While the usual explanation was that neuroproteins were actually not specific to the nervous system and were therefore also expressed in cancer cells, it now appears that the presence of neuroproteins in cancer is largely due to the infiltration of nerves in the tumor microenvironment. Given the newly identified function of nerves as promoters of cancer growth and metastasis, neuroproteins should be considered with great attention because they may actually represent innovative biomarkers and therapeutic targets in oncology.
Collapse
Affiliation(s)
- Xiang Li
- School of Biomedical Sciences and Pharmacy & Hunter Medical Research Institute, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Matt D Dun
- School of Biomedical Sciences and Pharmacy & Hunter Medical Research Institute, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Sam Faulkner
- School of Biomedical Sciences and Pharmacy & Hunter Medical Research Institute, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Hubert Hondermarck
- School of Biomedical Sciences and Pharmacy & Hunter Medical Research Institute, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia
| |
Collapse
|
40
|
Almazi JG, Pockney P, Gedye C, Smith ND, Hondermarck H, Verrills NM, Dun MD. Cell-Free DNA Blood Collection Tubes Are Appropriate for Clinical Proteomics: A Demonstration in Colorectal Cancer. Proteomics Clin Appl 2018; 12:e1700121. [PMID: 29476593 PMCID: PMC5947838 DOI: 10.1002/prca.201700121] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 02/14/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Optimized blood collection tubes (BCT) have been developed to expand the utility of plasma cell-free DNA (cfDNA) and are in clinical use. The appropriateness of plasma collected and stored in these tubes for proteomic analysis is unknown. METHODS Paired blood samples were collected in BCT and traditional K3EDTA (EDTA) tubes from healthy controls and from colorectal cancer (CRC) patients before and after surgery, and stored for between 45 min and 48 h at room temperature. Plasma proteins were analyzed following high-abundant plasma protein depletion in quantitative discovery and targeted proteomics by liquid chromatography tandem-mass spectrometry (LC-MS/MS). RESULTS BCT reduced cellular protein contamination in healthy controls over time, and increased the number of high confident low-abundant protein identifications in CRC blood samples compared to matched samples collected in EDTA tubes. The known CRC plasma protein biomarker, carcinoembryonic antigen (CEA), showed elevated levels across patients pre-operatively when collected and stored in BCT compared to EDTA tubes. Emerging CRC biomarkers, Dickkopf-3 (DKK3) and Gelsolin (GSN), showed elevated levels pre-operatively when collected in BCT. CONCLUSIONS Optimized BCT are appropriate for low-abundant plasma protein analysis and can be used with confidence for clinical proteomics.
Collapse
Affiliation(s)
- Juhura G. Almazi
- Priority Research Centre for Cancer ResearchInnovation and TranslationHunter Medical Research Institute Cancer Research ProgramSchool of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
| | - Peter Pockney
- Priority Research Centre for Cancer ResearchInnovation and TranslationHunter Medical Research Institute Cancer Research ProgramSchool of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
- School of Medicine and Public HealthFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
| | - Craig Gedye
- Priority Research Centre for Cancer ResearchInnovation and TranslationHunter Medical Research Institute Cancer Research ProgramSchool of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
- School of Medicine and Public HealthFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
| | - Nathan D. Smith
- Analytical and Biomolecular Research Facility Advanced Mass Spectrometry UnitUniversity of NewcastleCallaghanNSWAustralia
| | - Hubert Hondermarck
- Priority Research Centre for Cancer ResearchInnovation and TranslationHunter Medical Research Institute Cancer Research ProgramSchool of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
| | - Nicole M. Verrills
- Priority Research Centre for Cancer ResearchInnovation and TranslationHunter Medical Research Institute Cancer Research ProgramSchool of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
| | - Matthew D. Dun
- Priority Research Centre for Cancer ResearchInnovation and TranslationHunter Medical Research Institute Cancer Research ProgramSchool of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
| |
Collapse
|
41
|
Narkhede AA, Crenshaw JH, Manning RM, Rao SS. The influence of matrix stiffness on the behavior of brain metastatic breast cancer cells in a biomimetic hyaluronic acid hydrogel platform. J Biomed Mater Res A 2018; 106:1832-1841. [DOI: 10.1002/jbm.a.36379] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 02/04/2018] [Accepted: 02/15/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Akshay A. Narkhede
- Department of Chemical and Biological EngineeringThe University of AlabamaTuscaloosa Alabama
| | - James H. Crenshaw
- Department of Chemical and Biological EngineeringThe University of AlabamaTuscaloosa Alabama
| | - Riley M. Manning
- Department of Chemical and Biological EngineeringThe University of AlabamaTuscaloosa Alabama
| | - Shreyas S. Rao
- Department of Chemical and Biological EngineeringThe University of AlabamaTuscaloosa Alabama
| |
Collapse
|
42
|
Jamaluddin MFB, Ko YA, Kumar M, Brown Y, Bajwa P, Nagendra PB, Skerrett-Byrne DA, Hondermarck H, Baker MA, Dun MD, Scott RJ, Nahar P, Tanwar PS. Proteomic Profiling of Human Uterine Fibroids Reveals Upregulation of the Extracellular Matrix Protein Periostin. Endocrinology 2018; 159:1106-1118. [PMID: 29244110 DOI: 10.1210/en.2017-03018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 12/04/2017] [Indexed: 12/11/2022]
Abstract
The central characteristic of uterine fibroids is excessive deposition of extracellular matrix (ECM), which contributes to fibroid growth and bulk-type symptoms. Despite this, very little is known about patterns of ECM protein expression in fibroids and whether these are influenced by the most common genetic anomalies, which relate to MED12. We performed extensive genetic and proteomic analyses of clinically annotated fibroids and adjacent normal myometrium to identify the composition and expression patterns of ECM proteins in MED12 mutation-positive and mutation-negative uterine fibroids. Genetic sequencing of tissue samples revealed MED12 alterations in 39 of 65 fibroids (60%) from 14 patients. Using isobaric tagged-based quantitative mass spectrometry on three selected patients (n = 9 fibroids), we observed a common set of upregulated (>1.5-fold) and downregulated (<0.66-fold) proteins in small, medium, and large fibroid samples of annotated MED12 status. These two sets of upregulated and downregulated proteins were the same in all patients, regardless of variations in fibroid size and MED12 status. We then focused on one of the significant upregulated ECM proteins and confirmed the differential expression of periostin using western blotting and immunohistochemical analysis. Our study defined the proteome of uterine fibroids and identified that increased ECM protein expression, in particular periostin, is a hallmark of uterine fibroids regardless of MED12 mutation status. This study sets the foundation for further investigations to analyze the mechanisms regulating ECM overexpression and the functional role of upregulated ECM proteins in leiomyogenesis.
Collapse
Affiliation(s)
- M Fairuz B Jamaluddin
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - Yi-An Ko
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - Manish Kumar
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - Yazmin Brown
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - Preety Bajwa
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - Prathima B Nagendra
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - David A Skerrett-Byrne
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - Hubert Hondermarck
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - Mark A Baker
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia
| | - Matt D Dun
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - Pravin Nahar
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Department of Maternity and Gynaecology, John Hunter Hospital, New Lambton Heights, New South Wales, Australia
| | - Pradeep S Tanwar
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
43
|
Martinez-Garcia E, Lesur A, Devis L, Campos A, Cabrera S, van Oostrum J, Matias-Guiu X, Gil-Moreno A, Reventos J, Colas E, Domon B. Development of a sequential workflow based on LC-PRM for the verification of endometrial cancer protein biomarkers in uterine aspirate samples. Oncotarget 2018; 7:53102-53115. [PMID: 27447978 PMCID: PMC5288171 DOI: 10.18632/oncotarget.10632] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 06/07/2016] [Indexed: 11/25/2022] Open
Abstract
About 30% of endometrial cancer (EC) patients are diagnosed at an advanced stage of the disease, which is associated with a drastic decrease in the 5-year survival rate. The identification of biomarkers in uterine aspirate samples, which are collected by a minimally invasive procedure, would improve early diagnosis of EC. We present a sequential workflow to select from a list of potential EC biomarkers, those which are the most promising to enter a validation study. After the elimination of confounding contributions by residual blood proteins, 52 potential biomarkers were analyzed in uterine aspirates from 20 EC patients and 18 non-EC controls by a high-resolution accurate mass spectrometer operated in parallel reaction monitoring mode. The differential abundance of 26 biomarkers was observed, and among them ten proteins showed a high sensitivity and specificity (AUC > 0.9). The study demonstrates that uterine aspirates are valuable samples for EC protein biomarkers screening. It also illustrates the importance of a biomarker verification phase to fill the gap between discovery and validation studies and highlights the benefits of high resolution mass spectrometry for this purpose. The proteins verified in this study have an increased likelihood to become a clinical assay after a subsequent validation phase.
Collapse
Affiliation(s)
- Elena Martinez-Garcia
- Biomedical Research Group in Gynecology, Vall Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Antoine Lesur
- Luxembourg Clinical Proteomics Center (LCP), Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | - Laura Devis
- Biomedical Research Group in Gynecology, Vall Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alexandre Campos
- Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| | - Silvia Cabrera
- Gynecological Oncology Department, Vall Hebron University Hospital, Barcelona, Spain
| | - Jan van Oostrum
- Luxembourg Clinical Proteomics Center (LCP), Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | - Xavier Matias-Guiu
- Pathological Oncology Group and Pathology Department, Hospital Arnau de Vilanova, Lleida, Spain
| | - Antonio Gil-Moreno
- Biomedical Research Group in Gynecology, Vall Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.,Gynecological Oncology Department, Vall Hebron University Hospital, Barcelona, Spain
| | - Jaume Reventos
- Biomedical Research Group in Gynecology, Vall Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.,Basic Sciences Department, International University of Catalonia, Barcelona, Spain
| | - Eva Colas
- Biomedical Research Group in Gynecology, Vall Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.,Pathological Oncology Group and Pathology Department, Hospital Arnau de Vilanova, Lleida, Spain
| | - Bruno Domon
- Luxembourg Clinical Proteomics Center (LCP), Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| |
Collapse
|
44
|
Husa AM, Magić Ž, Larsson M, Fornander T, Pérez-Tenorio G. EPH/ephrin profile and EPHB2 expression predicts patient survival in breast cancer. Oncotarget 2017; 7:21362-80. [PMID: 26870995 PMCID: PMC5008291 DOI: 10.18632/oncotarget.7246] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 01/23/2016] [Indexed: 11/25/2022] Open
Abstract
The EPH and ephrins function as both receptor and ligands and the output on their complex signaling is currently investigated in cancer. Previous work shows that some EPH family members have clinical value in breast cancer, suggesting that this family could be a source of novel clinical targets. Here we quantified the mRNA expression levels of EPH receptors and their ligands, ephrins, in 65 node positive breast cancer samples by RT-PCR with TaqMan® Micro Fluidics Cards Microarray. Upon hierarchical clustering of the mRNA expression levels, we identified a subgroup of patients with high expression, and poor clinical outcome. EPHA2, EPHA4, EFNB1, EFNB2, EPHB2 and EPHB6 were significantly correlated with the cluster groups and particularly EPHB2 was an independent prognostic factor in multivariate analysis and in four public databases. The EPHB2 protein expression was also analyzed by immunohistochemistry in paraffin embedded material (cohort 2). EPHB2 was detected in the membrane and cytoplasmic cell compartments and there was an inverse correlation between membranous and cytoplasmic EPHB2. Membranous EPHB2 predicted longer breast cancer survival in both univariate and multivariate analysis while cytoplasmic EPHB2 indicated shorter breast cancer survival in univariate analysis. Concluding: the EPH/EFN cluster analysis revealed that high EPH/EFN mRNA expression is an independent prognostic factor for poor survival. Especially EPHB2 predicted poor breast cancer survival in several materials and EPHB2 protein expression has also prognostic value depending on cell localization.
Collapse
Affiliation(s)
- Anna-Maria Husa
- Department of Clinical and Experimental Medicine, Division of Oncology, Linköping University, Linköping, Sweden.,Current address: CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| | - Željana Magić
- Department of Clinical and Experimental Medicine, Division of Oncology, Linköping University, Linköping, Sweden
| | - Malin Larsson
- Bioinformatics Infrastructure for Life Sciences (BILS) and Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Tommy Fornander
- Department of Oncology, Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden
| | - Gizeh Pérez-Tenorio
- Department of Clinical and Experimental Medicine, Division of Oncology, Linköping University, Linköping, Sweden
| |
Collapse
|
45
|
An J, Wang L, Zhao Y, Hao Q, Zhang Y, Zhang J, Yang C, Liu L, Wang W, Fang D, Lu T, Gao Y. Effects of FSTL1 on cell proliferation in breast cancer cell line MDA‑MB‑231 and its brain metastatic variant MDA‑MB‑231‑BR. Oncol Rep 2017; 38:3001-3010. [PMID: 29048681 PMCID: PMC5780039 DOI: 10.3892/or.2017.6004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 09/04/2017] [Indexed: 12/23/2022] Open
Abstract
In the past decades, altered Follistatin-like 1 (FSTL1) expression has been documented in a variety of cancers, while its functional roles are poorly understood. Particularly in breast cancer, the expression of FSTL1 and its signaling pathway remain to be determined. In the present study, an elevated FSTL1 expression and a supressed cell proliferation were detected in a specific brain metastatic cell line MDA-MB-231-BR (231-BR), compared with its parental cell line MDA-MB-231. However, this protein was hardly detected in the other three breast cancer cell lines. Next, lentiviral vectors encoding FSTL1 or FSTL1 specific shRNAs were used to overexpress or knock down FSTL1 in MDA-MB-231 or 231-BR, respectively (MDA-MB-231FSTL1 or 231-BRsh FSTL1). Results showed that overexpression of FSTL1 inhibited MDA-MB-231 cell proliferation, while knockdown of FSTL1 in 231-BR cells promotes cell proliferation, compared with their corresponding control groups. These results were further confirmed in nude mouse xenografts. The tumor volume in 231-BR cell-bearing mice was significantly smaller than that of MDA-MB-231 group, and reduction of tumor volume was detected in MDA-MB-231FSTL1 cell-bearing mice compared with the control group. Previous studies revealed that TGF-β-Smad2/3 signaling pathway was activated in 231-BR and MDA-MB-231FSTL1 cells, which may contribute to the inhibited cell proliferation. In addition, Smad3 knockdown could restore the inhibition of cell proliferation induced by FSTL1 overexpression in MDA-MB-231FSTL1 cells, indicating that the anti-proliferative effect of FSTL1 overexpression may be associated with Smad3 involved TGF-β signaling pathway regulation. This study identified FSTL1 as an inhibitor of cell proliferation in MDA-MB-231 and 231-BR cell lines, which may provide new insights into the development and management of breast cancer.
Collapse
Affiliation(s)
- Jiaqiang An
- Department of Human Anatomy, Capital Medical University, Beijing 100069, P.R. China
| | - Lulu Wang
- Department of Human Anatomy, Capital Medical University, Beijing 100069, P.R. China
| | - Yuanli Zhao
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Qiang Hao
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Ying Zhang
- Department of Human Anatomy, Capital Medical University, Beijing 100069, P.R. China
| | - Jingyi Zhang
- Department of Human Anatomy, Capital Medical University, Beijing 100069, P.R. China
| | - Chun Yang
- Department of Human Anatomy, Capital Medical University, Beijing 100069, P.R. China
| | - Li Liu
- Department of Human Anatomy, Capital Medical University, Beijing 100069, P.R. China
| | - Wenjuan Wang
- Department of Human Anatomy, Capital Medical University, Beijing 100069, P.R. China
| | - Dongliang Fang
- Department of Human Anatomy, Capital Medical University, Beijing 100069, P.R. China
| | - Tao Lu
- Department of Human Anatomy, Capital Medical University, Beijing 100069, P.R. China
| | - Yan Gao
- Department of Human Anatomy, Capital Medical University, Beijing 100069, P.R. China
| |
Collapse
|
46
|
Hutcheon K, McLaughlin EA, Stanger SJ, Bernstein IR, Dun MD, Eamens AL, Nixon B. Analysis of the small non-protein-coding RNA profile of mouse spermatozoa reveals specific enrichment of piRNAs within mature spermatozoa. RNA Biol 2017; 14:1776-1790. [PMID: 28816603 DOI: 10.1080/15476286.2017.1356569] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Post-testicular sperm maturation and storage within the epididymis is a key determinant of gamete quality and fertilization competence. Here we demonstrate that mouse spermatozoa possess a complex small non-protein-coding RNA (sRNA) profile, the composition of which is markedly influenced by their epididymal transit. Thus, although microRNAs (miRNAs) are highly represented in the spermatozoa of the proximal epididymis, this sRNA class is largely diminished in mature spermatozoa of the distal epididymis. Coincident with this, a substantial enrichment in Piwi-interacting RNA (piRNA) abundance in cauda spermatozoa was detected. Further, features of cauda piRNAs, including; predominantly 29-31 nts in length; preference for uracil at their 5' terminus; no adenine enrichment at piRNA nt 10, and; predominantly mapping to intergenic regions of the mouse genome, indicate that these piRNAs are generated by the PIWIL1-directed primary piRNA production pathway. Accordingly, PIWIL1 was detected via immunoblotting and mass spectrometry in epididymal spermatozoa. These data provide insight into the complexity and dynamic nature of the sRNA profile of spermatozoa and raise the intriguing prospect that piRNAs are generated in situ in maturing spermatozoa. Such information is of particular interest in view of the potential role for paternal sRNAs in influencing conception, embryo development and intergenerational inheritance.
Collapse
Affiliation(s)
- Kate Hutcheon
- a School of Environmental and Life Sciences , The University of Newcastle , Callaghan , NSW , Australia
| | - Eileen A McLaughlin
- a School of Environmental and Life Sciences , The University of Newcastle , Callaghan , NSW , Australia.,b Priority Research Centre for Reproductive Biology , The University of Newcastle , Callaghan , NSW , Australia.,c School of Biological Sciences , University of Auckland , Auckland , New Zealand
| | - Simone J Stanger
- a School of Environmental and Life Sciences , The University of Newcastle , Callaghan , NSW , Australia.,b Priority Research Centre for Reproductive Biology , The University of Newcastle , Callaghan , NSW , Australia
| | - Ilana R Bernstein
- a School of Environmental and Life Sciences , The University of Newcastle , Callaghan , NSW , Australia.,b Priority Research Centre for Reproductive Biology , The University of Newcastle , Callaghan , NSW , Australia
| | - Matthew D Dun
- d Priority Research Centre for Cancer Research, Innovation and Translation , Hunter Medical Research Institute, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle , Callaghan , NSW , Australia
| | - Andrew L Eamens
- a School of Environmental and Life Sciences , The University of Newcastle , Callaghan , NSW , Australia
| | - Brett Nixon
- a School of Environmental and Life Sciences , The University of Newcastle , Callaghan , NSW , Australia.,b Priority Research Centre for Reproductive Biology , The University of Newcastle , Callaghan , NSW , Australia
| |
Collapse
|
47
|
Degryse S, de Bock CE, Demeyer S, Govaerts I, Bornschein S, Verbeke D, Jacobs K, Binos S, Skerrett-Byrne DA, Murray HC, Verrills NM, Van Vlierberghe P, Cools J, Dun MD. Mutant JAK3 phosphoproteomic profiling predicts synergism between JAK3 inhibitors and MEK/BCL2 inhibitors for the treatment of T-cell acute lymphoblastic leukemia. Leukemia 2017; 32:788-800. [PMID: 28852199 PMCID: PMC5843905 DOI: 10.1038/leu.2017.276] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/17/2017] [Accepted: 08/15/2017] [Indexed: 02/06/2023]
Abstract
Mutations in the interleukin-7 receptor (IL7R) or the Janus kinase 3 (JAK3) kinase occur frequently in T-cell acute lymphoblastic leukemia (T-ALL) and both are able to drive cellular transformation and the development of T-ALL in mouse models. However, the signal transduction pathways downstream of JAK3 mutations remain poorly characterized. Here we describe the phosphoproteome downstream of the JAK3(L857Q)/(M511I) activating mutations in transformed Ba/F3 lymphocyte cells. Signaling pathways regulated by JAK3 mutants were assessed following acute inhibition of JAK1/JAK3 using the JAK kinase inhibitors ruxolitinib or tofacitinib. Comprehensive network interrogation using the phosphoproteomic signatures identified significant changes in pathways regulating cell cycle, translation initiation, mitogen-activated protein kinase and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/AKT signaling, RNA metabolism, as well as epigenetic and apoptotic processes. Key regulatory proteins within pathways that showed altered phosphorylation following JAK inhibition were targeted using selumetinib and trametinib (MEK), buparlisib (PI3K) and ABT-199 (BCL2), and found to be synergistic in combination with JAK kinase inhibitors in primary T-ALL samples harboring JAK3 mutations. These data provide the first detailed molecular characterization of the downstream signaling pathways regulated by JAK3 mutations and provide further understanding into the oncogenic processes regulated by constitutive kinase activation aiding in the development of improved combinatorial treatment regimens.
Collapse
Affiliation(s)
- S Degryse
- VIB Center for Cancer Biology, Leuven, Belgium.,KU Leuven Center for Human Genetics, Leuven, Belgium
| | - C E de Bock
- VIB Center for Cancer Biology, Leuven, Belgium.,KU Leuven Center for Human Genetics, Leuven, Belgium
| | - S Demeyer
- VIB Center for Cancer Biology, Leuven, Belgium.,KU Leuven Center for Human Genetics, Leuven, Belgium
| | - I Govaerts
- VIB Center for Cancer Biology, Leuven, Belgium.,KU Leuven Center for Human Genetics, Leuven, Belgium
| | - S Bornschein
- VIB Center for Cancer Biology, Leuven, Belgium.,KU Leuven Center for Human Genetics, Leuven, Belgium
| | - D Verbeke
- VIB Center for Cancer Biology, Leuven, Belgium.,KU Leuven Center for Human Genetics, Leuven, Belgium
| | - K Jacobs
- VIB Center for Cancer Biology, Leuven, Belgium.,KU Leuven Center for Human Genetics, Leuven, Belgium
| | - S Binos
- Thermo Fisher Scientific, Scoresby, Victoria, Australia
| | - D A Skerrett-Byrne
- Faculty of Health and Medicine, University of Newcastle, Callaghan, New South Wales, Australia.,Cancer Research Program, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - H C Murray
- Faculty of Health and Medicine, University of Newcastle, Callaghan, New South Wales, Australia.,Cancer Research Program, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - N M Verrills
- Faculty of Health and Medicine, University of Newcastle, Callaghan, New South Wales, Australia.,Cancer Research Program, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - P Van Vlierberghe
- Department of Pediatrics and Genetics, Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - J Cools
- VIB Center for Cancer Biology, Leuven, Belgium.,KU Leuven Center for Human Genetics, Leuven, Belgium
| | - M D Dun
- Faculty of Health and Medicine, University of Newcastle, Callaghan, New South Wales, Australia.,Cancer Research Program, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
48
|
Murray HC, Dun MD, Verrills NM. Harnessing the power of proteomics for identification of oncogenic, druggable signalling pathways in cancer. Expert Opin Drug Discov 2017; 12:431-447. [PMID: 28286965 DOI: 10.1080/17460441.2017.1304377] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Genomic and transcriptomic profiling of tumours has revolutionised our understanding of cancer. However, the majority of tumours possess multiple mutations, and determining which oncogene, or even which pathway, to target is difficult. Proteomics is emerging as a powerful approach to identify the functionally important pathways driving these cancers, and how they can be targeted therapeutically. Areas covered: The authors provide a technical overview of mass spectrometry based approaches for proteomic profiling, and review the current and emerging strategies available for the identification of dysregulated networks, pathways, and drug targets in cancer cells, with a key focus on the ability to profile cancer kinomes. The potential applications of mass spectrometry in the clinic are also highlighted. Expert opinion: The addition of proteomic information to genomic platforms - 'proteogenomics' - is providing unparalleled insight in cancer cell biology. Application of improved mass spectrometry technology and methodology, in particular the ability to analyse post-translational modifications (the PTMome), is providing a more complete picture of the dysregulated networks in cancer, and uncovering novel therapeutic targets. While the application of proteomics to discovery research will continue to rise, improved workflow standardisation and reproducibility is required before mass spectrometry can enter routine clinical use.
Collapse
Affiliation(s)
- Heather C Murray
- a School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Priority Research Centre for Cancer Research, Innovation and Translation , University of Newcastle , Callaghan , NSW , Australia.,b Cancer Research Program , Hunter Medical Research Institute , Newcastle , NSW , Australia
| | - Matthew D Dun
- a School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Priority Research Centre for Cancer Research, Innovation and Translation , University of Newcastle , Callaghan , NSW , Australia.,b Cancer Research Program , Hunter Medical Research Institute , Newcastle , NSW , Australia
| | - Nicole M Verrills
- a School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Priority Research Centre for Cancer Research, Innovation and Translation , University of Newcastle , Callaghan , NSW , Australia.,b Cancer Research Program , Hunter Medical Research Institute , Newcastle , NSW , Australia
| |
Collapse
|
49
|
Faria SS, Morris CFM, Silva AR, Fonseca MP, Forget P, Castro MS, Fontes W. A Timely Shift from Shotgun to Targeted Proteomics and How It Can Be Groundbreaking for Cancer Research. Front Oncol 2017; 7:13. [PMID: 28265552 PMCID: PMC5316539 DOI: 10.3389/fonc.2017.00013] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 01/17/2017] [Indexed: 01/10/2023] Open
Abstract
The fact that cancer is a leading cause of death all around the world has naturally sparked major efforts in the pursuit of novel and more efficient biomarkers that could better serve as diagnostic tools, prognostic predictors, or therapeutical targets in the battle against this type of disease. Mass spectrometry-based proteomics has proven itself as a robust and logical alternative to the immuno-based methods that once dominated the field. Nevertheless, intrinsic limitations of classic proteomic approaches such as the natural gap between shotgun discovery-based methods and clinically applicable results have called for the implementation of more direct, hypothesis-based studies such as those made available through targeted approaches, that might be able to streamline biomarker discovery and validation as a means to increase survivability of affected patients. In fact, the paradigm shifting potential of modern targeted proteomics applied to cancer research can be demonstrated by the large number of advancements and increasing examples of new and more useful biomarkers found during the course of this review in different aspects of cancer research. Out of the many studies dedicated to cancer biomarker discovery, we were able to devise some clear trends, such as the fact that breast cancer is the most common type of tumor studied and that most of the research for any given type of cancer is focused on the discovery diagnostic biomarkers, with the exception of those that rely on samples other than plasma and serum, which are generally aimed toward prognostic markers. Interestingly, the most common type of targeted approach is based on stable isotope dilution-selected reaction monitoring protocols for quantification of the target molecules. Overall, this reinforces that notion that targeted proteomics has already started to fulfill its role as a groundbreaking strategy that may enable researchers to catapult the number of viable, effective, and validated biomarkers in cancer clinical practice.
Collapse
Affiliation(s)
- Sara S Faria
- Mastology Program, Federal University of Uberlandia (UFU) , Uberlandia , Brazil
| | - Carlos F M Morris
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia , Brasília , Brazil
| | - Adriano R Silva
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia , Brasília , Brazil
| | - Micaella P Fonseca
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Patrice Forget
- Department of Anesthesiology and Perioperative Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit of Brussel , Brussels , Belgium
| | - Mariana S Castro
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia , Brasília , Brazil
| | - Wagner Fontes
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia , Brasília , Brazil
| |
Collapse
|
50
|
Worthington J, Spain G, Timms JF. Effects of ErbB2 Overexpression on the Proteome and ErbB Ligand-specific Phosphosignaling in Mammary Luminal Epithelial Cells. Mol Cell Proteomics 2017; 16:608-621. [PMID: 28174229 PMCID: PMC5383782 DOI: 10.1074/mcp.m116.061267] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 01/30/2017] [Indexed: 12/12/2022] Open
Abstract
Most breast cancers arise from luminal epithelial cells, and 25–30% of these tumors overexpress the ErbB2/HER2 receptor that correlates with disease progression and poor prognosis. The mechanisms of ErbB2 signaling and the effects of its overexpression are not fully understood. Herein, stable isotope labeling by amino acids in cell culture (SILAC), expression profiling, and phosphopeptide enrichment of a relevant, non-transformed, and immortalized human mammary luminal epithelial cell model were used to profile ErbB2-dependent differences in protein expression and phosphorylation events triggered via EGF receptor (EGF treatment) and ErbB3 (HRG1β treatment) in the context of ErbB2 overexpression. Bioinformatics analysis was used to infer changes in cellular processes and signaling events. We demonstrate the complexity of the responses to oncogene expression and growth factor signaling, and we identify protein changes relevant to ErbB2-dependent altered cellular phenotype, in particular cell cycle progression and hyper-proliferation, reduced adhesion, and enhanced motility. Moreover, we define a novel mechanism by which ErbB signaling suppresses basal interferon signaling that would promote the survival and proliferation of mammary luminal epithelial cells. Numerous novel sites of growth factor-regulated phosphorylation were identified that were enhanced by ErbB2 overexpression, and we putatively link these to altered cell behavior and also highlight the importance of performing parallel protein expression profiling alongside phosphoproteomic analysis.
Collapse
Affiliation(s)
- Jenny Worthington
- From the ‡Women's Cancer, Institute for Women's Health, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Georgia Spain
- From the ‡Women's Cancer, Institute for Women's Health, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - John F Timms
- From the ‡Women's Cancer, Institute for Women's Health, University College London, Gower Street, London WC1E 6BT, United Kingdom
| |
Collapse
|