1
|
Wiśniewska K, Żabińska M, Gaffke L, Szulc A, Walter BM, Węgrzyn G, Pierzynowska K. Shared Gene Expression Dysregulation Across Subtypes of Sanfilippo and Morquio Diseases: The Role of PFN1 in Regulating Glycosaminoglycan Levels. FRONT BIOSCI-LANDMRK 2024; 29:415. [PMID: 39735993 DOI: 10.31083/j.fbl2912415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/27/2024] [Accepted: 10/31/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Mucopolysaccharidosis (MPS) is a class of hereditary metabolic diseases that demonstrate itself by accumulating incompletely degraded glycosaminoglycans (GAGs). MPS are classified according to the kind(s) of stored GAG(s) and specific genetic/enzymatic defects. Despite the accumulation of the same type of GAG, two MPS diseases, Sanfilippo (MPS III) and Morquio (MPS IV), are further distinguished into subclasses based on different enzymes that are deficient. Although genetic defects in MPS are known, molecular mechanisms of particular MPS types are still incomplete. This work aimed to investigate gene expression patterns in MPS III and MPS IV subtypes to identify dysregulated genes that could indicate unidentified molecular mechanisms of the diseases. METHODS Transcriptomic analyses were conducted to assess gene expression patterns in MPS and control cells. Western blotting and immunohistochemistry determined selected protein levels (products of the most significantly dysregulated genes). Effects of decreased levels of gene expression were investigated using small interferring RNA (siRNA)-mediated gene silencing. RESULTS Transcriptomic analyses indicated 45 commonly dysregulated genes among all MPS III subtypes and as many as 150 commonly dysregulated genes among both MPS IV subtypes. A few genes revealed particularly high levels of dysregulation, including PFN1, MFAP5, and MMP12. Intriguingly, elevated levels of profilin-1 (product of the PFN1 gene) could be reduced by decreasing GAG levels in genistein-treated MPS III and MPS IV cells, while silencing of PFN1 caused a significant decrease in GAG accumulation in these cells, indicating an interdependent correlation between profilin-1 and GAG levels. CONCLUSIONS A plethora of commonly dysregulated genes were identified in MPS subtypes III and IV. Some of these genes, like PFN1, MFAP5, and MMP12, revealed highly pronounced changes in expression relative to control cells. An interdependent correlation between GAG levels and the expression of the PFN1 gene was identified. Thus, PFN1 could be suggested as a potential new therapeutic target for MPS III and IV.
Collapse
Affiliation(s)
- Karolina Wiśniewska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland
| | - Magdalena Żabińska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland
| | - Lidia Gaffke
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland
| | - Aneta Szulc
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland
| | - Beata M Walter
- Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, 80-307 Gdansk, Poland
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland
| | - Karolina Pierzynowska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland
| |
Collapse
|
2
|
Ali F, Iqbal A, Azhar I, Qayyum A, Hassan SA, Hasan MSA, Jawi M, Hassan HM, Al-Emam A, Sajid M. Exploring a novel four-gene system as a diagnostic and prognostic biomarker for triple-negative breast cancer, using clinical variables. Comput Biol Chem 2024; 113:108247. [PMID: 39427606 DOI: 10.1016/j.compbiolchem.2024.108247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/25/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024]
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer with a poor prognosis. This research aims to find real hub genes for prognostic biomarkers of TNBC therapy. The GEO datasets GSE27447 and GSE233242 were analyzed using R package limma to explore DEGs. The PPI was generated using the STRING database. Cytoscape software plug-ins were used to screen the hub genes. Using the DAVID database, GO functional enrichment and KEGG pathway enrichment analysis were performed. Different online expression databases were employed to investigate the functions of real hub genes in tumor driving, diagnosis, and prognosis in TNBC patients with various clinicopathologic characteristics. A total of one hundred DEGs were identified between both datasets. The seven hub genes were identified after the topological parameter analysis of the PPI network. The KEGG pathway and GO analysis suggest that four genes (PSMB1, PSMC1, PSMF1, and PSMD8) are highly enriched in proteasome and were finally considered as real hub genes. Additionally, the expression analysis demonstrated that hub genes were notably up-regulated in TNBC patients compared to controls. Furthermore, correlational analyses revealed the positive and negative correlations among the expression of the real hub genes and various ancillary data, including tumor purity, promoter methylation status, overall survival (OS), genetic alterations, infiltration of CD8+ T and CD4+ immune cells, and a few more, across TNBC samples. Finally, our analysis identified a couple of significant chemotherapeutic drugs, miRNAs and transcription factors (TFS) with intriguing curative potential. In conclusion, we identified four real hub genes as novel biomarkers to overcome heterogenetic-particular challenges in diagnosis, prognosis, and therapy for TNBC patients.
Collapse
Affiliation(s)
- Faisal Ali
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan
| | - Azhar Iqbal
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan
| | - Iqra Azhar
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan
| | - Adiba Qayyum
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan
| | - Syed Ali Hassan
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan
| | - Md Sakib Al Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science And Technology University, Gopalgonj, Dhaka 8100, Bangladesh; Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj, 8100, Dhaka, Bangladesh.
| | - Motasim Jawi
- Department of Basic Medical Sciences, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Hesham M Hassan
- Department of Pathology, College of Medicine, King Khalid University, Asir 61421, Saudi Arabia
| | - Ahmed Al-Emam
- Department of Pathology, College of Medicine, King Khalid University, Asir 61421, Saudi Arabia.
| | - Muhammad Sajid
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan.
| |
Collapse
|
3
|
Swaminathan S, Karvembu R. Dichloro Ru(II)- p-cymene-1,3,5-triaza-7-phosphaadamantane (RAPTA-C): A Case Study. ACS Pharmacol Transl Sci 2023; 6:982-996. [PMID: 37470017 PMCID: PMC10353064 DOI: 10.1021/acsptsci.3c00085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Indexed: 07/21/2023]
Abstract
The use of organometallic compounds to treat various phenotypes of cancer has attracted increased interest in recent decades. Organometallic compounds, which are transitional between conventional inorganic and organic materials, have outstanding and one-of-a-kind features that offer fresh insight into the development of inorganic medicinal chemistry. The therapeutic potential of ruthenium(II)-arene RAPTA-type compounds is being thoroughly investigated, specifically owing to the excellent antimetastatic property of the initial candidate RAPTA-C. This review gives a thorough analysis of this complex and its evolution as a potential anticancer drug candidate. The numerous mechanistic investigations of RAPTA-C are discussed, and they are connected to the macroscopic biological characteristics that have been found. The "multitargeted" complex described here target enzymes, peptides, and intracellular proteins in addition to DNA that allow it to specifically target cancer cells. Understanding these may allow researchers to find specific targets and tune a new-generation organometallic complex accordingly.
Collapse
Affiliation(s)
- Srividya Swaminathan
- Department
of Chemistry, National Institute of Technology, Tiruchirappalli 620015, Tamil Nadu, India
- Center
for Computational Modeling, Chennai Institute
of Technology (CIT), Chennai 600069, India
| | - Ramasamy Karvembu
- Department
of Chemistry, National Institute of Technology, Tiruchirappalli 620015, Tamil Nadu, India
| |
Collapse
|
4
|
Swaminathan S, Haribabu J, Dharmasivam M, Maroli N, Jayadharini JP, Balakrishnan N, Bhuvanesh N, Echeverria C, Karvembu R. Hinged Bipodal Furoylthiourea-Based Ru(II)-Arene Complexes: Effect of ( ortho, meta, or para)-Substitution on Coordination and Anticancer Activity. Inorg Chem 2023; 62:3679-3691. [PMID: 36780329 DOI: 10.1021/acs.inorgchem.3c00073] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
We set out to design and synthesize bipodal ligands with the phenyl group as the spacer and varied the substitution on the spacer between ortho (L1), meta (L2), and para (L3). The respective ligands and complexes containing either p-cymene (PL1-PL3) or benzene (BL1-BL3) as the arene unit were synthesized and characterized successfully. The influence of the ligands due to substitution change on their coordination behavior was quite minimal; however, the differences were seen in the anticancer activity of the complexes. DFT studies revealed the structural variations between the three different substitutions, which was further confirmed by single-crystal X-ray diffraction studies. The anticancer activity of the complexes could be correlated with their rate of hydrolysis and their lipophilicity index as determined by UV-visible spectroscopy. The cell death mechanism of the active complexes was deduced to be apoptotic via staining assays, flow cytometry, and Western blot analysis.
Collapse
Affiliation(s)
- Srividya Swaminathan
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, Tamil Nadu, India
| | - Jebiti Haribabu
- Facultad de Medicina, Universidad de Atacama, Los Carreras 1579, 1532502 Copiapo, Chile
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia
| | - Nikhil Maroli
- Centre for Condensed Matter Theory, Department of Physics, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | | | - Nithya Balakrishnan
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, Tamil Nadu, India
| | - Nattamai Bhuvanesh
- Department of Chemistry, Texas A & M University, College Station, Texas 77842, United States
| | - Cesar Echeverria
- Facultad de Medicina, Universidad de Atacama, Los Carreras 1579, 1532502 Copiapo, Chile
| | - Ramasamy Karvembu
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, Tamil Nadu, India
| |
Collapse
|
5
|
Poljak A, Raftery M, Polly P. Evaluating Cellular Viability by iTRAQ Proteomic Profiling. Methods Mol Biol 2023; 2644:193-209. [PMID: 37142923 DOI: 10.1007/978-1-0716-3052-5_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Cellular health, functionality, response to environment, and other variables affecting cell, tissue, or organ viability are reflected in the cellular proteomes and metabolomes. These "omic" profiles are in constant flux even during normal cellular functioning, to maintain cellular homeostasis, in response to small environmental changes and maintenance of optimal cell viability. However proteomic "fingerprints" can also provide insight into cellular ageing, response to disease, adjustment to environmental changes, and other variables that impact cellular viability. A variety of proteomic methods can be used to determine qualitative and quantitative proteomic change. In this chapter, we will focus on a labeling method called isobaric tags for relative and absolute quantification (iTRAQ), which is frequently used to identify and quantify proteomic expression changes in cells and tissues.
Collapse
Affiliation(s)
- Anne Poljak
- Bioanalytical Mass Spectrometry Facility (BMSF), Mark Wainwright Analytical Centre (MWAC), University of New South Wales, Sydney, NSW, Australia
| | - Mark Raftery
- Bioanalytical Mass Spectrometry Facility (BMSF), Mark Wainwright Analytical Centre (MWAC), University of New South Wales, Sydney, NSW, Australia
| | - Patsie Polly
- Department of Pathology, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
6
|
Jayathirtha M, Whitham D, Alwine S, Donnelly M, Neagu AN, Darie CC. Investigating the Function of Human Jumping Translocation Breakpoint Protein (hJTB) and Its Interacting Partners through In-Solution Proteomics of MCF7 Cells. Molecules 2022; 27:8301. [PMID: 36500393 PMCID: PMC9740069 DOI: 10.3390/molecules27238301] [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: 09/20/2022] [Revised: 11/16/2022] [Accepted: 11/16/2022] [Indexed: 11/30/2022] Open
Abstract
Human jumping translocation breakpoint (hJTB) gene is located on chromosome 1q21 and is involved in unbalanced translocation in many types of cancer. JTB protein is ubiquitously present in normal cells but it is found to be overexpressed or downregulated in various types of cancer cells, where this protein and its isoforms promote mitochondrial dysfunction, resistance to apoptosis, genomic instability, proliferation, invasion and metastasis. Hence, JTB could be a tumor biomarker for different types of cancer, such as breast cancer (BC), and could be used as a drug target for therapy. However, the functions of the protein or the pathways through which it increases cell proliferation and invasiveness of cancer cells are not well-known. Therefore, we aim to investigate the functions of JTB by using in-solution digestion-based cellular proteomics of control and upregulated and downregulated JTB protein in MCF7 breast cancer cell line, taking account that in-solution digestion-based proteomics experiments are complementary to the initial in-gel based ones. Proteomics analysis allows investigation of protein dysregulation patterns that indicate the function of the protein and its interacting partners, as well as the pathways and biological processes through which it functions. We concluded that JTB dysregulation increases the epithelial-mesenchymal transition (EMT) potential and cell proliferation, harnessing cytoskeleton organization, apical junctional complex, metabolic reprogramming, and cellular proteostasis. Deregulated JTB expression was found to be associated with several proteins involved in mitochondrial organization and function, oxidative stress (OS), apoptosis, and interferon alpha and gamma signaling. Consistent and complementary to our previous results emerged by using in-gel based proteomics of transfected MCF7 cells, JTB-related proteins that are overexpressed in this experiment suggest the development of a more aggressive phenotype and behavior for this luminal type A non-invasive/poor-invasive human BC cell line that does not usually migrate or invade compared with the highly metastatic MDA-MB-231 cells. This more aggressive phenotype of MCF7 cells related to JTB dysregulation and detected by both in-gel and in-solution proteomics could be promoted by synergistic upregulation of EMT, Mitotic spindle and Fatty acid metabolism pathways. However, in both JTB dysregulated conditions, several downregulated JTB-interacting proteins predominantly sustain antitumor activities, attenuating some of the aggressive phenotypical and behavioral traits promoted by the overexpressed JTB-related partners.
Collapse
Affiliation(s)
- Madhuri Jayathirtha
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Danielle Whitham
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Shelby Alwine
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Mary Donnelly
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “AlexandruIoanCuza” University of Iasi, Carol I bvd. No. 20A, 700505 Iasi, Romania
| | - Costel C. Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| |
Collapse
|
7
|
Peng J, Pei S, Cui Y, Xia Y, Huang Y, Wu X, Zheng M, Weng M, Han X, Fu H, Yang L, Zhou W, Fu Z, Wang S, Xie H. Comparative analysis of transient receptor potential channel 5 opposite strand-induced gene expression patterns and protein-protein interactions in triple-negative breast cancer. Oncol Lett 2022; 24:259. [PMID: 35765270 PMCID: PMC9219028 DOI: 10.3892/ol.2022.13379] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/04/2022] [Indexed: 11/06/2022] Open
Abstract
In patients with triple-negative breast cancer (TNBC), high tumour mutation burden and aberrant oncogene expression profiles are some of the causes of poor prognosis. Therefore, it is necessary to identify aberrantly expressed oncogenes, since they have the potential to serve as therapeutic targets. Transient receptor potential channel 5 opposite strand (TRPC5OS) has been previously shown to function as a novel tumour inducer. However, the underlying mechanism of TRPC5OS function in TNBC remain to be elucidated. Therefore, in the present study TRPC5OS expression was first measured in tissue samples of patients with TNBC and a panel of breast cancer cell lines (ZR-75-1, MDA-MB-453, SK-BR-3, JIMT-1, BT474 and HCC1937) by using qRT-PCR and Western blotting. Subsequently, the possible effects of TRPC5OS on MDA-MB-231 cells proliferation were determined using Cell Counting Kit-8 and 5-Ethynyl-2′-deoxyuridine assays after Lentiviral transfection of MDA-MB-231. In addition, potential interaction partners of TRPC5OS were explored using liquid chromatography-mass spectrometry (LC-MS)/MS. Gene expression patterns following TRPC5OS overexpression were also detected in MDA-MB-231 cells by using High-throughput sequencing. Gene Ontology (GO) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) analysis were then used to systematically verify the potential interactions among the TRPC5OS-regulated genes. The potential relationship between TRPC5OS-interacting proteins and gene expression patterns were studied using Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) analysis. TRPC5OS expression was found to be significantly higher in TNBC tumour tissues and breast cancer cell lines compared with luminal tumour tissues and ZR-75-1. In addition, the overexpression of TRPC5OS significantly increased cell proliferation. High-throughput sequencing results revealed that 5,256 genes exhibited differential expression following TRPC5OS overexpression, including 3,269 upregulated genes and 1,987 downregulated genes. GO analysis results indicated that the functions of these differentially expressed genes were enriched in the categories of ‘cell division’ and ‘cell proliferation’ regulation. KEGG analysis showed that the TRPC5OS-regulated genes were associated with processes of ‘homologous recombination’ and ‘TNF signalling pathways’. Subsequently, 17 TRPC5OS-interacting proteins were found using LC-MS/MS and STRING analysis. The most important protein among interacting proteins was ENO1 which was associated with glycolysis and regulated proliferation of cancer. In summary, data from the present study suggest that TRPC5OS overexpression can increase TNBC cell proliferation and ENO1 may be a potential target protein mediated by TRPC5OS. Therefore, TRPC5OS may serve as a novel therapeutic target for TNBC.
Collapse
Affiliation(s)
- Jinghui Peng
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China.,Breast Disease Laboratory, Women and Children Central Laboratory, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Shengbin Pei
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China.,Breast Disease Laboratory, Women and Children Central Laboratory, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yangyang Cui
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yiqin Xia
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yue Huang
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xiaowei Wu
- Breast Disease Laboratory, Women and Children Central Laboratory, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Mingjie Zheng
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Miaomiao Weng
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xu Han
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China.,Breast Disease Laboratory, Women and Children Central Laboratory, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hongtao Fu
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China.,Breast Disease Laboratory, Women and Children Central Laboratory, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Lili Yang
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wenbin Zhou
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Ziyi Fu
- Breast Disease Laboratory, Women and Children Central Laboratory, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Shui Wang
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hui Xie
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
8
|
Predicting prognosis and clinical features of the tumor microenvironment based on ferroptosis score in patients with breast cancer. Sci Rep 2022; 12:10611. [PMID: 35739315 PMCID: PMC9226039 DOI: 10.1038/s41598-022-14964-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/15/2022] [Indexed: 12/20/2022] Open
Abstract
Ferroptosis genes have recently been reported to be involved in regulating the development of cancer, but their potential role in breast cancer (BRCA) is not fully understood. The purpose of this study is to systematically study the mechanism of ferroptosis in BRCA and its relationship with this cancer’s prognosis, cell infiltration, gene mutation, and other clinical features. In this study, The Cancer Genome Atlas breast cancer (TCGA-BRCA) database (UCSC Xena) was used to mine the ferroptosis genes related to BRCA patients, and the genes with prognostic value were screened by Cox regression analysis, which were then used to construct a prognostic model for scoring prognostic molecular risk. The relationships between ferroptosis score and prognosis, molecular typing, and clinical characteristics of BRCA were also analyzed. A total of 176 ferroptosis genes related to BRCA were retrieved from the database, 22 of which were found to be significantly related to BRCA prognosis after screening by single-factor Cox regression analysis (p < 0.01). Unsupervised clustering of samples was performed using factoextra, and two subgroups (ferroptosis cluster A and ferroptosis cluster B) with significant differences in prognosis were identified. Subsequently, single-factor Cox regression analysis and random forest dimensionality reduction were used to screen characteristic genes to construct a ferroptosis score model, which included a high ferroptosis score group and a low ferroptosis score group. The results showed that there were significant differences in ferroptosis scores between the ferroptosis cluster A and B groups. The prognosis of patients with low ferroptosis scores was poor, and the overall survival (OS) rate of patients with high ferroptosis scores was significantly higher, indicating that the prognosis of the sample can be well characterized based on calculated ferroptosis scores. Ferroptosis scores differed significantly according to patient age, TP53 and PIK3CA gene mutations, different PAM50 molecular types, and clinical stages. Ferroptosis activation plays a non-negligible role in tumor occurrence and development. Evaluating the ferroptosis score within BRCA will help advance our understanding of the infiltrating properties of cells in the tumor microenvironment and may guide more effective immunotherapy strategies.
Collapse
|
9
|
Ye Z, Ni W, Zhang J, Zhang Y, Yu L, Huang X. Molecular characterization of a profilin gene from a parasitic ciliate Cryptocaryon irritans. Exp Parasitol 2022; 236-237:108248. [DOI: 10.1016/j.exppara.2022.108248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 02/22/2022] [Accepted: 03/16/2022] [Indexed: 11/04/2022]
|
10
|
Mok BR, Kim AR, Baek SH, Ahn JH, Seok SH, Shin JU, Kim DH. Profilin-1 prevents psoriasis pathogenesis through IκBζ regulation. J Invest Dermatol 2022; 142:2455-2463.e9. [PMID: 35148999 DOI: 10.1016/j.jid.2022.01.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 01/03/2022] [Accepted: 01/05/2022] [Indexed: 12/14/2022]
Abstract
Profilin-1 (PFN-1) is an actin-binding protein that regulates actin polymerization, cell proliferation, apoptosis, angiogenesis, and carcinogenesis. Its dysregulation has been reported in diverse pathologic diseases; however, the role of PFN-1 in psoriasis has not yet been elucidated. In this study, we demonstrate that PFN-1 expression is increased in both skin and serum of patients with psoriasis. PFN-1 was markedly expressed in the epidermis of psoriatic lesions and its expression positively correlated with psoriasis severity. IL-17A treatment of keratinocytes increased the PFN-1 expression, whereas TNF-α induced the PFN-1 expression and secretion. In addition, knockdown of PFN-1 with shRNA resulted in an altered expression of psoriasis-associated inflammatory markers, HBD-2, S100A7, S100A9, and Ki67, and recombinant PFN-1 suppressed the IL-17A-induced inflammatory response in keratinocytes. Interestingly, recombinant PFN-1 also suppressed IL-17A-induced IκBζ, an important player in immune response in psoriasis. Collectively, our results show that PFN-1 acts as a negative regulator of psoriatic inflammation through suppression of IκBζ, and the balanced level of PFN-1 is important for the IκBζ regulation. Thus, the expression of PFN-1 can be used as a biomarker for psoriasis severity, and it can be considered as a possible target for the treatment of psoriasis.
Collapse
Affiliation(s)
- Bo Ram Mok
- Department of Biomedical Science, School of Medicine, CHA University, Seongnam, Korea
| | - A-Ram Kim
- Department of Biomedical Science, School of Medicine, CHA University, Seongnam, Korea
| | - Seung Hwa Baek
- Department of Biomedical Science, School of Medicine, CHA University, Seongnam, Korea
| | - Ji Hae Ahn
- Department of Dermatology, Bundang Medical Center, School of Medicine, CHA University, Seongnam, Korea
| | - Seung Hui Seok
- Department of Dermatology, Bundang Medical Center, School of Medicine, CHA University, Seongnam, Korea
| | - Jung U Shin
- Department of Dermatology, Bundang Medical Center, School of Medicine, CHA University, Seongnam, Korea
| | - Dong Hyun Kim
- Department of Dermatology, Bundang Medical Center, School of Medicine, CHA University, Seongnam, Korea.
| |
Collapse
|
11
|
Kao TJ, Wu CC, Phan NN, Liu YH, Ta HDK, Anuraga G, Wu YF, Lee KH, Chuang JY, Wang CY. Prognoses and genomic analyses of proteasome 26S subunit, ATPase (PSMC) family genes in clinical breast cancer. Aging (Albany NY) 2021; 13:17970. [PMID: 34329194 PMCID: PMC8351721 DOI: 10.18632/aging.203345] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/08/2021] [Indexed: 12/11/2022]
Abstract
Breast cancer is a complex disease, and several processes are involved in its development. Therefore, potential therapeutic targets need to be discovered for these patients. Proteasome 26S subunit, ATPase gene (PSMC) family members are well reported to be involved in protein degradation. However, their roles in breast cancer are still unknown and need to be comprehensively researched. Leveraging publicly available databases, such as cBioPortal and Oncomine, for high-throughput transcriptomic profiling to provide evidence-based targets for breast cancer is a rapid and robust approach. By integrating the aforementioned databases with the Kaplan–Meier plotter database, we investigated potential roles of six PSMC family members in breast cancer at the messenger RNA level and their correlations with patient survival. The present findings showed significantly higher expression profiles of PSMC2, PSMC3, PSMC4, PSMC5, and PSMC6 in breast cancer compared to normal breast tissues. Besides, positive correlations were also revealed between PSMC family genes and ubiquinone metabolism, cell cycle, and cytoskeletal remodeling. Meanwhile, we discovered that high levels of PSMC1, PSMC3, PSMC4, PSMC5, and PSMC6 transcripts were positively correlated with poor survival, which likely shows their importance in breast cancer development. Collectively, PSMC family members have the potential to be novel and essential prognostic biomarkers for breast cancer development.
Collapse
Affiliation(s)
- Tzu-Jen Kao
- The Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chung-Che Wu
- Division of Neurosurgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.,Division of Neurosurgery, Department of Surgery, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Nam Nhut Phan
- NTT Institute of Hi-Technology, Nguyen Tat Thanh University, Ho Chi Minh 700000, Vietnam
| | - Yen-Hsi Liu
- School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung 82445, Taiwan
| | - Hoang Dang Khoa Ta
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science, Taipei Medical University, Taipei 11031, Taiwan.,Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Gangga Anuraga
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science, Taipei Medical University, Taipei 11031, Taiwan.,Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.,Department of Statistics, Faculty of Science and Technology, PGRI Adi Buana University, Surabaya, East Java 60234, Indonesia
| | - Yung-Fu Wu
- Department of Medical Research, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei 11490, Taiwan
| | - Kuen-Haur Lee
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science, Taipei Medical University, Taipei 11031, Taiwan.,Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.,Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Jian-Ying Chuang
- The Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taipei 11031, Taiwan.,Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan.,Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chih-Yang Wang
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science, Taipei Medical University, Taipei 11031, Taiwan.,Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
12
|
Hiu JJ, Yap MKK. The effects of Naja sumatrana venom cytotoxin, sumaCTX on alteration of the secretome in MCF-7 breast cancer cells following membrane permeabilization. Int J Biol Macromol 2021; 184:776-786. [PMID: 34174307 DOI: 10.1016/j.ijbiomac.2021.06.145] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/21/2021] [Accepted: 06/21/2021] [Indexed: 12/24/2022]
Abstract
Naja sumatrana venom cytotoxin (sumaCTX) is a basic protein which belongs to three-finger toxin family. It has been shown to induce caspase-dependent, mitochondrial-mediated apoptosis in MCF-7 cells at lower concentrations. This study aimed to investigate the alteration of secretome in MCF-7 cells following membrane permeabilization by high concentrations of sumaCTX, using label-free quantitative (LFQ) approach. The degree of membrane permeabilization of sumaCTX was determined by lactate dehydrogenase (LDH) assay and calcein-propidium iodide (PI) assays. LDH and calcein-PI assays revealed time-dependent membrane permeabilization within a narrow concentration range. However, as toxin concentrations increased, prolonged exposure of MCF-7 cells to sumaCTX did not promote the progression of membrane permeabilization. The secretome analyses showed that membrane permeabilization was an event preceding the release of intracellular proteins. Bioinformatics analyses of the LFQ secretome revealed the presence of 105 significantly distinguished proteins involved in metabolism, structural supports, inflammatory responses, and necroptosis in MCF-7 cells treated with 29.8 μg/mL of sumaCTX. Necroptosis was presumably an initial stress response in MCF-7 cells when exposed to high sumaCTX concentration. Collectively, sumaCTX-induced the loss of membrane integrity in a concentration-dependent manner, whereby the cell death pattern of MCF-7 cells transformed from apoptosis to necroptosis with increasing toxin concentrations.
Collapse
Affiliation(s)
- Jia Jin Hiu
- School of Science, Monash University Malaysia, Bandar Sunway, Malaysia.
| | - Michelle Khai Khun Yap
- School of Science, Monash University Malaysia, Bandar Sunway, Malaysia; Tropical Medicine and Biology Multidisciplinary Platform, Monash University Malaysia, Bandar Sunway, Malaysia.
| |
Collapse
|
13
|
Profilin-1; a novel regulator of DNA damage response and repair machinery in keratinocytes. Mol Biol Rep 2021; 48:1439-1452. [PMID: 33590416 PMCID: PMC7925496 DOI: 10.1007/s11033-021-06210-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/29/2021] [Indexed: 10/25/2022]
Abstract
Profilin-1 (PFN1) regulates actin polymerization and cytoskeletal growth. Despite the essential roles of PFN1 in cell integration, its subcellular function in keratinocyte has not been elucidated yet. Here we characterize the specific regulation of PFN1 in DNA damage response and repair machinery. PFN1 depletion accelerated DNA damage-mediated apoptosis exhibiting PTEN loss of function instigated by increased phosphorylated inactivation followed by high levels of AKT activation. PFN1 changed its predominant cytoplasmic localization to the nucleus upon DNA damage and subsequently restored the cytoplasmic compartment during the recovery time. Even though γH2AX was recruited at the sites of DNA double strand breaks in response to DNA damage, PFN1-deficient cells failed to recruit DNA repair factors, whereas control cells exhibited significant increases of these genes. Additionally, PFN1 depletion resulted in disruption of PTEN-AKT cascade upon DNA damage and CHK1-mediated cell cycle arrest was not recovered even after the recovery time exhibiting γH2AX accumulation. This might suggest PFN1 roles in regulating DNA damage response and repair machinery to protect cells from DNA damage. Future studies addressing the crosstalk and regulation of PTEN-related DNA damage sensing and repair pathway choice by PFN1 may further aid to identify new mechanistic insights for various DNA repair disorders.
Collapse
|
14
|
Pedret A, Catalán Ú, Rubió L, Baiges I, Herrero P, Piñol C, Rodríguez-Calvo R, Canela N, Fernández-Castillejo S, Motilva MJ, Solà R. Phosphoproteomic Analysis and Protein-Protein Interaction of Rat Aorta GJA1 and Rat Heart FKBP1A after Secoiridoid Consumption from Virgin Olive Oil: A Functional Proteomic Approach. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:1536-1554. [PMID: 33502189 DOI: 10.1021/acs.jafc.0c07164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Protein functional interactions could explain the biological response of secoiridoids (SECs), main phenolic compounds in virgin olive oil (VOO). The aim was to assess protein-protein interactions (PPIs) of the aorta gap junction alpha-1 (GJA1) and the heart peptidyl-prolyl cis-trans isomerase (FKBP1A), plus the phosphorylated heart proteome, to describe new molecular pathways in the cardiovascular system in rats using nanoliquid chromatography coupled with mass spectrometry. PPIs modified by SECs and associated with GJA1 in aorta rat tissue were calpain, TUBA1A, and HSPB1. Those associated with FKBP1A in rat heart tissue included SUCLG1, HSPE1, and TNNI3. In the heart, SECs modulated the phosphoproteome through the main canonical pathways PI3K/mTOR signaling (AKT1S1 and GAB2) and gap junction signaling (GAB2 and GJA1). PPIs associated with GJA1 and with FKBP1A, the phosphorylation of GAB2, and the dephosphorylation of GJA1 and AKT1S1 in rat tissues are promising protein targets promoting cardiovascular protection to explain the health benefits of VOO.
Collapse
Affiliation(s)
- Anna Pedret
- Faculty of Medicine and Health Sciences, Medicine and Surgery Department, Functional Nutrition, Oxidation, and CVD Research Group (NFOC-Salut), Universitat Rovira i Virgili, Reus 43201, Spain
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus 43204, Spain
| | - Úrsula Catalán
- Faculty of Medicine and Health Sciences, Medicine and Surgery Department, Functional Nutrition, Oxidation, and CVD Research Group (NFOC-Salut), Universitat Rovira i Virgili, Reus 43201, Spain
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus 43204, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus 43204, Spain
| | - Laura Rubió
- Faculty of Medicine and Health Sciences, Medicine and Surgery Department, Functional Nutrition, Oxidation, and CVD Research Group (NFOC-Salut), Universitat Rovira i Virgili, Reus 43201, Spain
- Food Technology Department, Universitat de Lleida-AGROTECNIO Center, Lleida 25198, Spain
| | - Isabel Baiges
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, Reus 43204, Spain
| | - Pol Herrero
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, Reus 43204, Spain
| | - Carme Piñol
- Department of Medicine, Universitat de Lleida, Lleida 25008, Catalonia, Spain
- Institut de Recerca Biomèdica de Lleida Fundació Dr. Pifarré-IRBLLeida, Lleida 25198, Spain
| | - Ricardo Rodríguez-Calvo
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus 43204, Spain
- Research Unit on Lipids and Atherosclerosis, Vascular Medicine and Metabolism Unit, Universitat Rovira i Virgili, Reus 43204, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Institute of Health Carlos III, Madrid 28029, Spain
- Hospital Universitari Sant Joan de Reus (HUSJR), Reus 43204, Spain
| | - Núria Canela
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, Reus 43204, Spain
| | - Sara Fernández-Castillejo
- Faculty of Medicine and Health Sciences, Medicine and Surgery Department, Functional Nutrition, Oxidation, and CVD Research Group (NFOC-Salut), Universitat Rovira i Virgili, Reus 43201, Spain
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus 43204, Spain
| | - Maria-Jose Motilva
- Instituto de Ciencias de la Vid y del Vino-ICVV CSIC, Gobierno de La Rioja, Universidad de La Rioja, Logroño 26006, Spain
| | - Rosa Solà
- Faculty of Medicine and Health Sciences, Medicine and Surgery Department, Functional Nutrition, Oxidation, and CVD Research Group (NFOC-Salut), Universitat Rovira i Virgili, Reus 43201, Spain
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus 43204, Spain
- Hospital Universitari Sant Joan de Reus (HUSJR), Reus 43204, Spain
| |
Collapse
|
15
|
Pimm ML, Hotaling J, Henty-Ridilla JL. Profilin choreographs actin and microtubules in cells and cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 355:155-204. [PMID: 32859370 PMCID: PMC7461721 DOI: 10.1016/bs.ircmb.2020.05.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Actin and microtubules play essential roles in aberrant cell processes that define and converge in cancer including: signaling, morphology, motility, and division. Actin and microtubules do not directly interact, however shared regulators coordinate these polymers. While many of the individual proteins important for regulating and choreographing actin and microtubule behaviors have been identified, the way these molecules collaborate or fail in normal or disease contexts is not fully understood. Decades of research focus on Profilin as a signaling molecule, lipid-binding protein, and canonical regulator of actin assembly. Recent reports demonstrate that Profilin also regulates microtubule dynamics and polymerization. Thus, Profilin can coordinate both actin and microtubule polymer systems. Here we reconsider the biochemical and cellular roles for Profilin with a focus on the essential cytoskeletal-based cell processes that go awry in cancer. We also explore how the use of model organisms has helped to elucidate mechanisms that underlie the regulatory essence of Profilin in vivo and in the context of disease.
Collapse
Affiliation(s)
- Morgan L Pimm
- Department of Cell and Developmental Biology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Jessica Hotaling
- Department of Cell and Developmental Biology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Jessica L Henty-Ridilla
- Department of Cell and Developmental Biology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States; Department of Biochemistry and Molecular Biology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States.
| |
Collapse
|
16
|
Abstract
Profilin is a ubiquitously expressed protein well known as a key regulator of actin polymerisation. The actin cytoskeleton is involved in almost all cellular processes including motility, endocytosis, metabolism, signal transduction and gene transcription. Hence, profilin's role in the cell goes beyond its direct and essential function in regulating actin dynamics. This review will focus on the interactions of Profilin 1 and its ligands at the plasma membrane, in the cytoplasm and the nucleus of the cells and the regulation of profilin activity within those cell compartments. We will discuss the interactions of profilin in cell signalling pathways and highlight the importance of the cell context in the multiple functions that this small essential protein has in conjunction with its role in cytoskeletal organisation and dynamics. We will review some of the mechanisms that control profilin expression and the implications of changed expression of profilin in the light of cancer biology and other pathologies.
Collapse
|
17
|
Wang G, Zhong WC, Bi YH, Tao SY, Zhu H, Zhu HX, Xu AM. The Prognosis Of Peroxiredoxin Family In Breast Cancer. Cancer Manag Res 2019; 11:9685-9699. [PMID: 31814764 PMCID: PMC6861534 DOI: 10.2147/cmar.s229389] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 11/02/2019] [Indexed: 12/11/2022] Open
Abstract
Purpose PRDX (Peroxiredoxin) family has involved in breast cancer tumorigenesis from the evidence obtained from cell lines, human tissues and mouse models. Nonetheless, the diversified expression patterns, coupled with the prognostic values of PRDX family, still require explanation. This study aimed at investigating the clinical importance and biological of PRDXs in breast cancer. Patients and methods Specimens of paraffin sections used for immunohistochemistry were collected from the hospital and the remaining patient information was retrieved from online databases. The expression and survival data of PRDXs in patients with breast cancer were from ONCOMINE, GEPIA, Kaplan–Meier Plotter. cBioPortal, Metascape, String, Cytoscape and DAVID were used to predict functions and pathways of the changes in PRDXs and their frequently altered neighbor genes. Immunohistochemistry was used to detect the expression of PRDXs in breast cancer. Results We discovered the expression levels of PRDX1-5 were higher in breast cancer tissues than in normal tissues, whereas the expression level of PRDX6 was observed as lower in the former one in comparison with that of the latter one. There existed a correlation between the expression levels of PRDX4, 5 and the advanced tumor stage. Survival analysis revealed that the expression of PRDXs were all associated with relapse-free survival (RFS) in all of the patients with breast cancer. Eventually, we discovered significant regulation of the cellular oxidant detoxification and detoxification of ROS by the PRDX changes, together with obtaining the core modules of genes (TXN, TXN2, TXNRD1, TXNRD2, GPX1 and GPX2) linked to the PRDX family of genes in breast cancer. Conclusion The PRDX family is widely involved in the development of breast cancer and affects the prognosis of patients. The functions and pathways of the changes in PRDXs and their frequently altered neighbor genes can be further verified by wet experiments.
Collapse
Affiliation(s)
- Gang Wang
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei 230001, People's Republic of China
| | - Wan-Chao Zhong
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Yi-Hui Bi
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei 230001, People's Republic of China
| | - Si-Yue Tao
- Department of Orthopaedics, Anhui Provincial Hospital Affiliated with Anhui Medical University, Hefei, Anhui 230001, People's Republic of China
| | - Hai Zhu
- Department of Gastrointestinal Surgery, Department Of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei 230001, People's Republic of China
| | - Hai-Xing Zhu
- Department of Gastrointestinal Surgery, Anhui Provincial Cancer Hospital, Hefei 230001, People's Republic of China
| | - A-Man Xu
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei 230001, People's Republic of China
| |
Collapse
|
18
|
Zhao J, Xu J, Lv J. Identification of profilin 1 as the primary target for the anti-cancer activities of Furowanin A in colorectal cancer. Pharmacol Rep 2019; 71:940-949. [PMID: 31454697 DOI: 10.1016/j.pharep.2019.05.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 04/21/2019] [Accepted: 05/13/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Furowanin A (Fur A) is a flavonoid compound isolated from medicinal plant Millettia pachycarpa Benth. This study aims to explore the effect of Fur A on Colorectal cancer (CRC) and its molecular mechanisms. METHODS Cell proliferative capacity of CRC cells was assessed by CCK-8 assay. Cell apoptosis and cell cycle distribution were detected by flow cytometry. Cell migration and invasion were detected by wound healing and Transwell assay, respectively. EMT markers, apoptosis and profilin 1(Pfn1) expression were detected by immunohistochemistry (IHC). The protein expression levels were examined by western blotting. i-TRAQ analyses were conducted to identify the differentially expressed genes in CRC cells. CRC xenograft model was also used to validate the in vivo anti-cancer activity of Fur A. RESULTS Fur A exhibited anti-prolifertive, blocked cell cycle progression and promoted apoptotic cell death in CRC cells. Fur A suppressed the migration, invasion and epithelial-to-mesenchymal transition (EMT) in vitro, and tumor growth and pulmonary metastasis in vivo, without causing obvious toxicity. iTRAQ analysis identified Pfn1 as a gene up-regulated by Fur A. In xenograft tumor tissue, the expression of Pfn1 was also elevated by Fur A treatment. In clinical CRC samples, high expression of Pfn1 was correlated with lower stage and longer survival. Knockdown of Pfn1 significantly dampened the pro-apoptotic and anti-metastatic activities of Fur A in CRC cells. Ectopic Pfn1 expression augmented the anti-neoplastic activities of Fur A. CONCLUSION Fur A exhibited anti-cancer activities in vitro and in vivo in CRC by up-regulating Pfn1.
Collapse
Affiliation(s)
- Jinxia Zhao
- The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Junhua Xu
- The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jing Lv
- The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
19
|
Tomasello L, Coppola A, Pitrone M, Failla V, Cillino S, Pizzolanti G, Giordano C. PFN1 and integrin-β1/mTOR axis involvement in cornea differentiation of fibroblast limbal stem cells. J Cell Mol Med 2019; 23:7210-7221. [PMID: 31513338 PMCID: PMC6815913 DOI: 10.1111/jcmm.14438] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/26/2019] [Accepted: 05/15/2019] [Indexed: 12/14/2022] Open
Abstract
Ex vivo limbal stem cell transplantation is the main therapeutic approach to address a complete and functional re-epithelialization in corneal blindness, the second most common eye disorder. Although important key points were defined, the molecular mechanisms involved in the epithelial phenotype determination are unclear. Our previous studies have demonstrated the pluripotency and immune-modulatory of fibroblast limbal stem cells (f-LSCs), isolated from the corneal limbus. We defined a proteomic profile especially enriched in wound healing and cytoskeleton-remodelling proteins, including Profilin-1 (PFN1). In this study we postulate that pfn-1 knock down promotes epithelial lineage by inhibiting the integrin-β1(CD29)/mTOR pathway and subsequent NANOG down-expression. We showed that it is possible modulate pfn1 expression levels by treating f-LSCs with Resveratrol (RSV), a natural compound: pfn1 decline is accompanied with up-regulation of the specific differentiation epithelial genes pax6 (paired-box 6), sox17 (sex determining region Y-box 17) and ΔNp63-α (p63 splice variant), consistent with drop-down of the principle stem gene levels. These results contribute to understand the molecular biology of corneal epithelium development and suggest that pfn1 is a potential molecular target for the treatment of corneal blindness based on epithelial cell dysfunction.
Collapse
Affiliation(s)
- Laura Tomasello
- Laboratory of Regenerative Medicine "Aldo Galluzzo", Section of Endocrinology, Diabetology and Metabolism, Department of Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro" (ProMISE), University of Palermo, Palermo, Italy
| | - Antonina Coppola
- Laboratory of Regenerative Medicine "Aldo Galluzzo", Section of Endocrinology, Diabetology and Metabolism, Department of Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro" (ProMISE), University of Palermo, Palermo, Italy
| | - Maria Pitrone
- Laboratory of Regenerative Medicine "Aldo Galluzzo", Section of Endocrinology, Diabetology and Metabolism, Department of Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro" (ProMISE), University of Palermo, Palermo, Italy
| | - Valentina Failla
- Department of Ophthalmology, University of Palermo, Palermo, Italy
| | | | - Giuseppe Pizzolanti
- Laboratory of Regenerative Medicine "Aldo Galluzzo", Section of Endocrinology, Diabetology and Metabolism, Department of Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro" (ProMISE), University of Palermo, Palermo, Italy
| | - Carla Giordano
- Laboratory of Regenerative Medicine "Aldo Galluzzo", Section of Endocrinology, Diabetology and Metabolism, Department of Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro" (ProMISE), University of Palermo, Palermo, Italy
| |
Collapse
|
20
|
Ramdas P, Radhakrishnan AK, Abdu Sani AA, Abdul-Rahman PS. Tocotrienols Modulate Breast Cancer Secretomes and Affect Cancer-Signaling Pathways in MDA-MB-231 Cells: A Label-Free Quantitative Proteomic Analysis. Nutr Cancer 2019; 71:1263-1271. [PMID: 31084432 DOI: 10.1080/01635581.2019.1607407] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Tocotrienols (T3), a family of vitamin E, are reported to possess potent anti-cancer effects but the molecular mechanisms behind these effects still remain unclear. The aim of this study was to investigate how T3 exert anti-cancer effects on MDA-MB-231 human breast cancer cells. The MDA-MB-231 cells were chosen for this study as they are triple-negative and highly metastatic cells, which form aggressive tumors in experimental models. The MDA-MB-231 cells were treated with varying concentrations (0-20 µg mL-1) of gamma (γ) or delta (δ) T3 and the secretome profiles of these cells treated with half maximal inhibitory concentration (IC50) of γT3 (5.8 µg mL-1) or δT3 (4.0 µg mL-1) were determined using label-free quantitative proteomic strategy. A total of 103, 174 and 141 proteins were identified with ProteinLynx Global Server (PLGS) score of more than 200 and above 25% sequence coverage in the untreated control and T3-treated cell culture supernatant respectively. A total of 18 proteins were dysregulated between untreated control and T3 (δT3 or γT3) treated conditions. The results showed that T3 treatment downregulated the exogenous Cathepsin D and Serpine1 proteins but upregulated Profilin-1 protein, which play a key role in breast cancer in the MDA-MB-231 cells. These findings strongly suggest that T3 may induce differential expression of secreted proteins involved in the cytoskeletal regulation of RHO GTPase signaling pathway.
Collapse
Affiliation(s)
- Premdass Ramdas
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia.,Department of Biotechnology, School of Health Sciences, International Medical University , Kuala Lumpur , Malaysia
| | - Ammu Kutty Radhakrishnan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University , Bandar Sunway , Malaysia
| | | | | |
Collapse
|
21
|
Coumans JVF, Davey RJ, Moens PDJ. Cofilin and profilin: partners in cancer aggressiveness. Biophys Rev 2018; 10:1323-1335. [PMID: 30027463 DOI: 10.1007/s12551-018-0445-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/08/2018] [Indexed: 02/07/2023] Open
Abstract
This review covers aspects of cofilin and profilin regulations and their influence on actin polymerisation responsible for cell motility and metastasis. The regulation of their activity by phosphorylation and nitration, miRs, PI(4,5)P2 binding, pH, oxidative stress and post-translational modification is described. In this review, we have highlighted selected similarities, complementarities and differences between the two proteins and how their interplay affects actin filament dynamics.
Collapse
Affiliation(s)
- Joelle V F Coumans
- School of Rural Medicine, University of New England, Armidale, Australia
| | - Rhonda J Davey
- Centre for Bioactive Discovery in Health and Ageing, School of Science and Technology, University of New England, Armidale, Australia
| | - Pierre D J Moens
- Centre for Bioactive Discovery in Health and Ageing, School of Science and Technology, University of New England, Armidale, Australia.
| |
Collapse
|
22
|
Zhang H, Yang W, Yan J, Zhou K, Wan B, Shi P, Chen Y, He S, Li D. Loss of profilin 2 contributes to enhanced epithelial-mesenchymal transition and metastasis of colorectal cancer. Int J Oncol 2018; 53:1118-1128. [PMID: 30015842 PMCID: PMC6065425 DOI: 10.3892/ijo.2018.4475] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 06/07/2018] [Indexed: 12/14/2022] Open
Abstract
Profilin 2 (PFN2) functions as an actin cytoskeleton regulator and serves an important role in cell motility. However, a role for PFN2 in the progression of colorectal cancer (CRC), particularly in metastasis, has yet to be clarified. The aim of the present study was to investigate whether PFN2 served specific roles in the progression of human CRC. The results demonstrated that PFN2 was differentially expressed in CRC tissues and cell lines by reverse transcription-quantitative polymerase chain reaction and western blotting. PFN2 expression was also negatively associated with the degree of tumor metastasis. Low PFN2 expression in CRC cells was related with enhanced epithelial-mesenchymal transition (EMT) and, in turn, may increase migratory capabilities. Overexpression of PFN2 in CRC cell lines with a low level of endogenous PFN2 inhibited the EMT process, as well as the associated migration; in addition, myosin light chain (MLC) phosphorylation was upregulated. Inhibition of MLC phosphorylation attenuated the inhibition of EMT and cell migratory abilities induced by PFN2 overexpression in CRC cell lines, the results suggested that PFN2 may suppress cancer EMT and the subsequent metastasis by regulating cytoskeletal reorganization. These results demonstrated that PFN2 may serve a suppressive role in the metastasis of CRC and therefore may provide a new potential target for cancer therapeutics.
Collapse
Affiliation(s)
- Hui Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Weiqiang Yang
- Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai 201800, P.R. China
| | - Jinlong Yan
- Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai 201800, P.R. China
| | - Kaiping Zhou
- Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai 201800, P.R. China
| | - Boshun Wan
- Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai 201800, P.R. China
| | - Peidong Shi
- Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai 201800, P.R. China
| | - Yueyu Chen
- Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai 201800, P.R. China
| | - Songbing He
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Dechun Li
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
23
|
Zoppino FCM, Guerrero-Gimenez ME, Castro GN, Ciocca DR. Comprehensive transcriptomic analysis of heat shock proteins in the molecular subtypes of human breast cancer. BMC Cancer 2018; 18:700. [PMID: 29954368 PMCID: PMC6022707 DOI: 10.1186/s12885-018-4621-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 06/20/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Heat Shock Proteins (HSPs), a family of genes with key roles in proteostasis, have been extensively associated with cancer behaviour. However, the HSP family is quite large and many of its members have not been investigated in breast cancer (BRCA), particularly in relation with the current molecular BRCA classification. In this work, we performed a comprehensive transcriptomic study of the HSP gene family in BRCA patients from both The Cancer Genome Atlas (TCGA) and the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) cohorts discriminating the BRCA intrinsic molecular subtypes. METHODS We examined gene expression levels of 1097 BRCA tissue samples retrieved from TCGA and 1981 samples of METABRIC, focusing mainly on the HSP family (95 genes). Data were stratified according to the PAM50 gene expression (Luminal A, Luminal B, HER2, Basal, and Normal-like). Transcriptomic analyses include several statistical approaches: differential gene expression, hierarchical clustering and survival analysis. RESULTS Of the 20,531 analysed genes we found that in BRCA almost 30% presented deregulated expression (19% upregulated and 10% downregulated), while of the HSP family 25% appeared deregulated (14% upregulated and 11% downregulated) (|fold change| > 2 comparing BRCA with normal breast tissues). The study revealed the existence of shared HSP genes deregulated in all subtypes of BRCA while other HSPs were deregulated in specific subtypes. Many members of the Chaperonin subfamily were found upregulated while three members (BBS10, BBS12 and CCTB6) were found downregulated. HSPC subfamily had moderate increments of transcripts levels. Various genes of the HSP70 subfamily were upregulated; meanwhile, HSPA12A and HSPA12B appeared strongly downregulated. The strongest downregulation was observed in several HSPB members except for HSPB1. DNAJ members showed heterogeneous expression pattern. We found that 23 HSP genes correlated with overall survival and three HSP-based transcriptional profiles with impact on disease outcome were recognized. CONCLUSIONS We identified shared and specific HSP genes deregulated in BRCA subtypes. This study allowed the recognition of HSP genes not previously associated with BRCA and/or any cancer type, and the identification of three clinically relevant clusters based on HSPs expression patterns with influence on overall survival.
Collapse
Affiliation(s)
- Felipe C. M. Zoppino
- Laboratory of Oncology, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), Av. Dr. Ruiz Leal s/n, Parque General San Martín, 5500 Mendoza, Argentina
| | - Martin E. Guerrero-Gimenez
- Laboratory of Oncology, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), Av. Dr. Ruiz Leal s/n, Parque General San Martín, 5500 Mendoza, Argentina
| | - Gisela N. Castro
- Laboratory of Oncology, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), Av. Dr. Ruiz Leal s/n, Parque General San Martín, 5500 Mendoza, Argentina
| | - Daniel R. Ciocca
- Laboratory of Oncology, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), Av. Dr. Ruiz Leal s/n, Parque General San Martín, 5500 Mendoza, Argentina
| |
Collapse
|
24
|
Joy M, Gau D, Castellucci N, Prywes R, Roy P. The myocardin-related transcription factor MKL co-regulates the cellular levels of two profilin isoforms. J Biol Chem 2017; 292:11777-11791. [PMID: 28546428 DOI: 10.1074/jbc.m117.781104] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/24/2017] [Indexed: 12/26/2022] Open
Abstract
Megakaryoblastic leukemia (MKL)/serum-response factor (SRF)-mediated gene transcription is a highly conserved mechanism that connects dynamic reorganization of the actin cytoskeleton to regulation of expression of a wide range of genes, including SRF itself and many important structural and regulatory components of the actin cytoskeleton. In this study, we examined the possible role of MKL/SRF in the context of regulation of profilin (Pfn), a major controller of actin dynamics and actin cytoskeletal remodeling in cells. We demonstrated that despite being located on different genomic loci, two major isoforms of Pfn (Pfn1 and Pfn2) are co-regulated by a common mechanism involving the action of MKL that is independent of its SRF-related activity. We found that MKL co-regulates the expression of Pfn isoforms indirectly by modulating signal transducer and activator of transcription 1 (STAT1) and utilizing its SAP-domain function. Unexpectedly, our studies revealed that cellular externalization, rather than transcription of Pfn1, is affected by the perturbations of MKL. We further demonstrated that MKL can influence cell migration by modulating Pfn1 expression, indicating a functional connection between MKL and Pfn1 in actin-dependent cellular processes. Finally, we provide initial evidence supporting the ability of Pfn to influence MKL and SRF expression. Collectively, these findings suggest that Pfn may play a role in a possible feedback loop of the actin/MKL/SRF signaling circuit.
Collapse
Affiliation(s)
- Marion Joy
- Departments of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | - David Gau
- Departments of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | - Nevin Castellucci
- Departments of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | - Ron Prywes
- Department of Biological Sciences, Columbia University, New York, New York 10027
| | - Partha Roy
- Departments of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15219; Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15219; Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania 15219.
| |
Collapse
|
25
|
Yan J, Ma C, Gao Y. MicroRNA-30a-5p suppresses epithelial-mesenchymal transition by targeting profilin-2 in high invasive non-small cell lung cancer cell lines. Oncol Rep 2017; 37:3146-3154. [PMID: 28405690 DOI: 10.3892/or.2017.5566] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 04/03/2017] [Indexed: 11/05/2022] Open
Abstract
PFN2 is an invasion promoter in several cancers including lung cancer. However, the probable effects and underlying mechanisms of PFN2 in tumor cell epithelial-mesenchymal-transition (EMT) of non-small cell lung cancer (NSCLC) remain poorly understood. The protein and mRNA levels of PFN2 in human bronchial epithelial cell line 16HBE and three NSCLC cell lines A549, NCI-H520 and 95D were assessed. The gain-of-function (overexpression) and loss‑of-function (siRNA) experiments of PFN2 were performed in 95D cells. A dual-luciferase reporter assay, western blotting and real-time PCR were used to investigate the relationship between PFN2 and miR‑30a‑5p. PFN2 was upregulated in three NSCLC cell lines, and the highest in 95D cell line. Furthermore, the upregulation of PFN2 promoted, whereas the downregulation of PFN2 suppressed invasion and EMT in 95D. Dual-luciferase reporter assay showed that miR‑30a‑5p directly interacts with the 3'-untranslated region (3'-UTR) of PFN2 mRNA. Interestingly, miR‑30a‑5p negatively regulates the expression of PFN2 and suppresses EMT and invasion in 95D. In summary, the present study demonstrated that miR‑30a‑5p inhibits EMT and invasion in high invasive NSCLC cell lines via targeting PFN2. Suggesting the association of miR‑30a‑5p and PFN2 may play an essential role in the development of NSCLC by modulating EMT and cell invasion.
Collapse
Affiliation(s)
- Jiliang Yan
- Department of Clinical Laboratory, Kaifeng Central Hospital, Kaifeng, Henan 475000, P.R. China
| | - Chunyan Ma
- Department of Clinical Laboratory, Kaifeng Central Hospital, Kaifeng, Henan 475000, P.R. China
| | - Yue Gao
- Department of Clinical Laboratory, Beibei Traditional Chinese Medical Hospital, Chongqing 400700, P.R. China
| |
Collapse
|
26
|
Amith SR, Vincent KM, Wilkinson JM, Postovit LM, Fliegel L. Defining the Na +/H + exchanger NHE1 interactome in triple-negative breast cancer cells. Cell Signal 2016; 29:69-77. [PMID: 27751915 DOI: 10.1016/j.cellsig.2016.10.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/04/2016] [Accepted: 10/13/2016] [Indexed: 12/30/2022]
Abstract
Mounting evidence supports a major role for the Na+/H+ exchanger NHE1 in cancer progression and metastasis. NHE1 is hyperactive at the onset of oncogenic transformation, resulting in intracellular alkalinization and extracellular microenvironmental acidification. These conditions promote invasion and facilitate metastasis. However, the signal pathways governing the regulation of exchanger activity are still unclear. This is especially important in the aggressively metastatic, triple-negative basal breast cancer subtype. We used affinity chromatography followed by mass spectrometry to identify novel and putative interaction partners of NHE1 in MDA-MB-231 triple-negative breast cancer cells. NHE1 associated with several types of proteins including cytoskeletal proteins and chaperones. We validated protein interactions by co-immunoprecipitation for: 14-3-3, AKT, α-enolase, CHP1, HSP70 and HSP90. Additionally, we used The Cancer Genome Atlas (TCGA) to study NHE1 gene expression in primary patient breast tumours versus adjacent normal tissue. NHE1 expression was elevated in breast tumour samples and, when broken down by breast cancer subtype, NHE1 gene expression was significantly lower in tumours of the basal subtype compared to luminal and HER2+ subtypes. Reverse phase protein array (RPPA) analysis showed that NHE1 expression positively correlated with p90RSK expression in basal, but not luminal, primary tumours. Other proteins were negatively correlated with NHE1 expression in basal breast cancer tumours. Taken together, our data provides the first insight into the signalling molecules that form the NHE1 interactome in triple-negative breast cancer cells. These results will focus our search for novel targeted therapies.
Collapse
Affiliation(s)
- Schammim Ray Amith
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada.
| | - Krista Marie Vincent
- Department of Oncology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada; Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario N6A 3K7, Canada.
| | - Jodi Marie Wilkinson
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada.
| | - Lynne Marie Postovit
- Department of Oncology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada.
| | - Larry Fliegel
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada.
| |
Collapse
|
27
|
Girotra S, Yeghiazaryan K, Golubnitschaja O. Potential biomarker panels in overall breast cancer management: advancements by multilevel diagnostics. Per Med 2016; 13:469-484. [PMID: 29767597 DOI: 10.2217/pme-2016-0020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Breast cancer (BC) prevalence has reached an epidemic scale with half a million deaths annually. Current deficits in BC management include predictive and preventive approaches, optimized screening programs, individualized patient profiling, highly sensitive detection technologies for more precise diagnostics and therapy monitoring, individualized prediction and effective treatment of BC metastatic disease. To advance BC management, paradigm shift from delayed to predictive, preventive and personalized medical services is essential. Corresponding step forwards requires innovative multilevel diagnostics procuring specific panels of validated biomarkers. Here, we discuss current instrumental advancements including genomics, proteomics, epigenetics, miRNA, metabolomics, circulating tumor cells and cancer stem cells with a focus on biomarker discovery and multilevel diagnostic panels. A list of the recommended biomarker candidates is provided.
Collapse
|
28
|
Moens PDJ, Coumans JVF. Profilin-1 mediated cell-cycle arrest: searching for drug targets. Cell Cycle 2015; 14:3669-70. [PMID: 26313613 DOI: 10.1080/15384101.2015.1086204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Pierre D J Moens
- a Centre for Bioactive Discovery in Health and Aging; School of Science and Technology ; University of New England ; Armidale , NSW Australia
| | - Joelle V F Coumans
- b School of Rural Medicine ; University of New England ; Armidale , NSW Australia
| |
Collapse
|
29
|
Jiang C, Veon W, Li H, Hallows KR, Roy P. Epithelial morphological reversion drives Profilin-1-induced elevation of p27(kip1) in mesenchymal triple-negative human breast cancer cells through AMP-activated protein kinase activation. Cell Cycle 2015; 14:2914-23. [PMID: 26176334 DOI: 10.1080/15384101.2015.1069929] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Profilin-1 (Pfn1) is an important regulator of actin polymerization that is downregulated in human breast cancer. Previous studies have shown Pfn1 has a tumor-suppressive effect on mesenchymal-like triple-negative breast cancer cells, and Pfn1-induced growth suppression is partly mediated by upregulation of cell-cycle inhibitor p27(kip1) (p27). In this study, we demonstrate that Pfn1 overexpression leads to accumulation of p27 through promoting AMPK activation and AMPK-dependent phosphorylation of p27 on T198 residue, a post-translational modification that leads to increased protein stabilization of p27. This pathway is mediated by Pfn1-induced epithelial morphological reversion of mesenchymal breast cancer through cadherin-mediated restoration of adherens junctions. These findings not only elucidate a potential mechanism of how Pfn1 may inhibit proliferation of mesenchymal breast cancer cells, but also highlight a novel pathway of cadherin-mediated p27 induction and therefore cell-cycle control in cells.
Collapse
Affiliation(s)
- Chang Jiang
- a Department of Bioengineering ; University of Pittsburgh ; Pittsburgh PA
| | - William Veon
- a Department of Bioengineering ; University of Pittsburgh ; Pittsburgh PA
| | - Hui Li
- b Department of Medicine ; Renal Electrolyte Division; University of Pittsburgh ; Pittsburgh PA
| | - Kenneth R Hallows
- b Department of Medicine ; Renal Electrolyte Division; University of Pittsburgh ; Pittsburgh PA.,c Department of Cell Biology ; University of Pittsburgh ; Pittsburgh PA
| | - Partha Roy
- a Department of Bioengineering ; University of Pittsburgh ; Pittsburgh PA.,d Magee Women's Research Institute; University of Pittsburgh ; Pittsburgh PA.,e Department of Pathology ; University of Pittsburgh ; PA
| |
Collapse
|
30
|
|