1
|
Biyik-Sit R, Waigel S, Andreeva K, Rouchka E, Clem BF. Bioinformatics analysis of PSAT1 loss identifies downstream pathways regulated in EGFR mutant NSCLC and a selective gene signature for predicting the risk of relapse. Oncol Lett 2025; 29:9. [PMID: 39512505 PMCID: PMC11542166 DOI: 10.3892/ol.2024.14755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 09/25/2024] [Indexed: 11/15/2024] Open
Abstract
The majority of malignant tumors exhibit an altered metabolic phenotype that ultimately provides the required energy and molecular precursors necessary for unregulated cell division. Within this, phosphoserine aminotransferase 1 (PSAT1) is involved in de novo serine biosynthesis and its activity promotes various biochemical processes, including one-carbon metabolism. It also directly generates α-ketoglutarate (α-KG), a Kreb cycle intermediate and epigenetic-regulating metabolite. Prior studies examining PSAT1 depletion have identified individual affected downstream pathways, such as GSK3β and E2F, in several cancer types, including non-small-cell lung cancer (NSCLC). However, global gene expression examination in response to PSAT1 loss, particularly in EGFR mutant NSCLC, has not been unexplored. Transcriptional profiling of EGFR mutant NSCLC cells with or without stable knock-down of PSAT1 identified differentially expressed genes (DEGs) enriched in several metabolic pathways required for cell division, including amino acid and nucleotide biosynthesis. Supplementation studies involving non-essential amino acids, nucleosides and α-KG partially restored defects in anchorage-independent growth due to the knockdown of PSAT1. Kyoto Encyclopedia of Genes and Genomes and Gene Ontology enrichment analysis identified potential impacts on actin cytoskeleton arrangement and β-catenin activity, which were rescued by PSAT1 re-expression. Finally, a comparative analysis of PSAT1 DEGs against transcripts enriched in patient EGFR mutant lung tumors identified a gene signature that is associated with overall and relapse-free survival (RFS) and was able to distinguish low or high-risk populations for RFS in early-stage EGFR mutant NSCLC. Overall, investigating genes altered by PSAT1 loss confirmed known PSAT1-regulated cellular pathways, identified a previously unknown role in the mediation of cytoskeleton arrangement in EGFR mutant NSCLC cells and allowed for the characterization of a gene signature with putative predictive potential for RFS in early-stage disease.
Collapse
Affiliation(s)
- Rumeysa Biyik-Sit
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY 40202, USA
- Brown Cancer Center, Louisville, KY 40202, USA
| | - Sabine Waigel
- Brown Cancer Center, Louisville, KY 40202, USA
- Kentucky IDeA Network of Biomedical Research Excellence Bioinformatics Core, University of Louisville, Louisville, KY 40202, USA
| | - Kalina Andreeva
- Kentucky IDeA Network of Biomedical Research Excellence Bioinformatics Core, University of Louisville, Louisville, KY 40202, USA
- Department of Neuroscience Training, University of Louisville, Louisville, KY 40202, USA
| | - Eric Rouchka
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY 40202, USA
- Kentucky IDeA Network of Biomedical Research Excellence Bioinformatics Core, University of Louisville, Louisville, KY 40202, USA
| | - Brian F Clem
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY 40202, USA
- Brown Cancer Center, Louisville, KY 40202, USA
| |
Collapse
|
2
|
Chacko N, Ankri R. Non-invasive early-stage cancer detection: current methods and future perspectives. Clin Exp Med 2024; 25:17. [PMID: 39708168 DOI: 10.1007/s10238-024-01513-x] [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: 09/02/2024] [Accepted: 10/21/2024] [Indexed: 12/23/2024]
Abstract
This review paper explores the realm of non-invasive methods for early cancer detection. Early identification is crucial for effective therapeutic intervention, and non-invasive techniques have emerged as promising tools to enhance diagnostic accuracy and improve patient outcomes. The paper thoroughly examines the advantages, limitations, and prospects of various non-invasive approaches, including blood tests, non-blood-based tests, and diverse imaging modalities. It discusses the biomarkers found in blood for early-stage cancer detection, specifying the types of cancer associated with each biomarker. The non-blood-based tests focus on components in saliva, urine, and breath for cancer detection, alongside current studies and future perspectives on various cancers. Optical imaging methods covered in this review include fluorescence imaging in the near-infrared (NIR) region, bioluminescence imaging, and Raman spectroscopy for early-stage cancer detection. The review also highlights the pros and cons of ultrasound imaging in early-stage cancer detection. Additionally, the clinical implications of using AI for cancer detection, both present and future, are explored. This paper provides valuable insights for researchers and clinicians working in the field of non-invasive early-stage cancer detection.
Collapse
Affiliation(s)
- Neelima Chacko
- Department of Physics, Faculty of Natural Science, Ariel University, 40700, Ariel, Israel
| | - Rinat Ankri
- Department of Physics, Faculty of Natural Science, Ariel University, 40700, Ariel, Israel.
| |
Collapse
|
3
|
Wan Y, Luo W, Song X, Zhao Y, Han Z, Shen J, Xie F, Li Y, He J. A Targeted Proteomics Approach Reveals a Serum Protein Signature as a Diagnostic Biomarker for Colorectal Cancer. J Inflamm Res 2024; 17:10755-10768. [PMID: 39677294 PMCID: PMC11645893 DOI: 10.2147/jir.s492356] [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: 08/20/2024] [Accepted: 11/12/2024] [Indexed: 12/17/2024] Open
Abstract
Background Circulating proteins secreted by tumors are an important source of cancer biomarkers. This study aims to investigate the changes in the content of tumor immune-related circulating proteins in peripheral blood from patients with colorectal cancer (CRC). Methods Olink's proximity extension assay was used to detect the levels of 92 tumor immune-related circulating proteins in peripheral blood from CRC patients. An enzyme-linked immunosorbent assay was performed to detect the levels of six proteins. Elastic network regression was used to establish the model, and the performance of the model was verified by multiple iterations of cross-validation. Results The best serum protein signature that was composed of six proteins (IL7, CXCL12, IL10, IL15, CXCL1, and MCP-3) was selected. The area under the curve value of this signature was 0.9924 in the training set and 0.8992 in the total set. IL7 and IL15 levels were significantly higher in the ≥4 cm tumor volume group than in the <4 cm tumor volume group (P = 0.0113 and P = 0.004, respectively). MCP-3 levels were significantly higher in the distant metastasis group than in the non-distant metastasis group (P =0.0465). There was a significant difference in MCP-3 levels among different tumor, node, metastasis stages (P = 0.0496). CXCL1 levels were positively correlated with the absolute count of basophils (R = 0.3220, P = 0.0273), and IL10 levels were positively correlated with the absolute count of neutrophils (R = 0.38737, P = 0.0078). CXCL1, IL7, and IL15 were independent prognostic factors of CRC (hazard ratio [HR] = 0.62, P = 0.006; HR = 0.57, P = 0.006; and HR = 0.64, P = 0.011, respectively). Conclusion The best serum protein signature model (IL7, CXCL12, IL10, IL15, CXCL1, and MCP-3) was able to distinguish CRC patients from healthy controls. These proteins were also involved in the occurrence and development of CRC.
Collapse
Affiliation(s)
- Yu Wan
- Gastroenterology Department, The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511400, People’s Republic of China
| | - Wenfeng Luo
- Central Laboratory, The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511400, People’s Republic of China
| | - Xiaoyu Song
- Central Laboratory, The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511400, People’s Republic of China
| | - Ying Zhao
- Central Laboratory, The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511400, People’s Republic of China
| | - Zeping Han
- Central Laboratory, The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511400, People’s Republic of China
| | - Jian Shen
- Central Laboratory, The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511400, People’s Republic of China
| | - Fangmei Xie
- Central Laboratory, The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511400, People’s Republic of China
| | - Yuguang Li
- Panyu Maternal and Children Healthcare Hospital (Hexian Memorial Medical Hospital of Panyu District), Guangzhou, Guangdong, 511400, People’s Republic of China
| | - Jinhua He
- Central Laboratory, The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511400, People’s Republic of China
- Rehabilitation Medicine Institute, The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511400, People’s Republic of China
| |
Collapse
|
4
|
Shao IH, Chang TH, Chang YH, Hsieh YH, Sheng TW, Wang LJ, Chien YH, Huang LK, Chu YC, Kan HC, Lin PH, Yu KJ, Hsieh ML, Chuang CK, Wu CT, Hsieh CH, Pang ST. Periprostatic adipose tissue inhibits tumor progression by secreting apoptotic factors: A natural barrier induced by the immune response during the early stages of prostate cancer. Oncol Lett 2024; 28:485. [PMID: 39170882 PMCID: PMC11338243 DOI: 10.3892/ol.2024.14617] [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: 11/23/2023] [Accepted: 06/27/2024] [Indexed: 08/23/2024] Open
Abstract
Prostate cancer (PCa) is the second most prevalent malignancy in men worldwide. The risk factors for PCa include obesity, age and family history. Increased visceral fat has been associated with high PCa risk, which has prompted previous researchers to investigate the influence of body composition and fat distribution on PCa prognosis. However, there is a lack of studies focusing on the mechanisms and interactions between periprostatic adipose tissue (PPAT) and PCa cells. The present study investigated the association between the composition of pelvic adipose tissue and PCa aggressiveness to understand the role played by this tissue in PCa progression. Moreover, PPAT-conditioned medium (CM) was prepared to assess the influence of the PPAT secretome on the pathophysiology of PCa. The present study included 50 patients with localized PCa who received robot-assisted radical prostatectomy. Medical records were collected, magnetic resonance imaging scans were analyzed and body compositions were calculated to identify the associations between adipose tissue volume and clinical PCa aggressiveness. In addition, CM was prepared from PPAT and perivesical adipose tissue (PVAT) collected from 25 patients during surgery, and its effects on the PCa cell lines C4-2 and LNCaP, and the prostate epithelial cell line PZ-HPV-7, were investigated using a cell proliferation assay and RNA sequencing (RNA-seq). The results revealed that the initial prostate-specific antigen level was significantly correlated with pelvic and periprostatic adipose tissue volumes. In addition, PPAT volume was significantly higher in patients with extracapsular tumor extension. PCa cell proliferation was significantly reduced when the cells were cultured in PPAT-CM compared with when they were cultured in control- and PVAT-CM. RNA-seq revealed that immune responses, and the cell death and apoptosis pathways were enriched in PPAT-CM-cultured cells indicating that the cytokines or other factors secreted from PPAT-CM induced PCa cell apoptosis. These findings revealed that the PPAT secretome may inhibit PCa cell proliferation by activating immune responses and promoting cancer cell apoptosis. This mechanism may act as a first-line defense during the early stages of PCa.
Collapse
Affiliation(s)
- I-Hung Shao
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333423, Taiwan, R.O.C
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
- Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Tzu-Hsuan Chang
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333423, Taiwan, R.O.C
| | - Ying-Hsu Chang
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
- Division of Urology, Department of Surgery, New Taipei Municipal Tucheng Hospital, New Taipei 236017, Taiwan, R.O.C
| | - Yu-Hsin Hsieh
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333423, Taiwan, R.O.C
| | - Ting-Wen Sheng
- Department of Medical Imaging and Intervention, New Taipei Municipal Tucheng Hospital, New Taipei 236017, Taiwan, R.O.C
| | - Li-Jen Wang
- Department of Medical Imaging and Intervention, New Taipei Municipal Tucheng Hospital, New Taipei 236017, Taiwan, R.O.C
| | - Yu-Hsuan Chien
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333423, Taiwan, R.O.C
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Liang-Kang Huang
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333423, Taiwan, R.O.C
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Yuan-Cheng Chu
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333423, Taiwan, R.O.C
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Hung-Cheng Kan
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333423, Taiwan, R.O.C
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Po-Hung Lin
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333423, Taiwan, R.O.C
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Kai-Jie Yu
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333423, Taiwan, R.O.C
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Ming-Li Hsieh
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333423, Taiwan, R.O.C
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Cheng-Keng Chuang
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333423, Taiwan, R.O.C
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Chun-Te Wu
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333423, Taiwan, R.O.C
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Chin-Hsuan Hsieh
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333423, Taiwan, R.O.C
| | - See-Tong Pang
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333423, Taiwan, R.O.C
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| |
Collapse
|
5
|
Seyhan AA. Circulating Liquid Biopsy Biomarkers in Glioblastoma: Advances and Challenges. Int J Mol Sci 2024; 25:7974. [PMID: 39063215 PMCID: PMC11277426 DOI: 10.3390/ijms25147974] [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: 06/12/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Gliomas, particularly glioblastoma (GBM), represent the most prevalent and aggressive tumors of the central nervous system (CNS). Despite recent treatment advancements, patient survival rates remain low. The diagnosis of GBM traditionally relies on neuroimaging methods such as magnetic resonance imaging (MRI) or computed tomography (CT) scans and postoperative confirmation via histopathological and molecular analysis. Imaging techniques struggle to differentiate between tumor progression and treatment-related changes, leading to potential misinterpretation and treatment delays. Similarly, tissue biopsies, while informative, are invasive and not suitable for monitoring ongoing treatments. These challenges have led to the emergence of liquid biopsy, particularly through blood samples, as a promising alternative for GBM diagnosis and monitoring. Presently, blood and cerebrospinal fluid (CSF) sampling offers a minimally invasive means of obtaining tumor-related information to guide therapy. The idea that blood or any biofluid tests can be used to screen many cancer types has huge potential. Tumors release various components into the bloodstream or other biofluids, including cell-free nucleic acids such as microRNAs (miRNAs), circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), proteins, extracellular vesicles (EVs) or exosomes, metabolites, and other factors. These factors have been shown to cross the blood-brain barrier (BBB), presenting an opportunity for the minimally invasive monitoring of GBM as well as for the real-time assessment of distinct genetic, epigenetic, transcriptomic, proteomic, and metabolomic changes associated with brain tumors. Despite their potential, the clinical utility of liquid biopsy-based circulating biomarkers is somewhat constrained by limitations such as the absence of standardized methodologies for blood or CSF collection, analyte extraction, analysis methods, and small cohort sizes. Additionally, tissue biopsies offer more precise insights into tumor morphology and the microenvironment. Therefore, the objective of a liquid biopsy should be to complement and enhance the diagnostic accuracy and monitoring of GBM patients by providing additional information alongside traditional tissue biopsies. Moreover, utilizing a combination of diverse biomarker types may enhance clinical effectiveness compared to solely relying on one biomarker category, potentially improving diagnostic sensitivity and specificity and addressing some of the existing limitations associated with liquid biomarkers for GBM. This review presents an overview of the latest research on circulating biomarkers found in GBM blood or CSF samples, discusses their potential as diagnostic, predictive, and prognostic indicators, and discusses associated challenges and future perspectives.
Collapse
Affiliation(s)
- Attila A. Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA;
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI 02912, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
| |
Collapse
|
6
|
Zhuang S, Semenec L, Nagy SS, Cain AK, Inglis DW. High-precision screening and sorting of double emulsion droplets. Cytometry A 2024; 105:547-554. [PMID: 38634684 DOI: 10.1002/cyto.a.24842] [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: 12/14/2023] [Revised: 03/25/2024] [Accepted: 04/04/2024] [Indexed: 04/19/2024]
Abstract
Mounting evidence suggests that cell populations are extremely heterogeneous, with individual cells fulfilling different roles within the population. Flow cytometry (FC) is a high-throughput tool for single-cell analysis that works at high optical resolution. Sub-populations with unique properties can be screened, isolated and sorted through fluorescence-activated cell sorting (FACS), using intracellular fluorescent products or surface-tagged fluorescent products of interest. However, traditional FC and FACS methods cannot identify or isolate cells that secrete extracellular products of interest. Double emulsion (DE) droplets are an innovative approach to retaining these extracellular products so cells producing them can be identified and isolated with FC and FACS. The water-in-oil-in-water structure makes DE droplets compatible with the sheath flow of flow cytometry. Single cells can be encapsulated with other reagents into DEs, which act as pico-reactors. These droplets allow biological activities to take place while allowing for cell cultivation monitoring, rare mutant identification, and cellular events characterization. However, using DEs in FACS presents technical challenges, including rupture of DEs, poor accuracy and low sorting efficiency. This study presents high-performance sorting using fluorescent beads (as simulants for cells). This study aims to guide researchers in the use of DE-based flow cytometry, offering insights into how to resolve the technical difficulties associated with DE-based screening and sorting using FC.
Collapse
Affiliation(s)
- Siyuan Zhuang
- School of Engineering, Macquarie University, Sydney, New South Wales, Australia
| | - Lucie Semenec
- School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Stephanie S Nagy
- ARC Centre of Excellence in Synthetic Biology, School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Amy K Cain
- School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - David W Inglis
- School of Engineering, Macquarie University, Sydney, New South Wales, Australia
| |
Collapse
|
7
|
Chen YT, Tu WJ, Ye ZH, Wu CC, Ueng SH, Yu KJ, Chen CL, Peng PH, Yu JS, Chang YH. Integration of the cancer cell secretome and transcriptome reveals potential noninvasive diagnostic markers for bladder cancer. Proteomics Clin Appl 2024; 18:e202300033. [PMID: 38196148 DOI: 10.1002/prca.202300033] [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: 04/01/2023] [Revised: 11/27/2023] [Accepted: 12/21/2023] [Indexed: 01/11/2024]
Abstract
PURPOSE Bladder cancer (BLCA) is a major cancer of the genitourinary system. Although cystoscopy is the standard protocol for diagnosing BLCA clinically, this procedure is invasive and expensive. Several urine-based markers for BLCA have been identified and investigated, but none has shown sufficient sensitivity and specificity. These observations underscore the importance of discovering novel BLCA biomarkers and developing a noninvasive method for detection of BLCA. Exploring the cancer secretome is a good starting point for the development of noninvasive biomarkers for cancer diagnosis. EXPERIMENTAL DESIGN In this study, we established a comprehensive secretome dataset of five representative BLCA cell lines, BFTC905, TSGH8301, 5637, MGH-U1, and MGH-U4, by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Expression of BLCA-specific secreted proteins at the transcription level was evaluated using the Oncomine cancer microarray database. RESULTS The expressions of four candidates-COMT, EWSR1, FUSIP1, and TNPO2-were further validated in clinical human specimens. Immunohistochemical analyses confirmed that transportin-2 was highly expressed in tumor cells relative to adjacent noncancerous cells in clinical tissue specimens from BLCA patients, and was significantly elevated in BLCA urine compared with that in urine samples from aged-matched hernia patients (controls). CONCLUSIONS Collectively, our findings suggest TNPO2 as a potential noninvasive tumor-stage or grade discriminator for BLCA management.
Collapse
Affiliation(s)
- Yi-Ting Chen
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Kidney Research Center, Department of Nephrology, LinKou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Wei-Ju Tu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Zong-Han Ye
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Ching Wu
- Department of Medical Biotechnology and Laboratory Science College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shir-Hwa Ueng
- Department of Anatomic Pathology, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan, Taiwan
| | - Kai-Jie Yu
- Department of Urology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chien-Lun Chen
- Department of Urology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Pei-Hua Peng
- Cancer Genome Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Jau-Song Yu
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Liver Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Ying-Hsu Chang
- Department of Urology, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
8
|
Muliawan GK, Lee TKW. The roles of cancer stem cell-derived secretory factors in shaping the immunosuppressive tumor microenvironment in hepatocellular carcinoma. Front Immunol 2024; 15:1400112. [PMID: 38868769 PMCID: PMC11167126 DOI: 10.3389/fimmu.2024.1400112] [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: 03/13/2024] [Accepted: 05/15/2024] [Indexed: 06/14/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent malignancies worldwide and has a poor prognosis. Although immune checkpoint inhibitors have entered a new era of HCC treatment, their response rates are modest, which can be attributed to the immunosuppressive tumor microenvironment within HCC tumors. Accumulating evidence has shown that tumor growth is fueled by cancer stem cells (CSCs), which contribute to therapeutic resistance to the above treatments. Given that CSCs can regulate cellular and physical factors within the tumor niche by secreting various soluble factors in a paracrine manner, there have been increasing efforts toward understanding the roles of CSC-derived secretory factors in creating an immunosuppressive tumor microenvironment. In this review, we provide an update on how these secretory factors, including growth factors, cytokines, chemokines, and exosomes, contribute to the immunosuppressive TME, which leads to immune resistance. In addition, we present current therapeutic strategies targeting CSC-derived secretory factors and describe future perspectives. In summary, a better understanding of CSC biology in the TME provides a rational therapeutic basis for combination therapy with ICIs for effective HCC treatment.
Collapse
Affiliation(s)
- Gregory Kenneth Muliawan
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Terence Kin-Wah Lee
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
9
|
Lopez-Valcarcel M, Lopez-Campos F, Zafra J, Cienfuegos I, Ferri M, Barrado M, Hernando S, Counago F. Liquid biopsy to personalize treatment for metastatic prostate cancer. Am J Transl Res 2024; 16:1531-1549. [PMID: 38883349 PMCID: PMC11170619 DOI: 10.62347/dicu9510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 04/17/2024] [Indexed: 06/18/2024]
Abstract
Liquid biopsy is an innovative approach that provides a more complete understanding of treatment response and prognosis in monitoring metastatic prostate cancer. It complements invasive tissue biopsy and involves the assessment of various biomarkers in body fluids such as blood, semen, and urine. Liquid biopsy analyzes circulating tumor cells, extracellular vesicles, circulating tumor DNA, and the secretome. This is particularly important given the heterogeneity of prostate cancer and the need for better prognostic biomarkers. Liquid biopsy can personalize the treatment of homonosensitive and castration-resistant metastatic prostate cancer by acting as a predictive and prognostic tool. This review discusses various biomarkers, assay techniques, and potential applications in daily clinical practice, highlighting the exciting possibilities that this emerging field holds for improving patient outcomes.
Collapse
Affiliation(s)
- Marta Lopez-Valcarcel
- Department of Radiation Oncology, Puerta de Hierro University Hospital Madrid, Spain
| | | | - Juan Zafra
- Department of Radiation Oncology, Virgen de la Victoria University Hospital Málaga, Spain
| | - Irene Cienfuegos
- Department of Urology, Virgen del Puerto Hospital Plasencia, Cáceres, Extremadura, Spain
| | - Maria Ferri
- Department of Radiation Oncology, Marques de Valdecilla University Hospital Santander, Cantabria, Spain
| | - Marta Barrado
- Department of Radiation Oncology, Navarra University Hospital Pamplona, Navarra, Spain
| | - Susana Hernando
- Department of Clinical Oncology, Fundación Alcorcon University Hospital Alcorcón, Madrid, Spain
| | - Felipe Counago
- Department of Radiation Oncology, GenesisCare Madrid Clinical Director, San Francisco de Asis and La Milagrosa Hospitals, National Chair of Research and Clinical Trials GenesisCare, Madrid, Spain
| |
Collapse
|
10
|
Farris F, Elhagh A, Vigorito I, Alongi N, Pisati F, Giannattasio M, Casagrande F, Veghini L, Corbo V, Tripodo C, Di Napoli A, Matafora V, Bachi A. Unveiling the mechanistic link between extracellular amyloid fibrils, mechano-signaling and YAP activation in cancer. Cell Death Dis 2024; 15:28. [PMID: 38199984 PMCID: PMC10781709 DOI: 10.1038/s41419-024-06424-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 12/20/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024]
Abstract
The tumor microenvironment is a complex ecosystem that plays a critical role in cancer progression and treatment response. Recently, extracellular amyloid fibrils have emerged as novel components of the tumor microenvironment; however, their function remains elusive. In this study, we establish a direct connection between the presence of amyloid fibrils in the secretome and the activation of YAP, a transcriptional co-activator involved in cancer proliferation and drug resistance. Furthermore, we uncover a shared mechano-signaling mechanism triggered by amyloid fibrils in both melanoma and pancreatic ductal adenocarcinoma cells. Our findings highlight the crucial role of the glycocalyx protein Agrin which binds to extracellular amyloid fibrils and acts as a necessary factor in driving amyloid-dependent YAP activation. Additionally, we reveal the involvement of the HIPPO pathway core kinase LATS1 in this signaling cascade. Finally, we demonstrate that extracellular amyloid fibrils enhance cancer cell migration and invasion. In conclusion, our research expands our knowledge of the tumor microenvironment by uncovering the role of extracellular amyloid fibrils in driving mechano-signaling and YAP activation. This knowledge opens up new avenues for developing innovative strategies to modulate YAP activation and mitigate its detrimental effects during cancer progression.
Collapse
Affiliation(s)
- Francesco Farris
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - Alice Elhagh
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - Ilaria Vigorito
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - Nicoletta Alongi
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Federica Pisati
- Histopathology Unit, Cogentech S.C.a.R.L, 20139, Milan, Italy
| | - Michele Giannattasio
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy
| | - Francesca Casagrande
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Human Technopole, Milan, Italy
| | - Lisa Veghini
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, 37134, Verona, Italy
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, 37134, Verona, Italy
- ARC-Net Centre for Applied Research on Cancer, University of Verona, 37134, Verona, Italy
| | - Claudio Tripodo
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90133, Palermo, Italy
| | - Arianna Di Napoli
- Pathology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea University Hospital, Sapienza University of Rome, 00189, Rome, Italy
| | - Vittoria Matafora
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy.
| | - Angela Bachi
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy.
| |
Collapse
|
11
|
Wang Z, Shi P, Huang P, Xu C, He Y, Lei W. Identification of secretory pathway-related genes based on Random Forest algorithm to predict the prognosis and immunotherapy response of hepatocellular carcinoma. J Gene Med 2024; 26:e3593. [PMID: 37730948 DOI: 10.1002/jgm.3593] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/31/2023] [Accepted: 08/25/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND The dysfunction of secretory pathways may represent biomarkers or therapeutic targets of cancer. The hepatocellular carcinoma (HCC) phenotype was studied in relation to the genes in the secretory pathway and to screen for a combination of genes that may be a viable therapeutic target for HCC and connected to the pathophysiological features of the tumor. METHODS Using the HCC information from The Cancer Genome Atlas, somatic mutation and prognostic association analysis were performed on the secretory pathway genes. Based on prognostic genes in the secretory pathway, the samples were consensus clustered, and a Random Forest model was built. The clinical characteristics, tumor mutation burden, functional status and potential responses to immunotherapy and tumor suppressor medications of various subtypes and risk groups were discussed. RESULTS Of the 84 genes for secretory pathway, 32 were prognostic genes related to HCC, which divided HCC into two categories: C1 and C2. By comparing the two types of HCC samples, it was found that the survival outcome of C1 was inferior, with stronger adaptive and innate immunity, but less sensitive to immunotherapy than C2. The constructed prognostic signature included seven of the 32 prognostic genes in the secretory pathway, which showed significant correlation with the prognosis, somatic mutation, biological pathway status, potential response to immunotherapy and sensitivity of 72 tumor suppressor drugs from different HCC cohorts, and had a feasible prognostic effect for 31 types of cancer and immunotherapy cohorts. CONCLUSIONS In this study, HCC was divided into two molecular subtypes according to prognostic genes in the secretory pathway, and seven of them were combined into one signature, which produced significant results in evaluating the prognosis of different HCC cohorts, pan-cancer cohorts and immunotherapy cohorts, and had potential guiding significance for prophylactic immunotherapy in patients with HCC.
Collapse
Affiliation(s)
- Zanzhi Wang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pengwei Shi
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Huang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chun Xu
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yaoquan He
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenxiong Lei
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
12
|
Tay AHM, Cinotti R, Sze NSK, Lundqvist A. Inhibition of ERO1a and IDO1 improves dendritic cell infiltration into pancreatic ductal adenocarcinoma. Front Immunol 2023; 14:1264012. [PMID: 38187398 PMCID: PMC10766682 DOI: 10.3389/fimmu.2023.1264012] [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: 07/20/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024] Open
Abstract
Introduction Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal and treatment resistant cancers. Due to its desmoplastic and hypoxic nature along with an abundance of myeloid cell infiltration and scarce T cell infiltration, PDAC is considered a cold tumor. Methods Here we sought to investigate myeloid cell infiltration and composition in PDAC spheroids by targeting the hypoxia-associated pathways endoplasmic reticulum oxidoreductase 1 alpha (ERO1a) and indoleamine 2,3-dioxygenase 1 (IDO1). Using MiaPaCa2 spheroids with hypoxic core, we assessed the roles of ERO1a and IDO1 inhibition in modulating monocyte infiltration and differentiation, followed by characterizing immunomodulatory factors secreted using LC-MS/MS. Results Inhibition of ERO1a and IDO1 significantly improved monocyte infiltration and differentiation into dendritic cells. LC-MS/MS analysis of the PDAC spheroid secretome identified downregulation of hypoxia and PDAC pathways, and upregulation of antigen presentation pathways upon inhibition of ERO1a and IDO1. Furthermore, immunomodulatory factors involved in immune infiltration and migration including interleukin-8, lymphocyte cytosolic protein 1, and transgelin-2, were upregulated upon inhibition of ERO1a and IDO1. Discussion Collectively, our results show that inhibition of ERO1a and IDO1 modulates the tumor microenvironment associated with improved monocyte infiltration and differentiation into dendritic cells to potentially influence therapeutic responses in patients with PDAC.
Collapse
Affiliation(s)
- Apple Hui Min Tay
- School of Biological Science, Nanyang Technological University, Singapore, Singapore
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Riccardo Cinotti
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Newman Sui Kwan Sze
- School of Biological Science, Nanyang Technological University, Singapore, Singapore
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON, Canada
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
13
|
Bersano J, Lashuk K, Edinger A, Schueler J. A Subset of Colon Cancer Cell Lines Displays a Cytokine Profile Linked to Angiogenesis, EMT and Invasion Which Is Modulated by the Culture Conditions In Vitro. Cells 2023; 12:2539. [PMID: 37947617 PMCID: PMC10648033 DOI: 10.3390/cells12212539] [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: 09/26/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Colorectal cancer (CRC) is one of the deadliest cancers worldwide. The dysregulation of secretory pathways is a crucial driver of CRC progression, since it modulates cell proliferation, angiogenesis and survival. This study explores the changes in the CRC cytokine profile depending on the culture conditions and the presence of fibroblasts and macrophages as cellular components of the tumor microenvironment in 2D and in 3D formed spheroids. Upon analysis of 45 different cytokines, chemokines and growth factors, 20 CRC cell lines were categorized into high and low secretors. In the high secretor group cytokines related to angiogenesis, EMT and invasion were significantly upregulated. LIF and HFG were identified as the best discriminator between both groups. Independent of this grouping, the addition of normal as well as cancer-associated fibroblasts had a similar impact on the cytokine profile by increasing the total amount of secreted cytokines in most of the investigated cell lines. In contrast, the differentiation and polarization of macrophages was modulated differently by normal vs. cancer-associated fibroblasts. In summary, we identified two groups of CRC cell lines that differ in their cytokine profile. The dependance of this profile was analyzed in detail-not only from the tumor cell line but as well from the culture condition in vitro. Key cytokines that discriminate the two groups were identified and their importance as promising biomarker candidates for CRC discussed.
Collapse
Affiliation(s)
| | | | | | - Julia Schueler
- Charles River Discovery Research Services Germany GmbH, Am Flughafen 12–14, 79108 Freiburg, Germany; (J.B.); (K.L.); (A.E.)
| |
Collapse
|
14
|
Liu D, Paladino S, Zurzolo C, Lebreton S. Calcium-binding Cab45 regulates the polarized apical secretion of soluble proteins in epithelial cells. Mol Biol Cell 2023; 34:br12. [PMID: 37163315 PMCID: PMC10398874 DOI: 10.1091/mbc.e22-12-0549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/12/2023] [Accepted: 05/02/2023] [Indexed: 05/11/2023] Open
Abstract
Protein secretion is essential for epithelial tissue homoeostasis and therefore has to be tightly regulated. However, while the mechanisms regulating polarized protein sorting and trafficking have been widely studied in the past decade, those governing polarized secretion remain elusive. The calcium manganese pump SPCA1 and the calcium-binding protein Cab45 were recently shown to regulate the secretion of a subset of soluble cargoes in nonpolarized HeLa cells. Interestingly, we demonstrated that in polarized epithelial cells calcium levels in the trans-Golgi network (TGN), controlled by SPCA1, and Cab45 are critical for the apical sorting of glycosylphosphatidylinositol-anchored proteins (GPI-APs), a class of integral membrane proteins containing a soluble protein attached to the membrane by the GPI anchor, prompting us to investigate the mechanism regulating the polarized secretion of soluble cargoes. By reducing Cab45 expression level or overexpressing an inactive mutant of SPCA1, we found that Cab45 and calcium levels in the TGN drive the polarized apical secretion of a secretory form of placental alkaline phosphatase, exogenously expressed, and the endogenous soluble protein clusterin/Gp80 in Madin-Darby canine kidney (MDCK) cells. These data highlight the critical role of a calcium-dependent Cab45 mechanism regulating apical exocytosis in polarized MDCK cells.
Collapse
Affiliation(s)
- Dandan Liu
- Unité de Trafic Membranaire et Pathogenèse, Institut Pasteur, 75015 Paris, France
| | - Simona Paladino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Chiara Zurzolo
- Unité de Trafic Membranaire et Pathogenèse, Institut Pasteur, 75015 Paris, France
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Stéphanie Lebreton
- Unité de Trafic Membranaire et Pathogenèse, Institut Pasteur, 75015 Paris, France
| |
Collapse
|
15
|
Abuhamad AY, Mohamad Zamberi NN, Vanharanta S, Mohd Yusuf SNH, Mohtar MA, Syafruddin SE. Cancer Cell-Derived PDGFB Stimulates mTORC1 Activation in Renal Carcinoma. Int J Mol Sci 2023; 24:ijms24076447. [PMID: 37047421 PMCID: PMC10095210 DOI: 10.3390/ijms24076447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is a hypervascular tumor that is characterized by bi-allelic inactivation of the VHL tumor suppressor gene and mTOR signalling pathway hyperactivation. The pro-angiogenic factor PDGFB, a transcriptional target of super enhancer-driven KLF6, can activate the mTORC1 signalling pathway in ccRCC. However, the detailed mechanisms of PDGFB-mediated mTORC1 activation in ccRCC have remained elusive. Here, we investigated whether ccRCC cells are able to secrete PDGFB into the extracellular milieu and stimulate mTORC1 signalling activity. We found that ccRCC cells secreted PDGFB extracellularly, and by utilizing KLF6- and PDGFB-engineered ccRCC cells, we showed that the level of PDGFB secretion was positively correlated with the expression of intracellular KLF6 and PDGFB. Moreover, the reintroduction of either KLF6 or PDGFB was able to sustain mTORC1 signalling activity in KLF6-targeted ccRCC cells. We further demonstrated that conditioned media of PDGFB-overexpressing ccRCC cells was able to re-activate mTORC1 activity in KLF6-targeted cells. In conclusion, cancer cell-derived PDGFB can mediate mTORC1 signalling pathway activation in ccRCC, further consolidating the link between the KLF6-PDGFB axis and the mTORC1 signalling pathway activity in ccRCC.
Collapse
Affiliation(s)
- Asmaa Y. Abuhamad
- Bionanotechnology Research Group, Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Nurul Nadia Mohamad Zamberi
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| | - Sakari Vanharanta
- Department of Physiology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Siti Nur Hasanah Mohd Yusuf
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| | - M. Aiman Mohtar
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| | - Saiful Effendi Syafruddin
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
- Correspondence:
| |
Collapse
|
16
|
Secretome of Stromal Cancer-Associated Fibroblasts (CAFs): Relevance in Cancer. Cells 2023; 12:cells12040628. [PMID: 36831295 PMCID: PMC9953839 DOI: 10.3390/cells12040628] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
The cancer secretome reflects the assortment of proteins released by cancer cells. Investigating cell secretomes not only provides a deeper knowledge of the healthy and transformed state but also helps in the discovery of novel biomarkers. Secretomes of cancer cells have been studied in the past, however, the secretome contribution of stromal cells needs to be studied. Cancer-associated fibroblasts (CAFs) are one of the predominantly present cell populations in the tumor microenvironment (TME). CAFs play key role in functions associated with matrix deposition and remodeling, reciprocal exchange of nutrients, and molecular interactions and signaling with neighboring cells in the TME. Investigating CAFs secretomes or CAFs-secreted factors would help in identifying novel CAF-specific biomarkers, unique druggable targets, and an improved understanding for personalized cancer diagnosis and prognosis. In this review, we have tried to include all studies available in PubMed with the keywords "CAFs Secretome". We aim to provide a comprehensive summary of the studies investigating role of the CAF secretome on cancer development, progression, and therapeutic outcome. However, challenges associated with this process have also been addressed in the later sections. We have highlighted the functions and clinical relevance of secretome analysis in stromal CAF-rich cancer types. This review specifically discusses the secretome of stromal CAFs in cancers. A deeper understanding of the components of the CAF secretome and their interactions with cancer cells will help in the identification of personalized biomarkers and a more precise treatment plan.
Collapse
|
17
|
Selvaraj C, Panwar U, Ramalingam KR, Vijayakumar R, Singh SK. Exploring the macromolecules for secretory pathway in cancer disease. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 133:55-83. [PMID: 36707206 DOI: 10.1016/bs.apcsb.2022.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Secretory proteins play an important role in the tumor microenvironment and are widely distributed throughout tumor tissues. Tumor cells secrete a protein that mediates communication between tumor cells and stromal cells, thereby controlling tumor growth and affecting the success of cancer treatments in the clinic. The cancer secretome is produced by various secretory pathways and has a wide range of applications in oncoproteomics. Secretory proteins are involved in cancer development and tumor cell migration, and thus serve as biomarkers or effective therapeutic targets for a variety of cancers. Several proteomic strategies have recently been used for the analysis of cancer secretomes in order to gain a better understanding and elaborate interpretation. For instance, the development of exosome proteomics, degradomics, and tumor-host cell interaction provide clear information regarding the mechanism of cancer pathobiology. In this chapter, we emphasize the recent advances in secretory protein and the challenges in the field of secretome analysis and their clinical applications.
Collapse
Affiliation(s)
- Chandrabose Selvaraj
- Computer Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, Tamil Nadu, India.
| | - Umesh Panwar
- Computer Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Karthik Raja Ramalingam
- Department of Biotechnology, Division of Research and Innovation, Saveetha School of Engineering, SIMATS, Chennai, Tamil Nadu, India
| | - Rajendran Vijayakumar
- Department of Biology, College of Science in Zulfi, Majmaah University, Majmaah, Saudi Arabia
| | - Sanjeev Kumar Singh
- Computer Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, Tamil Nadu, India.
| |
Collapse
|
18
|
Zhuang S, Liu H, Inglis DW, Li M. Tuneable Cell-Laden Double-Emulsion Droplets for Enhanced Signal Detection. Anal Chem 2023; 95:2039-2046. [PMID: 36634052 DOI: 10.1021/acs.analchem.2c04697] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Water-in-oil-in-water (w/o/w) or double-emulsion (DE) droplets have been widely used for cellular assays at a single-cell level because of their stability and biocompatibility. The oil shell of w/o/w droplets plays the role of a semipermeable membrane that allows substances with low molecular weight (e.g., water) to travel through but restricts those with high molecular weight (e.g., fluorescent biomarkers). Therefore, the core of DEs can be manipulated using osmosis, resulting in the shrinking or swelling of the core. Water leaves the inner aqueous phase to the outer phase via the oil shell when the osmotic pressure of the outer phase is higher than that in the inner phase, causing the shrinkage of DEs and vice versa. These processes can be achieved by transferring the DEs to hypertonic or hypotonic solutions. Manipulation of the core size of DEs can be beneficial to cellular assays. First, due to the selectivity of the oil shell of DEs, the concentration of biomarkers in the core increases when the inner aqueous phase is shrunk, resulting in the enhancement of biosignals. We demonstrate this by encapsulating the Bgl3 enzyme-secreting yeast with a substrate that displays fluorescence after hydrolyzation. In a second application, a single GFP-tagged yeast cell was encapsulated in DEs. After swelling the core of DEs, we observe that the larger core of DEs promotes cell growth compared to those with the smaller cores, leading to more intracellular proteins (green-fluorescent protein) for screening. These osmotic manipulations provide new tools for droplet-based biochemistry.
Collapse
Affiliation(s)
- Siyuan Zhuang
- School of Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Hangrui Liu
- Department of Physics and Astronomy, Macquarie University, Sydney, New South Wales 2109, Australia
| | - David W Inglis
- School of Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Ming Li
- School of Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
- Biomolecular Discovery Research Centre, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
19
|
Dijkstra J, Neikes HK, Rezaeifard S, Ma X, Voest EE, Tauriello DVF, Vermeulen M. Multiomics of Colorectal Cancer Organoids Reveals Putative Mediators of Cancer Progression Resulting from SMAD4 Inactivation. J Proteome Res 2023; 22:138-151. [PMID: 36450103 PMCID: PMC9830641 DOI: 10.1021/acs.jproteome.2c00551] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
The development of metastasis severely reduces the life expectancy of patients with colorectal cancer (CRC). Although loss of SMAD4 is a key event in CRC progression, the resulting changes in biological processes in advanced disease and metastasis are not fully understood. Here, we applied a multiomics approach to a CRC organoid model that faithfully reflects the metastasis-supporting effects of SMAD4 inactivation. We show that loss of SMAD4 results in decreased differentiation and activation of pro-migratory and cell proliferation processes, which is accompanied by the disruption of several key oncogenic pathways, including the TGFβ, WNT, and VEGF pathways. In addition, SMAD4 inactivation leads to increased secretion of proteins that are known to be involved in a variety of pro-metastatic processes. Finally, we show that one of the factors that is specifically secreted by SMAD4-mutant organoids─DKK3─reduces the antitumor effects of natural killer cells (NK cells). Altogether, our data provide new insights into the role of SMAD4 perturbation in advanced CRC.
Collapse
Affiliation(s)
- Jelmer
J. Dijkstra
- Department
of Molecular Biology, Faculty of Science, Radboud Institute for Molecular
Life Sciences (RIMLS), Oncode Institute, Radboud University Nijmegen, Geert Grooteplein 26−28, 6525
GA Nijmegen, The
Netherlands
| | - Hannah K. Neikes
- Department
of Molecular Biology, Faculty of Science, Radboud Institute for Molecular
Life Sciences (RIMLS), Oncode Institute, Radboud University Nijmegen, Geert Grooteplein 26−28, 6525
GA Nijmegen, The
Netherlands
| | - Somayeh Rezaeifard
- Department
of Cell Biology, Radboud University Medical Center/Radboud Institute
for Molecular Life Sciences (RIMLS), Radboud
University Nijmegen, Geert Grooteplein 26−28, 6525
GA Nijmegen, The
Netherlands
| | - Xuhui Ma
- Department
of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Antoni van Leeuwenhoek
Hospital, 1066 CX Amsterdam, The Netherlands
| | - Emile E. Voest
- Department
of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Antoni van Leeuwenhoek
Hospital, 1066 CX Amsterdam, The Netherlands
| | - Daniele V. F. Tauriello
- Department
of Cell Biology, Radboud University Medical Center/Radboud Institute
for Molecular Life Sciences (RIMLS), Radboud
University Nijmegen, Geert Grooteplein 26−28, 6525
GA Nijmegen, The
Netherlands
| | - Michiel Vermeulen
- Department
of Molecular Biology, Faculty of Science, Radboud Institute for Molecular
Life Sciences (RIMLS), Oncode Institute, Radboud University Nijmegen, Geert Grooteplein 26−28, 6525
GA Nijmegen, The
Netherlands,
| |
Collapse
|
20
|
Quesnel A, Broughton A, Karagiannis GS, Filippou PS. Message in the bottle: regulation of the tumor microenvironment via exosome-driven proteolysis. Cancer Metastasis Rev 2022; 41:789-801. [PMID: 35394580 DOI: 10.1007/s10555-022-10030-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/29/2022] [Indexed: 01/25/2023]
Abstract
Exosomes comprise a subtype of extracellular vesicles involved in cell-to-cell communication, specifically by transporting biological molecules, such as proteins and nucleic acids, to either local or more distant recipient cells, thus triggering distinct biological behaviors. Included in the exosome cargo is frequently a wide range of proteolytic enzymes, such as the matrix metalloproteinases (MMPs), the disintegrin and metalloproteinases (ADAMs), and the ADAM with thrombospondin-like motifs (ADAMTSs), whose functions contribute to the development and progression of cancer. In recent years, extensive research on the potential use of exosomes in diagnostic and therapeutic applications for personalized medicine has emerged, but the targeting of the proteolytic cargo of exosomes has not been fully exploited in this direction. In this review, we aim to explore both the mechanistic and the translational importance of proteolytic enzymes carried by the tumor cell-derived exosomes, as well as their role in the acquisition and support of certain hallmarks of cancer.
Collapse
Affiliation(s)
- Agathe Quesnel
- School of Health & Life Sciences, Teesside University, Middlesbrough, TS1 3BX, UK.,National Horizons Centre, Teesside University, Darlington, DL1 1HG, UK
| | - Amy Broughton
- School of Health & Life Sciences, Teesside University, Middlesbrough, TS1 3BX, UK
| | - George S Karagiannis
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.,Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA.,Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY, USA.,Albert Einstein Cancer Center, Tumor Microenvironment and Metastasis Program, Bronx, NY, USA
| | - Panagiota S Filippou
- School of Health & Life Sciences, Teesside University, Middlesbrough, TS1 3BX, UK. .,National Horizons Centre, Teesside University, Darlington, DL1 1HG, UK.
| |
Collapse
|
21
|
Circulating proteins as predictive and prognostic biomarkers in breast cancer. Clin Proteomics 2022; 19:25. [PMID: 35818030 PMCID: PMC9275040 DOI: 10.1186/s12014-022-09362-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 06/28/2022] [Indexed: 11/22/2022] Open
Abstract
Breast cancer (BC) is the most common cancer and among the leading causes of cancer death in women. It is a heterogeneous group of tumours with numerous morphological and molecular subtypes, making predictions of disease evolution and patient outcomes difficult. Therefore, biomarkers are needed to help clinicians choose the best treatment for each patient. For the last years, studies have increasingly focused on biomarkers obtainable by liquid biopsy. Circulating proteins (from serum or plasma) can be used for inexpensive and minimally invasive determination of disease risk, early diagnosis, treatment adjusting, prognostication and disease progression monitoring. We provide here a review of the main published studies on serum proteins in breast cancer and elaborate on the potential of circulating proteins to be predictive and/or prognostic biomarkers in breast cancer.
Collapse
|
22
|
Rehman AU, Olsson PO, Akhtar A, Padhiar AA, Liu H, Dai Y, Gong Y, Zhou Y, Khan N, Yang H, Tang L. Systematic molecular analysis of the human secretome and membrane proteome in gastrointestinal adenocarcinomas. J Cell Mol Med 2022; 26:3329-3342. [PMID: 35488454 PMCID: PMC9189341 DOI: 10.1111/jcmm.17338] [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: 10/12/2021] [Revised: 03/12/2022] [Accepted: 04/09/2022] [Indexed: 11/27/2022] Open
Abstract
The human secretome and membrane proteome are a large source of cancer biomarkers. Membrane‐bound and secreted proteins are promising targets for many clinically approved drugs, including for the treatment of tumours. Here, we report a deep systematic analysis of 957 adenocarcinomas of the oesophagus, stomach, colon and rectum to examine the cancer‐associated human secretome and membrane proteome of gastrointestinal tract adenocarcinomas (GIACs). Transcriptomic data from these GIACs were applied to an innovative majority decision‐based algorithm. We quantified significantly expressed protein‐coding genes. Interestingly, we found a consistent pattern in a small group of genes found to be overexpressed in GIACs, which were associated with a cytokine–cytokine interaction pathway (CCRI) in all four cancer subtypes. These CCRI associated genes, which spanned both one secretory and one membrane isoform were further analysed, revealing a putative biomarker, interleukin‐1 receptor accessory protein (IL1RAP), which indicated a poor overall survival, a positive correlation with cancer stemness and a negative correlation with several kinds of T cells. These results were further validated in vitro through the knockdown of IL1RAP in two human gastric carcinoma cell lines, which resulted in a reduced indication of cellular proliferation, migration and markers of invasiveness. Following IL1RAP silencing, RNA seq results showed a consistent pattern of inhibition related to CCRI, proliferation pathways and low infiltration of regulatory T cells (Tregs) and CD8 naive cells. The significance of the human secretome and membrane proteome is elucidated by these findings, which indicate IL1RAP as a potential candidate biomarker for cytokine‐mediated cancer immunotherapy in gastric carcinoma.
Collapse
Affiliation(s)
- Adeel Ur Rehman
- Department of General Surgery, Changzhou No. 2 People's Hospital affiliated with Nanjing Medical University, Changzhou, China
| | - Per Olof Olsson
- UAE Biotech Research Center, Abu Dhabi, United Arab Emirates
| | | | - Arshad Ahmed Padhiar
- Department of Ecology and Evolutionary Biology, University of Connecticut, Storrs, Connecticut, USA
| | - Hanyang Liu
- Charité-University Medical Center, Department of Hematology, Oncology and Tumor Immunology, Virchow Campus, and Molecular Cancer Research Center, Berlin, Germany
| | - Yi Dai
- Department of General Surgery, Changzhou No. 2 People's Hospital affiliated with Nanjing Medical University, Changzhou, China
| | - Yu Gong
- Department of General Surgery, Changzhou No. 2 People's Hospital affiliated with Nanjing Medical University, Changzhou, China
| | - Yan Zhou
- Department of General Surgery, Changzhou No. 2 People's Hospital affiliated with Nanjing Medical University, Changzhou, China
| | - Naveed Khan
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Biological Sciences, University of Chinese Academy of Science, Chinese Academy of Science, Shanghai, China
| | - Haojun Yang
- Department of General Surgery, Changzhou No. 2 People's Hospital affiliated with Nanjing Medical University, Changzhou, China
| | - Liming Tang
- Department of General Surgery, Changzhou No. 2 People's Hospital affiliated with Nanjing Medical University, Changzhou, China
| |
Collapse
|
23
|
Bajaj R, Warner AN, Fradette JF, Gibbons DL. Dance of The Golgi: Understanding Golgi Dynamics in Cancer Metastasis. Cells 2022; 11:1484. [PMID: 35563790 PMCID: PMC9102947 DOI: 10.3390/cells11091484] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 12/17/2022] Open
Abstract
The Golgi apparatus is at the center of protein processing and trafficking in normal cells. Under pathological conditions, such as in cancer, aberrant Golgi dynamics alter the tumor microenvironment and the immune landscape, which enhances the invasive and metastatic potential of cancer cells. Among these changes in the Golgi in cancer include altered Golgi orientation and morphology that contribute to atypical Golgi function in protein trafficking, post-translational modification, and exocytosis. Golgi-associated gene mutations are ubiquitous across most cancers and are responsible for modifying Golgi function to become pro-metastatic. The pharmacological targeting of the Golgi or its associated genes has been difficult in the clinic; thus, studying the Golgi and its role in cancer is critical to developing novel therapeutic agents that limit cancer progression and metastasis. In this review, we aim to discuss how disrupted Golgi function in cancer cells promotes invasion and metastasis.
Collapse
Affiliation(s)
- Rakhee Bajaj
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.B.); (A.N.W.); (J.F.F.)
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Amanda N. Warner
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.B.); (A.N.W.); (J.F.F.)
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Jared F. Fradette
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.B.); (A.N.W.); (J.F.F.)
| | - Don L. Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.B.); (A.N.W.); (J.F.F.)
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
24
|
Aung TM, Ciin MN, Silsirivanit A, Jusakul A, Lert-Itthiporn W, Proungvitaya T, Roytrakul S, Proungvitaya S. Serum Angiopoietin-Like Protein 4: A Potential Prognostic Biomarker for Prediction of Vascular Invasion and Lymph Node Metastasis in Cholangiocarcinoma Patients. Front Public Health 2022; 10:836985. [PMID: 35392474 PMCID: PMC8980351 DOI: 10.3389/fpubh.2022.836985] [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: 12/16/2021] [Accepted: 02/23/2022] [Indexed: 11/26/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a tumor arising from cholangiocytes lining the bile ducts. Vascular invasion and lymph node metastasis are important prognostic factors for disease staging as well as clinical therapeutic decisions for CCA patients. In the present study, we applied CCA sera proteomic analysis to identify a potential biomarker for prognosis of CCA patients. Then, using bioinformatics tools, we identified angiopoietin-like protein 4 (ANGPTL4) which expressed highest signal intensity among candidate proteins in proteomic analysis of CCA sera. Expression of ANGPTL4 in CCA tissues was determined using immunohistochemistry. The results showed that ANGPTL4 was stained at higher level in CCA cells when compared with normal cholangiocytes. The high expression of ANGPTL4 was associated with lymph node metastasis and advanced tumor stage (p = 0.013 and p = 0.031, respectively). Furthermore, serum ANGPTL4 levels in CCA and healthy control (HC) were analyzed using a dot blot assay. And it was found that ANGPTL4 level was significantly higher in CCA than HC group (p < 0.0001). ROC curve analysis revealed that serum ANGPTL4 level was effectively distinguished CCA from healthy patients (cutoff = 0.2697 arbitrary unit (AU), 80.0% sensitivity, 72.7% specificity, AUC = 0.825, p < 0.0001). Serum ANGPTL4 level was associated with vascular invasion and lymph node metastasis (p = 0.0004 and p = 0.006), so that it differentiated CCA with vascular invasion from CCA without vascular invasion (cutoff = 0.5526 AU, 64.9% sensitivity, 92.9% specificity, AUC = 0.751, p = 0.006) and it corresponded to CCA with/without lymph node metastasis (cutoff = 0.5399 AU, 71.4% sensitivity, 70.8% specificity, AUC = 0.691, p = 0.01) by ROC analysis. Serum ANGPTL4 levels showed superior predictive efficiency compared with CA 19-9 and CEA for vascular invasion and lymph node metastasis. In addition, serum ANGPTL4 level was an independent predictive indicator by multivariate regression analysis. In conclusion, serum ANGPTL4 could be a novel prognostic biomarker for prediction of vascular invasion and lymph node metastasis of CCA patients.
Collapse
Affiliation(s)
- Tin May Aung
- Faculty of Associated Medical Sciences, Centre of Research and Development of Medical Diagnostic Laboratories (CMDL), Khon Kaen University, Khon Kaen, Thailand
| | - Mang Ngaih Ciin
- Faculty of Associated Medical Sciences, Centre of Research and Development of Medical Diagnostic Laboratories (CMDL), Khon Kaen University, Khon Kaen, Thailand
| | - Atit Silsirivanit
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand.,Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Apinya Jusakul
- Faculty of Associated Medical Sciences, Centre of Research and Development of Medical Diagnostic Laboratories (CMDL), Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Worachart Lert-Itthiporn
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand.,Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Tanakorn Proungvitaya
- Faculty of Associated Medical Sciences, Centre of Research and Development of Medical Diagnostic Laboratories (CMDL), Khon Kaen University, Khon Kaen, Thailand
| | - Sittiruk Roytrakul
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Siriporn Proungvitaya
- Faculty of Associated Medical Sciences, Centre of Research and Development of Medical Diagnostic Laboratories (CMDL), Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
25
|
Butera G, Manfredi M, Fiore A, Brandi J, Pacchiana R, De Giorgis V, Barberis E, Vanella V, Galasso M, Scupoli MT, Marengo E, Cecconi D, Donadelli M. Tumor Suppressor Role of Wild-Type P53-Dependent Secretome and Its Proteomic Identification in PDAC. Biomolecules 2022; 12:305. [PMID: 35204804 PMCID: PMC8869417 DOI: 10.3390/biom12020305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/03/2022] [Accepted: 02/09/2022] [Indexed: 12/10/2022] Open
Abstract
The study of the cancer secretome is gaining even more importance in cancers such as pancreatic ductal adenocarcinoma (PDAC), whose lack of recognizable symptoms and early detection assays make this type of cancer highly lethal. The wild-type p53 protein, frequently mutated in PDAC, prevents tumorigenesis by regulating a plethora of signaling pathways. The importance of the p53 tumor suppressive activity is not only primarily involved within cells to limit tumor cell proliferation but also in the extracellular space. Thus, loss of p53 has a profound impact on the secretome composition of cancer cells and marks the transition to invasiveness. Here, we demonstrate the tumor suppressive role of wild-type p53 on cancer cell secretome, showing the anti-proliferative, apoptotic and chemosensitivity effects of wild-type p53 driven conditioned medium. By using high-resolution SWATH-MS technology, we characterized the secretomes of p53-deficient and p53-expressing PDAC cells. We found a great number of secreted proteins that have known roles in cancer-related processes, 30 of which showed enhanced and 17 reduced secretion in response to p53 silencing. These results are important to advance our understanding on the link between wt-p53 and cancer microenvironment. In conclusion, this approach may detect a secreted signature specifically driven by wild-type p53 in PDAC.
Collapse
Affiliation(s)
- Giovanna Butera
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
| | - Marcello Manfredi
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.M.); (V.D.G.); (E.B.); (V.V.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy;
- ISALIT, Spin-off at the University of Piemonte Orientale, 28100 Novara, Italy
| | - Alessandra Fiore
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
| | - Jessica Brandi
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (J.B.); (D.C.)
| | - Raffaella Pacchiana
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
| | - Veronica De Giorgis
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.M.); (V.D.G.); (E.B.); (V.V.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy;
| | - Elettra Barberis
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.M.); (V.D.G.); (E.B.); (V.V.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy;
- ISALIT, Spin-off at the University of Piemonte Orientale, 28100 Novara, Italy
| | - Virginia Vanella
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.M.); (V.D.G.); (E.B.); (V.V.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy;
| | - Marilisa Galasso
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
- Department of Medicine, Section of Hematology, University of Verona, 37134 Verona, Italy
| | - Maria Teresa Scupoli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
- Research Center LURM, Interdepartmental Laboratory of Medical Research, University of Verona, 37134 Verona, Italy
| | - Emilio Marengo
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy;
- ISALIT, Spin-off at the University of Piemonte Orientale, 28100 Novara, Italy
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 28100 Novara, Italy
| | - Daniela Cecconi
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (J.B.); (D.C.)
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
| |
Collapse
|
26
|
In Vitro Neuroprotective Effect of the Bovine Umbilical Vein Endothelial Cell Conditioned Medium Mediated by Downregulation of IL-1β, Caspase-3, and Caspase-9 Expression. Vet Sci 2022; 9:vetsci9020048. [PMID: 35202301 PMCID: PMC8878894 DOI: 10.3390/vetsci9020048] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 02/04/2023] Open
Abstract
Mesenchymal stem cells (MSCs) and conditioned medium (CM) derived from human umbilical blood cord stem cells (HUBSC) are now being extensively utilized. Human umbilical vein endothelial cells (HUVECs) have the same ability as HUBSC as an option for autologous therapy. In addition, cell therapy using HUVECs may produce protective signals for cerebral vessels and promote neuronal survival after hypoxic–ischemic damage. HUVECs have the same anatomical and physiological structure as bovine umbilical vein endothelial cells (BUVECs). In this study, we aim to determine the ability of BUVEC-CM to reduce inflammation and apoptosis on in vitro neurodegeneration models (PC12 and SH-SY5Y cell lines). BUVEC-CM obtained from the third and fourth passages were analyzed using liquid chromatography–mass spectrometry (LC-MS) and high-resolution mass spectrometry (HR-MS), while the other part was used as a treatment for in vitro model neurodegeneration. The PC12 and SH-SY5Y cell lines were cultured and grouped into seven different treatments, including untreated cells. As the treatment group, cells were given TMT 10 µM in the presence of different doses of CM (25%, 50%, 75%, and 100%); as a control comparison of recent therapy, donepezil was used. In addition, cells with the administration of TMT 10 µM were run as a positive control. Cell viability assay (CCK-8) and enzyme-linked immunosorbent assay (ELISA) were performed to identify the viability and expression of interleukin-1β (IL-1β), caspase-3, and caspase-9 for both PC12 and SH-SY5Y cells. The results showed that BUVEC-CM could significantly reduce IL-1β expression and downregulate caspase-3 and caspase-9, as well as when compared to the donepezil group. Taken together, these results indicate that BUVEC-CM can be used as a potential candidate for neuroprotective agents by reducing the activity of IL-1β and the expression of caspase-9 and caspase-3 in PC12 and SH-SY5Y cells induced by TMT. However, further research still needs to be conducted.
Collapse
|
27
|
Kamble PR, Breed AA, Pawar A, Kasle G, Pathak BR. Prognostic utility of the ovarian cancer secretome: a systematic investigation. Arch Gynecol Obstet 2022; 306:639-662. [PMID: 35083554 DOI: 10.1007/s00404-021-06361-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 12/06/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND Ovarian cancer is usually detected at an advanced stage with frequent recurrence. The recurrence-free survival and overall survival is influenced by the age at diagnosis, tumor stage and histological subtype. Nonetheless, quantifiable prognostic biomarkers are needed for early identification of the high-risk patients and for personalized medicine. Several studies link tumor-specific dysregulated expression of certain proteins with ovarian cancer prognosis. However, careful investigation of presence of these prognostically relevant proteins in ovarian cancer secretome is lacking. OBJECTIVE To critically analyze the recent published data on prognostically relevant proteins for ovarian cancer and to carefully search how many of them are reported in the published ovarian cancer secretome datasets. DESIGN A search for relevant studies in the past 2 years was conducted in PubMed and a comprehensive list of proteins associated with the ovarian cancer prognosis was prepared. These were cross-referred to the published ovarian cancer secretome profiles. The proteins identified in the secretome were further shortlisted based on a scoring strategy employing stringent criteria. RESULTS A panel of seven promising secretory biomarkers associated with ovarian cancer prognosis is proposed. CONCLUSION Scanning the ovarian cancer secretome datasets provides the opportunity to identify if tumor-specific biomarkers could be tested as secretory biomarkers. Detecting their levels in the body fluid would be more advantageous than evaluating the expression in the tissue, since it could be monitored multiple times over the course of the disease to have a better judgment of the prognosis and response to therapy.
Collapse
Affiliation(s)
- Pradnya R Kamble
- Cellular and Structural Biology Division, National Institute for Research in Reproductive Health (ICMR), Jehangir Merwanji Street, Parel, Mumbai, 400012, India
| | - Ananya A Breed
- Cellular and Structural Biology Division, National Institute for Research in Reproductive Health (ICMR), Jehangir Merwanji Street, Parel, Mumbai, 400012, India
| | - Apoorva Pawar
- Cellular and Structural Biology Division, National Institute for Research in Reproductive Health (ICMR), Jehangir Merwanji Street, Parel, Mumbai, 400012, India
| | - Grishma Kasle
- Cellular and Structural Biology Division, National Institute for Research in Reproductive Health (ICMR), Jehangir Merwanji Street, Parel, Mumbai, 400012, India
- Division of Biological Sciences, IISER, Kolkata, India
| | - Bhakti R Pathak
- Cellular and Structural Biology Division, National Institute for Research in Reproductive Health (ICMR), Jehangir Merwanji Street, Parel, Mumbai, 400012, India.
| |
Collapse
|
28
|
Rado M, Flepisi B, Fisher D. The Effect of Normoxic and Hypoxic U-87 Glioblastoma Paracrine Secretion on the Modulation of Brain Endothelial Cells. Cells 2022; 11:276. [PMID: 35053392 PMCID: PMC8773645 DOI: 10.3390/cells11020276] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a highly invasive brain tumour, characterized by its ability to secrete factors promoting its virulence. Brain endothelial cells (BECs) in the GBM environment are physiologically modulated. The present study investigated the modulatory effects of normoxically and hypoxically induced glioblastoma U-87 cell secretions on BECs. METHODS Conditioned media (CM) were derived by cultivating U-87 cells under hypoxic incubation (5% O2) and normoxic incubation (21% O2). Treated bEnd.3 cells were evaluated for mitochondrial dehydrogenase activity, mitochondrial membrane potential (ΔΨm), ATP production, transendothelial electrical resistance (TEER), and endothelial tight-junction (ETJ) gene expression over 96 h. RESULTS The coculture of bEnd.3 cells with U-87 cells, or exposure to either hypoxic or normoxic U-87CM, was associated with low cellular viability. The ΔΨm in bEnd.3 cells was hyperpolarized after hypoxic U-87CM treatment (p < 0.0001). However, normoxic U-87CM did not affect the state of ΔΨm. BEC ATP levels were reduced after being cocultured with U-87 cells, or with hypoxic and normoxic CM (p < 0.05). Suppressed mitochondrial activity in bEnd.3 cells was associated with increased transendothelial permeability, while bEnd.3 cells significantly increased the gene expression levels of ETJs (p < 0.05) when treated with U-87CM. CONCLUSIONS Hypoxic and normoxic glioblastoma paracrine factors differentially suppressed mitochondrial activity in BECs, increasing the BECs' barrier permeability.
Collapse
Affiliation(s)
- Mariam Rado
- Medical Bioscience Department, Faculty of Natural Sciences, University of the Western Cape, Robert Sobukwe Road, Bellville 7535, South Africa;
| | - Brian Flepisi
- Department of Pharmacology, Faculty of Health Sciences, University of Pretoria, 9 Bophelo Road, Pretoria 0002, South Africa;
| | - David Fisher
- Medical Bioscience Department, Faculty of Natural Sciences, University of the Western Cape, Robert Sobukwe Road, Bellville 7535, South Africa;
| |
Collapse
|
29
|
Barbonari S, D'Amore A, Palombi F, De Cesaris P, Parrington J, Riccioli A, Filippini A. RELEVANCE OF LYSOSOMAL Ca2+ SIGNALLING MACHINERY IN CANCER. Cell Calcium 2022; 102:102539. [DOI: 10.1016/j.ceca.2022.102539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 12/23/2022]
|
30
|
Budiariati V, Rinendyaputri R, Noviantari A, Haq NMD, Budiono D, Pristihadi DN, Juliandi B, Fahrudin M, Boediono A. Conditioned medium of E17 rat brain cells induced differentiation of primary colony of mice blastocyst into neuron-like cells. J Vet Sci 2021; 22:e86. [PMID: 34854268 PMCID: PMC8636651 DOI: 10.4142/jvs.2021.22.e86] [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: 05/11/2021] [Revised: 08/01/2021] [Accepted: 08/24/2021] [Indexed: 11/29/2022] Open
Abstract
Background Conditioned medium is the medium obtained from certain cultured cells and contained secretome from the cells. The secretome, which can be in the form of growth factors, cytokines, exosomes, or other proteins secreted by the cells, can induce the differentiation of cells that still have pluripotent or multipotent properties. Objectives This study examined the effects of conditioned medium derived from E17 rat brain cells on cells with pluripotent properties. Methods The conditioned medium used in this study originated from E17 rat brain cells. The CM was used to induce the differentiation of primary colonies of mice blastocysts. Primary colonies were stained with alkaline phosphatase to analyze the pluripotency. The morphological changes in the colonies were examined, and the colonies were stained with GFAP and Neu-N markers on days two and seven after adding the conditioned medium. Results The conditioned medium could differentiate the primary colony, beginning with the formation of embryoid-body-like structure; round GFAP positive cells were identified. Finally, neuron-like cells testing positive for Neu-N were observed on the seventh day after adding the conditioned medium. Conclusions Conditioned medium from different species, in this case, E17 rat brain cells, induced and promoted the differentiation of the primary colony from mice blastocysts into neuron-like cells. The addition of CM mediated neurite growth in the differentiation process.
Collapse
Affiliation(s)
- Vista Budiariati
- Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia.
| | - Ratih Rinendyaputri
- Center for Research and Development of Biomedical and Basic Health Technology, National Institute of Health Research and Development, Ministry of Health Republic of Indonesia, Jakarta 10560, Indonesia
| | - Ariyani Noviantari
- Center for Research and Development of Biomedical and Basic Health Technology, National Institute of Health Research and Development, Ministry of Health Republic of Indonesia, Jakarta 10560, Indonesia
| | - Noer Muhammad Dliyaul Haq
- Department of Anatomy, Physiology, and Pharmacology, Faculty of Veterinary Medicine, IPB University, Bogor 16680, Indonesia
| | - Dwi Budiono
- Department of Anatomy, Physiology, and Pharmacology, Faculty of Veterinary Medicine, IPB University, Bogor 16680, Indonesia
| | - Diah Nugrahani Pristihadi
- Department of Anatomy, Physiology, and Pharmacology, Faculty of Veterinary Medicine, IPB University, Bogor 16680, Indonesia
| | - Berry Juliandi
- Department of Biology, Faculty of Mathematics and Natural Sciences, IPB University, Bogor 16680, Indonesia
| | - Mokhamad Fahrudin
- Department of Anatomy, Physiology, and Pharmacology, Faculty of Veterinary Medicine, IPB University, Bogor 16680, Indonesia
| | - Arief Boediono
- Department of Anatomy, Physiology, and Pharmacology, Faculty of Veterinary Medicine, IPB University, Bogor 16680, Indonesia
| |
Collapse
|
31
|
Zhang C, Li H, Huang Y, Tang Y, Wang J, Cheng Y, Wei Y, Zhu D, Cao Z, Zhou J. Integrative analysis of TNFRSF6B as a potential therapeutic target for pancreatic cancer. J Gastrointest Oncol 2021; 12:1673-1690. [PMID: 34532119 PMCID: PMC8421915 DOI: 10.21037/jgo-21-303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/23/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Pancreatic cancer is one of the most lethal malignant tumors worldwide with poor outcomes. Previous studies have shown that tumor necrosis factor receptor superfamily member 6b (TNFRSF6B) plays an important role in cancer progression and immunosuppression. However, the mechanisms by which TNFRSF6B influence pancreatic cancer, and the regulatory networks involved remain to be further studied. METHODS This study analyzed the mRNA information and clinical data of patients from The Cancer Genome Atlas (TCGA) and the ONCOMINE databases. The gene co-expression data regarding TNFRSF6B was obtained from the c-BioPortal and used to explore the functional network of TNFRSF6B in pancreatic cancer, as well as its function in tumor immunity. Short hairpin (sh) RNA knock-down experiments were performed to examine the functional roles of TNFRSF6B in pancreatic cancer cell lines. RESULTS The expression of TNFRSF6B was elevated in pancreatic cancer tissues compared to normal pancreatic tissues, and its high expression was associated with poor prognosis of patients with pancreatic cancer. TNFRSF6B was found to be widely involved in cell cycle processes, apoptosis, apoptosis signaling pathways, immune responses, and responses to interferon. Knock-down of TNFRSF6B expression inhibited pancreatic cancer cell proliferation and invasion in vitro. Moreover, carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) was found to be co-expressed with TNFRSF6B, and there was a positive correlation between these molecules in pancreatic cancer cells. CONCLUSIONS This report suggested that TNFRSF6B has a critical role in the progression and metastasis of pancreatic cancer. These findings provide novel insights into the role of TNFRSF6B in the functional network of pancreatic cancer, and suggest that TNFRSF6B may be a potential therapeutic target.
Collapse
Affiliation(s)
- Chen Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haoran Li
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yujie Huang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuchen Tang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jie Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yinxiang Cheng
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yijun Wei
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Dongming Zhu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhifei Cao
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
32
|
Li M, Liu H, Zhuang S, Goda K. Droplet flow cytometry for single-cell analysis. RSC Adv 2021; 11:20944-20960. [PMID: 35479393 PMCID: PMC9034116 DOI: 10.1039/d1ra02636d] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 06/06/2021] [Indexed: 01/22/2023] Open
Abstract
The interrogation of single cells has revolutionised biology and medicine by providing crucial unparalleled insights into cell-to-cell heterogeneity. Flow cytometry (including fluorescence-activated cell sorting) is one of the most versatile and high-throughput approaches for single-cell analysis by detecting multiple fluorescence parameters of individual cells in aqueous suspension as they flow past through a focus of excitation lasers. However, this approach relies on the expression of cell surface and intracellular biomarkers, which inevitably lacks spatial and temporal phenotypes and activities of cells, such as secreted proteins, extracellular metabolite production, and proliferation. Droplet microfluidics has recently emerged as a powerful tool for the encapsulation and manipulation of thousands to millions of individual cells within pico-litre microdroplets. Integrating flow cytometry with microdroplet architectures surrounded by aqueous solutions (e.g., water-in-oil-in-water (W/O/W) double emulsion and hydrogel droplets) opens avenues for new cellular assays linking cell phenotypes to genotypes at the single-cell level. In this review, we discuss the capabilities and applications of droplet flow cytometry (DFC). This unique technique uses standard commercially available flow cytometry instruments to characterise or select individual microdroplets containing single cells of interest. We explore current challenges associated with DFC and present our visions for future development.
Collapse
Affiliation(s)
- Ming Li
- School of Engineering, Macquarie University Sydney NSW 2109 Australia
- Biomolecular Discovery Research Centre, Macquarie University Sydney NSW 2109 Australia
| | - Hangrui Liu
- Department of Physics and Astronomy, Macquarie University Sydney NSW 2109 Australia
| | - Siyuan Zhuang
- School of Engineering, Macquarie University Sydney NSW 2109 Australia
| | - Keisuke Goda
- Department of Chemistry, The University of Tokyo Tokyo 113-0033 Japan
- Institute of Technological Sciences, Wuhan University 430072 Hubei PR China
- Department of Bioengineering, University of California Los Angeles CA 90095 USA
| |
Collapse
|
33
|
Role of IGFBP-2 in oral cancer metastasis. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166143. [PMID: 33864954 DOI: 10.1016/j.bbadis.2021.166143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/30/2021] [Accepted: 04/05/2021] [Indexed: 01/01/2023]
Abstract
Cancer metastasis is one of most main causes of failure in cancer treatment. Nonetheless, more than half of oral cancer patients were diagnosed as advanced oral cancer with dramatically decreased 5-year survival rate to lower than 20%, while the stages become more advanced. In order to improve oral cancer treatment, the identification of cancer metastatic biomarkers and mechanisms is critical. In the current study, two pairs of oral squamous cell carcinoma lines, OC3/C9, and invasive OC3-I5/C9-I5were used as model systems to investigate invasive mechanism as well as to identify potential therapy-associated targets. Based on our previous proteomic analysis, insulin-like growth factor-binding protein 2 (IGFBP-2) was reported participating in oral cancer metastasis. Subsequent studies have applied interference RNA as well as recombinant protein techniques to confirm the roles of IGFBP-2 in oral cancer metastasis and examine their potency in regulating invasion as well as the mechanism IGFBP-2 involved. The results demonstrated that expression of epithelial-mesenchymal transition (EMT) markers including Twist, Snail1, SIP1, profilin, vimentin, uPA and MMP9 were increased in both OC3-I5 and C9-I5 compared to OC3 and C9 cells, while E-cadherin expression was down-regulated in the OC3-I5 and C9-I5 cells. Moreover, IGFBP-2 is shown to affect not only migration and invasion but also wound healing ability and cell proliferation. Our results also revealed that uPA is a downstream target of IGFBP-2 to intermediate oral cancer metastasis. To sum up, the current studies indicated that elevated IGFBP-2 is strongly correlated with oral cancer metastasis and progression, and that it could potentially serve as a prognostic biomarker as well as an innovative target for the treatment of oral cancer invasion.
Collapse
|
34
|
Ardila DC, Aggarwal V, Singh M, Chattopadhyay A, Chaparala S, Sant S. Identifying Molecular Signatures of Distinct Modes of Collective Migration in Response to the Microenvironment Using Three-Dimensional Breast Cancer Models. Cancers (Basel) 2021; 13:cancers13061429. [PMID: 33804802 PMCID: PMC8004051 DOI: 10.3390/cancers13061429] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/09/2021] [Accepted: 03/16/2021] [Indexed: 01/16/2023] Open
Abstract
Simple Summary The objective of this study was to investigate the role of two microenvironmental factors, namely, tumor-intrinsic hypoxia and secretome in inducing collective migration. We utilized three-dimensional (3D) discrete-sized microtumor models, which recapitulate hallmarks of transition of ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC). Tumor-intrinsic hypoxia induced directional migration in large hypoxic microtumors while secretome from large microtumors induced radial migration in non-hypoxic microtumors. This highlights the emergence phenotypic heterogeneity and plasticity in cancer cells in response to different microenvironmental stimuli. To unravel mechanisms underlying these two distinct modes of migration, we performed differential gene expression analysis of hypoxia- and secretome-induced migratory phenotypes using non-migratory, non-hypoxic microtumors as controls. We proposed unique gene signature sets related to tumor-intrinsic hypoxia, hypoxia-induced epithelial-mesenchymal transition (EMT), as well as hypoxia-induced directional migration and secretome-induced radial migration. Abstract Collective cell migration is a key feature of transition of ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC) among many other cancers, yet the microenvironmental factors and underlying mechanisms that trigger collective migration remain poorly understood. Here, we investigated two microenvironmental factors, tumor-intrinsic hypoxia and tumor-secreted factors (secretome), as triggers of collective migration using three-dimensional (3D) discrete-sized microtumor models that recapitulate hallmarks of DCIS-IDC transition. Interestingly, the two factors induced two distinct modes of collective migration: directional and radial migration in the 3D microtumors generated from the same breast cancer cell line model, T47D. Without external stimulus, large (600 µm) T47D microtumors exhibited tumor-intrinsic hypoxia and directional migration, while small (150 µm), non-hypoxic microtumors exhibited radial migration only when exposed to the secretome of large microtumors. To investigate the mechanisms underlying hypoxia- and secretome-induced directional vs. radial migration modes, we performed differential gene expression analysis of hypoxia- and secretome-induced migratory microtumors compared with non-hypoxic, non-migratory small microtumors as controls. We propose unique gene signature sets related to tumor-intrinsic hypoxia, hypoxia-induced epithelial-mesenchymal transition (EMT), as well as hypoxia-induced directional migration and secretome-induced radial migration. Gene Set Enrichment Analysis (GSEA) and protein-protein interaction (PPI) network analysis revealed enrichment and potential interaction between hypoxia, EMT, and migration gene signatures for the hypoxia-induced directional migration. In contrast, hypoxia and EMT were not enriched in the secretome-induced radial migration, suggesting that complete EMT may not be required for radial migration. Survival analysis identified unique genes associated with low survival rate and poor prognosis in TCGA-breast invasive carcinoma dataset from our tumor-intrinsic hypoxia gene signature (CXCR4, FOXO3, LDH, NDRG1), hypoxia-induced EMT gene signature (EFEMP2, MGP), and directional migration gene signature (MAP3K3, PI3K3R3). NOS3 was common between hypoxia and migration gene signature. Survival analysis from secretome-induced radial migration identified ATM, KCNMA1 (hypoxia gene signature), and KLF4, IFITM1, EFNA1, TGFBR1 (migration gene signature) to be associated with poor survival rate. In conclusion, our unique 3D cultures with controlled microenvironments respond to different microenvironmental factors, tumor-intrinsic hypoxia, and secretome by adopting distinct collective migration modes and their gene expression analysis highlights the phenotypic heterogeneity and plasticity of epithelial cancer cells.
Collapse
Affiliation(s)
- Diana Catalina Ardila
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (D.C.A.); (V.A.); (M.S.)
| | - Vaishali Aggarwal
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (D.C.A.); (V.A.); (M.S.)
| | - Manjulata Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (D.C.A.); (V.A.); (M.S.)
| | - Ansuman Chattopadhyay
- Health Sciences Library System, University of Pittsburgh, Pittsburgh, PA 15219, USA; (A.C.); (S.C.)
| | - Srilakshmi Chaparala
- Health Sciences Library System, University of Pittsburgh, Pittsburgh, PA 15219, USA; (A.C.); (S.C.)
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (D.C.A.); (V.A.); (M.S.)
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
- UPMC-Hillman Cancer Center, Pittsburgh, PA 15260, USA
- Correspondence: ; Tel.: +1-412-6489804
| |
Collapse
|
35
|
Ritchie S, Reed DA, Pereira BA, Timpson P. The cancer cell secretome drives cooperative manipulation of the tumour microenvironment to accelerate tumourigenesis. Fac Rev 2021; 10:4. [PMID: 33659922 PMCID: PMC7894270 DOI: 10.12703/r/10-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cellular secretions are a fundamental aspect of cell-cell and cell-matrix interactions in vivo. In malignancy, cancer cells have an aberrant secretome compared to their non-malignant counterparts, termed the "cancer cell secretome". The cancer cell secretome can influence every stage of the tumourigenic cascade. At the primary site, cancer cells can secrete a multitude of factors that facilitate invasion into surrounding tissue, allowing interaction with the local tumour microenvironment (TME), driving tumour development and progression. In more advanced disease, the cancer cell secretome can be involved in extravasation and metastasis, including metastatic organotropism, pre-metastatic niche (PMN) preparation, and metastatic outgrowth. In this review, we will explore the latest advances in the field of cancer cell secretions, including its dynamic and complex role in activating the TME and potentiating invasion and metastasis, with comments on how these secretions may also promote therapy resistance.
Collapse
Affiliation(s)
- Shona Ritchie
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Daniel A Reed
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Brooke A Pereira
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Paul Timpson
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| |
Collapse
|
36
|
Iorio E, Podo F, Leach MO, Koutcher J, Blankenberg FG, Norfray JF. A novel roadmap connecting the 1H-MRS total choline resonance to all hallmarks of cancer following targeted therapy. Eur Radiol Exp 2021; 5:5. [PMID: 33447887 PMCID: PMC7809082 DOI: 10.1186/s41747-020-00192-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/29/2020] [Indexed: 11/15/2022] Open
Abstract
This review describes a cellular adaptive stress signalling roadmap connecting the 1H magnetic resonance spectroscopy (MRS) total choline peak at 3.2 ppm (tCho) to cancer response after targeted therapy (TT). Recent research on cell signalling, tCho metabolism, and TT of cancer has been retrospectively re-examined. Signalling research describes how the unfolded protein response (UPR), a major stress signalling network, transduces, regulates, and rewires the total membrane turnover in different cancer hallmarks after a TT stress. In particular, the UPR signalling maintains or increases total membrane turnover in all pro-survival hallmarks, whilst dramatically decreases turnover during apoptosis, a pro-death hallmark. Recent research depicts the TT-induced stress as a crucial event responsible for interrupting UPR pro-survival pathways, leading to an UPR-mediated cell death. The 1H-MRS tCho resonance represents the total mobile precursors and products during the enzymatic modification of phosphatidylcholine membrane abundance. The tCho profile represents a biomarker that noninvasively monitors TT-induced enzymatic changes in total membrane turnover in a wide variety of existing and new anticancer treatments targeting specific layers of the UPR signalling network. Our overview strongly suggests further evaluating and validating the 1H-MRS tCho peak as a powerful noninvasive imaging biomarker of cancer response in TT clinical trials.
Collapse
Affiliation(s)
- Egidio Iorio
- High Resolution NMR Unit-Core Facilities, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Roma, Italy.
| | - Franca Podo
- High Resolution NMR Unit-Core Facilities, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Roma, Italy
| | - Martin O Leach
- MRI Unit, Royal Marsden Hospital, Downs Road, Sutton, Surrey, SM2 5PT, UK
| | - Jason Koutcher
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | | | - Joseph F Norfray
- Emeritus, Chicago Northside MRI Center, 2818 N. Sheridan Rd, Chicago, IL, 60657, USA
| |
Collapse
|
37
|
Clinicopathologic Analysis of Cathepsin B as a Prognostic Marker of Thyroid Cancer. Int J Mol Sci 2020; 21:ijms21249537. [PMID: 33333840 PMCID: PMC7765333 DOI: 10.3390/ijms21249537] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/03/2020] [Accepted: 12/10/2020] [Indexed: 12/27/2022] Open
Abstract
Thyroid cancer incidence has increased worldwide; however, investigations of thyroid cancer-related factors as potential prognosis markers remain insufficient. Secreted proteins from the cancer secretome are regulators of several molecular mechanisms and are, thereby, ideal candidates for potential markers. We aimed to identify a specific factor for thyroid cancer by analyzing the secretome from normal thyroid cells, papillary thyroid cancer (PTC) cells, and anaplastic thyroid cancer cells using mass spectrometry (MS). Cathepsin B (CTSB) showed highest expression in PTC cells compared to other cell lines, and CTSB levels in tumor samples were higher than that seen in normal tissue. Further, among thyroid cancer patients, increased CTSB expression was related to higher risk of lymph node metastasis (LNM) and advanced N stage. Overexpression of CTSB in thyroid cancer cell lines activated cell migration by increasing the expression of vimentin and Snail, while its siRNA-mediated silencing inhibited cell migration by decreasing vimentin and Snail expression. Mechanistically, CTSB-associated enhanced cell migration and upregulation of vimentin and Snail occurred via increased phosphorylation of p38. As our results suggest that elevated CTSB in thyroid cancer induces the expression of metastatic proteins and thereby leads to LNM, CTSB may be a good and clinically relevant prognostic marker.
Collapse
|
38
|
Suri R, Zimmerman JW, Burkhart RA. Modeling human pancreatic ductal adenocarcinoma for translational research: current options, challenges, and prospective directions. ANNALS OF PANCREATIC CANCER 2020; 3:17. [PMID: 33889840 PMCID: PMC8059695 DOI: 10.21037/apc-20-29] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating malignancy with one of the lowest survival rates. Early detection, an improved understanding of tumor biology, and novel therapeutic discoveries are needed in order to improve overall patient survival. Scientific progress towards meeting these goals relies upon accurate modeling of the human disease. From two-dimensional (2D) cell lines to the advanced modeling available today, we aim to characterize the critical tools in efforts to further understand PDAC biology. The National Center for Biotechnology Information's PubMed and the Elsevier's SCOPUS were used to perform a comprehensive literature review evaluating preclinical human-derived PDAC models. Keywords included pancreatic cancer, PDAC, preclinical models, KRAS mutations, xenograft, co-culturing fibroblasts, co-culturing lymphocytes and PDAC immunotherapy Initial search was limited to articles about PDAC and was then expanded to include other gastrointestinal malignancies where information may complement our effort. A supervised review of the key literature's references was utilized to augment the capture of relevant data. The discovery and refinement of techniques enabling immortalized 2D cell culture provided the cornerstone for modern cancer biology research. Cell lines have been widely used to represent PDAC in vitro but are limited in capacity to model three-dimensional (3D) tumor attributes and interactions within the tumor microenvironment. Xenografts are an alternative method to model PDAC with improved capacity to understand certain aspects of 3D tumor biology in vivo while limited by the use of immunodeficient mice. Advances of in vitro modeling techniques have led to 3D organoid models for PDAC biology. Co-culturing models in the 3D environment have been proposed as an efficient modeling system for improving upon the limitations encountered in the standard 2D and xenograft tumor models. The integrated network of cells and stroma that comprise PDAC in vivo need to be accurately depicted ex vivo to continue to make progress in this disease. Recapitulating the complex tumor microenvironment in a preclinical model of human disease is an outstanding and urgent need in PDAC. Definitive characterization of available human models for PDAC serves to further the core mission of pancreatic cancer translational research.
Collapse
Affiliation(s)
- Reecha Suri
- Division of Hepatobiliary and Pancreatic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jacquelyn W. Zimmerman
- Department of Medical Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Richard A. Burkhart
- Division of Hepatobiliary and Pancreatic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
39
|
Rauter T, Burgstaller S, Gottschalk B, Ramadani-Muja J, Bischof H, Hay JC, Graier WF, Malli R. ER-to-Golgi Transport in HeLa Cells Displays High Resilience to Ca 2+ and Energy Stresses. Cells 2020; 9:E2311. [PMID: 33080790 PMCID: PMC7603030 DOI: 10.3390/cells9102311] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/09/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
One third of all human proteins are either transmembrane or soluble secretory proteins that first target the endoplasmic reticulum (ER). These proteins subsequently leave the ER and enter the Golgi apparatus via ER-Golgi intermediate vesicular structures. Live-cell imaging of cargos fused to fluorescent proteins (FPs) enables the high-resolution visualization and characterization of secretory transport processes. Here, we performed fluorescence time-lapse imaging to assess the Ca2+ and energy dependency of ER-to-Golgi transport in living HeLa cells, a cancer cell model which has been well investigated. Our data revealed that ER-to-Golgi transport remained highly efficient in the absence of ATP-generating substrates, despite clear reductions in cytosolic and mitochondrial ATP levels under these energy stress conditions. However, cell treatment with 2-deoxy-D-glucose (2-DG), which severely diminished subcellular ATP levels, abolished ER-to-Golgi transport. Interestingly, while 2-DG elevated cytosolic Ca2+ levels and reduced long-distance movements of glycosylphosphatidylinositol (GPI)-positive vesicles, robust short-term ER Ca2+ mobilizations, which strongly affected the motility of these vesicles, did not considerably impair ER-to-Golgi transport. In summary, we highlight that ER-to-Golgi transport in HeLa cells remains functional despite high energy and Ca2+ stress levels.
Collapse
Affiliation(s)
- Thomas Rauter
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (T.R.); (S.B.); (B.G.); (J.R.-M.); (H.B.); (W.F.G.)
| | - Sandra Burgstaller
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (T.R.); (S.B.); (B.G.); (J.R.-M.); (H.B.); (W.F.G.)
- Interfaculty Institute of Cell Biology, University of Tuebingen, Auf der Morgenstelle 15, 72076 Tuebingen, Germany
| | - Benjamin Gottschalk
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (T.R.); (S.B.); (B.G.); (J.R.-M.); (H.B.); (W.F.G.)
| | - Jeta Ramadani-Muja
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (T.R.); (S.B.); (B.G.); (J.R.-M.); (H.B.); (W.F.G.)
| | - Helmut Bischof
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (T.R.); (S.B.); (B.G.); (J.R.-M.); (H.B.); (W.F.G.)
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| | - Jesse C. Hay
- Division of Biological Sciences and Center for Structural and Functional Neuroscience, The University of Montana, 32 Campus Drive, HS 302A, Missoula, MT 59812-4824, USA;
| | - Wolfgang F. Graier
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (T.R.); (S.B.); (B.G.); (J.R.-M.); (H.B.); (W.F.G.)
- BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Roland Malli
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (T.R.); (S.B.); (B.G.); (J.R.-M.); (H.B.); (W.F.G.)
- BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria
| |
Collapse
|
40
|
Matafora V, Farris F, Restuccia U, Tamburri S, Martano G, Bernardelli C, Sofia A, Pisati F, Casagrande F, Lazzari L, Marsoni S, Bonoldi E, Bachi A. Amyloid aggregates accumulate in melanoma metastasis modulating YAP activity. EMBO Rep 2020; 21:e50446. [PMID: 32749065 PMCID: PMC7507035 DOI: 10.15252/embr.202050446] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/27/2020] [Accepted: 07/07/2020] [Indexed: 12/13/2022] Open
Abstract
Melanoma progression is generally associated with increased transcriptional activity mediated by the Yes-associated protein (YAP). Mechanical signals from the extracellular matrix are sensed by YAP, which then activates the expression of proliferative genes, promoting melanoma progression and drug resistance. Which extracellular signals induce mechanotransduction, and how this is mediated, is not completely understood. Here, using secretome analyses, we reveal the extracellular accumulation of amyloidogenic proteins, i.e. premelanosome protein (PMEL), in metastatic melanoma, together with proteins that assist amyloid maturation into fibrils. We also confirm the accumulation of amyloid-like aggregates, similar to those detected in Alzheimer disease, in metastatic cell lines, as well as in human melanoma biopsies. Mechanistically, beta-secretase 2 (BACE2) regulates the maturation of these aggregates, which in turn induce YAP activity. We also demonstrate that recombinant PMEL fibrils are sufficient to induce mechanotransduction, triggering YAP signaling. Finally, we demonstrate that BACE inhibition affects cell proliferation and increases drug sensitivity, highlighting the importance of amyloids for melanoma survival, and the use of beta-secretase inhibitors as potential therapeutic approach for metastatic melanoma.
Collapse
Affiliation(s)
| | | | - Umberto Restuccia
- IFOM‐ FIRC Institute of Molecular OncologyMilanItaly
- Present address:
ADIENNE Pharma & BiotechCaponagoItaly
| | - Simone Tamburri
- IFOM‐ FIRC Institute of Molecular OncologyMilanItaly
- Present address:
Department of Experimental OncologyIEO‐European Institute of Oncology IRCCSMilanItaly
| | | | - Clara Bernardelli
- IFOM‐ FIRC Institute of Molecular OncologyMilanItaly
- Present address:
Fondazione Politecnico di MilanoMilanItaly
| | - Andrea Sofia
- IFOM‐ FIRC Institute of Molecular OncologyMilanItaly
- University of InsubriaVareseItaly
| | - Federica Pisati
- IFOM‐ FIRC Institute of Molecular OncologyMilanItaly
- Cogentech SRL Benefit CorporationMilanItaly
| | | | - Luca Lazzari
- IFOM‐ FIRC Institute of Molecular OncologyMilanItaly
| | | | - Emanuela Bonoldi
- Department of Laboratory MedicineDivision of PathologyGrande Ospedale Metropolitano NiguardaMilanItaly
| | - Angela Bachi
- IFOM‐ FIRC Institute of Molecular OncologyMilanItaly
| |
Collapse
|
41
|
Fiocchetti M, Solar Fernandez V, Segatto M, Leone S, Cercola P, Massari A, Cavaliere F, Marino M. Extracellular Neuroglobin as a Stress-Induced Factor Activating Pre-Adaptation Mechanisms against Oxidative Stress and Chemotherapy-Induced Cell Death in Breast Cancer. Cancers (Basel) 2020; 12:cancers12092451. [PMID: 32872414 PMCID: PMC7564643 DOI: 10.3390/cancers12092451] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 08/26/2020] [Indexed: 12/16/2022] Open
Abstract
Components of tumor microenvironment, including tumor and/or stromal cells-derived factors, exert a critical role in breast cancer (BC) progression. Here we evaluated the possible role of neuroglobin (NGB), a monomeric globin that acts as a compensatory protein against oxidative and apoptotic processes, as part of BC microenvironment. The extracellular NGB levels were evaluated by immunofluorescence of BC tissue sections and by Western blot of the culture media of BC cell lines. Moreover, reactive oxygen species (ROS) generation, cell apoptosis, and cell migration were evaluated in different BC cells and non-tumorigenic epithelial mammary cells treated with BC cells (i.e., Michigan Cancer Foundation-7, MCF-7) conditioned culture media and extracellular NGB. Results demonstrate that NGB is a component of BC microenvironment. NGB is released in tumor microenvironment by BC cells only under oxidative stress conditions where it can act as autocrine/paracrine factor able to communicate cell resilience against oxidative stress and chemotherapeutic treatment.
Collapse
Affiliation(s)
- Marco Fiocchetti
- Department of Science, University Roma Tre, Viale Guglielmo Marconi 446, I-00146 Roma, Italy; (V.S.F.); (S.L.)
- Correspondence: (M.F.); (M.M.); Tel.: +39-06-5733-6455 (M.F.); +39-06-5733-6320 (M.M.); Fax: +39-06-5733-6321 (M.F. & M.M.)
| | - Virginia Solar Fernandez
- Department of Science, University Roma Tre, Viale Guglielmo Marconi 446, I-00146 Roma, Italy; (V.S.F.); (S.L.)
| | - Marco Segatto
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche (IS), Italy;
| | - Stefano Leone
- Department of Science, University Roma Tre, Viale Guglielmo Marconi 446, I-00146 Roma, Italy; (V.S.F.); (S.L.)
| | - Paolo Cercola
- Division of Senology, Belcolle Hospital, Str. Sammartinese, 01100 Viterbo, Italy; (P.C.); (A.M.); (F.C.)
| | - Annalisa Massari
- Division of Senology, Belcolle Hospital, Str. Sammartinese, 01100 Viterbo, Italy; (P.C.); (A.M.); (F.C.)
| | - Francesco Cavaliere
- Division of Senology, Belcolle Hospital, Str. Sammartinese, 01100 Viterbo, Italy; (P.C.); (A.M.); (F.C.)
| | - Maria Marino
- Department of Science, University Roma Tre, Viale Guglielmo Marconi 446, I-00146 Roma, Italy; (V.S.F.); (S.L.)
- Correspondence: (M.F.); (M.M.); Tel.: +39-06-5733-6455 (M.F.); +39-06-5733-6320 (M.M.); Fax: +39-06-5733-6321 (M.F. & M.M.)
| |
Collapse
|
42
|
Kosnopfel C, Sinnberg T, Sauer B, Niessner H, Muenchow A, Fehrenbacher B, Schaller M, Mertens PR, Garbe C, Thakur BK, Schittek B. Tumour Progression Stage-Dependent Secretion of YB-1 Stimulates Melanoma Cell Migration and Invasion. Cancers (Basel) 2020; 12:cancers12082328. [PMID: 32824741 PMCID: PMC7464723 DOI: 10.3390/cancers12082328] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/10/2020] [Accepted: 08/16/2020] [Indexed: 12/16/2022] Open
Abstract
Secreted factors play an important role in intercellular communication. Therefore, they are not only indispensable for the regulation of various physiological processes but can also decisively advance the development and progression of tumours. In the context of inflammatory disease, Y-box binding protein 1 (YB-1) is actively secreted and the extracellular protein promotes cell proliferation and migration. In malignant melanoma, intracellular YB-1 expression increases during melanoma progression and represents an unfavourable prognostic marker. Here, we show active secretion of YB-1 from melanoma cells as opposed to benign cells of the skin. Intriguingly, YB-1 secretion correlates with the stage of melanoma progression and depends on a calcium- and ATP-dependent non-classical secretory pathway leading to the occurrence of YB-1 in the extracellular space as a free protein. Along with an elevated YB-1 secretion of melanoma cells in the metastatic growth phase, extracellular YB-1 exerts a stimulating effect on melanoma cell migration, invasion, and tumourigenicity. Collectively, these data suggest that secreted YB-1 plays a functional role in melanoma cell biology, stimulating metastasis, and may serve as a novel biomarker in malignant melanoma that reflects tumour aggressiveness.
Collapse
Affiliation(s)
- Corinna Kosnopfel
- Department of Dermatology, University of Tübingen, 72076 Tübingen, Germany; (T.S.); (B.S.); (H.N.); (A.M.); (B.F.); (M.S.); (C.G.)
- Department of Dermatology, University Hospital Würzburg, 97080 Würzburg, Germany
- Correspondence: (C.K.); (B.S.); Tel.: +49-931-20126778 (C.K.); +49-7071-29-80832 (B.S.)
| | - Tobias Sinnberg
- Department of Dermatology, University of Tübingen, 72076 Tübingen, Germany; (T.S.); (B.S.); (H.N.); (A.M.); (B.F.); (M.S.); (C.G.)
| | - Birgit Sauer
- Department of Dermatology, University of Tübingen, 72076 Tübingen, Germany; (T.S.); (B.S.); (H.N.); (A.M.); (B.F.); (M.S.); (C.G.)
| | - Heike Niessner
- Department of Dermatology, University of Tübingen, 72076 Tübingen, Germany; (T.S.); (B.S.); (H.N.); (A.M.); (B.F.); (M.S.); (C.G.)
| | - Alina Muenchow
- Department of Dermatology, University of Tübingen, 72076 Tübingen, Germany; (T.S.); (B.S.); (H.N.); (A.M.); (B.F.); (M.S.); (C.G.)
| | - Birgit Fehrenbacher
- Department of Dermatology, University of Tübingen, 72076 Tübingen, Germany; (T.S.); (B.S.); (H.N.); (A.M.); (B.F.); (M.S.); (C.G.)
| | - Martin Schaller
- Department of Dermatology, University of Tübingen, 72076 Tübingen, Germany; (T.S.); (B.S.); (H.N.); (A.M.); (B.F.); (M.S.); (C.G.)
| | - Peter R. Mertens
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University, 39120 Magdeburg, Germany;
| | - Claus Garbe
- Department of Dermatology, University of Tübingen, 72076 Tübingen, Germany; (T.S.); (B.S.); (H.N.); (A.M.); (B.F.); (M.S.); (C.G.)
| | - Basant Kumar Thakur
- Department of Pediatric Hematology and Oncology, University Hospital Essen, 45147 Essen, Germany;
| | - Birgit Schittek
- Department of Dermatology, University of Tübingen, 72076 Tübingen, Germany; (T.S.); (B.S.); (H.N.); (A.M.); (B.F.); (M.S.); (C.G.)
- Correspondence: (C.K.); (B.S.); Tel.: +49-931-20126778 (C.K.); +49-7071-29-80832 (B.S.)
| |
Collapse
|
43
|
Matas-Nadal C, Bech-Serra JJ, Guasch-Vallés M, Fernández-Armenteros JM, Barceló C, Casanova JM, de la Torre Gómez C, Aguayo Ortiz R, Garí E. Evaluation of Tumor Interstitial Fluid-Extraction Methods for Proteome Analysis: Comparison of Biopsy Elution versus Centrifugation. J Proteome Res 2020; 19:2598-2605. [PMID: 31877049 DOI: 10.1021/acs.jproteome.9b00770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The analysis of tumor interstitial fluid (TIF) composition is a valuable procedure to identify antimetastatic targets, and different laboratories have set up techniques for TIF isolation and proteomic analyses. However, those methods had never been compared in samples from the same tumor and patient. In this work, we compared the two most used methods, elution and centrifugation, in pieces of the same biopsy samples of cutaneous squamous cell carcinoma (cSCC). First, we established that high G-force (10 000g) was required to obtain TIF from cSCC by centrifugation. Second, we compared the centrifugation method with the elution method in pieces of three different cSCC tumors. We found that the mean protein intensities based in the number of peptide spectrum matches was significantly higher in the centrifuged samples than in the eluted samples. Regarding the robustness of the methods, we observed higher overlapping between both methods (77-80%) than among samples (50%). These results suggest that there exists an elevated consistence of TIF composition independently of the method used. However, we observed a 3-fold increase of extracellular proteins in nonoverlapped proteome obtained by centrifugation. We therefore conclude that centrifugation is the method of choice to study the proteome of TIF from cutaneous biopsies.
Collapse
Affiliation(s)
- Clara Matas-Nadal
- Cell Cycle Laboratory, Institut de Recerca Biomèdica de Lleida (IRB Lleida), Lleida, 25198, Spain
| | - Joan Josep Bech-Serra
- Proteomics Unit, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
| | - Marta Guasch-Vallés
- Cell Cycle Laboratory, Institut de Recerca Biomèdica de Lleida (IRB Lleida), Lleida, 25198, Spain.,Department de Ciències Mèdiques Bàsiques. Facultat de Medicina, Universitat de Lleida, Lleida, 25003, Spain
| | - Josep Manel Fernández-Armenteros
- Cell Cycle Laboratory, Institut de Recerca Biomèdica de Lleida (IRB Lleida), Lleida, 25198, Spain.,Servei de Dermatologia, Hospital Universitari Arnau de Vilanova, Lleida, 25198, Spain
| | - Carla Barceló
- Cell Cycle Laboratory, Institut de Recerca Biomèdica de Lleida (IRB Lleida), Lleida, 25198, Spain
| | - Josep Manel Casanova
- Cell Cycle Laboratory, Institut de Recerca Biomèdica de Lleida (IRB Lleida), Lleida, 25198, Spain.,Department de Ciències Mèdiques Bàsiques. Facultat de Medicina, Universitat de Lleida, Lleida, 25003, Spain.,Servei de Dermatologia, Hospital Universitari Arnau de Vilanova, Lleida, 25198, Spain
| | | | - Rafael Aguayo Ortiz
- Cell Cycle Laboratory, Institut de Recerca Biomèdica de Lleida (IRB Lleida), Lleida, 25198, Spain.,Servei de Dermatologia, Hospital Universitari Arnau de Vilanova, Lleida, 25198, Spain
| | - Eloi Garí
- Cell Cycle Laboratory, Institut de Recerca Biomèdica de Lleida (IRB Lleida), Lleida, 25198, Spain.,Department de Ciències Mèdiques Bàsiques. Facultat de Medicina, Universitat de Lleida, Lleida, 25003, Spain
| |
Collapse
|
44
|
Rehman AU, Olof Olsson P, Khan N, Khan K. Identification of Human Secretome and Membrane Proteome-Based Cancer Biomarkers Utilizing Bioinformatics. J Membr Biol 2020; 253:257-270. [PMID: 32415382 DOI: 10.1007/s00232-020-00122-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 05/02/2020] [Indexed: 12/12/2022]
Abstract
Cellular secreted proteins (secretome), together with cellular membrane proteins, collectively referred to as secretory and membrane proteins (SMPs) are a large potential source of biomarkers as they can be used to indicate cell types and conditions. SMPs have been shown to be ideal candidates for several clinically approved drug regimens including for cancer. This study aimed at performing a functional analysis of SMPs within different cancer subtypes to provide great clinical targets for potential prognostic, diagnostic and the therapeutics use. Using an innovative majority decision-based algorithm and transcriptomic data spanning 5 cancer types and over 3000 samples, we quantified the relative difference in SMPs gene expression compared to normal adjacent tissue. A detailed deep data mining analysis revealed a consistent group of downregulated SMP isoforms, enriched in hematopoietic cell lineages (HCL), in multiple cancer types. HCL-associated genes were frequently downregulated in successive cancer stages and high expression was associated with good patient prognosis. In addition, we suggest a potential mechanism by which cancer cells suppress HCL signaling by reducing the expression of immune-related genes. Our data identified potential biomarkers for the cancer immunotherapy. We conclude that our approach may be applicable for the delineation of other types of cancer and illuminate specific targets for therapeutics and diagnostics.
Collapse
Affiliation(s)
- Adeel Ur Rehman
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.
| | | | - Naveed Khan
- Max Plank Partner Institute of Computational Biology, Shanghai Institute of Biological Sciences, Shanghai, 200032, China
| | - Khalid Khan
- Department of Respiratory and Critical Care Medicine, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen Institute of Respiratory Diseases, Shenzhen, China.,Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
| |
Collapse
|
45
|
Madden EC, Gorman AM, Logue SE, Samali A. Tumour Cell Secretome in Chemoresistance and Tumour Recurrence. Trends Cancer 2020; 6:489-505. [PMID: 32460003 DOI: 10.1016/j.trecan.2020.02.020] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 02/20/2020] [Accepted: 02/26/2020] [Indexed: 12/22/2022]
Abstract
Chemoresistance is a major factor driving tumour relapse and the high rates of cancer-related deaths. Understanding how cancer cells overcome chemotherapy-induced cell death is critical in promoting patient survival. One emerging mechanism of chemoresistance is the tumour cell secretome (TCS), an array of protumorigenic factors released by tumour cells. Chemotherapy exposure can also alter the composition of the TCS, known as therapy-induced TCS, and can promote tumour relapse and the formation of an immunosuppressive tumour microenvironment (TME). Here, we outline how the TCS can protect cancer cells from chemotherapy-induced cell death. We also highlight recent evidence describing how therapy-induced TCS can impact cancer stem cell (CSC) expansion and tumour-associated immune cells to enable tumour regrowth and antitumour immunity.
Collapse
Affiliation(s)
- Emma C Madden
- Apoptosis Research Centre, NUI Galway, Galway, Ireland; School of Natural Sciences, NUI Galway, Galway, Ireland
| | - Adrienne M Gorman
- Apoptosis Research Centre, NUI Galway, Galway, Ireland; School of Natural Sciences, NUI Galway, Galway, Ireland
| | - Susan E Logue
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.
| | - Afshin Samali
- Apoptosis Research Centre, NUI Galway, Galway, Ireland; School of Natural Sciences, NUI Galway, Galway, Ireland.
| |
Collapse
|
46
|
Fiorini E, Veghini L, Corbo V. Modeling Cell Communication in Cancer With Organoids: Making the Complex Simple. Front Cell Dev Biol 2020; 8:166. [PMID: 32258040 PMCID: PMC7094029 DOI: 10.3389/fcell.2020.00166] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/28/2020] [Indexed: 12/14/2022] Open
Abstract
Homotypic and heterotypic interactions between cells are of crucial importance in multicellular organisms for the maintenance of physiological functions. Accordingly, changes in cell-to-cell communication contribute significantly to tumor development. Cancer cells engage the different components of the tumor microenvironment (TME) to support malignant proliferation, escape immune control, and favor metastatic spreading. The interaction between cancerous and non-cancerous cell types within tumors occurs in many ways, including physical contact and paracrine signaling. Furthermore, local and long-range transfer of biologically active molecules (e.g., DNA, RNA, and proteins) can be mediated by small extracellular vesicles (EVs) and this has been shown to influence many aspects of tumor progression. As it stands, there is a critical need for suitable experimental systems that enable modeling the cell-to-cell communications occurring in cancer. Given their intrinsic complexity, animal models represent the ideal system to study cell-to-cell interaction between different cell types; however, they might make difficult to assess individual contribution to a given phenotype. On the other hand, simplest experimental models (i.e., in vitro culture systems) might be of great use when weighing individual contributions to a given phenomenon, yet it is imperative that they share a considerable number of features with human cancer. Of the many culture systems available to the scientific community, patient-derived organoids already proved to faithfully recapitulate many of the traits of patients’ disease, including genetic heterogeneity and response to therapy. The organoid technology offers several advantages over conventional monolayer cell cultures, including the preservation of the topology of cell-to-cell and cell-to-matrix interactions as observed in vivo. Several studies have shown that organoid cultures can be successfully used to study interaction between cancer cells and cellular components of the TME. Here, we discuss the potential of using organoids to model the interplay between cancer and non-cancer cells in order to unveil biological mechanisms involved in cancers initiation and progression, which might ultimately lead to the identification of novel intervention strategy for those diseases.
Collapse
Affiliation(s)
- Elena Fiorini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy.,Department of Medicine, University of Verona, Verona, Italy
| | - Lisa Veghini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy.,Department of Medicine, University of Verona, Verona, Italy
| | - Vincenzo Corbo
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy.,ARC-Net Research Centre, University of Verona, Verona, Italy
| |
Collapse
|
47
|
Wei Y, Chen X, Yang J, Yao J, Yin N, Zhang Z, Li D, Zhu D, Zhou J. DcR3 promotes proliferation and invasion of pancreatic cancer via a DcR3/STAT1/IRF1 feedback loop. Am J Cancer Res 2019; 9:2618-2633. [PMID: 31911850 PMCID: PMC6943350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 11/21/2019] [Indexed: 06/10/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most common gastrointestinal malignancies that are highly aggressive with a low 5-year survival rate. Accumulated evidence has indicated that decoy receptor 3 (DcR3) is involved in several pathologic processes and various cancers. However, the mechanisms underlying dysregulated DcR3 expression and activation in PC remain to be fully established. In this study, we investigate the function and regulatory network of DcR3 in PC. We found that DcR3 was upregulated in PC tissues and serum. High DcR3 expression was associated with aggressive clinicopathological features and poor prognosis. Functionally, DcR3 not only increased cell migration and invasion in vitro but also promoted tumour growth both in vitro and in vivo by loss-of-function and gain-of-function experiments. Mechanistically, DcR3 promoted the phosphorylation of signal transducers and activators of transcription 1 (STAT1), leading to a dramatic increase in interferon regulatory factor 1 (IRF1). IRF1 then increased the transcriptional activity of DcR3, forming a positive feedback loop to reinforce DcR3 expression. In addition, DcR3 promoted carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) expression through activated IRF1. In conclusion, our findings provided novel insights into the function and mechanism of DcR3 in the pathogenesis of PC, which may be a potential therapeutic target for PC.
Collapse
Affiliation(s)
- Yijun Wei
- Pancreatic Disease Research Centre, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
- Department of General Surgery, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Xingyu Chen
- Taizhou Fourth People’s HospitalTaizhou 225300, Jiangsu, China
| | - Jian Yang
- Pancreatic Disease Research Centre, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
- Department of General Surgery, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Jun Yao
- Pancreatic Disease Research Centre, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
- Department of General Surgery, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Ni Yin
- Department of Oncology, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Zixiang Zhang
- Pancreatic Disease Research Centre, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
- Department of General Surgery, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Dechun Li
- Pancreatic Disease Research Centre, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
- Department of General Surgery, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Dongming Zhu
- Pancreatic Disease Research Centre, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
- Department of General Surgery, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Jian Zhou
- Pancreatic Disease Research Centre, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
- Department of General Surgery, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| |
Collapse
|
48
|
Jeon JM, Kwon OK, Na AY, Sung EJ, Cho IJ, Kim M, Yea SS, Chun SY, Lee JH, Ha YS, Kwon TG, Lee S. Secretome profiling of PC3/nKR cells, a novel highly migrating prostate cancer subline derived from PC3 cells. PLoS One 2019; 14:e0220807. [PMID: 31404090 PMCID: PMC6690527 DOI: 10.1371/journal.pone.0220807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/23/2019] [Indexed: 11/19/2022] Open
Abstract
Prostate cancer (PCa) is the most common cancer among men worldwide. Most PCa cases are not fatal; however, the outlook is poor when PCa spreads to another organ. Bone is the target organ in about 80% of patients who experience metastasis from a primary PCa tumor. In the present study, we characterized the secretome of PC3/nKR cells, which are a new subline of PC3 cells that were originally isolated from nude mice that were implanted with PC3 cells without anti-natural killer (NK) cell treatment. Wound healing and Transwell assays revealed that PC3/nKR cells had increased migratory and invasive activities in addition to a higher resistance to NK cells-induced cytotoxicity as compared to PC3 cells. We quantitatively profiled the secreted proteins of PC3/nKR and PC3 cells by liquid chromatography-tandem mass spectrometry analysis coupled with 2-plex tandem mass tag labeling. In total, 598 secretory proteins were identified, and 561 proteins were quantified, among which 45 proteins were secreted more and 40 proteins were secreted less by PC3/nKR cells than by PC3 cells. For validation, the adapter molecule crk, serpin B3, and cystatin-M were analyzed by western blotting. PC3/nKR cells showed the selective secretion of NKG2D ligand 2, HLA-A, and IL-6, which may contribute to their NK cell-mediated cytotoxicity resistance, and had a high secretion of crk protein, which may contribute to their high migration and invasion properties. Based on our secretome analysis, we propose that PC3/nKR cells represent a new cell system for studying the metastasis and progression of PCa.
Collapse
Affiliation(s)
- Ju Mi Jeon
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Oh Kwang Kwon
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Ann-Yae Na
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Eun Ji Sung
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Il Je Cho
- College of Korean Medicine, Daegu Haany University, Gyeongsan, Gyeongsangbuk-do, Republic of Korea
| | - Mirae Kim
- Department of Biochemistry, College of Medicine, Inje University, Busan, Republic of Korea
| | - Sung Su Yea
- Department of Biochemistry, College of Medicine, Inje University, Busan, Republic of Korea
| | - So Young Chun
- Joint Institute for Regenerative Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jun Hyung Lee
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Urology, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Yun-Sok Ha
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Urology, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Tae Gyun Kwon
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Urology, Kyungpook National University Hospital, Daegu, Republic of Korea
- * E-mail: ;
| | - Sangkyu Lee
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
- * E-mail: ;
| |
Collapse
|
49
|
da Cunha BR, Domingos C, Stefanini ACB, Henrique T, Polachini GM, Castelo-Branco P, Tajara EH. Cellular Interactions in the Tumor Microenvironment: The Role of Secretome. J Cancer 2019; 10:4574-4587. [PMID: 31528221 PMCID: PMC6746126 DOI: 10.7150/jca.21780] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 05/25/2019] [Indexed: 02/06/2023] Open
Abstract
Over the past years, it has become evident that cancer initiation and progression depends on several components of the tumor microenvironment, including inflammatory and immune cells, fibroblasts, endothelial cells, adipocytes, and extracellular matrix. These components of the tumor microenvironment and the neoplastic cells interact with each other providing pro and antitumor signals. The tumor-stroma communication occurs directly between cells or via a variety of molecules secreted, such as growth factors, cytokines, chemokines and microRNAs. This secretome, which derives not only from tumor cells but also from cancer-associated stromal cells, is an important source of key regulators of the tumorigenic process. Their screening and characterization could provide useful biomarkers to improve cancer diagnosis, prognosis, and monitoring of treatment responses.
Collapse
Affiliation(s)
- Bianca Rodrigues da Cunha
- Department of Molecular Biology, School of Medicine of São José do Rio Preto/FAMERP, São José do Rio Preto, SP, Brazil
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, SP, Brazil
| | - Célia Domingos
- Department of Biomedical Sciences and Medicine, University of Algarve, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal
| | - Ana Carolina Buzzo Stefanini
- Department of Molecular Biology, School of Medicine of São José do Rio Preto/FAMERP, São José do Rio Preto, SP, Brazil
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, SP, Brazil
| | - Tiago Henrique
- Department of Molecular Biology, School of Medicine of São José do Rio Preto/FAMERP, São José do Rio Preto, SP, Brazil
| | - Giovana Mussi Polachini
- Department of Molecular Biology, School of Medicine of São José do Rio Preto/FAMERP, São José do Rio Preto, SP, Brazil
| | - Pedro Castelo-Branco
- Department of Biomedical Sciences and Medicine, University of Algarve, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal
- Algarve Biomedical Center, Gambelas, Faro, Portugal
| | - Eloiza Helena Tajara
- Department of Molecular Biology, School of Medicine of São José do Rio Preto/FAMERP, São José do Rio Preto, SP, Brazil
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, SP, Brazil
| |
Collapse
|
50
|
Singh M, Tian XJ, Donnenberg VS, Watson AM, Zhang J, Stabile LP, Watkins SC, Xing J, Sant S. Targeting the Temporal Dynamics of Hypoxia-Induced Tumor-Secreted Factors Halts Tumor Migration. Cancer Res 2019; 79:2962-2977. [PMID: 30952634 PMCID: PMC6548579 DOI: 10.1158/0008-5472.can-18-3151] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 02/01/2019] [Accepted: 03/29/2019] [Indexed: 12/17/2022]
Abstract
Targeting microenvironmental factors that foster migratory cell phenotypes is a promising strategy for halting tumor migration. However, lack of mechanistic understanding of the emergence of migratory phenotypes impedes pharmaceutical drug development. Using our three-dimensional microtumor model with tight control over tumor size, we recapitulated the tumor size-induced hypoxic microenvironment and emergence of migratory phenotypes in microtumors from epithelial breast cells and patient-derived primary metastatic breast cancer cells, mesothelioma cells, and lung cancer xenograft cells. The microtumor models from various patient-derived tumor cells and patient-derived xenograft cells revealed upregulation of tumor-secreted factors, including matrix metalloproteinase-9 (MMP9), fibronectin (FN), and soluble E-cadherin, consistent with clinically reported elevated levels of FN and MMP9 in patient breast tumors compared with healthy mammary glands. Secreted factors in the conditioned media of large microtumors induced a migratory phenotype in nonhypoxic, nonmigratory small microtumors. Subsequent mathematical analyses identified a two-stage microtumor progression and migration mechanism whereby hypoxia induces a migratory phenotype in the initialization stage, which then becomes self-sustained through a positive feedback loop established among the tumor-secreted factors. Computational and experimental studies showed that inhibition of tumor-secreted factors effectively halts microtumor migration despite tumor-to-tumor variation in migration kinetics, while inhibition of hypoxia is effective only within a time window and is compromised by tumor-to-tumor variation, supporting our notion that hypoxia initiates migratory phenotypes but does not sustain it. In summary, we show that targeting temporal dynamics of evolving microenvironments, especially tumor-secreted factors during tumor progression, can halt tumor migration. SIGNIFICANCE: This study uses state-of-the-art three-dimensional microtumor models and computational approaches to highlight the temporal dynamics of tumor-secreted microenvironmental factors in inducing tumor migration.
Collapse
Affiliation(s)
- Manjulata Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xiao-Jun Tian
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona
| | - Vera S Donnenberg
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, McGowan Institute for Regenerative Medicine, and UPMC-Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alan M Watson
- Center for Biologic Imaging, Center for Vaccine Research, and Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - JingYu Zhang
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Laura P Stabile
- Department of Pharmacology & Chemical Biology, UPMC-Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Simon C Watkins
- Center for Biologic Imaging and the Department of Cellular Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jianhua Xing
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.
- UPMC-Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania.
- UPMC-Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Bioengineering, Swanson School of Engineering, and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|