1
|
Zhang X, Liu SS, Ma J, Qu W. Secretory leukocyte protease inhibitor (SLPI) in cancer pathophysiology: Mechanisms of action and clinical implications. Pathol Res Pract 2023; 248:154633. [PMID: 37356220 DOI: 10.1016/j.prp.2023.154633] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/17/2023] [Accepted: 06/18/2023] [Indexed: 06/27/2023]
Abstract
Cancer is a multifaceted disorder frequently linked to the dysregulation of several biological processes. The SLPI is a multifunctional protein involved in the modulation of immunological response and the inhibition of protease activities. SLPI acts as an inhibitor of proteases, exerts antibacterial properties, and suppresses the transcription of proinflammatory genes through the nuclear factor-kappa B (NF-κB) pathway. The role of this protein as a regulatory agent has been implicated in various types of cancer. Recent research has revealed that SLPI upregulation in cancer cells enhances the metastatic capacity of epithelial malignancies, indicating the deleterious effects of this protein. Furthermore, SLPI interacts intricately with other cancer-promoting factors, including matrix metalloproteinase-2 (MMP-2), MMP-9, the NF-κB and Akt pathways, and the p53-upregulated modulator of apoptosis (PUMA). This review provides an overview of the role of SLPI in cancer pathophysiology, emphasizing its expression in cancer cells and tissues, its potential as a prognostic biomarker, and its therapeutic promise as a target in cancer treatment. The mechanisms of SLPI action in cancer, including its anti-inflammatory effects, regulation of cell proliferation and angiogenesis, and modulation of the tumor microenvironment, have been investigated. The clinical implications of SLPI in cancer have been discussed, including its potential as a diagnostic and prognostic biomarker, its role in chemoresistance, and its therapeutic potential in several types of cancer, such as hepatocellular carcinoma (HCC), colorectal cancer (CRC), pancreatic cancer, head and neck squamous cell carcinoma (HNSCC), ovarian cancer (OvCa), prostate cancer (PC), gastric cancer (GC), breast cancer, and other cancers. In addition, we emphasized the significance of SLPI in cancer, which offers fresh perspectives on potential targets for cancer therapy.
Collapse
Affiliation(s)
- Xiaohua Zhang
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun 130000, China
| | - Shan Shan Liu
- Department of General Medicine, the Second Hospital of Jilin University, Changchun 130000, China.
| | - Jingru Ma
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun 130000, China
| | - Wei Qu
- Department of General Medicine, the Second Hospital of Jilin University, Changchun 130000, China
| |
Collapse
|
2
|
Hao L, Zhao RT, Welch NL, Tan EKW, Zhong Q, Harzallah NS, Ngambenjawong C, Ko H, Fleming HE, Sabeti PC, Bhatia SN. CRISPR-Cas-amplified urinary biomarkers for multiplexed and portable cancer diagnostics. NATURE NANOTECHNOLOGY 2023; 18:798-807. [PMID: 37095220 PMCID: PMC10359190 DOI: 10.1038/s41565-023-01372-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 03/10/2023] [Indexed: 05/03/2023]
Abstract
Synthetic biomarkers, bioengineered sensors that generate molecular reporters in diseased microenvironments, represent an emerging paradigm in precision diagnostics. Despite the utility of DNA barcodes as a multiplexing tool, their susceptibility to nucleases in vivo has limited their utility. Here we exploit chemically stabilized nucleic acids to multiplex synthetic biomarkers and produce diagnostic signals in biofluids that can be 'read out' via CRISPR nucleases. The strategy relies on microenvironmental endopeptidase to trigger the release of nucleic acid barcodes and polymerase-amplification-free, CRISPR-Cas-mediated barcode detection in unprocessed urine. Our data suggest that DNA-encoded nanosensors can non-invasively detect and differentiate disease states in transplanted and autochthonous murine cancer models. We also demonstrate that CRISPR-Cas amplification can be harnessed to convert the readout to a point-of-care paper diagnostic tool. Finally, we employ a microfluidic platform for densely multiplexed, CRISPR-mediated DNA barcode readout that can potentially evaluate complex human diseases rapidly and guide therapeutic decisions.
Collapse
Affiliation(s)
- Liangliang Hao
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Renee T Zhao
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nicole L Welch
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Program in Virology, Division of Medical Sciences, Harvard Medical School, Boston, MA, USA
| | - Edward Kah Wei Tan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Qian Zhong
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nour Saida Harzallah
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Chayanon Ngambenjawong
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Henry Ko
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Heather E Fleming
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Pardis C Sabeti
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
- Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Sangeeta N Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Nugteren S, den Uil SH, Delis-van Diemen PM, Simons-Oosterhuis Y, Lindenbergh-Kortleve DJ, van Haaften DH, Stockmann HBAC, Sanders J, Meijer GA, Fijneman RJA, Samsom JN. High expression of secretory leukocyte protease inhibitor (SLPI) in stage III micro-satellite stable colorectal cancer is associated with reduced disease recurrence. Sci Rep 2022; 12:12174. [PMID: 35842496 PMCID: PMC9288430 DOI: 10.1038/s41598-022-16427-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/11/2022] [Indexed: 12/02/2022] Open
Abstract
Secretory leukocyte protease inhibitor (SLPI) is a pleiotropic protein produced by healthy intestinal epithelial cells. SLPI regulates NF-κB activation, inhibits neutrophil proteases and has broad antimicrobial activity. Recently, increased SLPI expression was found in various types of carcinomas and was suggested to increase their metastatic potential. Indeed, we demonstrated that SLPI protein expression in colorectal cancer (CRC) liver metastases and matched primary tumors is associated with worse outcome, suggesting that SLPI promotes metastasis in human CRC. However, whether SLPI plays a role in CRC before distant metastases have formed is unclear. Therefore, we examined whether SLPI expression is associated with prognosis in CRC patients with localized disease. Using a cohort of 226 stage II and 160 stage III CRC patients we demonstrate that high SLPI protein expression is associated with reduced disease recurrence in patients with stage III micro-satellite stable tumors treated with adjuvant chemotherapy, independently of established clinical risk factors (hazard rate ratio 0.54, P-value 0.03). SLPI protein expression was not associated with disease-free survival in stage II CRC patients. Our data suggest that the role of SLPI in CRC may be different depending on the stage of disease. In stage III CRC, SLPI expression may be unfavorable for tumors, whereas SLPI expression may be beneficial for tumors once distant metastases have established.
Collapse
Affiliation(s)
- Sandrine Nugteren
- Division Gastroenterology and Nutrition, Laboratory of Pediatrics, Erasmus University Medical Center, Sophia Children's Hospital, Room Ee1567A, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Sjoerd H den Uil
- Department of Surgery, Spaarne Gasthuis, Haarlem, The Netherlands
| | | | - Ytje Simons-Oosterhuis
- Division Gastroenterology and Nutrition, Laboratory of Pediatrics, Erasmus University Medical Center, Sophia Children's Hospital, Room Ee1567A, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Dicky J Lindenbergh-Kortleve
- Division Gastroenterology and Nutrition, Laboratory of Pediatrics, Erasmus University Medical Center, Sophia Children's Hospital, Room Ee1567A, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Daniëlle H van Haaften
- Division Gastroenterology and Nutrition, Laboratory of Pediatrics, Erasmus University Medical Center, Sophia Children's Hospital, Room Ee1567A, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | | | - Joyce Sanders
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Gerrit A Meijer
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Remond J A Fijneman
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Janneke N Samsom
- Division Gastroenterology and Nutrition, Laboratory of Pediatrics, Erasmus University Medical Center, Sophia Children's Hospital, Room Ee1567A, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
4
|
Douglas TC, Hannila SS. Working from within: how secretory leukocyte protease inhibitor regulates the expression of pro-inflammatory genes. Biochem Cell Biol 2021; 100:1-8. [PMID: 34555292 DOI: 10.1139/bcb-2021-0284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Secretory leukocyte protease inhibitor (SLPI) is a small but powerful member of the serine protease inhibitor family, which includes proteins such as elafin and α1-antitrypsin. These proteins all have similar structures and antiprotease abilities, but SLPI has been found to have an additional role as an anti-inflammatory factor. It can inhibit the production of pro-inflammatory cytokines in cells stimulated with lipopolysaccharide, prevent neutrophil infiltration in murine models of lung and liver injury, and regulate the activity of the transcription factor NF-κB. In this review, we will revisit SLPI's unique biochemistry, and then explore how its anti-inflammatory functions can be linked to more recent findings showing that SLPI can localize to the nuclei of cells, bind DNA, and act as a regulator of gene expression.
Collapse
Affiliation(s)
- Tinsley Claire Douglas
- Department of Human Anatomy and Cell Science, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB R3E 0J9, Canada.,Department of Human Anatomy and Cell Science, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB R3E 0J9, Canada
| | - Sari S Hannila
- Department of Human Anatomy and Cell Science, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB R3E 0J9, Canada.,Department of Human Anatomy and Cell Science, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
5
|
Mizutani Y, Omagari D, Hayatsu M, Nameta M, Komiyama K, Mikami Y, Ushiki T. SLPI facilitates cell migration by regulating lamellipodia/ruffles and desmosomes, in which Galectin4 plays an important role. Cell Adh Migr 2021; 14:195-203. [PMID: 33016205 PMCID: PMC7553583 DOI: 10.1080/19336918.2020.1829264] [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] [Indexed: 10/29/2022] Open
Abstract
To elucidate the underlying mechanism of secretory leukocyte protease inhibitor (SLPI)-induced cell migration, we compared SLPI-deleted human gingival carcinoma Ca9-22 (ΔSLPI) cells and original (wild-type: wt) Ca9-22 cells using several microscopic imaging methods and gene expression analysis. Our results indicated reduced migration of ΔSLPI cells compared to wtCa9-22 cells. The lamellipodia/dorsal ruffles were smaller and moved slower in ΔSLPI cells compared to wtCa9-22 cells. Furthermore, well-developed intermediate filament bundles were observed at the desmosome junction of ΔSLPI cells. In addition, Galectin4 was strongly expressed in ΔSLPI cells, and its forced expression suppressed migration of wtCa9-22 cells. Taken together, SLPI facilitates cell migration by regulating lamellipodia/ruffles and desmosomes, in which Galectin4 plays an important role.
Collapse
Affiliation(s)
- Yusuke Mizutani
- Division of Microscopic Anatomy, Niigata University Graduate School of Medical and Dental Sciences , Niigata-shi, Japan.,Office of Institutional Research, Hokkaido University , Kita-ku, Japan
| | - Daisuke Omagari
- Department of Pathology, Nihon University School of Dentistry , Tokyo, Japan
| | - Manabu Hayatsu
- Division of Microscopic Anatomy, Niigata University Graduate School of Medical and Dental Sciences , Niigata-shi, Japan
| | - Masaaki Nameta
- Electron Microscope Core Facility, Niigata University , Niigata-shi, Japan
| | - Kazuo Komiyama
- Department of Pathology, Nihon University School of Dentistry , Tokyo, Japan
| | - Yoshikazu Mikami
- Division of Microscopic Anatomy, Niigata University Graduate School of Medical and Dental Sciences , Niigata-shi, Japan
| | - Tatsuo Ushiki
- Division of Microscopic Anatomy, Niigata University Graduate School of Medical and Dental Sciences , Niigata-shi, Japan
| |
Collapse
|
6
|
Mirmohseni Namini N, Abdollahi A, Movahedi M, Emami Razavi A, Saghiri R. HE4, A New Potential Tumor Marker for Early Diagnosis and Predicting of Breast Cancer Progression. IRANIAN JOURNAL OF PATHOLOGY 2021; 16:284-296. [PMID: 34306124 PMCID: PMC8298057 DOI: 10.30699/ijp.2021.135323.2482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 06/12/2021] [Indexed: 12/25/2022]
Abstract
Background & Objective: This study examined the potential of human epididymis protein 4 (HE4) as a marker in early diagnosis or as a prognostic factor for breast cancer (BC) patients. Methods: A total of 31 patients diagnosed with BC were enrolled in the study between 2008 and 2018. The mRNA and protein expression levels of HE4 were analyzed by immunohistochemistry (IHC) and real-time polymerase chain reaction (PCR) in the BC tissue and the non-tumoral adjacent tissue. Using ELISA technique, HE4 plasma levels were also measured in 43 BC patients compared to 43 healthy individuals. The correlation between HE4 expression and clinicopathological features was then investigated. Results: An increase in HE4 expression was observed at mRNA and protein levels in the BC group compared to the control group (P<0.01, P<0.0001, respectively). In addition, the relative expression of HE4 mRNA in BC patients showed a significant correlation with the differentiation grade of cancer cells (P<0.001). Plasma levels of HE4 was also associated with grade (P<0.0001), stage, and tumor size in BC patients (for both P<0.01). Patients with metastatic BC (P<0.01), lymphatic invasion, and lymph node involvement (for both P<0.05) showed significantly higher plasma levels of HE4 expression than patients without metastasis. Conclusion: According to our findings, upregulation of HE4 may be related to invasive BC phenotype. Measuring plasma levels of HE4 could be useful as a screening test in early diagnosis of BC.
Collapse
Affiliation(s)
- Nazanin Mirmohseni Namini
- Department of Biochemistry, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Alireza Abdollahi
- Department of Pathology, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Monireh Movahedi
- Department of Biochemistry, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Amirnader Emami Razavi
- Iran National Tumor Bank, Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Saghiri
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
7
|
Nugteren S, Samsom JN. Secretory Leukocyte Protease Inhibitor (SLPI) in mucosal tissues: Protects against inflammation, but promotes cancer. Cytokine Growth Factor Rev 2021; 59:22-35. [PMID: 33602652 DOI: 10.1016/j.cytogfr.2021.01.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 01/24/2021] [Indexed: 12/20/2022]
Abstract
The immune system is continuously challenged with large quantities of exogenous antigens at the barriers between the external environment and internal human tissues. Antimicrobial activity is essential at these sites, though the immune responses must be tightly regulated to prevent tissue destruction by inflammation. Secretory Leukocyte Protease Inhibitor (SLPI) is an evolutionarily conserved, pleiotropic protein expressed at mucosal surfaces, mainly by epithelial cells. SLPI inhibits proteases, exerts antimicrobial activity and inhibits nuclear factor-kappa B (NF-κB)-mediated inflammatory gene transcription. SLPI maintains homeostasis at barrier tissues by preventing tissue destruction and regulating the threshold of inflammatory immune responses, while protecting the host from infection. However, excessive expression of SLPI in cancer cells may have detrimental consequences, as recent studies demonstrate that overexpression of SLPI increases the metastatic potential of epithelial tumors. Here, we review the varied functions of SLPI in the respiratory tract, skin, gastrointestinal tract and genitourinary tract, and then discuss the mechanisms by which SLPI may contribute to cancer.
Collapse
Affiliation(s)
- Sandrine Nugteren
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Janneke N Samsom
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
8
|
Serrero G. Progranulin/GP88, A Complex and Multifaceted Player of Tumor Growth by Direct Action and via the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:475-498. [PMID: 34664252 DOI: 10.1007/978-3-030-73119-9_22] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Investigation of the role of progranulin/GP88 on the proliferation and survival of a wide variety of cells has been steadily increasing. Several human diseases stem from progranulin dysregulation either through its overexpression in cancer or its absence as in the case of null mutations in some form of frontotemporal dementia. The present review focuses on the role of progranulin/GP88 in cancer development, progression, and drug resistance. Various aspects of progranulin identification, biology, and signaling pathways will be described. Information will be provided about its direct role as an autocrine growth and survival factor and its paracrine effect as a systemic factor as well as via interaction with extracellular matrix proteins and with components of the tumor microenvironment to influence drug resistance, migration, angiogenesis, inflammation, and immune modulation. This chapter will also describe studies examining progranulin/GP88 tumor tissue expression as well as circulating level as a prognostic factor for several cancers. Due to the wealth of publications in progranulin, this review does not attempt to be exhaustive but rather provide a thread to lead the readers toward more in-depth exploration of this fascinating and unique protein.
Collapse
|
9
|
Wang YL, Zhang YY. cg04448376, cg24387542, cg08548498, and cg14621323 as a Novel Signature to Predict Prognosis in Kidney Renal Papillary Cell Carcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4854390. [PMID: 33381555 PMCID: PMC7759405 DOI: 10.1155/2020/4854390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/19/2020] [Accepted: 11/28/2020] [Indexed: 10/26/2022]
Abstract
INTRODUCTION DNA methylation plays a vital role in prognosis prediction of cancers. In this study, we aimed to identify novel DNA methylation site biomarkers and create an efficient methylated site model for predicting survival in kidney renal papillary cell carcinoma (KIRP). METHODS DNA methylation and gene expression profile data were downloaded from The Cancer Genome Atlas (TCGA) database and the Gene Expression Omnibus (GEO) database. Differential methylated genes (DMGs) and differential expression genes (DEGs) were identified and then searched for the hub genes. Cox proportional hazards regression was applied to identify DNA methylated site biomarkers from the hub genes. Kaplan-Meier survival and ROC analyses were used to validate the effective prognostic ability of the methylation gene site biomarker. The biomarker sites were validated in the GEO cohorts. The GO and KEGG annotation was done to explore the biological function of DNA methylated site signature. RESULTS Nine DMGs with opposite expression patterns containing 47 methylated sites were identified. Finally, four methylated sites were identified using the hazard regression model (cg04448376, cg24387542, cg08548498, and cg14621323) located in UTY, LGALS9B, SLPI, and PFN3, respectively. These sites classified patients into high- and low-risk groups in the training cohort. The 5-year survival rates for patients with low-risk and high-risk scores were 97.5% and 75.9% (P < 0.001). The prognostic accuracy and signature methylation sites were validated in the test (TCGA, n = 87) and GEO cohorts (n = 14). Multivariate regression analysis showed that the signature was an independent prediction prognostic factor for KIRP. Based on this analysis, we developed methylated site signature nomogram that predicts an individual's risk of survival. Functional analysis suggested that these signature genes are involved in the biological processes of protein binding. CONCLUSIONS Our study demonstrated that the methylated gene site signature might be a powerful prognostic tool for evaluating survival rate and guiding tailored therapy for KIRP patients.
Collapse
Affiliation(s)
- Ying-Lei Wang
- Department of Urinary Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, China
| | - Ying-Ying Zhang
- Out-patient Department, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, China
| |
Collapse
|
10
|
Yang Z, Chen J, Xie H, Liu T, Chen Y, Ma Z, Pei X, Yang W, Li L. Androgen receptor suppresses prostate cancer metastasis but promotes bladder cancer metastasis via differentially altering miRNA525-5p/SLPI-mediated vasculogenic mimicry formation. Cancer Lett 2019; 473:118-129. [PMID: 31843555 DOI: 10.1016/j.canlet.2019.12.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/20/2022]
Abstract
Early studies suggest that the androgen receptor (AR) may play differential roles in influencing prostate cancer (PCa) and bladder cancer (BCa) metastasis, but the underlying mechanisms remain unclear. Here, we found that the AR might function via differentially altering vasculogenic mimicry (VM) formation to either decrease PCa metastasis or increase BCa metastasis. Mechanism dissection showed that the AR could differentially alter the expression of the VM marker SLPI through miR-525-5p to regulate SLPI; moreover, it could either increase miR-525-5p transcription in PCa or decrease it in BCa via binding to different androgen-response-elements (AREs) located at different positions in the miR-525 precursor promoter. Further, results from liquid chromatography-mass spectrometry (LC-MS) showed that the co-factors of AR in PCa and BCa are NFIX and HDAC2, respectively. Together, these results provide the first detailed mechanism of how the AR can differentially alter PCa and BCa metastasis; thus, targeting the newly identified AR-miR-525-5p-SLPI axis may help suppress metastasis.
Collapse
Affiliation(s)
- Zhao Yang
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jiaqi Chen
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Hongjun Xie
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Tianjie Liu
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yule Chen
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhenkun Ma
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xinqi Pei
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Wenjie Yang
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lei Li
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
11
|
Ajila V, Babu S, Shetty V, Hegde S, Shenoy S. Secretory Leukocyte Protease Inhibitor in Oral Potentially Malignant Disorders and Oral Squamous Cell Carcinoma. Indian J Med Paediatr Oncol 2019. [DOI: 10.4103/ijmpo.ijmpo_75_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Abstract
Objectives: Secretory leukocyte protease inhibitor (SLPI) is a protein found in body fluids and implicated as a prognostic factor in many cancers. There is limited research regarding serum and salivary SLPI in oral potentially malignant disorders (OPMDs) and oral squamous cell carcinoma (OSCC). Materials and Methods: A total of 90 participants were included in the study; of which 30 formed the control group, 30 included participants with OPMD, and 30 included participants with OSCC. Serum and salivary levels of SLPI were estimated using enzyme-linked immunosorbent assay. Results: Serum levels of SLPI showed progressive increase in potentially malignant disorders and SCC patients when compared with controls, while salivary levels were increased in both OPMD and OSCC groups as compared to controls. However, the difference between groups was not statistically significant (P < 0.05). Conclusion: The above results suggest that increase in SLPI levels in the serum may have a role in indicating progression of OPMD to OSCC.
Collapse
Affiliation(s)
- Vidya Ajila
- Department of Oral Medicine and Radiology, A. B. Shetty Memorial Institute of Dental Sciences, Nitte Deemed to be University
| | - Subhas Babu
- Department of Oral Medicine and Radiology, A. B. Shetty Memorial Institute of Dental Sciences, Nitte Deemed to be University
| | - Veena Shetty
- Department of Microbiology, K. S. Hegde Medical Academy, Nitte Deemed to be University
| | - Shruthi Hegde
- Department of Oral Medicine and Radiology, A. B. Shetty Memorial Institute of Dental Sciences, Nitte Deemed to be University
| | - Shilpa Shenoy
- Central Research Laboratory, K. S. Hegde Medical Academy, Nitte Deemed to be University, Mangalore, Karnataka, India
| |
Collapse
|
12
|
Chen BJ, Tang YJ, Tang YL, Liang XH. What makes cells move: Requirements and obstacles for leader cells in collective invasion. Exp Cell Res 2019; 382:111481. [PMID: 31247191 DOI: 10.1016/j.yexcr.2019.06.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/15/2019] [Accepted: 06/23/2019] [Indexed: 02/05/2023]
Abstract
Most recently, mounting evidence has shown that cancer cells can invade as a cohesive and multicellular group with coordinated movement, which is called collective invasion. In this cohesive cancer cell group, cancer cells at the front of collective invasion are defined as leader cell that are responsible for many aspects of collective invasion, including sensing the microenvironment, determining the invasion direction, modifying the path of invasion and transmitting information to other cells. To fulfill their dispensable roles, leader cells are required to embark on some specific phenotypes with unusual expression of some proteins and it's very important to investigate into these proteins as they may serve as potential therapeutic targets. Here, in this review we will summarize current knowledge on four emerging proteins highly expressed in leader cells including K14, ΔNp63α, Dll4 and cysteine protease cathepsin B (CTSB), with a focus on their important roles in collective invasion and special mechanisms by which they promote collective invasion.
Collapse
Affiliation(s)
- Bing-Jun Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, China.
| | - Ya-Jie Tang
- Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China.
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral Pathology, West China Hospital of Stomatology, Sichuan University.China.
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, China.
| |
Collapse
|
13
|
Wu Z, Wu Y, Fischer J, Bartels J, Schröder JM, Meyer-Hoffert U. Skin-Derived SPINK9 Kills Escherichia coli. J Invest Dermatol 2019; 139:1135-1142. [DOI: 10.1016/j.jid.2018.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 11/01/2018] [Accepted: 11/04/2018] [Indexed: 01/17/2023]
|
14
|
Alur VC, Raju V, Vastrad B, Vastrad C. Mining Featured Biomarkers Linked with Epithelial Ovarian CancerBased on Bioinformatics. Diagnostics (Basel) 2019; 9:diagnostics9020039. [PMID: 30970615 PMCID: PMC6628368 DOI: 10.3390/diagnostics9020039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/31/2019] [Accepted: 04/05/2019] [Indexed: 11/16/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the18th most common cancer worldwide and the 8th most common in women. The aim of this study was to diagnose the potential importance of, as well as novel genes linked with, EOC and to provide valid biological information for further research. The gene expression profiles of E-MTAB-3706 which contained four high-grade ovarian epithelial cancer samples, four normal fallopian tube samples and four normal ovarian epithelium samples were downloaded from the ArrayExpress database. Pathway enrichment and Gene Ontology (GO) enrichment analysis of differentially expressed genes (DEGs) were performed, and protein-protein interaction (PPI) network, microRNA-target gene regulatory network and TFs (transcription factors) -target gene regulatory network for up- and down-regulated were analyzed using Cytoscape. In total, 552 DEGs were found, including 276 up-regulated and 276 down-regulated DEGs. Pathway enrichment analysis demonstrated that most DEGs were significantly enriched in chemical carcinogenesis, urea cycle, cell adhesion molecules and creatine biosynthesis. GO enrichment analysis showed that most DEGs were significantly enriched in translation, nucleosome, extracellular matrix organization and extracellular matrix. From protein-protein interaction network (PPI) analysis, modules, microRNA-target gene regulatory network and TFs-target gene regulatory network for up- and down-regulated, and the top hub genes such as E2F4, SRPK2, A2M, CDH1, MAP1LC3A, UCHL1, HLA-C (major histocompatibility complex, class I, C), VAT1, ECM1 and SNRPN (small nuclear ribonucleoprotein polypeptide N) were associated in pathogenesis of EOC. The high expression levels of the hub genes such as CEBPD (CCAAT enhancer binding protein delta) and MID2 in stages 3 and 4 were validated in the TCGA (The Cancer Genome Atlas) database. CEBPD andMID2 were associated with the worst overall survival rates in EOC. In conclusion, the current study diagnosed DEGs between normal and EOC samples, which could improve our understanding of the molecular mechanisms in the progression of EOC. These new key biomarkers might be used as therapeutic targets for EOC.
Collapse
Affiliation(s)
- Varun Chandra Alur
- Department of Endocrinology, J.J. M Medical College, Davanagere, Karnataka 577004, India.
| | - Varshita Raju
- Department of Obstetrics and Gynecology, J.J. M Medical College, Davanagere, Karnataka 577004, India.
| | - Basavaraj Vastrad
- Department of Pharmaceutics, SET`S College of Pharmacy, Dharwad, Karnataka 580002, India.
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics,Chanabasava Nilaya, Bharthinagar,Dharwad, Karanataka 580001, India.
| |
Collapse
|
15
|
Ajila V, Shetty V, Subhas B, Hegde S. Secretory leukocyte protease inhibitor and its role in virus induced head and neck cancers. ACTA STOMATOLOGICA NAISSI 2019. [DOI: 10.5937/asn1979936a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
16
|
Takamura T, Suguro H, Mikami Y, Iwase T, Komiyama Y, Kuyama K, Komiyama K, Oki H. Comparison of gene expression profiles of gingival carcinoma Ca9-22 cells and colorectal adenocarcinoma HT-29 cells to identify potentially important mediators of SLPI-induced cell migration. J Oral Sci 2018. [PMID: 28637988 DOI: 10.2334/josnusd.16-0534] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Secretory leukocyte protease inhibitor (SLPI) is a serine protease inhibitor whose expression level is positively correlated with tumor aggressiveness and metastatic potential. However, the mechanism underlying SLPI-induced enhancement of malignant phenotype is not completely understood. The malignancy of cancer cells is highly dependent on cell migration activity. Our previous study revealed that gingival carcinoma Ca9-22 cells, but not colorectal adenocarcinoma HT-29 cells, expressed SLPI. Therefore, we investigated the migration activity of these two cell types to understand the nature of SLPI-mediated tumor aggressiveness and metastatic potential. In vitro wound healing assay indicated that HT-29 cells and SLPI-deleted Ca9-22 cells showed lower migration activity than wild-type Ca9-22 cells, suggesting that SLPI-induced cell migration plays an important role in tumor aggressiveness and metastatic potential. In addition, our gene expression profiling study based on microarray data for the three cell types identified a number of candidates, including LCP1 and GLI, that could be key molecules in the mechanism of SLPI-induced cell migration.
Collapse
Affiliation(s)
- Tsuyoshi Takamura
- Department of Oral and Maxillofacial Surgery, Nihon University School of Dentistry
| | - Hisashi Suguro
- Department of Endodontics, Nihon University School of Dentistry
| | - Yoshikazu Mikami
- Division of Microscopic Anatomy, Niigata University Graduate School of Medical and Dental Sciences
| | - Takashi Iwase
- Department of Pathology, Nihon University School of Dentistry
| | - Yusuke Komiyama
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine
| | - Kayo Kuyama
- Department of Oral Pathology, Nihon University School of Dentistry at Matsudo
| | - Kazuo Komiyama
- Department of Pathology, Nihon University School of Dentistry
| | - Hiderou Oki
- Department of Oral and Maxillofacial Surgery, Nihon University School of Dentistry
| |
Collapse
|
17
|
Kozin SV, Maimon N, Wang R, Gupta N, Munn L, Jain RK, Garkavtsev I. Secretory leukocyte protease inhibitor (SLPI) as a potential target for inhibiting metastasis of triple-negative breast cancers. Oncotarget 2017; 8:108292-108302. [PMID: 29312532 PMCID: PMC5752445 DOI: 10.18632/oncotarget.22660] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/30/2017] [Indexed: 01/29/2023] Open
Abstract
SLPI has been implicated in the progression and metastasis of certain cancers. However, the effects of SLPI seem to be tumor-specific and the mechanisms remain poorly defined. Here, we demonstrate that highly metastatic, triple-negative breast cancer (TNBC) 4T1 cells secreted more SLPI compared to their non-metastatic counterparts. Furthermore, SLPI secretion directly correlated with spontaneous lung metastasis from 4T1 tumors orthotopically implanted in mice. Consistent with our experimental results, we also found that higher SLPI expression levels correlate with worse clinical outcome in basal/TNBC patients. Using high-throughput screening we identified a novel compound, C74, which significantly inhibits SLPI secretion. C74 administration in our mouse model slows the growth of primary 4T1 tumors and inhibits their dissemination to the lung. We also discovered that SLPI physically interacts with the retinoblastoma tumor suppressor protein (Rb) and releases FoxM1 from the Rb-FoxM1 complex, which may activate FoxM1 target genes involved in breast cancer metastasis.
Collapse
Affiliation(s)
- Sergey V Kozin
- Department of Radiation Oncology, Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Nir Maimon
- Department of Radiation Oncology, Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Rong Wang
- Department of Radiation Oncology, Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Nisha Gupta
- Department of Radiation Oncology, Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Lance Munn
- Department of Radiation Oncology, Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Rakesh K Jain
- Department of Radiation Oncology, Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Igor Garkavtsev
- Department of Radiation Oncology, Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
18
|
Small DM, Doherty DF, Dougan CM, Weldon S, Taggart CC. The role of whey acidic protein four-disulfide-core proteins in respiratory health and disease. Biol Chem 2017; 398:425-440. [PMID: 27930359 DOI: 10.1515/hsz-2016-0262] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 10/13/2016] [Indexed: 11/15/2022]
Abstract
Members of the whey acidic protein (WAP) or WAP four-disulfide-core (WFDC) family of proteins are a relatively under-explored family of low molecular weight proteins. The two most prominent WFDC proteins, secretory leukocyte protease inhibitor (SLPI) and elafin (or the precursor, trappin-2), have been shown to possess multiple functions including anti-protease, anti-bacterial, anti-viral and anti-inflammatory properties. It is therefore of no surprise that both SLPI and elafin/trappin-2 have been developed as potential therapeutics. Given the abundance of SLPI and elafin/trappin-2 in the human lung, most work in the area of WFDC research has focused on the role of WFDC proteins in protecting the lung from proteolytic attack. In this review, we will outline the current evidence regarding the expanding role of WFDC protein function with a focus on WFDC activity in lung disease as well as emerging data regarding the function of some of the more recently described WFDC proteins.
Collapse
|
19
|
Chen Y, Huang L, Wang S, Liu T, Wu Y, Li JL, Li M. WAP four-disulfide core domain protein 2 promotes metastasis of human ovarian cancer by regulation of metastasis-associated genes. J Ovarian Res 2017; 10:40. [PMID: 28679402 PMCID: PMC5499000 DOI: 10.1186/s13048-017-0329-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 04/24/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND WAP four-disulfide core domain protein 2 (WFDC2) shows a tumor-restricted upregulated pattern of expression in ovarian cancer. METHODS In this study, we evaluated the role of WFCD2 in tumor mobility, invasion and metastasis of ovarian cancer in clinical tissue and in ovarian cancer cells, both in vitro and in vivo. RESULTS Our results revealed WFCD2 was overexpressed in ovarian tissues, and the expression level of WFCD2 was associated with metastasis and lymph node metastasis. Higher expression of WFCD2 was also observed in aggressive HO8910-PM cells than in HO8910 cells, and WFCD2 knockdown halted cell migration, invasion, tumorigenicity and metastasis in ovarian cancer cells, both in vitro and in vivo. Knockdown of WFDC2 induced the down-regulation of ICAM-1, CD44, and MMP2. CONCLUSION In summary, our work demonstrates that WFCD2 promotes metastasis in ovarian cancer. These findings suggest that WFCD2 plays a critical role in promoting metastasis and may constitute a potential therapeutic target of ovarian cancer.
Collapse
Affiliation(s)
- Yao Chen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China.,State Key Laboratory of Organ Failure, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou, 510515, China.,Institute of Antibody Engineering, Southern Medical University, 1023 Shatainan Road, Guangzhou, 510515, China
| | - Liping Huang
- Obstetrics and Gynecology Center, Nanfang Hospital, Guangzhou, 510515, China
| | - Suihai Wang
- Institute of Antibody Engineering, Southern Medical University, 1023 Shatainan Road, Guangzhou, 510515, China
| | - Tiancai Liu
- Institute of Antibody Engineering, Southern Medical University, 1023 Shatainan Road, Guangzhou, 510515, China
| | - Yingsong Wu
- Institute of Antibody Engineering, Southern Medical University, 1023 Shatainan Road, Guangzhou, 510515, China.
| | - Ji-Liang Li
- Institute of Antibody Engineering, Southern Medical University, 1023 Shatainan Road, Guangzhou, 510515, China. .,Department of Oncology, Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK.
| | - Ming Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China. .,State Key Laboratory of Organ Failure, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou, 510515, China. .,Institute of Antibody Engineering, Southern Medical University, 1023 Shatainan Road, Guangzhou, 510515, China.
| |
Collapse
|
20
|
Gadducci A, Cosio S, Zanca G, Genazzani AR. Evolving Role of Serum Biomarkers in the Management of Ovarian Cancer. WOMENS HEALTH 2016; 2:141-58. [DOI: 10.2217/17455057.2.1.141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The availability of an ideal serum tumor marker would be of great clinical benefit for both the diagnosis and management of patients with epithelial ovarian cancer. Serum cancer antigen 125 assay significantly increases the diagnostic reliability of ultrasound in discriminating a malignant from a benign ovarian mass, especially in postmenopausal women, and it is the only well validated tumor marker for monitoring disease course. Several other tumor-associated antigens have been assessed, including glycoprotein antigens other than cancer antigen 125, soluble cytokeratin fragments, kallikreins, cytokines and cytokine receptors, vascular endothelial growth factor, D-dimer, and lisophosphatidic acid. This article assesses the potential diagnostic and prognostic role of these novel biomarkers, both alone and in combination with cancer antigen 125. The future for serum tumor marker research is represented by the emerging technology of proteomics, which may allow scientific advances comparable to those achieved with the introduction of monoclonal antibody technology.
Collapse
Affiliation(s)
- Angiolo Gadducci
- Department of Procreative Medicine, Division of Gynecology and Obstetrics, University of Pisa, Via Roma 56, Pisa, 56127, Italy, Tel.: +39 50 992 609; Fax: +39 50 553 410
| | - Stefania Cosio
- Department of Procreative Medicine, Division of Gynecology and Obstetrics, University of Pisa, Via Roma 56, Pisa, 56127, Italy, Tel.: +39 50 992 609; Fax: +39 50 553 410
| | - Giulia Zanca
- Department of Procreative Medicine, Division of Gynecology and Obstetrics, University of Pisa, Via Roma 56, Pisa, 56127, Italy, Tel.: +39 50 992 609; Fax: +39 50 553 410
| | - Andrea Riccardo Genazzani
- Department of Procreative Medicine, Division of Gynecology and Obstetrics, University of Pisa, Via Roma 56, Pisa, 56127, Italy, Tel.: +39 50 992 609; Fax: +39 50 553 410
| |
Collapse
|
21
|
Rostoker R, Ben-Shmuel S, Rashed R, Shen Orr Z, LeRoith D. CD24 cell surface expression in Mvt1 mammary cancer cells serves as a biomarker for sensitivity to anti-IGF1R therapy. Breast Cancer Res 2016; 18:51. [PMID: 27179633 PMCID: PMC4867988 DOI: 10.1186/s13058-016-0711-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 04/26/2016] [Indexed: 01/14/2023] Open
Abstract
Background The pro-tumorigenic effects of the insulin-like growth factor receptor (IGF1R) are well described. IGF1R promotes cancer cell survival and proliferation and prevents apoptosis, and, additionally it was shown that IGF1R levels are significantly elevated in most common human malignancies including breast cancer. However, results from phase 3 clinical trials in unselected patients demonstrated lack of efficacy for anti-IGF1R therapy. These findings suggest that predictive biomarkers are greatly warranted in order to identify patients that will benefit from anti-IGF1R therapeutic strategies. Methods Using the delivery of shRNA vectors into the Mvt1 cell line, we tested the role of the IGF1R in the development of mammary tumors. Based on CD24 cell surface expression, control and IGF1R-knockdown (IGF1R-KD) cells were FACS sorted into CD24− and CD24+ subsets and further characterized in vitro. The tumorigenic capacity of each was determined following orthotopic inoculation into the mammary fat pad of female mice. Tumor cells were FACS characterized upon sacrifice to determine IGF1R effect on the plasticity of this cell’s phenotype. Metastatic capacity of the cells was assessed using the tail vein assay. Results In this study we demonstrate that downregulation of the IGF1R specifically in cancer cells expressing CD24 on the cell surface membrane affect both their morphology (from mesenchymal-like into epithelial-like morphology) and phenotype in vitro. Moreover, we demonstrate that IGF1R-KD abolished both CD24+ cells capacity to form mammary tumors and lung metastatic lesions. We found in both cells and tumors a marked upregulation in CTFG and a significant reduction of SLP1 expression in the CD24+/IGF1R-KD; tumor-suppressor and tumor-promoting genes respectively. Moreover, we demonstrate here that the IGF1R is essential for the maintenance of stem/progenitor-like cancer cells and we further demonstrate that IGF1R-KD induces in vivo differentiation of the CD24+ cells toward the CD24- phenotype. This further supports the antitumorigenic effects of IGF1R-KD, as we recently published that these differentiated cells demonstrate significantly lower tumorigenic capacity compared with their CD24+ counterparts. Conclusions Taken together these findings suggest that CD24 cell surface expression may serve as a valuable biomarker in order to identify mammary tumors that will positively respond to targeted IGF1R therapies. Electronic supplementary material The online version of this article (doi:10.1186/s13058-016-0711-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ran Rostoker
- Diabetes and Metabolism Clinical Research Center of Excellence, Clinical Research Institute at Rambam (CRIR), Rambam Medical Center, Haifa, Israel
| | - Sarit Ben-Shmuel
- Diabetes and Metabolism Clinical Research Center of Excellence, Clinical Research Institute at Rambam (CRIR), Rambam Medical Center, Haifa, Israel
| | - Rola Rashed
- Diabetes and Metabolism Clinical Research Center of Excellence, Clinical Research Institute at Rambam (CRIR), Rambam Medical Center, Haifa, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Zila Shen Orr
- Diabetes and Metabolism Clinical Research Center of Excellence, Clinical Research Institute at Rambam (CRIR), Rambam Medical Center, Haifa, Israel
| | - Derek LeRoith
- Diabetes and Metabolism Clinical Research Center of Excellence, Clinical Research Institute at Rambam (CRIR), Rambam Medical Center, Haifa, Israel. .,Department of Medicine, Icahn School of Medicine at Mt Sinai, New York City, NY, USA.
| |
Collapse
|
22
|
Secretory leukocyte protease inhibitor is a survival and proliferation factor for castration-resistant prostate cancer. Oncogene 2016; 35:4807-15. [DOI: 10.1038/onc.2016.13] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 12/23/2015] [Accepted: 12/27/2015] [Indexed: 12/22/2022]
|
23
|
Polyclonal breast cancer metastases arise from collective dissemination of keratin 14-expressing tumor cell clusters. Proc Natl Acad Sci U S A 2016; 113:E854-63. [PMID: 26831077 DOI: 10.1073/pnas.1508541113] [Citation(s) in RCA: 532] [Impact Index Per Article: 59.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Recent genomic studies challenge the conventional model that each metastasis must arise from a single tumor cell and instead reveal that metastases can be composed of multiple genetically distinct clones. These intriguing observations raise the question: How do polyclonal metastases emerge from the primary tumor? In this study, we used multicolor lineage tracing to demonstrate that polyclonal seeding by cell clusters is a frequent mechanism in a common mouse model of breast cancer, accounting for >90% of metastases. We directly observed multicolored tumor cell clusters across major stages of metastasis, including collective invasion, local dissemination, intravascular emboli, circulating tumor cell clusters, and micrometastases. Experimentally aggregating tumor cells into clusters induced a >15-fold increase in colony formation ex vivo and a >100-fold increase in metastasis formation in vivo. Intriguingly, locally disseminated clusters, circulating tumor cell clusters, and lung micrometastases frequently expressed the epithelial cytoskeletal protein, keratin 14 (K14). RNA-seq analysis revealed that K14(+) cells were enriched for desmosome and hemidesmosome adhesion complex genes, and were depleted for MHC class II genes. Depletion of K14 expression abrogated distant metastases and disrupted expression of multiple metastasis effectors, including Tenascin C (Tnc), Jagged1 (Jag1), and Epiregulin (Ereg). Taken together, our findings reveal K14 as a key regulator of metastasis and establish the concept that K14(+) epithelial tumor cell clusters disseminate collectively to colonize distant organs.
Collapse
|
24
|
Birse CE, Lagier RJ, FitzHugh W, Pass HI, Rom WN, Edell ES, Bungum AO, Maldonado F, Jett JR, Mesri M, Sult E, Joseloff E, Li A, Heidbrink J, Dhariwal G, Danis C, Tomic JL, Bruce RJ, Moore PA, He T, Lewis ME, Ruben SM. Blood-based lung cancer biomarkers identified through proteomic discovery in cancer tissues, cell lines and conditioned medium. Clin Proteomics 2015; 12:18. [PMID: 26279647 PMCID: PMC4537594 DOI: 10.1186/s12014-015-9090-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 07/07/2015] [Indexed: 12/18/2022] Open
Abstract
Background Support for early detection of lung cancer has emerged from the National Lung Screening Trial (NLST), in which low-dose computed tomography (LDCT) screening reduced lung cancer mortality by 20 % relative to chest x-ray. The US Preventive Services Task Force (USPSTF) recently recommended annual screening for the high-risk population, concluding that the benefits (life years gained) outweighed harms (false positive findings, abortive biopsy/surgery, radiation exposure). In making their recommendation, the USPSTF noted that the moderate net benefit of screening was dependent on the resolution of most false-positive results without invasive procedures. Circulating biomarkers may serve as a valuable adjunctive tool to imaging. Results We developed a broad-based proteomics discovery program, integrating liquid chromatography/mass spectrometry (LC/MS) analyses of freshly resected lung tumor specimens (n = 13), lung cancer cell lines (n = 17), and conditioned media collected from tumor cell lines (n = 7). To enrich for biomarkers likely to be found at elevated levels in the peripheral circulation of lung cancer patients, proteins were prioritized based on predicted subcellular localization (secreted, cell-membrane associated) and differential expression in disease samples. 179 candidate biomarkers were identified. Several markers selected for further validation showed elevated levels in serum collected from subjects with stage I NSCLC (n = 94), relative to healthy smoker controls (n = 189). An 8-marker model was developed (TFPI, MDK, OPN, MMP2, TIMP1, CEA, CYFRA 21–1, SCC) which accurately distinguished subjects with lung cancer (n = 50) from high risk smokers (n = 50) in an independent validation study (AUC = 0.775). Conclusions Integrating biomarker discovery from multiple sample types (fresh tissue, cell lines and conditioned medium) has resulted in a diverse repertoire of candidate biomarkers. This unique collection of biomarkers may have clinical utility in lung cancer detection and diagnoses. Electronic supplementary material The online version of this article (doi:10.1186/s12014-015-9090-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Charles E Birse
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Robert J Lagier
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - William FitzHugh
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Harvey I Pass
- Department of Cardiothoracic Surgery, NYU Langone Medical Center, 530 First Avenue, New York, NY USA
| | - William N Rom
- Division of Pulmonary, Critical Care, and Sleep Medicine, NYU School of Medicine, New York, NY USA
| | - Eric S Edell
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN USA
| | - Aaron O Bungum
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN USA
| | - Fabien Maldonado
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN USA
| | - James R Jett
- Division of Oncology, National Jewish Health, Denver, CO USA
| | - Mehdi Mesri
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Erin Sult
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Elizabeth Joseloff
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Aiqun Li
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Jenny Heidbrink
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Gulshan Dhariwal
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Chad Danis
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Jennifer L Tomic
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Robert J Bruce
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Paul A Moore
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Tao He
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Marcia E Lewis
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Steve M Ruben
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| |
Collapse
|
25
|
Nepomuceno AI, Shao H, Jing K, Ma Y, Petitte JN, Idowu MO, Muddiman DC, Fang X, Hawkridge AM. In-depth LC-MS/MS analysis of the chicken ovarian cancer proteome reveals conserved and novel differentially regulated proteins in humans. Anal Bioanal Chem 2015; 407:6851-63. [PMID: 26159569 DOI: 10.1007/s00216-015-8862-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 06/11/2015] [Accepted: 06/15/2015] [Indexed: 12/12/2022]
Abstract
Ovarian cancer (OVC) remains the most lethal gynecological malignancy in the world due to the combined lack of early-stage diagnostics and effective therapeutic strategies. The development and application of advanced proteomics technology and new experimental models has created unique opportunities for translational studies. In this study, we investigated the ovarian cancer proteome of the chicken, an emerging experimental model of OVC that develops ovarian tumors spontaneously. Matched plasma, ovary, and oviduct tissue biospecimens derived from healthy, early-stage OVC, and late-stage OVC birds were quantitatively characterized by label-free proteomics. Over 2600 proteins were identified in this study, 348 of which were differentially expressed by more than twofold (p ≤ 0.05) in early- and late-stage ovarian tumor tissue specimens relative to healthy ovarian tissues. Several of the 348 proteins are known to be differentially regulated in human cancers including B2M, CLDN3, EPCAM, PIGR, S100A6, S100A9, S100A11, and TPD52. Of particular interest was ovostatin 2 (OVOS2), a novel 165-kDa protease inhibitor found to be strongly upregulated in chicken ovarian tumors (p = 0.0005) and matched plasma (p = 0.003). Indeed, RT-quantitative PCR and Western blot analysis demonstrated that OVOS2 mRNA and protein were also upregulated in multiple human OVC cell lines compared to normal ovarian epithelia (NOE) cells and immunohistochemical staining confirmed overexpression of OVOS2 in primary human ovarian cancers relative to non-cancerous tissues. Collectively, these data provide the first evidence for involvement of OVOS2 in the pathogenesis of both chicken and human ovarian cancer.
Collapse
Affiliation(s)
- Angelito I Nepomuceno
- W.M. Keck FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, 2620 Yarbrough Dr., Box 8204, Raleigh, NC, 27695, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Secretory leukocyte protease inhibitor is a proliferation and survival factor for pancreatic cancer cells. Clin Transl Oncol 2014; 17:314-21. [PMID: 25319722 DOI: 10.1007/s12094-014-1232-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Accepted: 09/20/2014] [Indexed: 12/20/2022]
Abstract
OBJECTIVES A variety of inflammatory cytokines have been demonstrated to participate in tumorigenesis and progression. Secretory leukocyte protease inhibitor (SLPI) has been demonstrated to show a broad-spectrum of anti-inflammatory effects. This study investigates the expression of SLPI in human pancreatic cancer tissues and cells as well as its biological effects in human pancreatic cancer cells. METHODS Reverse transcription-polymerase chain reaction, immunohistochemistry, and Western blot were used to detect SLPI mRNA and protein levels in human pancreatic cancer tissues, adjacent tissues, and pancreatic cancer Bxpc-3 and Panc-1 cells. Knockout of SLPI expression was established by recombinant viral vector expressing short hairpin RNA (shRNA) targeting SLPI. Cell viability was analyzed by MTT assay. Cell apoptosis was detected by Hochest33258 staining and flow cytometry assay. RESULTS Higher SLPI expression was observed in pancreatic tissues, Bxpc-3 cells, and Panc-1 cells compared to the peritumoral tissues (p < 0.01). SLPI expression in Bxpc-3 and Panc-1 cells was effectively silenced by shRNA (p < 0.001). Silencing of SLPI expression significantly reduced cell viability, inhibited cell proliferation, and induced cell apoptosis (p < 0.001). CONCLUSIONS Abnormal over-expression of SLPI in pancreatic cancer cells may be associated with the development of disease through its roles in promoting cancer cell survival and proliferation as well as anti-apoptosis. SLPI can be used as a target for developing targeted therapy of pancreatic cancer.
Collapse
|
27
|
Sayers KT, Brooks AD, Sayers TJ, Chertov O. Increased secretory leukocyte protease inhibitor (SLPI) production by highly metastatic mouse breast cancer cells. PLoS One 2014; 9:e104223. [PMID: 25110884 PMCID: PMC4128660 DOI: 10.1371/journal.pone.0104223] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 07/11/2014] [Indexed: 02/04/2023] Open
Abstract
The precise molecular mechanisms enabling cancer cells to metastasize from the primary tumor to different tissue locations are still largely unknown. Secretion of some proteins by metastatic cells could facilitate metastasis formation. The comparison of secreted proteins from cancer cells with different metastatic capabilities in vivo might provide insight into proteins involved in the metastatic process. Comparison of the secreted proteins from the mouse breast cancer cell line 4T1 and its highly metastatic 4T1.2 clone revealed a prominent differentially secreted protein which was identified as SLPI (secretory leukocyte protease inhibitor). Western blotting indicated higher levels of the protein in both conditioned media and whole cell lysates of 4T1.2 cells. Additionally higher levels of SLPI were also observed in 4T1.2 breast tumors in vivo following immunohistochemical staining. A comparison of SLPI mRNA levels by gene profiling using microarrays and RT-PCR did not detect major differences in SLPI gene expression between the 4T1 and 4T1.2 cells indicating that SLPI secretion is regulated at the protein level. Our results demonstrate that secretion of SLPI is drastically increased in highly metastatic cells, suggesting a possible role for SLPI in enhancing the metastatic behavior of breast cancer cell line 4T1.
Collapse
Affiliation(s)
- Kevin T. Sayers
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Alan D. Brooks
- Cancer and Inflammation Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Thomas J. Sayers
- Cancer and Inflammation Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Oleg Chertov
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| |
Collapse
|
28
|
Jan Treda C, Fukuhara T, Suzuki T, Nakamura A, Zaini J, Kikuchi T, Ebina M, Nukiwa T. Secretory leukocyte protease inhibitor modulates urethane-induced lung carcinogenesis. Carcinogenesis 2013; 35:896-904. [PMID: 24282288 DOI: 10.1093/carcin/bgt382] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Secretory leukocyte protease inhibitor (SLPI), 11.7 kDa serine protease inhibitor, is produced primarily in the respiratory tract, but it is often elevated in lung, head/neck and ovarian cancers. SLPI expression in relation to cancer progression, metastasis and invasion has been studied extensively in non-small cell lung cancer. However, the role of SLPI during the early stages of carcinogenesis remains unknown. We hypothesized that SLPI is required from the initiation and promotion to the progression of lung carcinogenesis. A skin allograft model using SLPI-knockout (SLPI-KO) mice and short hairpin RNA-treated cells was used to demonstrate that SLPI expression in tumor cells is crucial for tumor formation. Moreover, lung tumorigenesis induced by urethane, a chemical lung carcinogen, was significantly suppressed in SLPI-KO mice in association with decreased nuclear factor-kappaB (NF-κB) activity. SLPI deficiency also resulted in decreased cell numbers and decreased production of inflammatory cytokines in bronchoalveolar lavage fluids. The suppression of NF-κB activation in SLPI-KO mice was associated with lower expression of NF-κB-related survival genes and DNA repair genes. Our findings demonstrate that SLPI plays an important role from the initial stages of lung carcinogenesis to the progression of lung cancer in an NF-κB-dependent manner.
Collapse
Affiliation(s)
- Cezary Jan Treda
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan and
| | | | | | | | | | | | | | | |
Collapse
|
29
|
HE4 transcription- and splice variants-specific expression in endometrial cancer and correlation with patient survival. Int J Mol Sci 2013; 14:22655-77. [PMID: 24252907 PMCID: PMC3856083 DOI: 10.3390/ijms141122655] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Revised: 10/14/2013] [Accepted: 10/15/2013] [Indexed: 12/14/2022] Open
Abstract
We investigated the HE4 variant-specific expression patterns in various normal tissues as well as in normal and malignant endometrial tissues. The relationships between mRNA variants and age, body weight, or survival are analyzed. ICAT-labeled normal and endometrial cancer (EC) tissues were analyzed with multidimensional liquid chromatography followed by tandem mass spectrometry. Levels of HE4 mRNA variants were measured by real-time PCR. Mean mRNA levels were compared among 16 normal endometrial samples, 14 grade 1 and 14 grade 3 endometrioid EC, 15 papillary serous EC, and 14 normal human tissue samples. The relationship between levels of HE4 variants and EC patient characteristics was analyzed with the use of Pearson correlation test. We found that, although all five HE4 mRNA variants are detectable in normal tissue samples, their expression is highly tissue-specific, with epididymis, trachea, breast and endometrium containing the highest levels. HE4-V0, -V1, and -V3 are the most abundant variants in both normal and malignant tissues. All variants are significantly increased in both endometrioid and papillary serous EC, with higher levels observed in grade 3 endometrioid EC. In the EC group, HE4-V1, -V3, and -V4 levels inversely correlate with EC patient survival, whereas HE4-V0 levels positively correlate with age. HE4 variants exhibit tissue-specific expression, suggesting that each variant may exert distinct functions in normal and malignant cells. HE4 levels appear to correlate with EC patient survival in a variant-specific manner. When using HE4 as a biomarker for EC management, the effects of age should be considered.
Collapse
|
30
|
Carlson AM, Maurer MJ, Goergen KM, Kalli KR, Erskine CL, Behrens MD, Knutson KL, Block MS. Utility of progranulin and serum leukocyte protease inhibitor as diagnostic and prognostic biomarkers in ovarian cancer. Cancer Epidemiol Biomarkers Prev 2013; 22:1730-5. [PMID: 23878295 DOI: 10.1158/1055-9965.epi-12-1368] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) is the fifth leading cause of cancer-related death in females and leading gynecologic cause of cancer-related death. Despite the identification of a number of serum biomarkers, methods to identify early-stage disease and predict prognosis remain scarce. We have evaluated two biologically connected serum biomarkers, serum leukocyte protease inhibitor (SLPI) and progranulin (PGRN). METHODS Two-hundred frozen plasma samples were acquired from the Mayo Clinic Biospecimen Repository for Ovarian Cancer Research. Samples were obtained from 50 patients with benign conditions, 50 with American Joint Committee on Cancer (AJCC) stage I and II EOC, and 100 with AJCC stage III and IV EOC. Samples were obtained before surgical resection of a mass and were analyzed for absolute levels of SLPI and PGRN using ELISA assays. Receiver-operator characteristic curves were generated for SLPI and PGRN. Median follow-up was 48 months. RESULTS Absolute levels of SLPI were significantly elevated in patients with EOC compared with benign disease and predicted the presence of EOC (AUC of 0.812; P = 0.04); SLPI remained elevated in the subset of patients with normal CA-125. PGRN levels were not significantly increased in patients with early-stage or late-stage EOC as a whole, but an increase in PGRN levels was associated with decreased overall survival in advanced EOC. CONCLUSIONS SLPI levels are elevated in EOC, and SLPI shows promise as a diagnostic biomarker for patients with both elevated and normal CA-125 levels. An increase in PGRN is associated with decreased overall survival. IMPACT SLPI is elevated in EOC and warrants investigation in a screening study in women at risk for EOC.
Collapse
Affiliation(s)
- Aaron M Carlson
- Authors' Affiliations: Mayo Medical School, Biomedical Statistics and Informatics, and Departments of Medical Oncology and Immunology, Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Bidinotto LT, de Cicco RL, Vanegas JE, Santucci-Pereira J, Vanden Heuvel JP, Washington S, Aliaga C, Xu H, Russo IH, Manni A, El-Bayoumy K, Russo J. Fish oil alters tamoxifen-modulated expression of mRNAs that encode genes related to differentiation, proliferation, metastasis, and immune response in rat mammary tumors. Nutr Cancer 2013; 64:991-9. [PMID: 23061905 DOI: 10.1080/01635581.2012.712736] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We have previously shown that a fish oil (FO)-rich diet increased the chemopreventive efficacy of tamoxifen (Tam) against N-methyl-N-nitrosourea (MNU)-induced rat mammary carcinogenesis. Herein, we provide evidence that Tam treatment modifies gene expression of mammary tumors depending upon the type of dietary fat fed to the animals. Rats initiated with MNU and treated with Tam were fed a diet rich in corn oil or FO. After 8 wk, cribriform tumors were collected and gene expression analysis was performed. Increased RNA expression of genes such as SerpinB10, Wisp2, and Apod in tumors from FO-treated rats is indicative of highly differentiated tumors. Decreased expression of H19 and Igf2 mRNA in Tam-treated groups, and Gamma Synuclein mRNA in the FO + Tam group may be related to tumor growth impairment and lower metastatic capacity. Change in the expression of genes associated with immunity in animals in the FO + Tam group may suggest a shift in the immune response. These data show that, although Tam modulates the expression of genes leading to tumor growth impairment, further modulations of genes are influenced by FO. FO modulation of Tam changes in gene expression accounts for its enhancing chemopreventive effect against MNU-induced mammary carcinogenesis. Supplemental materials are available for this article. Go to the publisher's online edition of Nutrition and Cancer to view the supplemental file.
Collapse
Affiliation(s)
- Lucas Tadeu Bidinotto
- Breast Cancer Research Laboratory, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
HE4 (WFDC2) Promotes Tumor Growth in Endometrial Cancer Cell Lines. Int J Mol Sci 2013; 14:6026-43. [PMID: 23502467 PMCID: PMC3634435 DOI: 10.3390/ijms14036026] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 02/07/2013] [Accepted: 02/25/2013] [Indexed: 12/13/2022] Open
Abstract
HE4, also known as WFDC2, is a useful biomarker for ovarian cancer when either used alone or in combination with CA125. HE4 is also overexpressed in endometrial cancer (EC), but its function in cancer cells is not clear. In this study, we investigate the role of HE4 in EC progression. An HE4-overexpression system was established by cloning the HE4 prototypic mRNA variant (HE4-V0) into a eukaryotic expression vector. Following transfection, stable clones in two EC cell lines were selected. The effects of HE4 overexpression on cell growth and function were measured with the use of cell proliferation assay, matrigel invasion, and soft agar gel colony formation assays. HE4-induced cancer cell proliferation in vivo was examined in a mouse xenograft model. HE4 overexpression significantly enhanced EC cell proliferation, matrigel invasion, and colony formation in soft agar. Moreover, HE4 overexpression promoted tumor growth in the mouse xenograft model. HE4 overexpression enhanced several malignant phenotypes in cell culture and in a mouse model. These results are consistent with our previous observation that high levels of serum HE4 closely correlate with the stage, myometrial invasion and tumor size in patients with EC. This study provides evidence that HE4 overexpression directly impacts tumor progression in endometrial cancer.
Collapse
|
33
|
Amiano NO, Costa MJ, Reiteri RM, Payés C, Guerrieri D, Tateosian NL, Sánchez ML, Maffia PC, Diament M, Karas R, Orqueda A, Rizzo M, Alaniz L, Mazzolini G, Klein S, Sallenave JM, Chuluyan HE. Anti-tumor effect of SLPI on mammary but not colon tumor growth. J Cell Physiol 2013; 228:469-75. [PMID: 22767220 DOI: 10.1002/jcp.24153] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Secretory leukocyte protease inhibitor (SLPI) is a serine protease inhibitor that was related to cancer development and metastasis dissemination on several types of tumors. However, it is not known the effect of SLPI on mammary and colon tumors. The aim of this study was to examine the effect of SLPI on mammary and colon tumor growth. The effect of SLPI was tested on in vitro cell apoptosis and in vivo tumor growth experiments. SLPI over-expressing human and murine mammary and colon tumor cells were generated by gene transfection. The administration of murine mammary tumor cells over-expressing high levels of SLPI did not develop tumors in mice. On the contrary, the administration of murine colon tumor cells over-expressing SLPI, developed faster tumors than control cells. Intratumoral, but not intraperitoneal administration of SLPI, delayed the growth of tumors and increased the survival of mammary but not colon tumor bearing mice. In vitro culture of mammary tumor cell lines treated with SLPI, and SLPI producer clones were more prone to apoptosis than control cells, mainly under serum deprivation culture conditions. Herein we demonstrated that SLPI induces the apoptosis of mammary tumor cells in vitro and decreases the mammary but not colon tumor growth in vivo. Therefore, SLPI may be a new potential therapeutic tool for certain tumors, such as mammary tumors.
Collapse
Affiliation(s)
- Nicolás O Amiano
- Department of Pharmacology, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Chhikara N, Saraswat M, Tomar AK, Dey S, Singh S, Yadav S. Human epididymis protein-4 (HE-4): a novel cross-class protease inhibitor. PLoS One 2012; 7:e47672. [PMID: 23139753 PMCID: PMC3489902 DOI: 10.1371/journal.pone.0047672] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 09/18/2012] [Indexed: 02/07/2023] Open
Abstract
Epididymal proteins represent the factors necessary for maturation of sperm and play a crucial role in sperm maturation. HE-4, an epididymal protein, is a member of whey acidic protein four-disulfide core (WFDC) family with no known function. A WFDC protein has a conserved WFDC domain of 50 amino acids with eight conserved cystine residue. HE-4 is a 124 amino acid long polypeptide with two WFDC domains. Here, we show that HE-4 is secreted in the human seminal fluid as a disulfide-bonded homo-trimer and is a cross-class protease inhibitor inhibits some of the serine, aspartyl and cysteine proteases tested using hemoglobin as a substrate. Using SPR we have also observed that HE-4 shows a significant binding with all these proteases. Disulfide linkages are essential for this activity. Moreover, HE-4 is N-glycosylated and highly stable on a wide range of pH and temperature. Taken together this suggests that HE-4 is a cross-class protease inhibitor which might confer protection against microbial virulence factors of proteolytic nature.
Collapse
Affiliation(s)
- Nirmal Chhikara
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Mayank Saraswat
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Anil Kumar Tomar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Sharmistha Dey
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Sarman Singh
- Department of Lab Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Savita Yadav
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
35
|
Chen Y, Mu X, Wang S, Zhao L, Wu Y, Li J, Li M. WAP four-disulfide core domain protein 2 mediates the proliferation of human ovarian cancer cells through the regulation of growth- and apoptosis-associated genes. Oncol Rep 2012; 29:288-96. [PMID: 23129262 DOI: 10.3892/or.2012.2114] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 09/28/2012] [Indexed: 11/06/2022] Open
Abstract
The WAP four-disulfide core domain protein 2 (WFDC2) is frequently overexpressed in epithelial ovarian cancer cells and has been proposed as a potential biomarker. The biological function of WFDC2 in tumor progression remains unclear. In this study, the stable expression of short hairpin RNA (shRNA) against WFDC2 in the human ovarian SKOV3 cell line was established. Cell proliferation in vitro was determined by MTT assay. Cell cycle and apoptosis were analyzed by FACS. The expression of genes related to cell proliferation and survival was detected by real-time RT-PCR and western blotting. In vivo tumor growth assay was performed by establishing WFDC2-knockdown xenografts in nude mice and monitoring tumor growth. The expression of WFDC2, Ki67 and activated caspase-3 was analyzed by immunohistochemistry in order to determine the role of WFDC2 in proliferation and apoptosis. Our results revealed that the silencing of WFDC2 abolished ovarian cancer cell proliferation, suppressing tumor formation and growth in ovarian cancer cells both in vitro and in vivo. The knockdown of WFDC2 induced upregulation of Fasl and the downregulation of cyclin D1 activated caspase-3 and Ki67. These results indicate that WFDC2 plays a crucial role in tumor formation and growth in ovarian cancer cells. WFDC2 may be a potential therapeutic target for epithelial ovarian cancer.
Collapse
Affiliation(s)
- Yao Chen
- School of Biotechnology, Southern Medical University, Guangzhou 510515, PR China
| | | | | | | | | | | | | |
Collapse
|
36
|
CCL2, galectin-3, and SMRP combination improves the diagnosis of mesothelioma in pleural effusions. J Thorac Oncol 2012; 7:883-9. [PMID: 22729037 DOI: 10.1097/jto.0b013e31824c9272] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Malignant pleural mesothelioma (MPM) is a highly aggressive tumor with poor prognosis. One major challenge for this disease is the development of new, early, and highly reliable diagnostic markers. The aim of this study was to compare the diagnostic value of the chemokine chemokine (C-C motif) ligand 2 (CCL2), galectin-3, and the secretory leukocyte peptidase inhibitor (SLPI) with soluble mesothelin-related peptides (SMRP), and to evaluate the diagnostic performance of marker combinations. METHODS The levels of the different markers were measured by enzyme-linked immunosorbent assay in pleural fluids from patients with MPM (n = 61), adenocarcinomas (ADCA, n = 25), or with benign pleural effusions (BPE, n = 15). RESULTS SMRP, SLPI, and CCL2 concentrations were significantly higher in pleural effusions from mesothelioma patients. Conversely, galectin-3 levels seemed to be elevated in patients with pulmonary ADCA. Receiver operating characteristic curve analysis revealed that SMRP (area under the curve [AUC] = 0.9059), CCL2 (AUC = 0.7912), galectin-3 (AUC = 0.7584), and SLPI (AUC = 0.7219) were potentially interesting biomarkers for the differentiation of MPM patients from those with BPE or ADCA. Of interest, we showed that the combination of SMRP/CCL2/galectin-3 greatly improved MPM diagnosis (AUC = 0.9680), when compared with SMRP alone. CONCLUSION The combination of SMRP/CCL2/galectin-3 seems to represent a promising panel of biomarkers for the reliable diagnosis of MPM in pleural fluids.
Collapse
|
37
|
Abstract
Background Large-scale tumor sequencing projects are now underway to identify genetic mutations that drive tumor initiation and development. Most studies take a gene-based approach to identifying driver mutations, highlighting genes mutated in a large percentage of tumor samples as those likely to contain driver mutations. However, this gene-based approach usually does not consider the position of the mutation within the gene or the functional context the position of the mutation provides. Here we introduce a novel method for mapping mutations to distinct protein domains, not just individual genes, in which they occur, thus providing the functional context for how the mutation contributes to disease. Furthermore, aggregating mutations from all genes containing a specific protein domain enables the identification of mutations that are rare at the gene level, but that occur frequently within the specified domain. These highly mutated domains potentially reveal disruptions of protein function necessary for cancer development. Results We mapped somatic mutations from the protein coding regions of 100 colon adenocarcinoma tumor samples to the genes and protein domains in which they occurred, and constructed topographical maps to depict the “mutational landscapes” of gene and domain mutation frequencies. We found significant mutation frequency in a number of genes previously known to be somatically mutated in colon cancer patients including APC, TP53 and KRAS. In addition, we found significant mutation frequency within specific domains located in these genes, as well as within other domains contained in genes having low mutation frequencies. These domain “peaks” were enriched with functions important to cancer development including kinase activity, DNA binding and repair, and signal transduction. Conclusions Using our method to create the domain landscapes of mutations in colon cancer, we were able to identify somatic mutations with high potential to drive cancer development. Interestingly, the majority of the genes involved have a low mutation frequency. Therefore, themethod shows good potential for identifying rare driver mutations in current, large-scale tumor sequencing projects. In addition, mapping mutations to specific domains provides the necessary functional context for understanding how the mutations contribute to the disease, and may reveal novel or more refined gene and domain target regions for drug development.
Collapse
Affiliation(s)
- Nathan L Nehrt
- Department of Biological Sciences, University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| | | | | | | |
Collapse
|
38
|
Bose D, Zimmerman LJ, Pierobon M, Petricoin E, Tozzi F, Parikh A, Fan F, Dallas N, Xia L, Gaur P, Samuel S, Liebler DC, Ellis LM. Chemoresistant colorectal cancer cells and cancer stem cells mediate growth and survival of bystander cells. Br J Cancer 2011; 105:1759-67. [PMID: 22045189 PMCID: PMC3242606 DOI: 10.1038/bjc.2011.449] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Revised: 09/27/2011] [Accepted: 10/04/2011] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Recent studies suggest that cancer stem cells (CSCs) mediate chemoresistance, but interestingly, only a small percentage of cells in a resistant tumour are CSCs; this suggests that non-CSCs survive by other means. We hypothesised that chemoresistant colorectal cancer (CRC) cells generate soluble factors that enhance survival of chemonaive tumour cells. METHODS Chemoresistant CRC cells were generated by serial passage in oxaliplatin (Ox cells). Conditioned media (CM) was collected from parental and oxaliplatin-resistant (OxR) cells. CRC cells were treated with CM and growth and survival were assessed. Tumour growth rates were determined in nude mice after cells were treated with CM. Mass spectrometry (MS) identified proteins in CM. Reverse phase protein microarray assays determined signalling effects of CM in parental cells. RESULTS Oxaliplatin-resistant CM increased survival of chemo-naive cells. CSC CM also increased growth of parental cells. Parental and OxR mixed tumours grew larger than tumours composed of parental or OxR cells alone. Mass spectrometry detected unique survival-promoting factors in OxR CM compared with parental CM. Cells treated with OxR CM demonstrated early phosphorylation of EGFR and MEK1, with later upregulation of total Akt .We identified progranulin as a potential mediator of chemoresistance. CONCLUSION Chemoresistant tumour cells and CSCs may promote resistance through soluble factors that mediate survival in otherwise chemosensitive tumour cells.
Collapse
Affiliation(s)
- D Bose
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - L J Zimmerman
- Jim Ayers Institute for Precancer Detection and Diagnosis, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | - M Pierobon
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Fairfax, VA 22030, USA
| | - E Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Fairfax, VA 22030, USA
| | - F Tozzi
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - A Parikh
- Jim Ayers Institute for Precancer Detection and Diagnosis, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | - F Fan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - N Dallas
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - L Xia
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - P Gaur
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - S Samuel
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - D C Liebler
- Jim Ayers Institute for Precancer Detection and Diagnosis, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | - L M Ellis
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| |
Collapse
|
39
|
Bandiera E, Romani C, Specchia C, Zanotti L, Galli C, Ruggeri G, Tognon G, Bignotti E, Tassi RA, Odicino F, Caimi L, Sartori E, Santin AD, Pecorelli S, Ravaggi A. Serum human epididymis protein 4 and risk for ovarian malignancy algorithm as new diagnostic and prognostic tools for epithelial ovarian cancer management. Cancer Epidemiol Biomarkers Prev 2011; 20:2496-506. [PMID: 22028406 DOI: 10.1158/1055-9965.epi-11-0635] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The aim of this work was to analyze the diagnostic and prognostic value of serum human epididymis protein 4 (HE4) and Risk for Ovarian Malignancy Algorithm (ROMA) in epithelial ovarian cancer (EOC). METHODS Preoperative serum samples of 419 women (140 healthy controls, 131 ovarian benign cysts, 34 endometriosis, and 114 EOC) were tested for CA125 and HE4 using fully automated methods (Abbott ARCHITECT) and validated cutoff values. RESULTS For the discrimination of benign masses from EOC, in premenopausal women, the sensitivity and specificity were 92.3% and 59.4% for CA125, 84.6% and 94.2% for HE4, and 84.6% and 81.2% for ROMA, whereas in postmenopausal women, the sensitivity and specificity were 94.3% and 82.3% for CA125, 78.2% and 99.0% for HE4, and 93.1% and 84.4% for ROMA. In patients with EOC, elevated CA125, HE4, and ROMA levels were associated with advanced Federation of Gynaecologists and Obstetricians (FIGO) stage, suboptimally debulking, ascites, positive cytology, lymph node involvement, and advanced age (all P ≤ 0.05). Elevated HE4 and ROMA (both P ≤ 0.01), but not CA125 (P = 0.0579), were associated with undifferentiated tumors. In multivariable analysis, elevated HE4 and ROMA (all P ≤ 0.05) were independent prognostic factors for shorter overall, disease-free, and progression-free survival. CONCLUSIONS AND IMPACT This study underlines the high specificity of HE4 in discriminating endometriosis and ovarian benign cysts from EOC and the high sensitivity of CA125 in detecting EOC. We showed HE4 and ROMA as independent prognostic factors. Multicenter studies are needed to draw firm conclusions about the applicability of HE4 and ROMA in clinical practice.
Collapse
Affiliation(s)
- Elisabetta Bandiera
- Angelo Nocivelli, Institute of Molecular Medicine, Division of Gynecologic Oncology, University of Brescia, Brescia, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Tyan YC, Yang MH, Chen SCJ, Jong SB, Chen WC, Yang YH, Chung TW, Liao PC. Urinary protein profiling by liquid chromatography/tandem mass spectrometry: ADAM28 is overexpressed in bladder transitional cell carcinoma. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2011; 25:2851-2862. [PMID: 21913264 DOI: 10.1002/rcm.5169] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Bladder cancer is the most common urological cancer with higher incidence rate in the endemic areas of Blackfoot disease (BFD) in southern Taiwan. The aim of this study was to utilize the proteomic approach to establish urinary protein patterns of bladder cancer. The experimental results showed that most patients with bladder cancer had proteinuria or albuminuria. The urine arsenic concentrations of bladder cancer patients in BFD areas were significantly higher than those patients from non-BFD areas. In the proteomic analysis, the urinary proteome was identified by nano-high-performance liquid chromatography/electrospray ionization tandem mass spectrometry (nano-HPLC/ESI-MS/MS) followed by peptide fragmentation pattern analysis. We categorized 2782 unique proteins of which 89 proteins were identified with at least three unique matching peptide sequences. Among these 89 proteins, thirteen of them were not found in the control group and may represent proteins specific for bladder cancer. In this study, three proteins, SPINK5, ADAM28 and PTP1, were also confirmed by Western blotting and showed significant differential expression compared with the control group. ADAM28 may be used as a possible biomarker of bladder cancer.
Collapse
Affiliation(s)
- Yu-Chang Tyan
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Taiwan.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Hoskins E, Rodriguez-Canales J, Hewitt SM, Elmasri W, Han J, Han S, Davidson B, Kohn EC. Paracrine SLPI secretion upregulates MMP-9 transcription and secretion in ovarian cancer cells. Gynecol Oncol 2011; 122:656-62. [PMID: 21676452 PMCID: PMC3152651 DOI: 10.1016/j.ygyno.2011.04.052] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Revised: 04/27/2011] [Accepted: 04/30/2011] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Secretory leukocyte protease inhibitor (SLPI) is amplified in serous ovarian cancer. We have dissected its function, showing it is a survival factor for ovarian cancer and promotes tumorigenesis and paclitaxel-resistance. We hypothesized that the protease inhibitory function was responsible for modulating SLPI's invasive capacity. METHODS Stable HEYA8 ovarian cancer transfectants expressing vector, wild type SLPI, and protease inhibitor null (F-)SLPI were examined in vitro and in xenografts. Invasion, enzyme activity, and MMP production and function assays were applied. SLPI and MMP immunoexpression was graded on tissue microarray and clinical samples. Statistical comparisons used unpaired t test and ANOVA, where appropriate. RESULTS SLPI and F-SLPI cells caused greater parenchymal and peritoneal dissemination over control cells in xenografts and invasion assays (p<0.001). MMP-9 protease activity was increased in SLPI and F-SLPI cells over control. SLPI, but not F-SLPI, inhibited plasmin activity, necessary for MMP-9 activation and release, and inhibited activation of MMP-9. However, paradoxically, both induced quantitative MMP-9 transcription (p<0.05) and protein (p<0.008), yielding an increased net MMP-9 activity in the face of plasmin inhibition. SLPI and MMP-9 expression were strongly correlated in serous ovarian cancers (r(2)=0.986) and a set of ovarian cancers (p<0.02). SLPI expression was greater in serous than endometrioid ovarian cancers (p=0.04). CONCLUSIONS SLPI stimulates ovarian cancer invasion, modulated in part by its serine protease inhibitory activity attenuating MMP-9 release. However, SLPI induction of MMP-9, independent of protease inhibition activity, is greater yielding a net pro-invasive behavior. These findings further support SLPI as a molecular target for ovarian cancer.
Collapse
Affiliation(s)
- Ebony Hoskins
- Molecular Signaling Section, Medical Oncology Branch, National Cancer Institute, Bethesda, Maryland 20892
| | - Jaime Rodriguez-Canales
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892
| | - Stephen M. Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892
| | - Wafic Elmasri
- Molecular Signaling Section, Medical Oncology Branch, National Cancer Institute, Bethesda, Maryland 20892
| | - Jasmine Han
- Molecular Signaling Section, Medical Oncology Branch, National Cancer Institute, Bethesda, Maryland 20892
| | - Shing Han
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892
| | - Ben Davidson
- Division of Pathology, Norwegian Radium Hospital, Oslo University Hospital; The Medical Faculty, University of Oslo, Norway
| | - Elise C. Kohn
- Molecular Signaling Section, Medical Oncology Branch, National Cancer Institute, Bethesda, Maryland 20892
| |
Collapse
|
42
|
Wang G, Lim DS, Choi BD, Park JJ, Jeong SJ, Kim JS, Kim JD, Park JS, Kim EK, Kim BH, Ham JH, Jeong MJ. Effect of secretory leukocyte protease inhibitor on migration and invasion of human KB oral carcinoma cells. Anim Cells Syst (Seoul) 2011. [DOI: 10.1080/19768354.2011.577573] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
43
|
Tomita T, Sakurai Y, Ishibashi S, Maru Y. Imbalance of Clara cell-mediated homeostatic inflammation is involved in lung metastasis. Oncogene 2011; 30:3429-39. [PMID: 21399660 DOI: 10.1038/onc.2011.53] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We have previously shown that tumor necrosis factor (TNF)α produced from primary tumor-induced expression of two endogenous Toll-like receptor 4 (TLR4) ligands, S100A8 and serum amyloid A3 (SAA3), in pre-metastatic lungs. However, mechanistic details of the signaling network and relevance to pulmonary physiology are poorly understood. Here, we identify Clara cells as a control tower of the network. Clara cell ablation by naphthalene suppressed pulmonary recruitment of CD11b+TLR4+ cells and spontaneous lung metastasis. Clara cells turned out to express TLR4 through which SAA3 was auto-amplified. Reciprocal bone marrow transplantation between wild-type and TLR4 knockout mice demonstrated that pulmonary TLR4+ Clara cells could be derived from bone marrow. SAA3-induced TNFα expression in both alveolar type II cells and macrophages. Primary co-cultures of alveolar type II cells and Clara cells revealed that the induction of TNFα in alveolar type II cells was dependent on the Clara cell-mediated amplification of SAA3. SAA3 induction by bacterial endotoxin also required both Clara cells and TLR4. Thus, pulmonary metastatic soil may feature deregulation of homeostatic inflammatory responses to constant assaults of microbes with endotoxin.
Collapse
Affiliation(s)
- T Tomita
- Department of Pharmacology, Tokyo Women's Medical University, Tokyo, Japan
| | | | | | | |
Collapse
|
44
|
Progranulin is a potential prognostic biomarker in advanced epithelial ovarian cancers. Gynecol Oncol 2010; 120:5-10. [PMID: 20950846 DOI: 10.1016/j.ygyno.2010.09.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Revised: 09/07/2010] [Accepted: 09/09/2010] [Indexed: 12/22/2022]
Abstract
OBJECTIVE There are few validated relapse prediction biomarkers for epithelial ovarian cancer (EOC). We have shown progranulin (PGRN) and secretory leukocyte protease inhibitor (SLPI) are up regulated, overexpressed survival factors in EOC. We hypothesized they would predict presence of occult EOC. METHOD PGRN, SLPI, and the known biomarker HE4 were measured in EOC patient plasma samples, prospectively collected every 3 months from initial remission until relapse. Clinical data and CA125 results were incorporated into statistical analyses. Exploratory Kaplan-Meier estimates, dividing markers at median values, evaluated association with progression-free survival (PFS) and overall survival (OS). Area-under-the-curve (AUC) statistics were computed from receiver operating characteristic (ROC) curves to evaluate discrimination ability. A Cox proportional hazards model assessed the association between PFS, OS, and biomarkers, adjusting for clinical prognostic factors. RESULTS Samples from 23 advanced stage EOC patients were evaluated. PGRN at 3 months was the only biomarker independently associated with PFS (P<0.0001) and OS (P<0.003). When used to predict progression by 18 months, sensitivity and specificity were 93% and 100%, respectively, with AUC=0.944. The Cox model hazard ratio for PFS, divided at 59 ng/ml by ROC analysis and adjusted for clinical factors, was 23.5 (95% CI: 2.49-220). Combinations with SLPI, HE4, and/or CA125 did not improve the model. CONCLUSIONS We report pilot data indicating a potential independent association of PGRN on EOC patient PFS and OS. A validation study will be required to confirm this finding and to inform whether PGRN warrants evaluation as a potential screening biomarker.
Collapse
|
45
|
Yamashita SI, Tokuishi K, Hashimoto T, Moroga T, Kamei M, Ono K, Miyawaki M, Takeno S, Chujo M, Yamamoto S, Kawahara K. Prognostic significance of HE4 expression in pulmonary adenocarcinoma. Tumour Biol 2010; 32:265-71. [PMID: 20953751 DOI: 10.1007/s13277-010-0118-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 10/01/2010] [Indexed: 10/18/2022] Open
Abstract
We investigated the possibility of human epididymis 4(HE4) to predict survival for patients with pulmonary adenocarcinoma. One hundred and thirty-seven patients with pulmonary adenocarcinoma underwent surgery in our institute from 2000 to 2008. We used immunohistochemical analysis to determine the expression of HE4 and compared with the clinicopathological factors and survival. Serum levels of HE4 in lung adenocarcinoma were investigated by enzyme immunometric assay. Fifty-seven of 137 cases (41.6%) were HE4 positive. It was found that there was no correlation between HE4 expression by immunohistochemistry and clinicopathological factors, however, adenocarcinoma subtype was significantly associated with HE4 expression. Sera in lung adenocarcinoma were significantly higher than in healthy control. Five-year disease-free survival in the HE4-positive group (44.6%) was significantly different from that in the negative group (82.3%, p = 0.001) by immunohistochemistry. The five-year overall survival rate was 60.1% in the HE4-positive group, as compared with 90.8% in the HE4-negative group (p = 0.001). In multivariate Cox regression analysis, positive HE4 protein expression was a worse prognosis factor of disease-free and overall survival (HR = 3.7, 95%CI = [1.7-8.4], p = 0.001; HR = 5.5, 95%CI = [1.8-17.2], p = 0.003, respectively), in addition to nodal status as a powerful value. When HE4 expression in adenocarcinoma cases except the BAC were analyzed, nodal status and HE4 expression were independent prognostic factors in disease-free and overall survivals. These data showed that HE4 expression is associated with a worse prognosis and is a possible prognostic factor of lung adenocarcinoma.
Collapse
Affiliation(s)
- Shin-Ichi Yamashita
- Department of Surgery II, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama, Yufu, Oita 879-5593, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Xi Z, LinLin M, Ye T. Human epididymis protein 4 is a biomarker for transitional cell carcinoma in the urinary system. J Clin Lab Anal 2010; 23:357-61. [PMID: 19927341 DOI: 10.1002/jcla.20329] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE To investigate human epididymis protein 4 (HE4) levels in transitional cell carcinoma (TCC) of the urinary system and its relationship with clinicopathological features. METHODS 102 patients with TCC, 60 with benign urinary diseases, and 60 healthy controls were included in this study. The HE4 levels were used to analyze different clinicopathologic characteristics and changes between pre- and postsurgical operation. RESULTS The HE4 level was significantly increased in patients with TCC compared to patients with benign urinary diseases patients (P<0.01) and healthy controls (P<0.01), and the level of HE4 in patients with superficial TCC (Tis Ta T1) was significantly higher than that of the benign urogenital group (P<0.05)and healthy controls (P<0.05). There was a significant difference between HE4 levels in patients before and after operation (P<0.05). There was no difference between HE4 levels based on tumor recurrence, clinical TNM stage, lymph node metastasis, or pathological stage (P>0.05). The HE4 level was also different between patients with a single tumor versus patients with multiple tumors. The area under the curves of HE4 is 0.821. The sensitivity and specificity of HE4 at a cutoff value of 45.7 pM were 67.6 and 88.3%, respectively. CONCLUSIONS HE4 may be a screening tool for early diagnosis of TCC in the urinary system, and may become a prognostic marker for TCC in the urinary system.
Collapse
Affiliation(s)
- Zhu Xi
- Department of Urology, Friendship Hospital, Capital Medical University, Beijing, China
| | | | | |
Collapse
|
47
|
Rasool N, LaRochelle W, Zhong H, Ara G, Cohen J, Kohn EC. Secretory leukocyte protease inhibitor antagonizes paclitaxel in ovarian cancer cells. Clin Cancer Res 2010; 16:600-9. [PMID: 20068074 DOI: 10.1158/1078-0432.ccr-09-1979] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Ovarian cancer recurrence with the development of paclitaxel resistance is an obstacle to long-term survival. We showed that secretory leukocyte protease inhibitor (SLPI) is a survival factor for ovarian cancer. We hypothesize that SLPI may antagonize paclitaxel injury. EXPERIMENTAL DESIGN Differential SLPI induction in response to paclitaxel and in response to stable forced expression of SLPI was shown in A2780-1A9 cells and their paclitaxel-resistant sublines, PTX10 and PTX22, and confirmed with HEY-A8 cells. SLPI-mediated survival was reduced by the MAP/extracellular signal-regulated kinase (ERK) kinase inhibitor, U0126, and a humanized neutralizing monoclonal anti-SLPI antibody, CR012. OVCAR3 xenographs tested the role of CR012 in vivo. RESULTS SLPI expression was lower in A2780-1A9 ovarian cancer cells than in PTX10 and PTX22, and SLPI was induced by paclitaxel exposure. Stable SLPI expression yielded a proliferation advantage (P = 0.01); expression of and response to SLPI in OVCAR3 cells were abrogated by exposure to CR012. SLPI reduced the paclitaxel susceptibility of 1A9 and HEY-A8 cells (P <or= 0.05), and SLPI expression did not increase the resistance of PTX10 and PTX22 cells. Both paclitaxel and SLPI overexpression induced ERK activation. Inhibition of MAP/ERK kinase with U0126 increased paclitaxel injury and overcame SLPI-mediated cell protection. It did not reinstate PTX10 sensitivity to paclitaxel, which was associated with AKT activation. Significant inhibition of OVCAR3 xenograft growth was observed with CR012 and paclitaxel, over single agents (P <or= 0.001). CONCLUSIONS A two-pronged approach confirmed that SLPI overcomes paclitaxel in part through activation of ERK1/2. These results credential SLPI as a molecular target for ovarian cancer and suggest CR012 as a tool for proof of concept.
Collapse
Affiliation(s)
- Nabila Rasool
- Molecular Signaling Section, Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | | | | | | | | | | |
Collapse
|
48
|
Temporal induction of secretory leukocyte protease inhibitor (SLPI) in odontoblasts by lipopolysaccharide and wound infection. J Endod 2009; 35:997-1002. [PMID: 19567322 DOI: 10.1016/j.joen.2009.04.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Revised: 04/08/2009] [Accepted: 04/10/2009] [Indexed: 11/22/2022]
Abstract
INTRODUCTION The secretory leukocyte protease inhibitor (SLPI) is a bacterial lipopolysaccharide (LPS)-induced product of macrophages that antagonizes the LPS-induced activation of a number of proinflammatory signaling factors. From our previous experiments, it was found that SLPI was expressed slightly in odontoblast-like cells (MDPC-23). Therefore, these experiments were designed to determine the function of SLPI in MDPC-23 and odontoblasts during the inflammatory response caused by infections and wounds. METHODS MDPC-23 cells were exposed to 100 ng/mL Escherichia coli LPS, and artificial wounds were induced in the right first molar of the maxillary of rats. In addition, a morphological change in the MDPC-23 cells was observed after LPS treatment. MDPC-23 cells were transfected transiently with the nuclear factor kappa-B (NF-kappaB) promoter binding vector. RESULTS The level of SLPI expression increased strongly 30 minutes after the LPS treatment. Scanning electron microscopy revealed many extensions of the cytoplasmic processes after LPS stimulation. SLPI was expressed along the dentinal tubules and odontoblasts layer in rat teeth after an artificial wound. SLPI also inhibited the LPS-induced activation of NF-kappaB in MDPC-23. CONCLUSIONS We report for the first time that SLPI is expressed temporally in infected odontoblasts and may participate in the anti-inflammatory response through NF-kappaB signaling in odontoblast-like cells.
Collapse
|
49
|
High expression of tumour-associated trypsin inhibitor correlates with liver metastasis and poor prognosis in colorectal cancer. Br J Cancer 2009; 100:1540-8. [PMID: 19384300 PMCID: PMC2696764 DOI: 10.1038/sj.bjc.6605047] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Increased expression of tumour-associated trypsin inhibitor (TATI) in tumour tissue and/or serum has been associated with poor survival in various cancer forms. Moreover, a proinvasive function of TATI has been shown in colon cancer cell lines. In this study, we have examined the prognostic significance of tumour-specific TATI expression in colorectal cancer, assessed by immunohistochemistry (IHC) on tissue microarrays (TMAs) with tumour specimens from two independent patient cohorts. Kaplan–Meier analysis and Cox proportional hazards modelling were used to estimate time to recurrence, disease-free survival and overall survival. In both cohorts, a high (>50% of tumour cells) TATI expression was an independent predictor of a significantly shorter overall survival. In cohort II, in multivariate analysis including age, gender, disease stage, differentiation grade, vascular invasion and carcinoembryonal antigen (CEA), high TATI expression was associated with a significantly decreased overall survival (HR=1.82; 95% CI=1.19–2.79) and disease-free survival (HR=1.56; 95% CI=1.05–2.32) in curatively treated patients. Moreover, there was an increased risk for liver metastasis in both cohorts that remained significant in multivariate analysis in cohort II (HR=2.85; 95% CI=1.43–5.66). In conclusion, high TATI expression is associated with liver metastasis and is an independent predictor of poor prognosis in patients with colorectal cancer.
Collapse
|
50
|
Devoogdt N, Rasool N, Hoskins E, Simpkins F, Tchabo N, Kohn EC. Overexpression of protease inhibitor-dead secretory leukocyte protease inhibitor causes more aggressive ovarian cancer in vitro and in vivo. Cancer Sci 2009; 100:434-40. [PMID: 19154415 DOI: 10.1111/j.1349-7006.2009.01076.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The alarm anti-protease secretory leukocyte protease inhibitor (SLPI) is frequently overexpressed in ovarian cancer cells and has been proposed for inclusion in biomarker panels but function remains unclear. We hypothesized that SLPI overexpression promotes ovarian cancer growth and survival. Low SLPI-expressing Hey-A8 ovarian cancer cells were engineered to produce functional (WT) or protease inhibitor-null (PI-) mutant SLPI; lack of PI activity was confirmed by enzymatic assay. WT/SLPI and PI- mutants stimulated significant proliferation and survival of Hey-A8 ovarian cancer cells under basal culture conditions (P < or = 0.02), in soft agar colony number and size (P < or = 0.05), and in anoikis resistance (P < or = 0.005). SLPI protected the ovarian cancer survival factor, progranulin (PRGN), and HEY-A8 cells from degradation and apoptosis due to neutrophil elastase. PI-/SLPI cells had greater protective activity than WT/SLPI cells. HEY-A8 murine xenografts revealed enhanced solid tumor formation, dissemination, and invasion in WT/SLPI and PI-/SLPI mutants. Increased proliferation was demonstrated by Ki-67 staining (P < or = 0.02). Increased secreted PRGN was seen in culture and was also observed by immunohistochemistry in the SLPI transfectant xenografts. This study describes a PI-independent function for SLPI in ovarian cancer growth and dissemination.
Collapse
Affiliation(s)
- Nick Devoogdt
- Molecular Signaling Section, Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892-1906, USA
| | | | | | | | | | | |
Collapse
|