1
|
HUANG BAOXING, JIA ZICHANG, FU CHENCHEN, CHEN MOXIAN, SU ZEZHUO, CHEN YUNSHENG. Oncogenic and tumor-suppressive roles of Lipocalin 2 (LCN2) in tumor progression. Oncol Res 2025; 33:567-575. [PMID: 40109857 PMCID: PMC11915076 DOI: 10.32604/or.2024.051672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/17/2024] [Indexed: 03/22/2025] Open
Abstract
Lipocalin-2 (LCN2) is a member of the lipocalin superfamily with multiple functions and can participate in the transport of a variety of small lipophilic ligands in vivo. LCN2 is significantly expressed in various tumors and plays an important role in regulating tumor cell proliferation, invasion, and metastasis. The specific actions of LCN2 in tumors may vary depending on the particular type of cancer involved. In this review, we provide an extensive overview of the transcriptional and post-transcriptional regulation of LCN2 in health and disease. Furthermore, we summarize the impact of LCN2 dysregulation in a broad range of tumors. Lastly, we examine the mechanisms of action of LCN2 during tumorigenesis, progression, and metastasis. Understanding the complex relationships between LCN2 and tumor development, progression, and metastasis is vital for advancing our knowledge of cancer biology, developing biomarkers for diagnosis and clinical decision-making, and creating therapeutic strategies to improve the management of patients with cancer.
Collapse
Affiliation(s)
- BAOXING HUANG
- Clinical Laboratory, Shenzhen Children’s Hospital, Shenzhen, 518038, China
| | - ZICHANG JIA
- Clinical Laboratory, Shenzhen Children’s Hospital, Shenzhen, 518038, China
| | - CHENCHEN FU
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, Key Laboratory of Tree Genetics and Biotechnology of Educational Department of China, Key Laboratory of State Forestry and Grassland Administration on Subtropical Forest Biodiversity Conservation, College of Life Sciences, Nanjing Forestry University, Nanjing, 210037, China
| | - MOXIAN CHEN
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, Key Laboratory of Tree Genetics and Biotechnology of Educational Department of China, Key Laboratory of State Forestry and Grassland Administration on Subtropical Forest Biodiversity Conservation, College of Life Sciences, Nanjing Forestry University, Nanjing, 210037, China
| | - ZEZHUO SU
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - YUNSHENG CHEN
- Clinical Laboratory, Shenzhen Children’s Hospital, Shenzhen, 518038, China
| |
Collapse
|
2
|
Radhakrishnan SK, Nath D, Russ D, Merodio LB, Lad P, Daisi FK, Acharjee A. Machine learning-based identification of proteomic markers in colorectal cancer using UK Biobank data. Front Oncol 2025; 14:1505675. [PMID: 39839775 PMCID: PMC11746037 DOI: 10.3389/fonc.2024.1505675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/02/2024] [Indexed: 01/23/2025] Open
Abstract
Colorectal cancer is one of the leading causes of cancer-related mortality in the world. Incidence and mortality are predicted to rise globally during the next several decades. When detected early, colorectal cancer is treatable with surgery and medications. This leads to the requirement for prognostic and diagnostic biomarker development. Our study integrates machine learning models and protein network analysis to identify protein biomarkers for colorectal cancer. Our methodology leverages an extensive collection of proteome profiles from both healthy and colorectal cancer individuals. To identify a potential biomarker with high predictive ability, we used three machine learning models. To enhance the interpretability of our models, we quantify each protein's contribution to the model's predictions using SHapley Additive exPlanations values. Three classifiers-LASSO, XGBoost, and LightGBM were evaluated for predictive performance along with hyperparameter tuning of each model using grid search, with LASSO achieving the highest AUC of 75% in the UK Biobank dataset and the AUCs for LightGBM and XGBoost are 69.61% and 71.42%, respectively. Using SHapley Additive exPlanations values, TFF3, LCN2, and CEACAM5 were found to be key biomarkers associated with cell adhesion and inflammation. Protein quantitative trait loci analyze studies provided further evidence for the involvement of TFF1, CEACAM5, and SELE in colorectal cancer, with possible connections to the PI3K/Akt and MAPK signaling pathways. By offering insights into colorectal cancer diagnostics and targeted therapeutics, our findings set the stage for further biomarker validation.
Collapse
Affiliation(s)
| | - Dipanwita Nath
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Dominic Russ
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- Institute of Translational Medicine, University Hospitals Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Centre for Health Data Research, University of Birmingham, Birmingham, United Kingdom
| | - Laura Bravo Merodio
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- Institute of Translational Medicine, University Hospitals Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Centre for Health Data Research, University of Birmingham, Birmingham, United Kingdom
| | - Priyani Lad
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Folakemi Kola Daisi
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Animesh Acharjee
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- Institute of Translational Medicine, University Hospitals Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Centre for Health Data Research, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
3
|
Zhang ZX, Peng J, Ding WW. Lipocalin-2 and intestinal diseases. World J Gastroenterol 2024; 30:4864-4879. [PMID: 39679305 PMCID: PMC11612708 DOI: 10.3748/wjg.v30.i46.4864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 09/25/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
Dysfunction of the intestinal barrier is a prevalent phenomenon observed across a spectrum of diseases, encompassing conditions such as mesenteric artery dissection, inflammatory bowel disease, cirrhosis, and sepsis. In these pathological states, the integrity of the intestinal barrier, which normally serves to regulate the selective passage of substances between the gut lumen and the bloodstream, becomes compromised. This compromised barrier function can lead to a range of adverse consequences, including increased permeability to harmful substances, the translocation of bacteria and their products into systemic circulation, and heightened inflammatory responses within the gut and beyond. Understanding the mechanisms underlying intestinal barrier dysfunction in these diverse disease contexts is crucial for the development of targeted therapeutic interventions aimed at restoring barrier integrity and ameliorating disease progression. Lipocalin-2 (LCN2) expression is significantly upregulated during episodes of intestinal inflammation, making it a pivotal indicator for gauging the extent of such inflammatory processes. Notably, however, LCN2 derived from distinct cellular sources, whether intestinal epithelial cells or immune cells, exhibits notably divergent functional characteristics. Furthermore, the multifaceted nature of LCN2 is underscored by its varying roles across different diseases, sometimes even demonstrating contradictory effects.
Collapse
Affiliation(s)
- Zhong-Xu Zhang
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Jian Peng
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Wei-Wei Ding
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu Province, China
| |
Collapse
|
4
|
Shahgoli VK, Dubik M, Pilecki B, Skallerup S, Schmidt SG, Detlefsen S, Sorensen GL, Holmskov U, Baradaran B, Moeller JB. Expression of FIBCD1 by intestinal epithelial cells alleviates inflammation-driven tumorigenesis in a mouse model of colorectal cancer. Front Oncol 2023; 13:1280891. [PMID: 38090485 PMCID: PMC10715588 DOI: 10.3389/fonc.2023.1280891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/06/2023] [Indexed: 01/05/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) ranks as the third most prevalent cancer globally, highlighting the pressing need to address its development. Inflammation plays a crucial role in augmenting the risk of CRC and actively contributes to all stages of tumorigenesis. Consequently, targeting early inflammatory responses in the intestinal tract to restore homeostasis holds significant potential for preventing and treating CRC. Fibrinogen C domain-containing 1 (FIBCD1), a chitin-binding transmembrane protein predominantly found on human intestinal epithelial cells (IECs), has garnered attention in previous research for its ability to effectively suppress inflammatory responses and promote tissue homeostasis at mucosal barriers. METHODS In this study, we investigated the role of FIBCD1 in CRC development using transgenic mice that mimic human expression of FIBCD1 at the intestinal mucosal barrier. To model aspects of CRC, we employed the azoxymethane/dextran sodium sulfate (AOM/DSS) mouse model. Additionally, we examined the expression pattern of FIBCD1 in surgical specimens obtained from human CRC patients by immunohistochemical methods. By accessing public data repositories, we further evaluated FIBCD1 expression in colon adenocarcinoma and explored survival outcomes associated with FIBCD1 expression. RESULTS Here, we demonstrate that FIBCD1 substantially impacts CRC development by significantly reducing intestinal inflammation and suppressing colorectal tumorigenesis in mice. Furthermore, we identify a soluble variant of FIBCD1 that is significantly increased in feces during acute inflammation. Finally, we demonstrate increased expression of FIBCD1 by immunohistochemistry in human CRC specimens at more developed tumor stages. These results are further supported by bioinformatic analyses of publicly available repositories, indicating increased FIBCD1 expression in tumor tissues, where higher expression is associated with unfavorable prognosis. CONCLUSION Collectively, these findings suggest that FIBCD1 influences early inflammatory responses in the AOM/DSS model, leading to a reduction in tumor size and burden. The increased expression of FIBCD1 in human CRC samples raises intriguing questions regarding its role in CRC, positioning it as a compelling candidate and novel molecular target for future research.
Collapse
Affiliation(s)
- Vahid Khaze Shahgoli
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Magdalena Dubik
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Bartosz Pilecki
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Sofie Skallerup
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Sandra Gaedt Schmidt
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Sönke Detlefsen
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Grith L. Sorensen
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Uffe Holmskov
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jesper B. Moeller
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Danish Institute for Advanced Study, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
5
|
Choudhary BS, Chaudhary N, Shah M, Dwivedi N, P K S, Das M, Dalal SN. Lipocalin 2 inhibits actin glutathionylation to promote invasion and migration. FEBS Lett 2023; 597:1086-1097. [PMID: 36650979 DOI: 10.1002/1873-3468.14572] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/06/2022] [Accepted: 12/14/2022] [Indexed: 01/19/2023]
Abstract
Invasive and metastatic tumor cells show an increase in migration and invasion, making the processes contributing to these phenotypes potential therapeutic targets. Lipocalin 2 (LCN2; also known as neutrophil gelatinase-associated lipocalin) is a putative therapeutic target in multiple tumor types and promotes invasion and migration, although the mechanisms underlying these phenotypes are unclear. The data in this report demonstrate that LCN2 promotes actin polymerization, invasion, and migration by inhibiting actin glutathionylation. LCN2 inhibits actin glutathionylation by decreasing the levels of reactive oxygen species (ROS) and by reducing intracellular iron levels. Inhibiting LCN2 function leads to increased actin glutathionylation, decreased migration, and decreased invasion. These results suggest that LCN2 is a potential therapeutic target in invasive tumors.
Collapse
Affiliation(s)
- Bhagya Shree Choudhary
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India.,Homi Bhabha National Institute, Mumbai, India
| | - Nazia Chaudhary
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India.,Homi Bhabha National Institute, Mumbai, India
| | - Manya Shah
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Nehanjali Dwivedi
- Molecular Immunology, Mazumdar Shaw Medical Foundation, Bommasandra, Bangalore, India
| | - Smitha P K
- Product Research Group, Mazumdar Shaw Medical Foundation, Bommasandra, Bangalore, India
| | - Manjula Das
- Molecular Immunology, Mazumdar Shaw Medical Foundation, Bommasandra, Bangalore, India
| | - Sorab Nariman Dalal
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India.,Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
6
|
Pali‐Schöll I, Bianchini R, Afify SM, Hofstetter G, Winkler S, Ahlers S, Altemeier T, Mayerhofer H, Hufnagl K, Korath ADJ, Pranger C, Widhalm R, Hann S, Wittek T, Kasper‐Giebl A, Pacios LF, Roth‐Walter F, Vercelli D, von Mutius E, Jensen‐Jarolim E. Secretory protein beta-lactoglobulin in cattle stable dust may contribute to the allergy-protective farm effect. Clin Transl Allergy 2022; 12:e12125. [PMID: 35169442 PMCID: PMC8840802 DOI: 10.1002/clt2.12125] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 01/22/2022] [Accepted: 02/01/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Growing up on a cattle farm and consuming raw cow's milk protects against asthma and allergies. We expect a cattle-specific protein as active component in this farm effect. METHODS Dust was collected from cattle and poultry stables and from mattresses of households. Urine was obtained from cattle, and ambient aerosols were sampled. Samples were analysed for BLG by SDS PAGE/immunoblot and mass spectrometry, and for association with metals by SEC-ICP-MS. PBMC of healthy donors were incubated with BLG +/- zinc, and proliferation and cytokines determined. BALB/c mice were pre-treated intranasally with stable dust extract containing BLG or depleted of BLG, and subsequent allergy response after sensitization was evaluated on antibody and symptom level. RESULTS A major protein in dust from cattle farms and ambient air was identified as BLG. Urine from female and male cattle is a major source of BLG. In dust samples, BLG was associated with zinc. In vitro, zinc-BLG provoked significantly lower proliferation of CD4+ and CD8+ cells while inducing significantly higher levels of IFN-γ and IL-6 than the apo-BLG devoid of zinc. In vivo, pre-treatment of mice with dust extract containing BLG resulted in lower allergy symptom scores to BLG and unrelated Bet v 1 than pre-treatment with extract depleted of BLG. These in vitro and in vivo effects were independent of endotoxin. CONCLUSION The lipocalin BLG is found in large amounts in cattle urine, accumulates in bovine dust samples and is aerosolized around farms. Its association with zinc favorably shapes the human cellular immune response towards Th1-cytokines in vitro. BLG together with zinc in stable dust protects mice from allergic sensitization. BLG with its associated ligands may in an innate manner contribute to the allergy-protective farm effect.
Collapse
Affiliation(s)
- Isabella Pali‐Schöll
- The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University ViennaViennaAustria
- Institute of Pathophysiology and Allergy Research; Center of Physiology, Pathophysiology and Immunology; Medical University ViennaViennaAustria
| | - Rodolfo Bianchini
- The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University ViennaViennaAustria
| | - Sheriene Moussa Afify
- The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University ViennaViennaAustria
- Laboratory Medicine and Immunology DepartmentFaculty of MedicineMenoufia UniversityMenoufiaEgypt
| | - Gerlinde Hofstetter
- The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University ViennaViennaAustria
| | - Simona Winkler
- The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University ViennaViennaAustria
| | - Stella Ahlers
- The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University ViennaViennaAustria
| | - Theresa Altemeier
- The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University ViennaViennaAustria
| | - Hanna Mayerhofer
- The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University ViennaViennaAustria
| | - Karin Hufnagl
- The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University ViennaViennaAustria
| | - Anna D. J. Korath
- The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University ViennaViennaAustria
| | - Christina Pranger
- The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University ViennaViennaAustria
- Institute of Pathophysiology and Allergy Research; Center of Physiology, Pathophysiology and Immunology; Medical University ViennaViennaAustria
| | - Raimund Widhalm
- Institute of Medical GeneticsMedical University of ViennaViennaAustria
- Karl‐Landsteiner Private University for Health SciencesKremsAustria
| | - Stephan Hann
- Division of Analytical Chemistry, Department of Chemistry, University of Natural Resources and Life Sciences, BOKU‐ViennaViennaAustria
| | - Thomas Wittek
- University Clinic for RuminantsUniversity of Veterinary Medicine ViennaViennaAustria
| | - Anne Kasper‐Giebl
- Institute of Chemical Technologies and Analytics, TU‐WienViennaAustria
| | - Luis F. Pacios
- Centro de Biotecnología y Genómica de Plantas (CBGP, UPM‐INIA), Campus de Montegancedo UPM and Departmento de Biotecnología‐Biología Vegetal, ETSIAAB, Universidad Politécnica de Madrid (UPM)MadridSpain
| | - Franziska Roth‐Walter
- The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University ViennaViennaAustria
- Institute of Pathophysiology and Allergy Research; Center of Physiology, Pathophysiology and Immunology; Medical University ViennaViennaAustria
| | - Donata Vercelli
- Arizona Respiratory CenterUniversity of Arizona College of MedicineTucsonArizonaUSA
| | - Erika von Mutius
- Asthma and Allergy DepartmentDr. von Hauner Children's HospitalUniversity of MunichMunichGermany
| | - Erika Jensen‐Jarolim
- The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University ViennaViennaAustria
- Institute of Pathophysiology and Allergy Research; Center of Physiology, Pathophysiology and Immunology; Medical University ViennaViennaAustria
| |
Collapse
|
7
|
Chaudhary N, Choudhary BS, Shah SG, Khapare N, Dwivedi N, Gaikwad A, Joshi N, Raichanna J, Basu S, Gurjar M, P K S, Saklani A, Gera P, Ramadwar M, Patil P, Thorat R, Gota V, Dhar SK, Gupta S, Das M, Dalal SN. Lipocalin 2 expression promotes tumor progression and therapy resistance by inhibiting ferroptosis in colorectal cancer. Int J Cancer 2021; 149:1495-1511. [PMID: 34146401 DOI: 10.1002/ijc.33711] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/13/2021] [Accepted: 06/08/2021] [Indexed: 01/14/2023]
Abstract
Lipocalin 2 is a siderophore-binding protein that regulates iron homeostasis. Lipocalin 2 expression is elevated in multiple tumor types; however, the mechanisms that drive tumor progression upon Lipocalin 2 expression remain unclear. When Lipocalin 2 is over-expressed, it leads to resistance to 5-fluorouracil in colon cancer cell lines in vitro and in vivo by inhibiting ferroptosis. Lipocalin 2 inhibits ferroptosis by decreasing intracellular iron levels and stimulating the expression of glutathione peroxidase4 and a component of the cysteine glutamate antiporter, xCT. The increase in xCT levels is dependent on increased levels of ETS1 in Lipocalin 2 over-expressing cells. Inhibiting Lipocalin 2 function with a monoclonal antibody leads to a decrease in chemo-resistance and transformation in vitro, and a decrease in tumor progression and chemo-resistance in xenograft mouse models. Lipocalin 2 and xCT levels exhibit a positive correlation in human tumor samples suggesting that the pathway we have identified in cell lines is operative in human tumor samples. These results indicate that Lipocalin 2 is a potential therapeutic target and that the monoclonal antibody described in our study can serve as the basis for a potential therapeutic in patients who do not respond to chemotherapy.
Collapse
Affiliation(s)
- Nazia Chaudhary
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Life Sciences, Homi Bhabha National Institute, Mumbai, India
| | - Bhagya Shree Choudhary
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Life Sciences, Homi Bhabha National Institute, Mumbai, India
| | - Sanket Girish Shah
- Life Sciences, Homi Bhabha National Institute, Mumbai, India
- Epigenetics and Chromatin Biology Group, Gupta Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Nileema Khapare
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Nehanjali Dwivedi
- Tumor Immunology Program, Mazumdar Shaw Medical Foundation, Mazumdar Shaw Medical Centre, Narayana Health City, Bangalore, India
| | - Anagha Gaikwad
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Neha Joshi
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Jinsy Raichanna
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Srikanta Basu
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Murari Gurjar
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Smitha P K
- Tumor Immunology Program, Mazumdar Shaw Medical Foundation, Mazumdar Shaw Medical Centre, Narayana Health City, Bangalore, India
| | - Avanish Saklani
- Department of Gastrointestinal Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Poonam Gera
- Department of Biorepository, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Mukta Ramadwar
- Department of Pathology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Prachi Patil
- Department of Digestive Disease and Clinical Nutrition, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Rahul Thorat
- Laboratory Animal Facility, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Vikram Gota
- Life Sciences, Homi Bhabha National Institute, Mumbai, India
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Sujan K Dhar
- Beyond Antibody, InCyte Laboratory, Mazumdar Shaw Medical Foundation, Mazumdar Shaw Medical Centre, Narayana Health City, Bangalore, India
| | - Sanjay Gupta
- Life Sciences, Homi Bhabha National Institute, Mumbai, India
- Epigenetics and Chromatin Biology Group, Gupta Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Manjula Das
- Tumor Immunology Program, Mazumdar Shaw Medical Foundation, Mazumdar Shaw Medical Centre, Narayana Health City, Bangalore, India
- Beyond Antibody, InCyte Laboratory, Mazumdar Shaw Medical Foundation, Mazumdar Shaw Medical Centre, Narayana Health City, Bangalore, India
| | - Sorab N Dalal
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Life Sciences, Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
8
|
Porter RJ, Murray GI, Brice DP, Petty RD, McLean MH. Novel biomarkers for risk stratification of Barrett's oesophagus associated neoplastic progression-epithelial HMGB1 expression and stromal lymphocytic phenotype. Br J Cancer 2019; 122:545-554. [PMID: 31831860 PMCID: PMC7028982 DOI: 10.1038/s41416-019-0685-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 10/30/2019] [Accepted: 11/28/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The incidence of oesophageal adenocarcinoma is increasing globally. Barrett's oesophagus (BO) is a pre-malignant condition with no biomarker to risk stratify those at highest risk of dysplasia and malignant transformation. METHODS Subcellular epithelial protein (HMGB1, p53, RUNX3) expression, alongside expression of CD20, CD4, CD8 and Foxp3 to characterise stromal B lymphocyte, and helper, cytotoxic and regulatory T-lymphocyte cell infiltrate, respectively, was assessed by immunohistochemistry in 218 human tissue samples including normal oesophageal/gastric biopsies (n = 39), BO (non-dysplasia, dysplasia, non-dysplastic background from progressors to dysplasia or cancer, n = 121) and oesophageal adenocarcinoma (n = 58). RESULTS There is a dynamic subcellular epithelial expression of HMGB1 (loss of nuclear, emergence of cytoplasmic), associated with epithelial p53 expression and differential immune cell phenotype in oesophageal neoplastic progression. We identify a protein signature and lymphocyte infiltrate in non-dysplastic BO when progressive disease (dysplasia or adenocarcinoma) is present but not histologically represented in the biopsied field. There is a dynamic stromal lymphocytic infiltrate in oesophageal neoplastic progression. CONCLUSIONS This data reveals novel insights into the microenvironment of BO and progression towards cancer and identifies a novel high-risk biomarker of disease progression to aid surveillance strategies to identify early progression and impact future incidence of oesophageal cancer.
Collapse
Affiliation(s)
- Ross J Porter
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Graeme I Murray
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Daniel P Brice
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Russell D Petty
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, DD1 1GZ, UK
| | - Mairi H McLean
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| |
Collapse
|
9
|
Du Z, Wu B, Xia Q, Zhao Y, Lin L, Cai Z, Wang S, Li E, Xu L, Li Y, Xu H, Yin D. LCN2-interacting proteins and their expression patterns in brain tumors. Brain Res 2019; 1720:146304. [PMID: 31233712 DOI: 10.1016/j.brainres.2019.146304] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/01/2019] [Accepted: 06/20/2019] [Indexed: 02/08/2023]
Abstract
Lipocalin 2 (LCN2) is a member of the lipocalin family. Elevated expression of LCN2 has been observed in many human tumors, suggesting it might be a potential biomarker and/or therapeutic target in malignancies. In this study, we aimed to explore LCN2 interacting proteins through bioinformatics, as well as their biological functions. Protein-protein interaction networks (PPIN) were constructed using LCN2 and its interacting proteins as the core node. These PPINs were scale free biological networks in which LCN2 and its interacting proteins could connect or cross-talk with at least one partner protein. Both functional and KEGG pathway enrichment analyses identified the known and potential biological functions of the PPIN, such as cell migration and cancer-related pathways. Expression levels of the PPIN proteins, as well as their expression correlations, in five types of brain tumor, were analyzed and integrated into the PPIN to illustrate a dynamic change. A significant correlation was found between the survival time of glioblastoma patients and the expression level of 10 genes (LCN2, MMP9, MMP2, PDE4DIP, L2HGDH, HNRNPA1, DDX31, LOXL2, FAM60A and RNF25). Taken together, our results suggest that LCN2 and its interacting proteins are mostly differentially expressed and have a distinguishing co-expression pattern. They might promote proliferation and migration via cell migration signaling and cancer-related pathways. LCN2 and its interacting proteins might be potential biomarkers in glioblastoma.
Collapse
Affiliation(s)
- Zepeng Du
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Genes Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China; Department of Pathology, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, Guangdong, China
| | - Bingli Wu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Qiaoxi Xia
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Yan Zhao
- Department of Pathology, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, Guangdong, China
| | - Ling Lin
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Zhixiong Cai
- Department of Cardiology, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, Guangdong, China
| | - Shaohong Wang
- Department of Pathology, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, Guangdong, China
| | - Enmin Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Liyan Xu
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Yun Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Genes Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China
| | - Haixiong Xu
- Department of Neurosurgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, Guangdong, China.
| | - Dong Yin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Genes Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China.
| |
Collapse
|
10
|
Miki M, Oono T, Fujimori N, Takaoka T, Kawabe K, Miyasaka Y, Ohtsuka T, Saito D, Nakamura M, Ohkawa Y, Oda Y, Suyama M, Ito T, Ogawa Y. CLEC3A, MMP7, and LCN2 as novel markers for predicting recurrence in resected G1 and G2 pancreatic neuroendocrine tumors. Cancer Med 2019; 8:3748-3760. [PMID: 31129920 PMCID: PMC6639196 DOI: 10.1002/cam4.2232] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/23/2019] [Accepted: 04/25/2019] [Indexed: 12/23/2022] Open
Abstract
Although the postoperative recurrence rate for pancreatic neuroendocrine tumors (PNETs) is reported to be 13.5%-30%, the paucity of valuable biomarkers to predict recurrence poses a problem for the early detection of relapse. Hence, this study aimed to identify new biomarkers to predict the recurrence of PNETs. We performed RNA sequencing (RNA-Seq) on RNA isolated from frozen primary tumors sampled from all localized G1/G2 PNETs resected curatively from 1998 to 2015 in our institution. We calculated differentially expressed genes (DEGs) in tumor with and without recurrence (≥3 years) for the propensity-matched cohort. Gene ontology analysis for the identified DEGs was also performed. Furthermore, we evaluated the expression levels of candidate genes as recurrence predictors via immunostaining. Comparison of transcriptional levels in tumors with and without recurrence identified 166 DEGs. Up- and downregulated genes with high significance in these tumors were mainly related to extracellular organization and cell adhesion, respectively. We observed the top three upregulated genes, C-type lectin domain family 3 member A (CLEC3A), matrix metalloproteinase-7 (MMP7), and lipocalin2 (LCN2) immunohistochemically and compared their levels in recurrent and nonrecurrent tumors. Significantly higher recurrence rate was shown in patients with positive expression of CLEC3A (P = 0.028), MMP7 (P = 0.003), and LCN2 (P = 0.040) than that with negative expression. We identified CLEC3A, MMP7, and LCN2 known to be associated with the phosphatidylinositol-3-kinase/Akt pathway, as potential novel markers to predict the postoperative recurrence of PNETs.
Collapse
Affiliation(s)
- Masami Miki
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takamasa Oono
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nao Fujimori
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takehiro Takaoka
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken Kawabe
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Miyasaka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takao Ohtsuka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daisuke Saito
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomical Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mikita Suyama
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Tetsuhide Ito
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Neuroendocrine Tumor Centre, Fukuoka Sanno Hospital, Internal University of Health and Welfare, Fukuoka, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Molecular and Cellular Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
11
|
Basu S, Chaudhary N, Shah S, Braggs C, Sawant A, Vaz S, Thorat R, Gupta S, Dalal SN. Plakophilin3 loss leads to an increase in lipocalin2 expression, which is required for tumour formation. Exp Cell Res 2018; 369:251-265. [PMID: 29803740 DOI: 10.1016/j.yexcr.2018.05.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 12/17/2022]
Abstract
An increase in tumour formation and metastasis are observed upon plakophilin3 (PKP3) loss. To identify pathways downstream of PKP3 loss that are required for increased tumour formation, a gene expression analysis was performed, which demonstrated that the expression of lipocalin2 (LCN2) was elevated upon PKP3 loss and this is consistent with expression data from human tumour samples suggesting that PKP3 loss correlates with an increase in LCN2 expression. PKP3 loss leads to an increase in invasion, tumour formation and metastasis and these phenotypes were dependent on the increase in LCN2 expression. The increased LCN2 expression was due to an increase in the activation of p38 MAPK in the HCT116 derived PKP3 knockdown clones as LCN2 expression decreased upon inhibition of p38 MAPK. The phosphorylated active form of p38 MAPK is translocated to the nucleus upon PKP3 loss and is dependent on complex formation between p38 MAPK and PKP3. WT PKP3 inhibits LCN2 reporter activity in PKP3 knockdown cells but a PKP3 mutant that fails to form a complex with p38 MAPK cannot suppress LCN2 promoter activity. Further, LCN2 expression is decreased upon loss of p38β, but not p38α, in the PKP3 knockdown cells. These results suggest that PKP3 loss leads to an increase in the nuclear translocation of p38 MAPK and p38β MAPK is required for the increase in LCN2 expression.
Collapse
Affiliation(s)
- Srikanta Basu
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Nazia Chaudhary
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Sanket Shah
- Epigenetics and Chromatin Biology Group, Gupta Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Carol Braggs
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Aakanksha Sawant
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Simone Vaz
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Rahul Thorat
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Sanjay Gupta
- Epigenetics and Chromatin Biology Group, Gupta Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Sorab N Dalal
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India.
| |
Collapse
|
12
|
Can NGAL be employed as prognostic and diagnostic biomarker in human cancers? A systematic review of current evidence. Int J Biol Markers 2017; 32:e53-e61. [PMID: 28106227 DOI: 10.5301/jbm.5000245] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2016] [Indexed: 12/30/2022]
Abstract
BACKGROUND Some studies have reported differentially altered neutrophil gelatinase-associated lipocalin (NGAL) levels in several malignancies. We evaluated NGAL measured in plasma or urine as both prognostic and diagnostic marker for different types of human tumors. METHODS We performed systematic electronic searches in Medline, Embase and CRDTAS. Studies were included if they evaluated NGAL as a prognostic or diagnostic marker for human cancers. The selection of the studies, screening of the full texts and data extraction were conducted independently by 2 authors. We used the random-effects model for the meta-analyses. A methodological assessment was completed. RESULTS We included 35 studies dedicated to colorectal, pancreas, breast, thyroid, gastric, kidney, endometrial, brain, liver, lung, esophageal, oral and ovarian cancers. Our meta-analyses showed that, in patients with colorectal and breast cancer, positive NGAL expression was associated with a decrease of disease-free survival (hazard ratio [HR] = 2.27, 95% confidence interval [95% CI], 1.54-3.36; HR = 1.78, 95% CI, 1.33-2.38, respectively). NGAL was a negative prognostic marker of overall survival in colorectal (HR = 2.37, 95% CI, 1.68-3.34) and endometrial (HR = 4.38, 95% CI, 1.9-10.12) cancers. Discriminative power of NGAL between cancer patients and control was moderate in colorectal cancer (area under the curve [AUC] = 0.6; pooled sensitivity 0.56; pooled specificity 0.72), acceptable in pancreatic cancer (AUC = 0.8; pooled sensitivity 0.6; pooled specificity 0.8) and good in thyroid cancer (AUC = 0.9; pooled sensitivity 0.85; pooled specificity 0.96). CONCLUSIONS NGAL determination in plasma and urine could be useful in the prognosis of colorectal and breast cancer, but its prognostic accuracy remains uncertain for other human tumors.
Collapse
|
13
|
Meng L, Wang M, Du Z, Fang Z, Wu B, Wu J, Xie W, Shen J, Zhu T, Xu X, Liao L, Xu L, Li E, Lan B. Cell Signaling Pathway in 12-O-Tetradecanoylphorbol-13-acetate-Induced LCN2 Gene Transcription in Esophageal Squamous Cell Carcinoma. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9592501. [PMID: 29098164 PMCID: PMC5642883 DOI: 10.1155/2017/9592501] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 08/22/2017] [Indexed: 02/06/2023]
Abstract
LCN2 is involved in various cellular functions, including transport of small hydrophobic molecules, protection of MMP9 from proteolytic degradation, and regulating innate immunity. LCN2 is elevated in multiple human cancers, frequently being associated with tumor size, stage, and invasiveness. Our previous studies have shown that LCN2 expression could be induced by 12-O-tetradecanoylphorbol-13-acetate (TPA) in esophageal squamous cell carcinoma (ESCC) by the binding of five nucleoproteins (MISP, KLF10, KLF15, PPP1R18, and RXRβ) at a novel TPA-responsive element (TRE), at -152~-60 bp of the 5' flanking region of the LCN2 promoter. However, much is unknown about whether these proteins can respond to TPA stimulation to regulate LCN2 transactivation and which cell signaling pathways mediate this process. In this study, expression plasmids encoding these five nucleoproteins were stably transfected into EC109 cells. Then, stable transfectant was characterized by a Dual-Luciferase Reporter Assay System. RT-PCR, real-time PCR, western blotting, specific kinase inhibitor treatment, and bioinformatics analyses were applied in this study. We found that MISP, KLF10, KLF15, PPP1R18, and RXRβ proteins could strongly respond to TPA stimulation and activate LCN2 transcriptional expression. MEK, ERK, JNK, and P38 kinases were involved in the LCN2 transactivation. Furthermore, the MEK-ERK signal pathway plays a major role in this biological process but does not involve PKCα signaling.
Collapse
Affiliation(s)
- Lingying Meng
- Department of Cardiothoracic Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong 515041, China
| | - Muting Wang
- Department of Cardiothoracic Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong 515041, China
| | - Zepeng Du
- Department of Pathology, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong 515041, China
| | - Zhongmin Fang
- Department of Cardiothoracic Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong 515041, China
| | - Bingli Wu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Jianyi Wu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Wenming Xie
- Network and Information Center, Shantou University Medical College, Shantou 515041, China
| | - Jian Shen
- Department of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Tianxiang Zhu
- Department of Cardiothoracic Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong 515041, China
| | - XieE Xu
- Department of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Liandi Liao
- Department of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Liyan Xu
- Department of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Enmin Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Bin Lan
- Department of Cardiothoracic Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong 515041, China
| |
Collapse
|
14
|
Lourdusamy V, Tharian B, Navaneethan U. Biomarkers in bile-complementing advanced endoscopic imaging in the diagnosis of indeterminate biliary strictures. World J Gastrointest Endosc 2015; 7:308-317. [PMID: 25901209 PMCID: PMC4400619 DOI: 10.4253/wjge.v7.i4.308] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 09/24/2014] [Accepted: 01/20/2015] [Indexed: 02/05/2023] Open
Abstract
Biliary strictures present a diagnostic challenge and a conundrum, particularly when an initial work up including abdominal imaging and endoscopic retrograde cholangiopancreatography based sampling are non-diagnostic. Advances in endoscopic imaging have helped us diagnose these strictures better. However, even with modern technology, some strictures remain a diagnostic challenge. The proximity of bile fluid to the bile duct epithelia makes it an attractive option to investigate for bio-markers, which might be representative of the functions/abnormal changes taking place in the biliary system. A number of biomarkers in bile have been discovered recently in approaching biliary strictures with their potential future diagnostic utility, further supported by the immunohistochemical analysis of the resected tissue specimens. Novel biliary biomarkers especially carcinoembryonic cell adhesion molecule 6 and neutrophil gelatinase-associated lipocalin seem promising in differentiating malignant from benign biliary strictures. Recent developments in lipidomic profiling of bile are also very promising. Biliary biomarkers appear to complement endoscopic imaging in diagnosing malignant etiologies of biliary stricture. Future studies addressing these biomarkers need to be incorporated to the current endoscopic techniques to determine the best approach in determining the etiology of biliary strictures.
Collapse
|
15
|
Diverse functional roles of lipocalin-2 in the central nervous system. Neurosci Biobehav Rev 2015; 49:135-56. [DOI: 10.1016/j.neubiorev.2014.12.006] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 11/28/2014] [Accepted: 12/04/2014] [Indexed: 12/16/2022]
|
16
|
Candido S, Maestro R, Polesel J, Catania A, Maira F, Signorelli SS, McCubrey JA, Libra M. Roles of neutrophil gelatinase-associated lipocalin (NGAL) in human cancer. Oncotarget 2015; 5:1576-94. [PMID: 24742531 PMCID: PMC4039233 DOI: 10.18632/oncotarget.1738] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cancer remains one of the major cause of death in the Western world. Although, it has been demonstrated that new therapies can improve the outcome of cancer patients, still many patients relapse after treatment. Therefore, there is a need to identify novel factors involved in cancer development and/or progression. Recently, neutrophil gelatinase-associated lipocalin (NGAL) has been suggested as a key player in different cancer types. Its oncogenic effect may be related to the complex NGAL/MMP-9. In the present study, NGAL was analyzed at both transcript and protein levels in different cancer types by analysing 38 public available microarray datasets and the Human Protein Atlas tool. NGAL transcripts were significantly higher in the majority of solid tumors compared to the relative normal tissues for every dataset analyzed. Furthermore, concordance of NGAL at both mRNA and protein levels was observed for 6 cancer types including bladder, colorectal, liver, lung, ovarian, and pancreatic. All metastatic tumors showed a decrease of NGAL expression when compared to matched primary lesions. According to these results, NGAL is a candidate marker for tumor growth in a fraction of solid tumors. Further investigations are required to elucidate the function of NGAL in tumor development and metastatic processes.
Collapse
Affiliation(s)
- Saverio Candido
- Department of Bio-medical Sciences, Section of Pathology and Oncology, Laboratory of Translational Oncology and Functional Genomics, University of Catania, Catania, (Italy)
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Duvillard L, Ortega-Deballon P, Bourredjem A, Scherrer ML, Mantion G, Delhorme JB, Deguelte-Lardière S, Petit JM, Bonithon-Kopp C. A case-control study of pre-operative levels of serum neutrophil gelatinase-associated lipocalin and other potential inflammatory markers in colorectal cancer. BMC Cancer 2014; 14:912. [PMID: 25472811 PMCID: PMC4289261 DOI: 10.1186/1471-2407-14-912] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 11/20/2014] [Indexed: 12/30/2022] Open
Abstract
Background Chronic inflammation is a key feature of colorectal cancer (CRC), meaning that inflammatory biomarkers may be useful for its diagnosis. In particular, high neutrophil gelatinase-associated lipocalin (NGAL) expression has been reported in CRC. Thus, we investigated whether serum NGAL and NGAL/MMP-9 could be potential biomarkers for the early detection of CRC. Concurrently, we studied other inflammatory biomarkers such as soluble tumor necrosis factor receptor 1 and 2 (sTNFR-1, sTNFR-2), and C reactive protein (CRP). Methods The AGARIC multicenter case–control study was performed in eastern France and included patients admitted for elective surgery either for a priori non-metastatic incident CRC (n = 224) or for benign causes (n = 252). Pre-operative serum levels of NGAL, NGAL/MMP-9, sTNFR-1, sTNFR-2 and CRP were measured. Results Median values of serum NGAL, NGAL/MMP-9, sTNFR-1, sTNFR-2 and CRP were significantly higher in CRC patients than in controls. Receiver Operating Characteristic analysis provided relatively poor values of area under the curve, ranging from 0.65 to 0.58. Except for NGAL/MMP-9, all biological parameters were strongly correlated in CRC cases and, less strongly in controls. Multivariate odds ratio (OR) of CRC comparing the extreme tertiles of serum NGAL was 2.76 (95% confidence interval (CI): 1.59-4.78; p < 0.001),. Lower but significant multivariate associations were observed for sTNFR-1, and sTNFR-2: OR = 2.44 (95% CI : 1.34-4.45, p = 0.015) and 1.93 (95% : CI 1.12-3.31), respectively. No independent association was found between case–control status and NGAL/MMP-9. Among CRC cases, maximal tumor size was an independent determinant of serum NGAL (p = 0.028) but this association was reduced after adjustment for CRP (p = 0.11). Conclusion Despite a significant increase in serum NGAL and other inflammatory markers among CRC patients, our findings suggest that they may not be suitable biomarkers for the diagnosis and especially early detection of CRC.
Collapse
|
18
|
Lippi G, Meschi T, Nouvenne A, Mattiuzzi C, Borghi L. Neutrophil gelatinase-associated lipocalin in cancer. Adv Clin Chem 2014; 64:179-219. [PMID: 24938019 DOI: 10.1016/b978-0-12-800263-6.00004-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL), also known as lipocalin-2, is a 178-amino acid protein which exists in three molecular forms, including a 25-kDa monomer, a 45-kDa homodimer, and a 135-kDa heterodimer complexed with matrix metalloproteinase 9 (MMP-9). Polymorphonuclear neutrophils and tubular cells of the kidney are the most representative cellular sources. As such, NGAL is now considered the biochemical gold standard for early diagnosis of acute kidney injury. Recent evidence suggests, however, that ectopic or enhanced expression of NGAL may occur in many other pathologic conditions including cancer. Several epidemiologic studies, as reviewed in this chapter, showed that a variety of malignant tumors consistently overexpressed NGAL with increased concentration in blood, urine, and other biologic fluids. In addition, NGAL was frequently associated with tumor size, stage, and invasiveness. These features thus make it a potential biomarker for malignancy. A number of experimental studies also demonstrated that the ability to bind MMP-9, to scavenge iron into cancer cells along with the effect on subcellular localization of transmembrane proteins such as cadherins and catenins, confers this protein the potential to enhance can cer aggressiveness and makes it an appealing target of future anticancer research.
Collapse
|
19
|
Maier HT, Aigner F, Trenkwalder B, Zitt M, Vallant N, Perathoner A, Margreiter C, Moser P, Pratschke J, Amberger A. Up-regulation of Neutrophil Gelatinase-Associated Lipocalin in Colorectal Cancer Predicts Poor Patient Survival. World J Surg 2014; 38:2160-7. [DOI: 10.1007/s00268-014-2499-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|