1
|
Drobiova H, Al-Mulla F, Al-Temaimi R. ADAM9 Genetic Variants and Their Role in Modulating Enzyme Activity in Diabetes and Metabolic Traits. J Diabetes Res 2025; 2025:5519447. [PMID: 40330740 PMCID: PMC12052454 DOI: 10.1155/jdr/5519447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
A disintegrin and metalloproteinase Domain 9 (ADAM9) is a zinc-dependent proteinase involved in various biological processes. However, its role in the pathophysiology of metabolic syndrome remains unclear, and studies exploring the association between ADAM9 polymorphisms and metabolic traits are limited. In this study, we investigated the potential link between ADAM9 variants and metabolic syndrome traits in a cohort of adult participants from Kuwait. Using a genome-wide association study (GWAS), followed by a replication study, we identified two ADAM9 variants-ADAM9-E76K (rs61753672) and ADAM9-P750L (rs144750648)-that were associated with various metabolic traits. The replication phase confirmed the association of ADAM9-P750L with HbA1c levels and revealed new associations with systolic blood pressure, waist-to-hip ratio, fasting blood glucose, triglycerides, and cholesterol. Functional analysis showed that both variants exhibited reduced proteolytic activity, potentially contributing to the pathogenesis of Type 2 diabetes. These findings suggest that ADAM9 variants may play a significant role in metabolic health and diabetes risk.
Collapse
Affiliation(s)
- Hana Drobiova
- Department of Pathology, College of Medicine, Kuwait University, Jabriya, Kuwait
| | - Fahd Al-Mulla
- Translational Medicine Department, Dasman Diabetes Institute, Dasman, Kuwait
| | - Rabeah Al-Temaimi
- Department of Pathology, College of Medicine, Kuwait University, Jabriya, Kuwait
| |
Collapse
|
2
|
Alonso-Nocelo M, Ruiz-Cañas L, Sancho P, Görgülü K, Alcalá S, Pedrero C, Vallespinos M, López-Gil JC, Ochando M, García-García E, David Trabulo SM, Martinelli P, Sánchez-Tomero P, Sánchez-Palomo C, Gonzalez-Santamaría P, Yuste L, Wörmann SM, Kabacaoğlu D, Earl J, Martin A, Salvador F, Valle S, Martin-Hijano L, Carrato A, Erkan M, García-Bermejo L, Hermann PC, Algül H, Moreno-Bueno G, Heeschen C, Portillo F, Cano A, Sainz B. Macrophages direct cancer cells through a LOXL2-mediated metastatic cascade in pancreatic ductal adenocarcinoma. Gut 2023; 72:345-359. [PMID: 35428659 PMCID: PMC9872246 DOI: 10.1136/gutjnl-2021-325564] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 03/21/2022] [Indexed: 01/27/2023]
Abstract
OBJECTIVE The lysyl oxidase-like protein 2 (LOXL2) contributes to tumour progression and metastasis in different tumour entities, but its role in pancreatic ductal adenocarcinoma (PDAC) has not been evaluated in immunocompetent in vivo PDAC models. DESIGN Towards this end, we used PDAC patient data sets, patient-derived xenograft in vivo and in vitro models, and four conditional genetically-engineered mouse models (GEMMS) to dissect the role of LOXL2 in PDAC. For GEMM-based studies, K-Ras +/LSL-G12D;Trp53 LSL-R172H;Pdx1-Cre mice (KPC) and the K-Ras +/LSL-G12D;Pdx1-Cre mice (KC) were crossed with Loxl2 allele floxed mice (Loxl2Exon2 fl/fl) or conditional Loxl2 overexpressing mice (R26Loxl2 KI/KI) to generate KPCL2KO or KCL2KO and KPCL2KI or KCL2KI mice, which were used to study overall survival; tumour incidence, burden and differentiation; metastases; epithelial to mesenchymal transition (EMT); stemness and extracellular collagen matrix (ECM) organisation. RESULTS Using these PDAC mouse models, we show that while Loxl2 ablation had little effect on primary tumour development and growth, its loss significantly decreased metastasis and increased overall survival. We attribute this effect to non-cell autonomous factors, primarily ECM remodelling. Loxl2 overexpression, on the other hand, promoted primary and metastatic tumour growth and decreased overall survival, which could be linked to increased EMT and stemness. We also identified tumour-associated macrophage-secreted oncostatin M (OSM) as an inducer of LOXL2 expression, and show that targeting macrophages in vivo affects Osm and Loxl2 expression and collagen fibre alignment. CONCLUSION Taken together, our findings establish novel pathophysiological roles and functions for LOXL2 in PDAC, which could be potentially exploited to treat metastatic disease.
Collapse
Affiliation(s)
- Marta Alonso-Nocelo
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Laura Ruiz-Cañas
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Patricia Sancho
- Translational Research Unit, Hospital Miguel Servet, Instituto de Investigacion Sanitaria Aragon, Zaragoza, Spain
| | - Kıvanç Görgülü
- Comprehensive Cancer Center München, Klinikum rechts der Isar der Technischen Universität München, München, Germany
| | - Sonia Alcalá
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Coral Pedrero
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Mireia Vallespinos
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Juan Carlos López-Gil
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Marina Ochando
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Elena García-García
- Departamento de Anatomía Patológica, Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| | - Sara Maria David Trabulo
- Stem Cells and Cancer Group, Molecular Pathology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Paola Martinelli
- Institute for Cancer Research, Comprehensive Cancer Center, Medizinische Universitat Wien, Wien, Austria
| | - Patricia Sánchez-Tomero
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Carmen Sánchez-Palomo
- Departamento de Anatomía, Histologia y Neurociencia, Universidad Autónoma de Madrid, Madrid, Spain
| | - Patricia Gonzalez-Santamaría
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
- Cancer and Human Molecular Genetics, Instituto de Investigación Sanitaria IdiPAZ, Madrid, Spain
| | - Lourdes Yuste
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- Cancer and Human Molecular Genetics, Instituto de Investigación Sanitaria IdiPAZ, Madrid, Spain
| | - Sonja Maria Wörmann
- Ahmed Cancer Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Derya Kabacaoğlu
- Comprehensive Cancer Center München, Klinikum rechts der Isar der Technischen Universität München, München, Germany
| | - Julie Earl
- Molecular Epidemiology and Predictive Tumor Markers Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain, Madrid, Spain
- Gastrointestinal Tumours Research Programme, Biomedical Research Network in Cancer (CIBERONC), Madrid, Spain
| | - Alberto Martin
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
| | - Fernando Salvador
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
| | - Sandra Valle
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Laura Martin-Hijano
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Alfredo Carrato
- Molecular Epidemiology and Predictive Tumor Markers Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain, Madrid, Spain
- Gastrointestinal Tumours Research Programme, Biomedical Research Network in Cancer (CIBERONC), Madrid, Spain
- Alcala University, Madrid, Spain
| | - Mert Erkan
- University Research Center for Translational Medicine - KUTTAM, Istanbul, Turkey
| | - Laura García-Bermejo
- Biomarkers and Therapeutic Targets Group, Area 4, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | | | - Hana Algül
- Comprehensive Cancer Center München, Klinikum rechts der Isar der Technischen Universität München, München, Germany
| | - Gema Moreno-Bueno
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
- Cancer and Human Molecular Genetics, Instituto de Investigación Sanitaria IdiPAZ, Madrid, Spain
- Breast Cancer Research Programme, Biomedical Research Network in Cancer (CIBERONC), Madrid, Spain
- Fundación MD Anderson Internacional, Madrid, Spain
| | - Christopher Heeschen
- Stem Cells and Cancer Group, Molecular Pathology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Center for Single-Cell Omics and Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Francisco Portillo
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
- Breast Cancer Research Programme, Biomedical Research Network in Cancer (CIBERONC), Madrid, Spain
| | - Amparo Cano
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
- Cancer and Human Molecular Genetics, Instituto de Investigación Sanitaria IdiPAZ, Madrid, Spain
- Breast Cancer Research Programme, Biomedical Research Network in Cancer (CIBERONC), Madrid, Spain
| | - Bruno Sainz
- Departament of Biochemistry, Universidad Autónoma de Madrid (UAM), Departament of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- Gastrointestinal Tumours Research Programme, Biomedical Research Network in Cancer (CIBERONC), Madrid, Spain
| |
Collapse
|
3
|
Xu F, Wang H, Pei H, Zhang Z, Liu L, Tang L, Wang S, Ren BC. SLC1A5 Prefers to Play as an Accomplice Rather Than an Opponent in Pancreatic Adenocarcinoma. Front Cell Dev Biol 2022; 10:800925. [PMID: 35419359 PMCID: PMC8995533 DOI: 10.3389/fcell.2022.800925] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/10/2022] [Indexed: 12/24/2022] Open
Abstract
Background: SLC1A5, a ferroptosis regulator gene, plays a dual role in cancer regulation. However, the roles of SLC1A5 in pancreatic adenocarcinoma (PAAD) remain elusive. Methods: SLC1A5’s expression and somatic mutation information were determined by TCGA, GEO, Oncomine, and cBioPortal databases. Its prognostic value was assessed in TCGA cohort and was validated in three independent cohorts. The effects of SLC1A5 on the tumor immune microenvironment were analyzed by the CIBERSORT algorithm, ssGSEA method, and TISIDB and TIMER databases. The “oncoPredict” R package, TIDE algorithm, ImmuCellAI online tool, and GSE35141 and GSE59357 datasets were used to ascertain its therapeutic correlations. GSEA and Western blot were applied to reveal the effects of SLC1A5 on the mTORC1 signaling pathway and ferroptosis process. The biofunctions of SLC1A5 were assessed by MTT, wound-healing, Transwell, and xenograft assays. Results: SLC1A5 was significantly upregulated in the PAAD samples but was not commonly accompanied with somatic mutation (2.3%). Overexpression of SLC1A5 led to a poor prognosis and was identified as an independent prognostic factor. Moreover, high SLC1A5 expression suppressed the antitumor immune process by changing the infiltrating levels of immune cells. As for therapeutic correlations, SLC1A5 was related to the efficacy of dasatinib, sunitinib, sorafenib, and imatinib but may not predict that of radiotherapy, chemotherapeutic drugs, and immune checkpoints inhibitors (ICIs). Notably, the overexpression of SLC1A5 could activate the mTORC1 signaling pathway and may increase the cellular sensitivity to ferroptosis. Finally, the overexpression of SLC1A5 markedly promoted proliferation, migration, and invasion of pancreatic cancer cells. At the in vivo level, SLC1A5 deletion inhibited tumor growth in a mice xenograft model. Conclusions: SLC1A5 prefers to play as an accomplice rather than an opponent in PAAD. Our findings provide novel insights into PAAD treatment.
Collapse
Affiliation(s)
- Fangshi Xu
- Department of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Hai Wang
- Department of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Honghong Pei
- Department of Emergency, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhengliang Zhang
- Department of Emergency, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liangliang Liu
- Department of Emergency, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Long Tang
- Department of Emergency, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Shuang Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Bin-Cheng Ren
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
4
|
Dong G, Lin LR, Xu LY, Li EM. Reaction mechanism of lysyl oxidase-like 2 (LOXL2) studied by computational methods. J Inorg Biochem 2020; 211:111204. [PMID: 32801097 DOI: 10.1016/j.jinorgbio.2020.111204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 02/05/2023]
Abstract
Lysyl oxidase-like 2 (LOXL2) is a copper-dependent amine oxidase that catalyzes the oxidative deamination of the ε-amino group of lysines/hydroxylysines on substrate proteins (collagen and elastin) to form aldehyde groups. The generated aldehyde groups are of significance in crosslinking with the adjacent aldehyde or ε-amino group on proteins in extracellular matrix. In this paper, we have studied the reaction mechanism of LOXL2 by means of quantum mechanics (QM) and combined QM and molecular mechanics (QM/MM) methods. This study is divided into two parts, i.e. the biosynthesis of lysine tyrosylquinone (LTQ) cofactor and oxidative deamination of ε-amino group of lysine by LTQ. For the former part, the reaction is driven by a large exothermicity of about 284 kJ/mol. Dopaquinone radical (DPQr) is suggested to be an intermediate state in this reaction. In addition, His652 residue is predicted to serve as proton acceptor. The rate-determining step for the biosynthesis of LTQ is found to be hydrogen-atom abstraction from the benzene ring on substrate by Cu2+-hydroxide, which is a proton-coupled electron transfer (PCET) process with an energy barrier of 84 kJ/mol. For the latter part, the reaction is exothermic by about 145 kJ/mol, and the copper ion is proposed to play a role of redox catalyst in the last step to generate the product of aldehyde. However, the copper ion might not be indispensable for the latter part, which is consistent with the previous study.
Collapse
Affiliation(s)
- Geng Dong
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, PR China; Medical Informatics Research Center, Shantou University Medical College, Shantou 515041, PR China.
| | - Li-Rui Lin
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, PR China; Medical Informatics Research Center, Shantou University Medical College, Shantou 515041, PR China
| | - Li-Yan Xu
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, PR China; Cancer Research Center, Shantou University Medical College, Shantou 515041, PR China
| | - En-Min Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, PR China; Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, PR China.
| |
Collapse
|
5
|
Khatri I, Bhasin MK. A Transcriptomics-Based Meta-Analysis Combined With Machine Learning Identifies a Secretory Biomarker Panel for Diagnosis of Pancreatic Adenocarcinoma. Front Genet 2020; 11:572284. [PMID: 33133160 PMCID: PMC7511758 DOI: 10.3389/fgene.2020.572284] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 08/12/2020] [Indexed: 12/22/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is generally incurable due to the late diagnosis and absence of markers that are concordant with expression in several sample sources (i.e., tissue, blood, plasma) and platforms (i.e., Microarray, sequencing). We optimized meta-analysis of 19 PDAC (tissue and blood) transcriptome studies from multiple platforms. The key biomarkers for PDAC diagnosis with secretory potential were identified and validated in different cohorts. Machine learning approach i.e., support vector machine supported by leave-one-out cross-validation was used to build and test the classifier. We identified a 9-gene panel (IFI27, ITGB5, CTSD, EFNA4, GGH, PLBD1, HTATIP2, IL1R2, CTSA) that achieved ∼0.92 average sensitivity and ∼0.90 average specificity in distinguishing PDAC from healthy samples in five training sets using cross-validation. These markers were also validated in proteomics and single-cell transcriptomics studies suggesting their prognostic role in the diagnosis of PDAC. Our 9-gene classifier can not only clearly discriminate between better and poor survivors but can also precisely discriminate PDAC from chronic pancreatitis (AUC = 0.95), early stages of progression [Stage I and II (AUC = 0.82), IPMA and IPMN (AUC = 1), and IPMC (AUC = 0.81)]. The 9-gene marker outperformed the previously known markers in blood studies particularly (AUC = 0.84). The discrimination of PDAC from early precursor lesions in non-malignant tissue (AUC > 0.81) and peripheral blood (AUC > 0.80) may assist in an early diagnosis of PDAC in blood samples and thus will also facilitate risk stratification upon validation in clinical trials.
Collapse
Affiliation(s)
- Indu Khatri
- Division of IMBIO, Department of Medicine, Beth Israel Lahey Health, Harvard Medical School, Boston, MA, United States.,Department of Immunology and Leiden Computational Biology Center, Leiden University Medical Center, Leiden, Netherlands
| | - Manoj K Bhasin
- Division of IMBIO, Department of Medicine, Beth Israel Lahey Health, Harvard Medical School, Boston, MA, United States.,Department of Pediatrics and Biomedical Informatics, Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, United States
| |
Collapse
|
6
|
Weidle UH, Birzele F, Nopora A. Pancreatic Ductal Adenocarcinoma: MicroRNAs Affecting Tumor Growth and Metastasis in Preclinical In Vivo Models. Cancer Genomics Proteomics 2020; 16:451-464. [PMID: 31659100 DOI: 10.21873/cgp.20149] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 02/08/2023] Open
Abstract
Patients with pancreatic ductal adenocarcinoma have a dismall prognosis because at the time of diagnosis, in the vast majority of patients the tumor has already disseminated to distant organs and the therapeutic benefit of approved agents such as gemcitabine is limited. Therefore, the identification and preclinical and clinical validation of therapeutic agents covering new targets is of paramount importance. In this review we have summarized microRNAs and corresponding targets which affect growth and metastasis of pancreatic tumors in preclinical mouse in vivo models. We identified four up-regulated and 16 down-regulated miRs in PDAC in comparison to corresponding normal tissues. Three sub-categories of miRs have emerged: miRs affecting tumor growth and miRs with an impact on both, tumor growth and metastasis or metastasis only. Finally, we discuss technical and therapeutic aspects of miR-related therapeutic agents for the treatment of pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Fabian Birzele
- Roche Innovation Center Basel, F. Hofman La Roche, Basel, Switzerland
| | - Adam Nopora
- Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| |
Collapse
|
7
|
Chen Z, Downing S, Tzanakakis ES. Four Decades After the Discovery of Regenerating Islet-Derived (Reg) Proteins: Current Understanding and Challenges. Front Cell Dev Biol 2019; 7:235. [PMID: 31696115 PMCID: PMC6817481 DOI: 10.3389/fcell.2019.00235] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/30/2019] [Indexed: 12/15/2022] Open
Abstract
Regenerating islet-derived (Reg) proteins have emerged as multifunctional agents with pro-proliferative, anti-apoptotic, differentiation-inducing and bactericidal properties. Over the last 40 years since first discovered, Reg proteins have been implicated in a gamut of maladies including diabetes, various types of cancer of the digestive tract, and Alzheimer disease. Surprisingly though, a consensus is still absent on the regulation of their expression, and molecular underpinning of their function. Here, we provide a critical appraisal of recent findings in the field of Reg protein biology. Specifically, the structural characteristics are reviewed particularly in connection with established or purported functions of different members of the Reg family. Moreover, Reg expression patterns in different tissues both under normal and pathophysiological conditions are summarized. Putative receptors and cascades reported to relay Reg signaling inciting cellular responses are presented aiming at a better appreciation of the biological activities of the distinct Reg moieties. Challenges are also discussed that have hampered thus far the rapid progress in this field such as the use of non-standard nomenclature for Reg molecules among various research groups, the existence of multiple Reg members with significant degree of homology and possibly compensatory modes of action, and the need for common assays with robust readouts of Reg activity. Coordinated research is warranted going forward, given that several research groups have independently linked Reg proteins to diseased states and raised the possibility that these biomolecules can serve as therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Zijing Chen
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, United States
| | - Shawna Downing
- Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA, United States
| | - Emmanuel S Tzanakakis
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, United States.,Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA, United States
| |
Collapse
|
8
|
Amendola PG, Reuten R, Erler JT. Interplay Between LOX Enzymes and Integrins in the Tumor Microenvironment. Cancers (Basel) 2019; 11:cancers11050729. [PMID: 31130685 PMCID: PMC6562985 DOI: 10.3390/cancers11050729] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/21/2019] [Accepted: 05/22/2019] [Indexed: 12/24/2022] Open
Abstract
Members of the lysyl oxidase (LOX) family are secreted copper-dependent amine oxidases that catalyze the covalent crosslinking of collagens and elastin in the extracellular matrix (ECM), an essential process for the structural integrity of all tissues. LOX enzymes can also remodel the tumor microenvironment and have been implicated in all stages of tumor initiation and progression of many cancer types. Changes in the ECM can influence several cancer cell phenotypes. Integrin adhesion complexes (IACs) physically connect cells with their microenvironment. This review article summarizes the main findings on the role of LOX proteins in modulating the tumor microenvironment, with a particular focus on how ECM changes are integrated by IACs to modulate cells behavior. Finally, we discuss how the development of selective LOX inhibitors may lead to novel and effective therapies in cancer treatment.
Collapse
Affiliation(s)
- Pier Giorgio Amendola
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Raphael Reuten
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Janine Terra Erler
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
9
|
Park JS, Lee JH, Lee YS, Kim JK, Dong SM, Yoon DS. Emerging role of LOXL2 in the promotion of pancreas cancer metastasis. Oncotarget 2018; 7:42539-42552. [PMID: 27285767 PMCID: PMC5173154 DOI: 10.18632/oncotarget.9918] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 05/04/2016] [Indexed: 12/20/2022] Open
Abstract
Lysyl oxidase-like 2 (LOXL2) is associated with invasiveness and metastasis in cancer. We analyzed the prognostic impact of LOXL2 in pancreatic cancer patients and investigated the role of LOXL2 in pancreatic cancer cell lines. Immunohistochemical analysis was performed in samples from 80 patients and showed LOXL2 expression in 81.2% of patients with pancreatic cancer. Regarding recurrence patterns, LOXL2-positive tumors showed a significantly higher rate of distant recurrence. The 1-year and 3-year disease-free survival rates were 84.6% and 0.0%, respectively, for LOXL2-negative patients, and 27.8 % and 0.0 %, respectively, for LOXL2-positive patients. On univariate analysis, combined resection of major vessels, depth of invasion, tumor stage, and LOXL2- positive status were significant factors for poor prognosis. After identification of LOXL2 expression in pancreatic cancer cell lines, LOXL2-silenced and LOXL2-overexpressed cell lines were used to perform transwell invasion and transendothelial migration assays. In vitro studies indicated that LOXL2 silencing in MIA PaCa-2 and PANC-1 cells induced a mesenchymal–epithelial transition (MET)-like process associated with decreased invasive and migratory properties. LOXL2 overexpression in AsPC-1 and BxPC-3 cells enhanced the epithelial-mesenchymal transition (EMT)-like process and increased migratory and invasive activity. These clinical and preclinical data confirm that higher LOXL2 expression is associated with the invasiveness of pancreatic cancer cells and the low survival rate of pancreatic cancer patients. Our results suggest the clinical value of LOXL2 as a therapeutic target in pancreatic cancer.
Collapse
Affiliation(s)
- Joon Seong Park
- Pancreatobiliary Cancer Clinic, Department of Surgery, Gangnam Severance Hospital, Yonsei University, Seoul, Korea
| | - Ji-Hae Lee
- Research Institute and Hospital, National Cancer Center, Goyang, Gyeonggi, Korea
| | - Yun Sun Lee
- Pancreatobiliary Cancer Clinic, Department of Surgery, Gangnam Severance Hospital, Yonsei University, Seoul, Korea
| | - Jae Keun Kim
- Pancreatobiliary Cancer Clinic, Department of Surgery, Gangnam Severance Hospital, Yonsei University, Seoul, Korea
| | - Seung Myung Dong
- Research Institute and Hospital, National Cancer Center, Goyang, Gyeonggi, Korea
| | - Dong Sup Yoon
- Pancreatobiliary Cancer Clinic, Department of Surgery, Gangnam Severance Hospital, Yonsei University, Seoul, Korea
| |
Collapse
|
10
|
Yuan P, He XH, Rong YF, Cao J, Li Y, Hu YP, Liu Y, Li D, Lou W, Liu MF. KRAS/NF-κB/YY1/miR-489 Signaling Axis Controls Pancreatic Cancer Metastasis. Cancer Res 2017; 77:100-111. [PMID: 27793842 DOI: 10.1158/0008-5472.can-16-1898] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 10/15/2016] [Accepted: 10/19/2016] [Indexed: 11/16/2022]
Abstract
KRAS activation occurring in more than 90% of pancreatic ductal adenocarcinomas (PDAC) drives progression and metastasis, but the underlying mechanisms involved in these processes are still poorly understood. Here, we show how KRAS acts through inflammatory NF-κB signaling to activate the transcription factor YY1, which represses expression of the tumor suppressor gene miR-489. In PDAC cells, repression of miR-489 by KRAS signaling inhibited migration and metastasis by targeting the extracellular matrix factors ADAM9 and MMP7. miR-489 downregulation elevated levels of ADAM9 and MMP7, thereby enhancing the migration and metastasis of PDAC cells. Together, our results establish a pivotal mechanism of PDAC metastasis and suggest miR-489 as a candidate therapeutic target for their attack. Cancer Res; 77(1); 100-11. ©2016 AACR.
Collapse
Affiliation(s)
- Peng Yuan
- Center for RNA Research, State Key Laboratory of Molecular Biology-University of Chinese Academy of Sciences, CAS Center for Excellence in Molecular Cell Science, Shanghai, China
- Shanghai Key Laboratory of Molecular Andrology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiao-Hong He
- Center for RNA Research, State Key Laboratory of Molecular Biology-University of Chinese Academy of Sciences, CAS Center for Excellence in Molecular Cell Science, Shanghai, China
- Shanghai Key Laboratory of Molecular Andrology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ye-Fei Rong
- Department of Pancreatic Surgery, Zhong Shan Hospital, Shanghai, China
| | - Jing Cao
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Yong Li
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Yun-Ping Hu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingbin Liu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dangsheng Li
- Shanghai Information Center for Life Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wenhui Lou
- Department of Pancreatic Surgery, Zhong Shan Hospital, Shanghai, China.
| | - Mo-Fang Liu
- Center for RNA Research, State Key Laboratory of Molecular Biology-University of Chinese Academy of Sciences, CAS Center for Excellence in Molecular Cell Science, Shanghai, China.
- Shanghai Key Laboratory of Molecular Andrology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| |
Collapse
|
11
|
Majeski HE, Yang J. The 2016 John J. Abel Award Lecture: Targeting the Mechanical Microenvironment in Cancer. Mol Pharmacol 2016; 90:744-754. [PMID: 27742780 PMCID: PMC5118638 DOI: 10.1124/mol.116.106765] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/13/2016] [Indexed: 12/14/2022] Open
Abstract
Past decades of cancer research have mainly focused on the role of various extracellular and intracellular biochemical signals on cancer progression and metastasis. Recent studies suggest an important role of mechanical forces in regulating cellular behaviors. This review first provides an overview of the mechanobiology research field. Then we specially focus on mechanotransduction pathways in cancer progression and describe in detail the key signaling components of such mechanotransduction pathways and extracellular matrix components that are altered in cancer. Although our understanding of mechanoregulation in cancer is still in its infancy, some agents against key mechanoregulators have been developed and will be discussed to explore the potential of pharmacologically targeting mechanotransduction in cancer.
Collapse
Affiliation(s)
- Hannah E Majeski
- Department of Pharmacology (H.E.M., J.Y.), Department of Pediatrics (J.Y.), and Biomedical Sciences Graduate Program (H.E.M., J.Y.), Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Jing Yang
- Department of Pharmacology (H.E.M., J.Y.), Department of Pediatrics (J.Y.), and Biomedical Sciences Graduate Program (H.E.M., J.Y.), Moores Cancer Center, University of California, San Diego, La Jolla, California
| |
Collapse
|
12
|
Erin N, İpekçi T, Akkaya B, Özbudak İH, Baykara M. Changes in expressions of ADAM9, 10, and 17 as well as α-secretase activity in renal cell carcinoma. Urol Oncol 2016; 35:36.e15-36.e22. [PMID: 27692848 DOI: 10.1016/j.urolonc.2016.08.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/17/2016] [Accepted: 08/19/2016] [Indexed: 01/10/2023]
Abstract
BACKGROUND ADAM9, 10, and 17 are a class of disintegrins and metallproteinases with α-secretase activity. There are conflicting results regarding the role(s) of ADAM9, 10, and 17 in carcinogenesis, and only a few studies have examined their levels and cellular localization in renal cell carcinoma (RCC). Studies examining changes in α-secretase activity in RCC compared to enzymatic activity of the uninvolved kidney are lacking. METHOD A cross-sectional study was conducted in 56 patients undergoing radical nephrectomy after the diagnosis of RCC. α-Secretase activity was determined using flourogenic substrate in freshly frozen tumor tissues as well as similarly treated tissues from the neighboring kidney. Immunohistochemical analyses of ADAM9, 10, and 17 were also performed. RESULTS α-Secretase activity decreased markedly in all types of RCC as compared to neighboring uninvolved kidney tissue having 5 to 10 times higher levels of α-secretase activity. Although type-dependent variations were observed, tumoral expressions of ADAMs, except for ADAM17, were lower in the tumors compared to that of neighboring tissues, but the changes in α-secretase activity were greater. In RCC tissue, ADAM9 expressions were localized in nuclear and cytoplasmic compartments, whereas ADAM10 and 17 were present predominately in the cytoplasm potentially explaining the markedly decreased enzyme activity. Membranous localization of ADAMs was noted in uninvolved kidney tissue. CONCLUSIONS The loss of α-secretase activity observed here in conjunction with previous findings argue against tumorigenic effects of ADAM9, 10, and 17 supporting that increased nuclear and cytoplasmic expression may be an attempt to compensate for loss of function.
Collapse
Affiliation(s)
- Nuray Erin
- Department of Medical Pharmacology, School of Medicine, Akdeniz University, Antalya, Turkey.
| | - Tümay İpekçi
- Department of Urology, Başkent University, Alanya, Turkey
| | - Bahar Akkaya
- Department of Urology, School of Medicine, Akdeniz University, Antalya, Turkey
| | - İrem Hicran Özbudak
- Department of Urology, School of Medicine, Akdeniz University, Antalya, Turkey; Department of Urology, Başkent University, Alanya, Turkey
| | - Mehmet Baykara
- Department of Pathology, School of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
13
|
Abstract
INTRODUCTION ADAM9 is a membrane-anchored metalloprotease that is markedly upregulated in several human carcinomas. We previously showed that ADAM9 staining was increased in colon cancer tissues. In the present work, we investigated the expression pattern of ADAM9 in colon cell lines, and the effects of overexpression ADAM9 on colon cancer proliferation and invasion. MATERIALS AND METHODS ADAM9 mRNA expression was determined in colon cancer tissues by RT-PCR assay, meanwhile, ADAM9 DNA and protein levels were measured in colon cancer cell lines by PCR and western blot, respectively. ADAM9 transfection was carried out in HT-29 colon cancer cells, and cell growth was measured by MTT assay, cell invasion ability was assessed by Matrigel assay. RESULTS ADAM9 mRNA was upregulated in colon cancer tissues, ADAM9 DNA and protein was overexpressed in Lovo cell lines, while the ADAM9 expression levels in HT-29, HCT-8, and Ls174t cells were below the levels of detection. Enhanced ADAM9 expression by ADAM9 transfection in HT-29 cells promoted cell invasion without increasing cell proliferation. CONCLUSIONS These results suggest that ADAM9 contributes to cell invasion and progression in colon cancer.
Collapse
Affiliation(s)
- Junsheng Li
- a Department of General Surgery, Affiliated Zhongda Hospital, Southeast University , Nanjing, China
| | - Zhenling Ji
- a Department of General Surgery, Affiliated Zhongda Hospital, Southeast University , Nanjing, China
| | - Cunzhong Qiao
- a Department of General Surgery, Affiliated Zhongda Hospital, Southeast University , Nanjing, China
| | - Yuhua Qi
- b Center of Disease Control , Jiangsu Province, China
| | - Wen Shi
- a Department of General Surgery, Affiliated Zhongda Hospital, Southeast University , Nanjing, China
| |
Collapse
|
14
|
Abstract
The new opportunities of modern assays of molecular biology can only be exploited fully if the results can be accurately correlated to the tissue phenotype under investigation. This is a general problem of non-in situ techniques, whereas results from in situ techniques are often difficult to quantify. The use of bulk tissue, which is not precisely characterized in terms of histology, has long been the basis for molecular analysis. It has, however, become apparent, that this simple approach is not sufficient for a detailed analysis of molecular alterations, which might be restricted to a specific tissue phenotype (e.g., tumor or normal tissue, stromal or epithelial cells). Microdissection is a method to provide minute amounts of histologically characterized tissues for molecular analysis with non-in situ techniques and has become an indispensable research tool. If tissue diversity is moderate and negligible, manual microdissection can be an easy and cost-efficient method of choice. In contrast, the advantage of laser microdissection is a very exact selection down to the level of a single cell, but often with a considerable time exposure to get enough material for the following analyses. The latter issue and the method of tissue preparation needed for laser microdissection are the main problems to solve if RNA, highly sensitive to degradation, shall be analyzed. This chapter focuses on optimized procedures for manual microdissection and laser microdissection to analyze RNA of malignant and nonmalignant prostate tissue.
Collapse
Affiliation(s)
- Anja Rabien
- Research Division, Department of Urology, Charité-Universitätsmedizin Berlin, Campus Charité Mitte, Charitéplatz 1, 10117, Berlin, Germany.
| | - Glen Kristiansen
- Universitätsklinikum Bonn (AöR), Institut für Pathologie, Bonn, Germany
| |
Collapse
|
15
|
Sempere LF. Tissue slide-based microRNA characterization of tumors: how detailed could diagnosis become for cancer medicine? Expert Rev Mol Diagn 2014; 14:853-69. [PMID: 25090088 PMCID: PMC4364265 DOI: 10.1586/14737159.2014.944507] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
miRNAs are short, non-coding, regulatory RNAs that exert cell type-dependent, context-dependent, transcriptome-wide gene expression control under physiological and pathological conditions. Tissue slide-based assays provide qualitative (tumor compartment) and semi-quantitative (expression levels) information about altered miRNA expression at single-cell resolution in clinical tumor specimens. Reviewed here are key technological advances in the last 5 years that have led to implementation of fully automated, robust and reproducible tissue slide-based assays for in situ miRNA detection on US FDA-approved instruments; recent tissue slide-based discovery studies that suggest potential clinical applications of specific miRNAs in cancer medicine are highlighted; and the challenges in bringing tissue slide-based miRNA assays into the clinic are discussed, including clinical validation, biomarker performance, biomarker space and integration with other biomarkers.
Collapse
Affiliation(s)
- Lorenzo F Sempere
- Laboratory of microRNA Diagnostics and Therapeutics, Van Andel Research Institute, 333 Bostwick Ave, N.E, Grand Rapids, MI 49503, USA
| |
Collapse
|
16
|
Lili LN, Matyunina LV, Walker LD, Daneker GW, McDonald JF. Evidence for the importance of personalized molecular profiling in pancreatic cancer. Pancreas 2014; 43:198-211. [PMID: 24518497 PMCID: PMC4206352 DOI: 10.1097/mpa.0000000000000020] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 10/28/2013] [Indexed: 12/11/2022]
Abstract
OBJECTIVES There is a growing body of evidence that targeted gene therapy holds great promise for the future treatment of cancer. A crucial step in this therapy is the accurate identification of appropriate candidate genes/pathways for targeted treatment. One approach is to identify variant genes/pathways that are significantly enriched in groups of afflicted individuals relative to control subjects. However, if there are multiple molecular pathways to the same cancer, the molecular determinants of the disease may be heterogeneous among individuals and possibly go undetected by group analyses. METHODS In an effort to explore this question in pancreatic cancer, we compared the most significantly differentially expressed genes/pathways between cancer and control patient samples as determined by group versus personalized analyses. RESULTS We found little to no overlap between genes/pathways identified by gene expression profiling using group analyses relative to those identified by personalized analyses. CONCLUSIONS Our results indicate that personalized and not group molecular profiling is the most appropriate approach for the identification of putative candidates for targeted gene therapy of pancreatic and perhaps other cancers with heterogeneous molecular etiology.
Collapse
Affiliation(s)
- Loukia N. Lili
- From the *Integrated Cancer Research Center, School of Biology, and Parker H. Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta; and †Cancer Treatment Centers of America SE Regional Facility, Newnan, GA
| | - Lilya V. Matyunina
- From the *Integrated Cancer Research Center, School of Biology, and Parker H. Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta; and †Cancer Treatment Centers of America SE Regional Facility, Newnan, GA
| | - L. DeEtte Walker
- From the *Integrated Cancer Research Center, School of Biology, and Parker H. Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta; and †Cancer Treatment Centers of America SE Regional Facility, Newnan, GA
| | - George W. Daneker
- From the *Integrated Cancer Research Center, School of Biology, and Parker H. Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta; and †Cancer Treatment Centers of America SE Regional Facility, Newnan, GA
| | - John F. McDonald
- From the *Integrated Cancer Research Center, School of Biology, and Parker H. Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta; and †Cancer Treatment Centers of America SE Regional Facility, Newnan, GA
| |
Collapse
|
17
|
Prognostic microRNAs in cancer tissue from patients operated for pancreatic cancer--five microRNAs in a prognostic index. World J Surg 2013; 36:2699-707. [PMID: 22851141 DOI: 10.1007/s00268-012-1705-y] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The aim of the present study was to identify a panel of microRNAs (miRNAs) that can predict overall survival (OS) in non micro-dissected cancer tissues from patients operated for pancreatic cancer (PC). METHODS MiRNAs were purified from formalin-fixed paraffin embedded (FFPE) cancer tissue from 225 patients operated for PC. Only a few of those patients received adjuvant chemotherapy. Expressions of miRNAs were determined with the TaqMan MicroRNA Array v2.0. Two statistical methods, univariate selection and the Lasso (Least Absolute Shrinkage and Selection Operator) method, were applied in conjunction with the Cox proportional hazard model to relate miRNAs to OS. RESULTS High expression of miR-212 and miR-675 and low expression of miR-148a, miR-187, and let-7g predicted short OS independent of age, gender, calendar year of operation, KRAS mutation status, tumor stage, American Society of Anesthesiologists (ASA) score, localization (not miR-148a), and differentiation of tumor. A prognostic index (PI) based on these five miRNAs was calculated for each patient. The median survival was 1.09 years (Confidence Interval [CI] 0.98-1.43) for PI > median PI compared to 2.23 years (CI 1.84-4.36) for PI < median. MiR-212, miR-675, miR-187, miR-205, miR-944, miR-431, miR-194, miR-148a, and miR-769-5p showed the strongest prediction ability by the Lasso method. Thus miR-212, miR-675, miR-187, and miR-148a were predictors for OS in both statistical methods. CONCLUSIONS The combination of five miRNAs expression in non micro-dissected FFPE PC tissue can identify patients with short OS after radical surgery. The results are independent of chemotherapy treatment. Patients with a prognostic index > median had a very short median OS of only 1 year.
Collapse
|
18
|
Abstract
The therapeutic targeting of extracellular proteins is becoming hugely attractive in light of evidence implicating the tumour microenvironment as pivotal in all aspects of tumour initiation and progression. Members of the lysyl oxidase (LOX) family of proteins are secreted by tumours and are the subject of much effort to understand their roles in cancer. In this Review we discuss the roles of members of this family in the remodelling of the tumour microenvironment and their paradoxical roles in tumorigenesis and metastasis. We also discuss how targeting this family of proteins might lead to a new avenue of cancer therapeutics.
Collapse
Affiliation(s)
- Holly E Barker
- Hypoxia & Metastasis Team, The Institute of Cancer Research, London SW3 6JB, UK
| | | | | |
Collapse
|
19
|
Gress TM, Kestler HA, Lausser L, Fiedler L, Sipos B, Michalski CW, Werner J, Giese N, Scarpa A, Buchholz M. Differentiation of multiple types of pancreatico-biliary tumors by molecular analysis of clinical specimens. J Mol Med (Berl) 2011; 90:457-64. [PMID: 22119958 DOI: 10.1007/s00109-011-0832-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 10/11/2011] [Accepted: 10/28/2011] [Indexed: 12/30/2022]
Abstract
Timely and accurate diagnosis of pancreatic ductal adenocarcinoma (PDAC) is critical in order to provide adequate treatment to patients. However, the clinical signs and symptoms of PDAC are shared by several types of malignant or benign tumors which may be difficult to differentiate from PDAC with conventional diagnostic procedures. Among others, these include ampullary cancers, solid pseudopapillary tumors, and adenocarcinomas of the distant bile duct, as well as inflammatory masses developing in chronic pancreatitis. Here, we report an approach to accurately differentiate between these different types of pancreatic masses based on molecular analysis of biopsy material. A total of 156 bulk tissue and fine needle aspiration biopsy samples were analyzed using a dedicated diagnostic cDNA array and a composite classification algorithm developed based on linear support vector machines. All five histological subtypes of pancreatic masses were clearly separable with 100% accuracy when using all 156 individual samples for classification. Generalized performance of the classification system was tested by 10 × 10-fold cross validation (100 test runs). Correct classification into the five diagnostic groups was demonstrated for 81.5% of 1,560 test set predictions. Performance increased to 85.3% accuracy when PDAC and distant bile duct carcinomas were combined in a single diagnostic class. Importantly, overall sensitivity of detection of malignant disease was 92.2%. The molecular diagnostic approach presented here is suitable to significantly aid in the differential diagnosis of undetermined pancreatic masses. To our knowledge, this is the first study reporting accurate differentiation between several types of pancreatico-biliary tumors in a single molecular analytical procedure.
Collapse
Affiliation(s)
- Thomas M Gress
- Division of Gastroenterology, University Hospital, Philipps-Universitaet Marburg, Marburg, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Barker HE, Erler JT. The potential for LOXL2 as a target for future cancer treatment. Future Oncol 2011; 7:707-10. [PMID: 21675833 DOI: 10.2217/fon.11.46] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
21
|
MicroRNA-148a is down-regulated in human pancreatic ductal adenocarcinomas and regulates cell survival by targeting CDC25B. J Transl Med 2011; 91:1472-9. [PMID: 21709669 DOI: 10.1038/labinvest.2011.99] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs: short non-coding RNAs) are emerging as a class of potential novel tumor markers, as their dysregulation is being increasingly reported in various types of cancers. In the present study, we investigated the transcription status of miRNA-148a (miR-148a) in human pancreatic ductal adenocarcinoma (PDAC) and its role in the regulation of the dual specificity protein phosphatase CDC25B. We observed that miR-148a exhibited a significant 4-fold down-regulation in PDAC as opposed to normal pancreatic ductal cells. In addition, we observed that stable lentiviral-mediated overexpression of miR-148a in the pancreatic cancer cell line IMIM-PC2, inhibited tumor cell growth and colony formation. Furthermore, CDC25B was identified as a potential target of miR-148a by in silico analysis using PicTar, Targetscan and miRanda in conjunction with gene ontology analysis. The proposed interaction between miR-148a and the 3' untranslated region (UTR) of CDC25B was verified by in-vitro luciferase assays. We demonstrate that the activity of a luciferase reporter containing the 3'UTR of CDC25B was repressed in the presence of miR-148a mimics, confirming that miR-148a targets the 3'UTR of CDC25B. Finally, CDC25B was down-regulated at the protein level in miR-148a overexpressing IMIM-PC2-cells, and in transiently transfected pancreatic cell lines (as detected by Western blot analysis), as well as in patient tumor samples (as detected by immunohistochemistry). In summary, we identified CDC25B as a novel miR-148a target which may confer a proliferative advantage in PDAC.
Collapse
|
22
|
Tissue and serum microRNAs in the Kras(G12D) transgenic animal model and in patients with pancreatic cancer. PLoS One 2011; 6:e20687. [PMID: 21738581 PMCID: PMC3124473 DOI: 10.1371/journal.pone.0020687] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Accepted: 05/06/2011] [Indexed: 12/30/2022] Open
Abstract
microRNAs (miRs) modulate the expression levels of mRNAs and proteins and can thus contribute to cancer initiation and progression. In addition to their intracelluar function, miRs are released from cells and shed into the circulation. We postulated that circulating miRs could provide insight into pathways altered during cancer progression and may indicate responses to treatment. Here we focus on pancreatic cancer malignant progression. We report that changes in miR expression patterns during progression of normal tissues to invasive pancreatic adenocarcinoma in the p48-Cre/LSL-KrasG12D mouse model mirrors the miR changes observed in human pancreatic cancer tissues. miR-148a/b and miR-375 expression were found decreased whereas miR-10, miR-21, miR-100 and miR-155 were increased when comparing normal tissues, premalignant lesions and invasive carcinoma in the mouse model. Predicted target mRNAs FGFR1 (miR-10) and MLH1 (miR-155) were found downregulated. Quantitation of nine microRNAs in plasma samples from patients distinguished pancreatic cancers from other cancers as well as non-cancerous pancreatic disease. Finally, gemcitabine treatment of control animals and p48-Cre/LSL-KrasG12D animals with pancreatic cancer caused distinct and up to 60-fold changes in circulating miRs that indicate differential drug effects on normal and cancer tissues. These findings support the significance of detecting miRs in the circulation and suggests that circulating miRs could serve as indicators of drug response.
Collapse
|
23
|
Rückert F, Aust D, Böhme I, Werner K, Brandt A, Diamandis EP, Krautz C, Hering S, Saeger HD, Grützmann R, Pilarsky C. Five primary human pancreatic adenocarcinoma cell lines established by the outgrowth method. J Surg Res 2011; 172:29-39. [PMID: 21683373 DOI: 10.1016/j.jss.2011.04.021] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2011] [Revised: 03/22/2011] [Accepted: 04/08/2011] [Indexed: 12/18/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma is an aggressive tumor; treatment remains a challenge because of the lack of effective therapeutic strategies. Basic research in this field is dependent on the availability of model systems. New pancreatic cancer cell lines are therefore important for the study of its biology. In the present study, we report the establishment and characterization of five new pancreatic cancer cell lines (PaCaDD-43, -60, -119, -135, -137). MATERIAL AND METHODS All cell lines were derived from pancreatic ductal adenocarcinomas by the Dresden outgrowth protocol. The five cell lines originated from primary pancreatic tumors, lymph node metastases, or malignant pleural effusions. We characterized the cell lines by examining their morphology and their cytostructural and functional profiles. RESULTS All cell lines grew as adherent monolayers and were cultured in optimized Dresden-medium. The doubling time ranged from 22 to 47 h. v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations were detected in four of the five cell lines. KRAS mutations were identical between each primary tumor and the cell line derived from it. Immunohistochemical staining showed cytoplasmic expression of CK8/18, mostly membrane and partially cytoplasmic expression of E-cadherin and strong expression of ezrin in all cell lines. Three cell lines showed nuclear p53 accumulation and heterogeneous expression of vimentin. SMAD4 was heterogeneously expressed in four of the cell lines. CONCLUSIONS We were able to establish five new primary pancreatic carcinoma cell lines. As applicable tools for basic research, these cell lines might contribute to a better understanding and treatment of this aggressive tumor.
Collapse
Affiliation(s)
- Felix Rückert
- Department of Visceral, Thoracic, and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Maker AV, Katabi N, Gonen M, DeMatteo RP, D'Angelica MI, Fong Y, Jarnagin WR, Brennan MF, Allen PJ. Pancreatic cyst fluid and serum mucin levels predict dysplasia in intraductal papillary mucinous neoplasms of the pancreas. Ann Surg Oncol 2011; 18:199-206. [PMID: 20717734 PMCID: PMC4241376 DOI: 10.1245/s10434-010-1225-7] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Indexed: 02/06/2023]
Abstract
INTRODUCTION There are no reliable markers of dysplasia in patients with incidentally discovered intraductal papillary mucinous neoplasms of the pancreas (IPMN). IPMN dysplasia may be associated with mucin protein (MUC) expression and histopathologic subtype. We hypothesize that MUC expression in cyst fluid and serum can identify lesions with high risk of malignancy. METHODS Cyst fluid and serum were collected from 40 patients during pancreatectomy for IPMN between 2005 and 2009. Samples were grouped into low-risk (low-grade or moderate dysplasia, n = 21) and high-risk groups (high-grade dysplasia or carcinoma, n = 19). Mucin expression (MUC1, MUC2, MUC4, and MUC5AC) was assessed utilizing enzyme-linked immunosorbent assays. RESULTS MUC2 and MUC4 cyst fluid concentrations were elevated in high-risk versus low-risk groups (10 ± 3.0 ng/ml vs. 4.4 ± 1.2 ng/ml, p = 0.03; 20.6 ± 10.6 ng/ml vs. 4.5 ± 1.4 ng/ml, p = 0.03, respectively). Corresponding serum samples revealed higher levels of MUC5AC in high-risk compared with low-risk patients (19.9 ± 9.3 ng/ml vs. 2.2 ± 1.1 ng/ml, p = 0.04). Histopathologic subtype was significantly associated with grade of dysplasia, and the intestinal subtype displayed increased MUC2 cyst fluid concentrations (13.8 ± 6.5 ng/ml vs. 4.1 ± 0.9 ng/ml, p = 0.02). CONCLUSIONS In this study, high-risk IPMN showed elevated cyst fluid concentrations of MUC2 and MUC4, and increased serum levels of MUC5AC. High-risk IPMN also displayed a distinct mucin expression profile in specific histologic subtypes. These data, if validated, may allow surgeons to more appropriately select patients for operative resection.
Collapse
MESH Headings
- Adenocarcinoma, Mucinous/diagnosis
- Adenocarcinoma, Mucinous/metabolism
- Adenocarcinoma, Mucinous/surgery
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/metabolism
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/surgery
- Carcinoma, Papillary/diagnosis
- Carcinoma, Papillary/metabolism
- Carcinoma, Papillary/surgery
- Cysts/chemistry
- Enzyme-Linked Immunosorbent Assay
- Female
- Humans
- Immunoenzyme Techniques
- Male
- Mucin 5AC/metabolism
- Mucin-1/metabolism
- Mucin-2/metabolism
- Mucin-4/metabolism
- Pancreatectomy
- Pancreatic Juice/metabolism
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/surgery
- Prognosis
- Prospective Studies
Collapse
Affiliation(s)
- Ajay V Maker
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Serum tumor markers in pancreatic cancer-recent discoveries. Cancers (Basel) 2010; 2:1107-24. [PMID: 24281109 PMCID: PMC3835121 DOI: 10.3390/cancers2021107] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Revised: 05/21/2010] [Accepted: 05/24/2010] [Indexed: 12/25/2022] Open
Abstract
The low prevalence of pancreatic cancer remains an obstacle to the development of effective screening tools in an asymptomatic population. However, development of effective serologic markers still offers the potential for improvement of diagnostic capabilities, especially for subpopulations of patients with high risk for pancreatic cancer. The accurate identification of patients with pancreatic cancer and the exclusion of disease in those with benign disorders remain important goals. While clinical experience largely dismissed many candidate markers as useful markers of pancreatic cancer, CA19-9 continues to show promise. The present review highlights the development and the properties of different tumor markers in pancreatic cancer and their impact on the diagnostic and treatment of this aggressive disease.
Collapse
|
26
|
Tao K, Qian N, Tang Y, Ti Z, Song W, Cao D, Dou K. Increased expression of a disintegrin and metalloprotease-9 in hepatocellular carcinoma: implications for tumor progression and prognosis. Jpn J Clin Oncol 2010; 40:645-51. [PMID: 20388695 DOI: 10.1093/jjco/hyq030] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE A disintegrin and metalloprotease-9 has been involved in the carcinogenesis of various solid tumors. However, its role in hepatocellular carcinoma remains unknown. The aim of this study was to investigate the clinicopathological and prognostic relevance of a disintegrin and metalloprotease-9 by immunohistochemistry. METHODS The expression profile of a disintegrin and metalloprotease-9 in association with the clinicopathological factors was determined by immunohistochemical analysis in hepatocellular carcinoma patients, and its potential prognostic value was investigated by comparing the survival rate between a disintegrin and metalloprotease-9-positive and a disintegrin and metalloprotease-9-negative patients. RESULTS Hepatocellular carcinoma tissues with positive a disintegrin and metalloprotease-9 expression were larger and less differentiated than those with negative expression (P = 0.02 and 0.008, respectively). Portal venous invasion, hepatic venous invasion, bile duct invasion and intrahepatic metastasis were detected significantly more frequently in a disintegrin and metalloprotease-9-positive group (P = 0.009, 0.01, 0.03 and 0.02, respectively). In addition, high alpha-fetoprotein levels were significantly associated with the expression of a disintegrin and metalloprotease-9 in hepatocellular carcinoma (P = 0.01). Moreover, a disintegrin and metalloprotease-9-positive group had significantly poorer outcomes than a disintegrin and metalloprotease-9-negative group (P = 0.01) and was an independent prognostic factor for overall survival. CONCLUSIONS A disintegrin and metalloprotease-9 is over-expressed in hepatocellular carcinoma tissues, consistent with findings in other tumor entities, and is an independent prognostic marker of overall survival following hepatectomy. Further studies are needed to investigate the precise function of a disintegrin and metalloprotease-9 in the progression of hepatocellular carcinoma.
Collapse
Affiliation(s)
- KaiShan Tao
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
OBJECTIVES Lysyl oxidase-like 2 (LOXL2) plays a part in epithelial-mesenchymal transition (EMT) by stabilizing the transcription factor SNAI1. Previous studies showed that LOXL2 is one of the most highly and specifically upregulated genes in pancreatic cancer. LOXL2 was also found to be strongly upregulated in the secretome of established pancreatic cancer cell lines. To get more insight into the aggressive growth and infiltrating nature of pancreatic cancer, we evaluated the functional role of LOXL2 in pancreatic cancer cells. METHODS Gene inhibition by small interfering RNAs was used to silence LOXL2 in pancreatic cancer cell lines MiaPaCa-2 and Panc1. Cell death, proliferation, and morphology of transfected cells were determined. Cell characteristics under cell stress and gemcitabine treatment were analyzed. Gene expression analysis of transfected cells by DNA microarray was used to understand the processes of chemosensitization. RESULTS Silencing of LOXL2 in pancreatic cancer cells resulted in an augmented sensitivity toward gemcitabine treatment, with significantly elevated cell death and reduced viable cells. However, transfection had no direct effect on morphology or growth pattern of Mia PaCa-2 and Panc1 cell lines. Gene expression analysis identified among others the transcription factor E2F5 as possible target of LOXL2. CONCLUSIONS Gene inhibition of the EMT regulatory gene LOXL2 resulted in a distinct sensitization toward gemcitabine. Additionally, gene expression analysis showed a role for LOXL2 in the regulation of different transcription factors associated with invasion and metastasis. Our results suggest that the improved response toward chemotherapy in LOLX2-silenced pancreatic cancer cells is possibly mediated by the transcription factor E2F5.
Collapse
|
28
|
Takahashi N, Fukushima T, Yorita K, Tanaka H, Chijiiwa K, Kataoka H. Dickkopf-1 is overexpressed in human pancreatic ductal adenocarcinoma cells and is involved in invasive growth. Int J Cancer 2010; 126:1611-20. [PMID: 19711349 DOI: 10.1002/ijc.24865] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The protein products of the Dickkopf (DKK) genes are antagonists of Wnt glycoproteins, which participate in tumor development and progression by binding to frizzled receptors. In this study, the expression of DKK-1 was analyzed in a panel of 43 human cultured carcinoma cell lines. DKK-1 expression was consistently and significantly upregulated in pancreatic carcinoma cell lines. Low level of DKK-3 expression was also seen. In contrast, the expression of DKK-2 and -4 was not detectable in most pancreatic carcinoma cell lines. The overexpression of DKK-1 was confirmed in surgically resected human pancreatic cancer tissues, in which the mRNA level was evaluated in paired samples from cancerous and noncancerous pancreatic tissues. In ductal adenocarcinomas (23 cases), DKK-1 mRNA levels were significantly upregulated compared to corresponding noncancerous tissues in a statistically significant level. To test the biological role of DKK-1 in pancreatic carcinoma cells, we performed a knockdown of DKK-1 in SUIT-2 human pancreatic adenocarcinoma cell line and S2-CP8, its metastatic subline, using a retroviral short hairpin RNA expression vector. DKK-1 knockdown resulted in reduced migratory activity of SUIT-2 in vitro. The in vitro growth rate and Matrigel invasion were also suppressed by DKK-1 knockdown in S2-CP8 cells. Collectively, the evidence suggests that, despite of its presumed antagonistic role in Wnt signaling, DKK-1 may have a role in the aggressiveness of pancreatic carcinoma cells and could, therefore, serve as a novel biomarker of pancreatic cancer.
Collapse
Affiliation(s)
- Nobuyasu Takahashi
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Kiyotake, Miyazaki, Japan
| | | | | | | | | | | |
Collapse
|
29
|
Radulovich N, Pham NA, Strumpf D, Leung L, Xie W, Jurisica I, Tsao MS. Differential roles of cyclin D1 and D3 in pancreatic ductal adenocarcinoma. Mol Cancer 2010; 9:24. [PMID: 20113529 PMCID: PMC2824633 DOI: 10.1186/1476-4598-9-24] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Accepted: 02/01/2010] [Indexed: 01/24/2023] Open
Abstract
Background The cyclin D1 (CCND1) and cyclin D3 (CCND3) are frequently co-overexpressed in pancreatic ductal adenocarcinoma (PDAC). Here we examine their differential roles in PDAC. Results CCND1 and CCND3 expression were selectively suppressed by shRNA in PDAC cell lines with expression levels of equal CCND1 and CCND3 (BxPC3), enhanced CCND1 (HPAC) or enhanced CCND3 (PANC1). Suppression of cell proliferation was greater with CCND3 than CCND1 downregulation. CCND3 suppression led to a reduced level of phosphorylated retinoblastoma protein (Ser795p-Rb/p110) and resulted in decreased levels of cyclin A mRNA and protein. A global gene expression analysis identified deregulated genes in D1- or D3-cyclin siRNA-treated PANC1 cells. The downregulated gene targets in CCND3 suppressed cells were significantly enriched in cell cycle associated processes (p < 0.005). In contrast, focal adhesion/actin cytoskeleton, MAPK and NF B signaling appeared to characterize the target genes and their interacting proteins in CCND1 suppressed PANC1 cells. Conclusions Our results suggest that CCND3 is the primary driver of the cell cycle, in cooperation with CCND1 that integrates extracellular mitogenic signaling. We also present evidence that CCND1 plays a role in tumor cell migration. The results provide novel insights for common and differential targets of CCND1 and CCND3 overexpression during pancreatic duct cell carcinogenesis.
Collapse
Affiliation(s)
- Nikolina Radulovich
- Ontario Cancer Institute and Princess Margaret Hospital, University Health Network, Toronto, Ontario M5G 2M9, Canada
| | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
The new opportunities of modern assays of molecular biology can only be exploited fully if the results can be accurately correlated to the tissue phenotype under investigation. This is a general problem of non-in situ techniques, whereas results from in situ techniques are often difficult to quantitate. The use of bulk tissue, which is not precisely characterized in terms of histology, has long been the basis for molecular analysis. It has, however, become apparent, that this simple approach is not sufficient for a detailed analysis of molecular alterations, which might be restricted to a specific tissue phenotype (e.g., tumor or healthy tissue, stromal or epithelial cells). Microdissection is a method to provide minute amounts of histologically characterized tissues for molecular analysis with non-in situ techniques and has become an indispensable research tool. Here, we describe a very simple technique for microdissection of tissues that can easily and cost effectively be established.
Collapse
Affiliation(s)
- Glen Kristiansen
- Department of Pathology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
31
|
Buchholz M, Gress TM. Molecular changes in pancreatic cancer. Expert Rev Anticancer Ther 2009; 9:1487-97. [PMID: 19828010 DOI: 10.1586/era.09.107] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
As with many human malignancies, pancreatic cancer is a complex genetic disorder. Several thousand disease-associated alterations on the DNA, mRNA, miRNA and protein levels have been reported to date. Some of these alterations, including a number of gatekeeper mutations, which are of pre-eminent importance for the onset and progression of the disease, have been extensively studied in primary tissues, in vitro experiments and transgenic mouse models. For the vast majority of alterations, however, data about the functional significance are lacking. The situation is complicated by the fact that no certainty exists concerning the identity of the cells that originally undergo malignant transformation nor about the precise nature and fate of premalignant lesions that are observed in pancreatic tissues.
Collapse
Affiliation(s)
- Malte Buchholz
- Klinik f. Innere Medizin, SP Gastroenterologie, Universitätsklinikum Marburg, Baldingerstrasse 35043 Marburg, Germany.
| | | |
Collapse
|
32
|
Streit S, Michalski CW, Erkan M, Friess H, Kleeff J. Confirmation of DNA microarray-derived differentially expressed genes in pancreatic cancer using quantitative RT-PCR. Pancreatology 2009; 9:577-82. [PMID: 19657213 DOI: 10.1159/000212084] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The fact that pancreatic ductal adenocarcinoma (PDAC) is still an exceptionally lethal disease with an annual mortality almost equivalent to its annual incidence has stimulated intense research efforts directed at understanding the underlying molecular mechanisms. By enabling simultaneous expression analysis of thousands of genes, microarray technology has significantly contributed to illuminating the pathophysiology of PDAC. Gene expression profiling studies have been performed for molecular classification of clinically relevant tumor subtypes and have shed light on various signaling pathways associated with tumor progression. Altered expression levels of several genes have been identified as correlating with functional in vitro data as well as patient survival, indicating the potential clinical value of transcriptional profiling. However, broad clinical use of array techniques for patient characterization has been hampered by their cost intensity and by limited inter-study comparability. Quantitative real-time reverse transcription-polymerase chain reaction (RT-PCR), as the most sensitive technique for mRNA detection and quantification, will complement arrays for the confirmation of individual transcripts in larger sample cohorts. This review highlights recent studies that addressed gene expression analysis with both methodologies and that identified components of the TGF-beta signaling pathway, BNIP3, or periostin to be differentially expressed in PDAC. These studies demonstrated that the combination of microarray and RT-PCR technologies is a highly efficient and reliable approach for the identification of clinically important diagnostic and prognostic biomarkers, as well as for the discovery of novel therapeutic target candidates.
Collapse
Affiliation(s)
- Sylvia Streit
- Department of Surgery, Technische Universitat Munchen, Munich, Germany
| | | | | | | | | |
Collapse
|
33
|
Abstract
Pancreatic cancer is a deadly disease with an annual incidence rate nearly equal to its mortality rate. Incremental improvement in outcome has been seen in the last 25 years, illustrating the critical need for novel approaches and intensive research investment. Expression profiling of pancreatic cancers has led to an explosion of informative gene-expression changes and the identification of new diagnostic and prognostic markers. However, the search for genes that are of functional significance in these large datasets continues to be much more challenging. One approach to focusing on genes or pathways that are likely to be more biologically relevant is to study those that are of prognostic significance. This review will therefore focus on the advantages of a prognostic gene signature for pancreatic cancer, the advances that have been made thus far, the approaches used and the challenges that remain.
Collapse
Affiliation(s)
- Jen Jen Yeh
- University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, Division of Surgical Oncology, Campus Box 7213, 1150 POB, 170 Manning Drive, Chapel Hill, NC 27599-7213, USA.
| |
Collapse
|
34
|
Yamazaki K, Takamura M, Masugi Y, Mori T, Du W, Hibi T, Hiraoka N, Ohta T, Ohki M, Hirohashi S, Sakamoto M. Adenylate cyclase-associated protein 1 overexpressed in pancreatic cancers is involved in cancer cell motility. J Transl Med 2009; 89:425-32. [PMID: 19188911 DOI: 10.1038/labinvest.2009.5] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Pancreatic cancer has the worst prognosis among cancers due to the difficulty of early diagnosis and its aggressive behavior. To characterize the aggressiveness of pancreatic cancers on gene expression, pancreatic cancer xenografts transplanted into severe combined immunodeficient mice served as a panel for gene-expression profiling. As a result of profiling, the adenylate cyclase-associated protein 1 (CAP1) gene was shown to be overexpressed in all of the xenografts. The expression of CAP1 protein in all 73 cases of pancreatic cancer was recognized by immunohistochemical analyses. The ratio of CAP1-positive tumor cells in clinical specimens was correlated with the presence of lymph node metastasis and neural invasion, and also with the poor prognosis of patients. Immunocytochemical analyses in pancreatic cancer cells demonstrated that CAP1 colocalized to the leading edge of lamellipodia with actin. Knockdown of CAP1 by RNA interference resulted in the reduction of lamellipodium formation, motility, and invasion of pancreatic cancer cells. This is the first report demonstrating the overexpression of CAP1 in pancreatic cancers and suggesting the involvement of CAP1 in the aggressive behavior of pancreatic cancer cells.
Collapse
Affiliation(s)
- Ken Yamazaki
- Department of Pathology, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Ranganathan P, Harsha HC, Pandey A. Molecular alterations in exocrine neoplasms of the pancreas. Arch Pathol Lab Med 2009; 133:405-12. [PMID: 19260746 DOI: 10.5858/133.3.405] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2008] [Indexed: 11/06/2022]
Abstract
CONTEXT Pancreatic cancer is one of the leading causes of cancer-related deaths. Most cases are diagnosed at an advanced stage when the disease is beyond surgical intervention. Molecular studies during the past decade have contributed greatly to our understanding of this disease. Various germ-line and somatic mutations associated with pancreatic cancers have been characterized, along with abnormal variations in the gene expression patterns. A thorough characterization of molecular alterations such as genetic and epigenetic changes, alterations in the expression of genes and changes in proteins, and posttranslational modifications in pancreatic cancer could lead to a better understanding of its pathogenesis. OBJECTIVE To provide an overview of the various molecular alterations in pancreatic cancer and the methodologies used to catalog such alterations. DATA SOURCES Published studies about various molecular alterations at the genomic, epigenetic, transcriptomic, and proteomic levels in pancreatic cancer. CONCLUSIONS The available data from pancreatic cancer suggests that there are a large number of molecular alterations at genomic, epigenetic, transcriptomic, and proteomic levels. It is now possible to initiate a systems approach to studying pancreatic cancer especially in light of newer initiatives to dissect the pancreatic cancer genome.
Collapse
|
36
|
Sipos B, Frank S, Gress T, Hahn S, Klöppel G. Pancreatic intraepithelial neoplasia revisited and updated. Pancreatology 2008; 9:45-54. [PMID: 19077454 DOI: 10.1159/000178874] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Most pancreatic neoplasms are classified as ductal adenocarcinoma because they show a ductal phenotype, making a ductal origin very likely. The duct lesions that may give rise to pancreatic ductal adenocarcinoma have been called pancreatic intraepithelial neoplasia (PanIN). A classification system for these lesions distinguishes between three grades of PanIN. Molecular studies revealed that PanIN-2 and PanIN-3 lesions represent a distinct step towards invasive carcinoma. While high-grade PanINs are extremely rare in the normal pancreas, low-grade PanINs are common in individuals older than 40 years and may be associated with lobular fibrosis and intraductal papillary mucinous neoplasms of the gastric type. This disease spectrum has also been described in members of kindreds with familial pancreatic cancer. The natural history and cause of PanINs are unknown. As PanIN-1 lesions entail little risk, while PanIN-3 lesions are high-risk lesions, it would be of interest to target PanIN-2 lesions, which can be regarded as the starting point of progressive neoplastic changes that lead to invasive pancreatic ductal adenocarcinoma. Global gene expression analysis identified several differentially expressed genes which show enhanced expression in PanINs and may be used as potential biomarkers to facilitate diagnosis and therapy.
Collapse
Affiliation(s)
- B Sipos
- Department of Pathology, University of Kiel, Kiel, Germany.
| | | | | | | | | |
Collapse
|
37
|
ADAM9 Expression is a Significant and Independent Prognostic Marker of PSA Relapse in Prostate Cancer. Eur Urol 2008; 54:1097-106. [DOI: 10.1016/j.eururo.2007.11.034] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2007] [Accepted: 11/12/2007] [Indexed: 01/13/2023]
|
38
|
Rückert F, Hennig M, Petraki CD, Wehrum D, Distler M, Denz A, Schröder M, Dawelbait G, Kalthoff H, Saeger HD, Diamandis EP, Pilarsky C, Grützmann R. Co-expression of KLK6 and KLK10 as prognostic factors for survival in pancreatic ductal adenocarcinoma. Br J Cancer 2008; 99:1484-92. [PMID: 18854834 PMCID: PMC2579692 DOI: 10.1038/sj.bjc.6604717] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Kallikreins play an important role in tumour microenvironment and as cancer biomarkers in different cancer entities. Previous studies suggested an upregulation of KLK10 and KLK6 in pancreatic ductal adenocarcinoma (PDAC). Therefore, we evaluated the clinicopathological role of these kallikreins and their value as biomarkers in PDAC. Differential expression was validated by DNA-microarrays and immunohistochemistry in normal and malignant pancreatic tissues. Sera concentrations of both kallikreins were evaluated using ELISA. In silico analysis of possible protein interactions and gene silencing of KLK10 in vitro using siRNAs gave further insights in the pathomechanisms. Gene expression analysis and immunohistochemistry demonstrated a strong expression for KLK10 and KLK6 in PDAC. Statistical analysis showed that co-expression of these kallikreins correlated with an R1-resection status (P=0.017) and worse outcome for overall survival (P=0.031). Multivariate analysis proofed that co-expression is an independent prognostic factor for survival (P=0.043). Importantly, KLK10 knockdown in AsPC-1 cells significantly reduced cell migration, whereas computational analysis suggested interaction of KLK6 with angiogenetic factors as an important mechanism. Co-expression of KLK10 and KLK6 plays an unfavourable role in PDAC. Our results suggest that this effect is likely mediated by an interaction with the factors of the extracellular matrix and enhancement of cancer cell motility.
Collapse
Affiliation(s)
- F Rückert
- Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technical University of Dresden, Fetscherstrasse 74, Dresden 01307, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Buchholz M, Kestler H, Gress TM. Differential diagnosis of pancreatic tumors by molecular analysis of clinical specimens. Pancreatology 2008; 8:551-7. [PMID: 18818507 DOI: 10.1159/000159213] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To describe the capabilities and limitations of the current state of the art in pancreatic cancer diagnostics and to discuss recent progress in the development of novel, highly accurate molecular diagnostic approaches. RESULTS Molecular analyses currently under evaluation as novel diagnostic tests include detection of point mutations, genomic imbalances, aberrant methylation patterns and gene expression changes on the mRNA and protein levels in pancreatic juice, fine needle aspiration biopsies and brush cytologies. CONCLUSIONS In exploratory studies, several candidate molecular markers show great potential to serve as general indicators of malignancy, but need to be validated in large, controlled, prospective studies. Multiplexing of diagnostic tests, e.g. in the form of specialized DNA microarrays, may provide more differentiated diagnoses such as the distinction of various tumor types or prognostic information for individual patients. The MolDiag-Paca consortium is strongly engaged in advancing these developments on a European level.
Collapse
Affiliation(s)
- Malte Buchholz
- Division of Gastroenterology, University Hospital, Philipps-Universität Marburg, Marburg, Germany
| | | | | |
Collapse
|
40
|
Fritzsche FR, Wassermann K, Jung M, Tölle A, Kristiansen I, Lein M, Johannsen M, Dietel M, Jung K, Kristiansen G. ADAM9 is highly expressed in renal cell cancer and is associated with tumour progression. BMC Cancer 2008; 8:179. [PMID: 18582378 PMCID: PMC2442841 DOI: 10.1186/1471-2407-8-179] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Accepted: 06/26/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A Disintegrin And Metalloprotease (ADAM) 9 has been implicated in tumour progression of various solid tumours, however, little is known about its role in renal cell carcinoma. We evaluated the expression of ADAM9 on protein and transcript level in a clinico-pathologically characterized renal cell cancer cohort. METHODS 108 renal cancer cases were immunostained for ADAM9 on a tissue-micro-array. For 30 additional cases, ADAM9 mRNA of microdissected tumour and normal tissue was analyzed via quantitative RT-PCR. SPSS 14.0 was used to apply crosstables (Fisher's exact test and chi2-test), correlations and univariate as well as multivariate survival analyses. RESULTS ADAM9 was significantly up-regulated in renal cancer in comparison to the adjacent normal tissue on mRNA level. On protein level, ADAM9 was significantly associated with higher tumour grade, positive nodal status and distant metastasis. Furthermore, ADAM9 protein expression was significantly associated with shortened patient survival in the univariate analysis. CONCLUSION ADAM9 is strongly expressed in a large proportion of renal cell cancers, concordant with findings in other tumour entities. Additionally, ADAM9 expression is significantly associated with markers of unfavourable prognosis. Whether the demonstrated prognostic value of ADAM9 is independent from other tumour parameters will have to be verified in larger study cohorts.
Collapse
Affiliation(s)
- Florian R Fritzsche
- Institute of Surgical Pathology, Universitätsmedizin Berlin, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
NQO1 expression in pancreatic cancer and its potential use as a biomarker. Appl Immunohistochem Mol Morphol 2008; 16:24-31. [PMID: 18091324 DOI: 10.1097/pai.0b013e31802e91d0] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is rarely curable due to regional/metastatic spread at diagnosis. Identification of molecular markers may enhance diagnosis and early detection of PDA. The 2-electron reductase, NAD(P)H:quinone oxidoreductase (NQO1) has been found to be overexpressed in many solid tumors including PDA, and may be a useful clinically relevant diagnostic marker of malignancy. For this study, we used 37 surgical resection cases: 24 PDAs and 13 benign pancreatic tissue specimens. An additional 16 specimens from pancreatic endoscopic ultrasound-guided fine needle aspiration (EUS-FNA) were included as a pilot series. NQO1 was detected by avidin-biotin based immunohistochemical and immunocytochemical methods. Both staining intensity and proportion of NQO1 positive tumor cells were scored. Moderate to strong (2 to 3+) staining for NQO1 was detected in 22/24 (92%) surgically resected PDAs, 9/9 (100%) EUS-FNAs with malignant diagnoses, one cytologically atypical but not diagnostic for malignancy EUS-FNA, and 1/6 (17%) EUS-FNAs initially diagnosed as negative for malignancy. Subsequent histologic assessment confirmed malignancy in all 9 cytologically positive EUS-FNAs and in the atypical case. The NQO1 positive case initially diagnosed as negative for malignancy showed no evidence of carcinoma on subsequent tissue biopsy. NQO1 staining was also observed in some benign ducts/cells; however, correlation of NQO1 expression with cellular morphology assessment minimizes the risk of false positive diagnosis. NQO1 is consistently overexpressed in PDA. Although NQO1 is observed in some benign tissue components, this marker may be a clinically useful diagnostic adjunct for detection of PDA, independent of tumor grade/stage.
Collapse
|
42
|
Karanjawala ZE, Illei PB, Ashfaq R, Infante JR, Murphy K, Pandey A, Schulick R, Winter J, Sharma R, Maitra A, Goggins M, Hruban RH. New markers of pancreatic cancer identified through differential gene expression analyses: claudin 18 and annexin A8. Am J Surg Pathol 2008; 32:188-96. [PMID: 18223320 PMCID: PMC2678811 DOI: 10.1097/pas.0b013e31815701f3] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND New markers to distinguish benign reactive glands from infiltrating ductal adenocarcinoma of the pancreas are needed. DESIGN The gene expression patterns of 24 surgically resected primary infiltrating ductal adenocarcinomas of the pancreas were compared with 18 non-neoplastic samples using the Affymetrix U133 Plus 2.0 Arrays and the Gene Logic GeneExpress Software System. Gene fragments from 4 genes (annexin A8, claudin 18, CXCL5, and S100 A2) were selected from the fragments found to be highly expressed in infiltrating adenocarcinomas when compared with normal tissues. The protein expression of these genes was examined using immunohistochemical labeling of tissue microarrays. RESULTS Claudin 18 labeled infiltrating carcinomas in a membranous pattern. When compared with normal and reactive ducts, claudin 18 was overexpressed, at least focally, in 159 of 166 evaluable carcinomas (96%). Strong and diffuse claudin 18 overexpression was most often seen in well-differentiated carcinomas (P=0.02). Claudin 18 was overexpressed in 51 of 52 cases (98%) of pancreatic intraepithelial neoplasia. Annexin A8 was at least focally overexpressed in 149 of 154 evaluable infiltrating carcinomas (97%). S100 A2 was at least focally overexpressed in 118 of 154 evaluable infiltrating carcinomas (77%). Non-neoplastic glands also frequently expressed S100 A2 diminishing its potential diagnostic utility. Immunolabeling with antibodies directed against CXCL5 did not reveal any significant differences in protein expression between infiltrating adenocarcinomas and normal pancreatic ducts. CONCLUSIONS Claudin 18 and annexin A8 are frequently highly overexpressed in infiltrating ductal adenocarcinomas when compared with normal reactive ducts, suggesting a role for these molecules in pancreatic ductal adenocarcinomas. Furthermore, these may serve as diagnostic markers, as screening tests and as therapeutic targets.
Collapse
MESH Headings
- Aged
- Annexins/genetics
- Annexins/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/mortality
- Carcinoma, Pancreatic Ductal/pathology
- Claudins
- Gene Expression
- Gene Expression Regulation, Neoplastic
- Humans
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Middle Aged
- Pancreas/metabolism
- Pancreas/pathology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/mortality
- Pancreatic Neoplasms/pathology
- Pancreatitis, Chronic/metabolism
- Pancreatitis, Chronic/pathology
- RNA, Messenger/metabolism
- RNA, Neoplasm/analysis
- Survival Rate
Collapse
Affiliation(s)
- Zarir E. Karanjawala
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Peter B. Illei
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Raheela Ashfaq
- Department of Pathology, The University of Texas Southwestern, Dallas, TX
| | - Jeffrey R. Infante
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
- The Sol Goldman Pancreatic Cancer Research Center, Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Kathleen Murphy
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Akhilesh Pandey
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
- The Sol Goldman Pancreatic Cancer Research Center, Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD
- The Sol Goldman Pancreatic Cancer Research Center, Institute for Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Richard Schulick
- The Sol Goldman Pancreatic Cancer Research Center, Department of Surgery, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Jordan Winter
- The Sol Goldman Pancreatic Cancer Research Center, Department of Surgery, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Rajni Sharma
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Anirban Maitra
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
- The Sol Goldman Pancreatic Cancer Research Center, Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD
- The Sol Goldman Pancreatic Cancer Research Center, Institute for Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Michael Goggins
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
- The Sol Goldman Pancreatic Cancer Research Center, Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Ralph H. Hruban
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
- The Sol Goldman Pancreatic Cancer Research Center, Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD
| |
Collapse
|
43
|
Dahl E, Kristiansen G, Gottlob K, Klaman I, Ebner E, Hinzmann B, Hermann K, Pilarsky C, Dürst M, Klinkhammer-Schalke M, Blaszyk H, Knuechel R, Hartmann A, Rosenthal A, Wild PJ. Molecular profiling of laser-microdissected matched tumor and normal breast tissue identifies karyopherin alpha2 as a potential novel prognostic marker in breast cancer. Clin Cancer Res 2007; 12:3950-60. [PMID: 16818692 DOI: 10.1158/1078-0432.ccr-05-2090] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The aim of the present study was to identify human genes that might prove useful in the diagnosis and therapy of primary breast cancer. EXPERIMENTAL DESIGN Twenty-four matched pairs of invasive ductal breast cancer and corresponding benign breast tissue were investigated by a combination of laser microdissection and gene expression profiling. Differential expression of candidate genes was validated by dot blot analysis of cDNA in 50 pairs of matching benign and malignant breast tissue. Cellular expression of candidate genes was further validated by RNA in situ hybridization, quantitative reverse transcription-PCR, and immunohistochemistry using tissue microarray analysis of 272 nonselected breast cancers. Multivariate analysis of factors on overall survival and recurrence-free survival was done. RESULTS Fifty-four genes were found to be up-regulated and 78 genes were found to be down-regulated. Dot blot analysis reduced the number of up-regulated genes to 15 candidate genes that showed at least a 2-fold overexpression in >15 of 50 (30%) tumor/normal pairs. We selected phosphatidic acid phosphatase type 2 domain containing 1A (PPAPDC1A) and karyopherin alpha2 (KPNA2) for further validation. PPAPDC1A and KPNA2 RNA was up-regulated (fold change >2) in 84% and 32% of analyzed tumor/normal pairs, respectively. Nuclear protein expression of KPNA2 was significantly associated with shorter overall survival and recurrence-free survival. Testing various multivariate Cox regression models, KPNA2 expression remained a highly significant, independent and adverse risk factor for overall survival. CONCLUSIONS Gene expression profiling of laser-microdissected breast cancer tissue revealed novel genes that may represent potential molecular targets for breast cancer therapy and prediction of outcome.
Collapse
Affiliation(s)
- Edgar Dahl
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Cordes N, Frick S, Brunner TB, Pilarsky C, Grützmann R, Sipos B, Klöppel G, McKenna WG, Bernhard EJ. Human pancreatic tumor cells are sensitized to ionizing radiation by knockdown of caveolin-1. Oncogene 2007; 26:6851-62. [PMID: 17471232 DOI: 10.1038/sj.onc.1210498] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2007] [Revised: 03/28/2007] [Accepted: 03/29/2007] [Indexed: 01/05/2023]
Abstract
Caveolin-1 (Cav-1) is an integral transmembrane protein and a critical component in interactions of integrin receptors with cytoskeleton-associated and signaling molecules. Since integrin-mediated cell adhesion generates signals conferring radiation resistance, we examined the effects of small interfering RNA-mediated knockdown of Cav-1 alone or in combination with beta1-integrin or focal adhesion kinase (FAK) on radiation survival and proliferation of pancreatic carcinoma cell lines. Irradiation induced Cav-1 expression in PATU8902, MiaPaCa2 and Panc1 cell lines. The cell lines showed significant radiosensitization after knockdown of Cav-1, beta1-integrin or FAK and cholesterol depletion by beta-cyclodextrin relative to nonspecific controls. Under knockdown conditions, proliferation of non-irradiated and irradiated cells was significantly attenuated relative to controls. These findings correlated with changes in expression or phosphorylation of Akt, glycogen synthase kinase 3beta, Paxillin, Src, c-Jun N-terminal kinase and mitogen-activated protein kinase. Analysis of DNA microarray data revealed a Cav-1 overexpression in a subset of pancreatic ductal adenocarcinoma samples. The data presented show, for the first time, that disruption of interactions of Cav-1 with beta1-integrin or FAK affects radiation survival and proliferation of pancreatic carcinoma cells and suggest that Cav-1 is critical to these processes. These results indicate that strategies targeting Cav-1 may be useful as an approach to improve conventional therapies, including radiotherapy, for pancreatic cancer.
Collapse
Affiliation(s)
- N Cordes
- OncoRay - Center for Radiation Research in Oncology, Medical Faculty Carl Gustav Carus, Dresden, University of Technology, Dresden, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Dawelbait G, Winter C, Zhang Y, Pilarsky C, Grützmann R, Heinrich JC, Schroeder M. Structural templates predict novel protein interactions and targets from pancreas tumour gene expression data. ACTA ACUST UNITED AC 2007; 23:i115-24. [PMID: 17646287 DOI: 10.1093/bioinformatics/btm188] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
MOTIVATION Pancreatic ductal adenocarcinoma (PDAC) eludes early detection and is characterized by its aggressiveness and resistance to current therapies. A number of gene expression screens have been carried out to identify genes differentially expressed in cancerous tissue. To identify molecular markers and suitable targets, these genes have been mapped to protein interactions to gain an understanding at systems level. RESULTS Here, we take such a network-centric approach to pancreas cancer by re-constructing networks from known interactions and by predicting novel protein interactions from structural templates. The pathways we find to be largely affected are signal transduction, actin cytoskeleton regulation, cell growth and cell communication. Our analysis indicates that the alteration of the calcium pathway plays an important role in pancreas-specific tumorigenesis. Furthermore, our structural prediction method identifies 40 novel interactions including the tissue factor pathway inhibitor 2 (TFPI2) interacting with the transmembrane protease serine 4 (TMPRSS4). Since TMPRSS4 is involved in metastasis formation, we hypothesize that the upregulation of TMPRSS4 and the downregulation of its predicted inhibitor TFPI2 plays an important role in this process. Moreover, we examine the potential role of BVDU (RP101) as an inhibitor of TMPRSS4. BDVU is known to support apoptosis and prevent the acquisition of chemoresistance. Our results suggest that BVDU might bind to the active site of TMPRSS4, thus reducing its assistance in metastasis. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Gihan Dawelbait
- Bioinformatics Group, Biotechnological Centre, TU Dresden, Dresden, Germany
| | | | | | | | | | | | | |
Collapse
|
46
|
Dhar G, Mehta S, Banerjee S, Gardner A, McCarty BM, Mathur SC, Campbell DR, Kambhampati S, Banerjee SK. Loss of WISP-2/CCN5 signaling in human pancreatic cancer: A potential mechanism for epithelial-mesenchymal-transition. Cancer Lett 2007; 254:63-70. [PMID: 17383817 DOI: 10.1016/j.canlet.2007.02.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2007] [Revised: 02/09/2007] [Accepted: 02/13/2007] [Indexed: 12/30/2022]
Abstract
The objective of this study was to explore the pathophysiological relevance of WISP-2/CCN5 in progression of human pancreatic adenocarcinoma (PAC). We found WISP-2/CCN5 mRNA and protein expression was faint and sporadic in PAC and detected in only 8.7-20% of the samples with varying grades as compared to adjacent normal and chronic pancreatitis samples where expression was very high in the ducts and acini. Colocalization studies in tissue-microarray slides revealed WISP-2/CCN5 mRNA loss was associated with p53 overexpression in PAC. Like tissue samples, p53 mutant-PAC cell lines show loss of WISP-2/CCN5. Moreover, functional analysis studies demonstrate exposure of pancreatic cancer cells to WISP-2/CCN5 recombinant protein enhances mesenchymal-epithelial-transition (MET). Collectively, we suggest WISP-2/CCN5 silencing may be a critical event during differentiation and progression of PAC and mutant p53 is possibly an important player in pursuing this episode.
Collapse
Affiliation(s)
- Gopal Dhar
- Cancer Research Unit, VA Medical Center, Kansas City, MO 64128, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Loos M, Bergmann F, Bauer A, Hoheisel JD, Esposito I, Kleeff J, Schirmacher P, Büchler MW, Klöppel G, Friess H. Solid type clear cell carcinoma of the pancreas: differential diagnosis of an unusual case and review of the literature. Virchows Arch 2007; 450:719-26. [PMID: 17453235 DOI: 10.1007/s00428-007-0416-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Revised: 03/29/2007] [Accepted: 04/02/2007] [Indexed: 10/23/2022]
Abstract
Pancreatic neoplasms have been reliably classified on the basis of their histopathology and immunophenotype. In this study, we report on a pancreatic tumor whose phenotype and genotype could not be assigned to any known tumor entity. The tumor was observed in the pancreatic head of a 54-year-old woman. It was found to be a solid infiltrating carcinoma with abundant clear cells. Apart from cytokeratin, the tumor cells expressed vimentin, S100, and MUC-1. DNA microarray analysis revealed a transcription profile clearly differing from that of normal pancreatic tissue and pancreatic ductal adenocarcinoma. Despite metastatic behavior, the tumor displayed a more favorable course than conventional pancreatic ductal adenocarcinoma. We suggest that this tumor be called solid type clear cell carcinoma of the pancreas.
Collapse
Affiliation(s)
- Martin Loos
- Department of General Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Jesnowski R, Zubakov D, Faissner R, Ringel J, Hoheisel JD, Lösel R, Schnölzer M, Löhr M. Genes and proteins differentially expressed during in vitro malignant transformation of bovine pancreatic duct cells. Neoplasia 2007; 9:136-46. [PMID: 17356710 PMCID: PMC1819583 DOI: 10.1593/neo.06754] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2006] [Revised: 01/15/2007] [Accepted: 01/16/2007] [Indexed: 02/06/2023] Open
Abstract
Pancreatic carcinoma has an extremely bad prognosis due to lack of early diagnostic markers and lack of effective therapeutic strategies. Recently, we have established an in vitro model recapitulating the first steps in the carcinogenesis of the pancreas. SV40 large T antigen-immortalized bovine pancreatic duct cells formed intrapancreatic adenocarcinoma tumors on k-ras(mut) transfection after orthotopic injection in the nude mouse pancreas. Here we identified genes and proteins differentially expressed in the course of malignant transformation using reciprocal suppression subtractive hybridization and 2D gel electrophoresis and mass spectrometry, respectively. We identified 34 differentially expressed genes, expressed sequence tags, and 15 unique proteins. Differential expression was verified for some of the genes or proteins in samples from pancreatic carcinoma. Among these genes and proteins, the majority had already been described either to be influenced by a mutated ras or to be differentially expressed in pancreatic adenocarcinoma, thus proving the feasibility of our model. Other genes and proteins (e.g., BBC1, GLTSCR2, and rhoGDIalpha), up to now, have not been implicated in pancreatic tumor development. Thus, we were able to establish an in vitro model of pancreatic carcinogenesis, which enabled us to identify genes and proteins differentially expressed during the early steps of malignant transformation.
Collapse
MESH Headings
- Animals
- Antigens, Polyomavirus Transforming/physiology
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Cattle
- Cell Line, Transformed/metabolism
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Viral/genetics
- Chronic Disease
- Disease Progression
- Electrophoresis, Gel, Two-Dimensional
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Genes, ras
- Humans
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/blood
- Neoplasm Proteins/genetics
- Oligonucleotide Array Sequence Analysis
- Pancreatic Ducts/cytology
- Pancreatic Ducts/metabolism
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Pancreatitis/genetics
- Pancreatitis/metabolism
- Polymerase Chain Reaction
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
- Subtraction Technique
Collapse
Affiliation(s)
- R Jesnowski
- Clinical Cooperation Unit Molecular Gastroenterology (E180), German Cancer Research Center Heidelberg and Department of Medicine II, Medical Faculty of Mannheim, University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Löhr JM. Pancreatic cancer — Outlook: gene therapy. THE CHINESE-GERMAN JOURNAL OF CLINICAL ONCOLOGY 2007; 6:181-186. [DOI: 10.1007/s10330-007-0049-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
|
50
|
Kanno A, Satoh K, Kimura K, Hirota M, Umino J, Masamune A, Satoh A, Asakura T, Egawa S, Sunamura M, Endoh M, Shimosegawa T. The expression of MUC4 and MUC5AC is related to the biologic malignancy of intraductal papillary mucinous neoplasms of the pancreas. Pancreas 2006; 33:391-6. [PMID: 17079945 DOI: 10.1097/01.mpa.0000236742.92606.c1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Intraductal papillary mucinous neoplasms (IPMNs) of the pancreas show heterogeneous proliferations with latent malignancy. Mucins (MUC) are high-molecular-weight glycoproteins, with an aberrant expression profile in various malignancies. Recently, MUC4 and MUC5AC expressions have been demonstrated to correlate with the unfavorable and the favorable prognosis of pancreatic duct cell carcinoma, respectively. However, little is known about these mucin expressions in IPMNs. METHODS To clarify the role of MUC4 and MUC5AC expressions in IPMNs, the expression profiles of MUC4 and MUC5AC were investigated in 50 lesions from 17 specimens with 16 IPMNs by immunohistochemistry, using each of their specific antibodies. RESULTS The expression of MUC4 was found in the lesions ranging from adenoma to cancer lesions of IPMNs, whereas it was undetectable in normal and hyperplastic lesions. Frequent expression of MUC4 is found in the higher grade of IPMNs (borderline and cancer lesions; 16/18 lesions, 94%). The differences were independently significant (P < 0.001) when the cutoff point was set between adenoma and borderline IPMNs. Similarly, frequent expression of MUC5AC was detected in the lesions from adenoma to cancer of IPMNs (32/34, 94%), whereas no intense expression was detected in normal or hyperplastic lesions. The significant difference was found when the cutoff point was set between hyperplasia and adenoma of IPMNs (P < 0.001). CONCLUSIONS These results indicated that the expressions of MUC4 and MUC5AC are potential markers to distinguish adenoma or above malignant lesions of IPMNs from lesser malignant ones, respectively.
Collapse
Affiliation(s)
- Atsushi Kanno
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|