1
|
Wedig J, Jasani S, Mukherjee D, Lathrop H, Matreja P, Pfau T, D'Alesio L, Guenther A, Fenn L, Kaiser M, Torok MA, McGue J, Sizemore GM, Noonan AM, Dillhoff ME, Blaser BW, Frankel TL, Culp S, Hart PA, Cruz-Monserrate Z, Mace TA. CD200 is overexpressed in the pancreatic tumor microenvironment and predictive of overall survival. Cancer Immunol Immunother 2024; 73:96. [PMID: 38619621 PMCID: PMC11018596 DOI: 10.1007/s00262-024-03678-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 03/15/2024] [Indexed: 04/16/2024]
Abstract
Pancreatic cancer is an aggressive disease with a 5 year survival rate of 13%. This poor survival is attributed, in part, to limited and ineffective treatments for patients with metastatic disease, highlighting a need to identify molecular drivers of pancreatic cancer to target for more effective treatment. CD200 is a glycoprotein that interacts with the receptor CD200R and elicits an immunosuppressive response. Overexpression of CD200 has been associated with differential outcomes, depending on the tumor type. In the context of pancreatic cancer, we have previously reported that CD200 is expressed in the pancreatic tumor microenvironment (TME), and that targeting CD200 in murine tumor models reduces tumor burden. We hypothesized that CD200 is overexpressed on tumor and stromal populations in the pancreatic TME and that circulating levels of soluble CD200 (sCD200) have prognostic value for overall survival. We discovered that CD200 was overexpressed on immune, stromal, and tumor populations in the pancreatic TME. Particularly, single-cell RNA-sequencing indicated that CD200 was upregulated on inflammatory cancer-associated fibroblasts. Cytometry by time of flight analysis of PBMCs indicated that CD200 was overexpressed on innate immune populations, including monocytes, dendritic cells, and monocytic myeloid-derived suppressor cells. High sCD200 levels in plasma correlated with significantly worse overall and progression-free survival. Additionally, sCD200 correlated with the ratio of circulating matrix metalloproteinase (MMP) 3: tissue inhibitor of metalloproteinase (TIMP) 3 and MMP11/TIMP3. This study highlights the importance of CD200 expression in pancreatic cancer and provides the rationale for designing novel therapeutic strategies that target this protein.
Collapse
Affiliation(s)
- Jessica Wedig
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Molecular, Cellular and Developmental Biology Program, The Ohio State University, Columbus, USA
| | - Shrina Jasani
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Debasmita Mukherjee
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Molecular, Cellular and Developmental Biology Program, The Ohio State University, Columbus, USA
| | - Hannah Lathrop
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Priya Matreja
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Timothy Pfau
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Liliana D'Alesio
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Abigail Guenther
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Lexie Fenn
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Morgan Kaiser
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Molly A Torok
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Jake McGue
- Department of Surgical Oncology, University of Michigan, Ann Arbor, USA
| | - Gina M Sizemore
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Department of Radiation Oncology, The Ohio State University, Columbus, USA
| | - Anne M Noonan
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Wexner Medical Center, Columbus, USA
| | - Mary E Dillhoff
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Department of Internal Medicine, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, USA
| | - Bradley W Blaser
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Department of Internal Medicine, Division of Hematology, The Ohio State University Wexner Medical Center, Columbus, USA
| | - Timothy L Frankel
- Department of Surgical Oncology, University of Michigan, Ann Arbor, USA
| | - Stacey Culp
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Department of Biomedical Informatics, The Ohio State University, Columbus, USA
| | - Phil A Hart
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, 420 W. 12th Ave., Columbus, OH, 43210, USA
| | - Zobeida Cruz-Monserrate
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, 420 W. 12th Ave., Columbus, OH, 43210, USA
| | - Thomas A Mace
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA.
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, 420 W. 12th Ave., Columbus, OH, 43210, USA.
| |
Collapse
|
2
|
Roy A, Chauhan S, Bhattacharya S, Jakhmola V, Tyagi K, Sachdeva A, Wasai A, Mandal S. Runt-related transcription factors in human carcinogenesis: a friend or foe? J Cancer Res Clin Oncol 2023; 149:9409-9423. [PMID: 37081242 DOI: 10.1007/s00432-023-04769-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 04/08/2023] [Indexed: 04/22/2023]
Abstract
PURPOSE Cancer is one of the deadliest pathologies with more than 19 million new cases and 10 million cancer-related deaths across the globe. Despite development of advanced therapeutic interventions, cancer remains as a fatal pathology due to lack of early prognostic biomarkers, therapy resistance and requires identification of novel drug targets. METHODS Runt-related transcription factors (Runx) family controls several cellular and physiological functions including osteogenesis. Recent literatures from PubMed was mined and the review was written in comprehensive manner RESULTS: Recent literature suggests that aberrant expression of Runx contributes to tumorigenesis of many organs. Conversely, cell- and tissue-specific tumor suppressor roles of Runx are also reported. In this review, we have provided the structural/functional properties of Runx isoforms and its regulation in context of human cancer. Moreover, in an urgent need to discover novel therapeutic interventions against cancer, we comprehensively discussed the reported oncogenic and tumor suppressive roles of Runx isoforms in several tumor types and discussed the discrepancies that may have risen on Runx as a driver of malignant transformation. CONCLUSION Runx may be a novel therapeutic target against a battery of deadly human cancers.
Collapse
Affiliation(s)
- Adhiraj Roy
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India.
| | - Shivi Chauhan
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India
| | - Sujata Bhattacharya
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India
| | - Vibhuti Jakhmola
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India
| | - Komal Tyagi
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India
| | - Abha Sachdeva
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India
| | - Abdul Wasai
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India
| | - Supratim Mandal
- Department of Microbiology, University of Kalyani, Kalyani, Nadia, West Bengal, 741235, India
| |
Collapse
|
3
|
Pen SL, Shan YS, Hsiao CF, Liu TW, Chen JS, Ho CL, Chou WC, Hsieh RK, Chen LT, Ch'ang HJ. High expression of krüppel-like factor 10 or Smad4 predicts clinical benefit of adjuvant chemoradiotherapy in curatively resected pancreatic adenocarcinoma: From a randomized phase III trial. Radiother Oncol 2021; 158:146-154. [PMID: 33667587 DOI: 10.1016/j.radonc.2021.02.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 02/09/2021] [Accepted: 02/24/2021] [Indexed: 12/30/2022]
Abstract
PURPOSE Our previous studies have demonstrated that Krüppel-like factor 10 (Klf10) modulated tumor radiation resistance and helps to predict clinical outcomes of pancreatic adenocarcinoma (PDAC). This study aimed to evaluate whether the expression levels of Klf10, Smad4 and Runx3 can help predict the benefits of adjuvant chemoradiotherapy (CRT) in resected PDAC. METHODS AND MATERIALS Tissue specimens were collected from 111 patients with curatively resected PDAC who were enrolled into a randomized trial comparing adjuvant gemcitabine with or without CRT. Immunohistochemical expression of biomarkers was quantified by pathologists blinded to patient outcomes through a grading system based on the extent and intensity of staining. The predictive value of biomarkers was analyzed using SAS statistical software. RESULTS In total, 56 and 55 patients received adjuvant gemcitabine alone and additional CRT, respectively. The expression levels of Klf10, Smad4 and postoperative CA19-9 were significantly correlated with overall survival (OS) (p = 0.013, 0.045, and 0.047, respectively). Multivariable analysis showed that the expression level of postoperative serum CA19-9 and tumor tissue Klf10 expression level were significant predictors for OS (p = 0.038, and 0.028, respectively). Patients with high Klf10 or Smad4 (n = 55), had a significantly better local recurrence-free survival (∞ vs 19.8 months; p = 0.026) and a longer OS (33.0 vs 23.0 months; p = 0.12) if they received additional adjuvant CRT than gemcitabine only. The results were similar after adjusted by postoperative level of CA19-9. CONCLUSION Patients with curatively resected PDAC and a high expression of either Klf10 or Smad4 have high chances of benefiting from adjuvant CRT. Combining Klf10 and Smad4 to predict the benefits of adjuvant CRT in resected PDAC deserves further validation.
Collapse
Affiliation(s)
- Shu-Ling Pen
- Department of Pathology, National Cheng Kung University, Tainan, Taiwan
| | - Yan-Shen Shan
- Department of Surgery, National Cheng Kung University, Tainan, Taiwan
| | - Chin-Fu Hsiao
- Institute of Public Health Sciences, National Health Research Institutes, Miaoli, Taiwan.
| | - Tsang-Wu Liu
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Jen-Shi Chen
- Department of Hematology-Oncology, Linkou Chang-Gung Memorial Hospital, Chang-Gung University, Tao-Yuan, Taiwan
| | - Ching-Liang Ho
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wen-Chi Chou
- Department of Hematology-Oncology, Linkou Chang-Gung Memorial Hospital, Chang-Gung University, Tao-Yuan, Taiwan
| | - Ruey-Kuen Hsieh
- Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan; Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Taiwan
| | - Hui-Ju Ch'ang
- Department of Radiation Oncology, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan; Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, and Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
4
|
Rossi E, Bagalà C, Inzani F, Leoncini E, Brunelli C, Lanza P, Basso M, Mattiucci GC, Cassano A, Rindi G, Barone C, Schinzari G. RUNX3 as a Potential Predictor of Metastasis in Human Pancreatic Cancer. ACTA ACUST UNITED AC 2018; 31:833-840. [PMID: 28882948 DOI: 10.21873/invivo.11136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 07/19/2017] [Accepted: 07/24/2017] [Indexed: 01/13/2023]
Abstract
BACKGROUND/AIM In genetically engineered murine models of pancreatic ductal adenocarcinomas (PDAC), high levels of Runx3 increase the metastatic potential of cancer cells. In this study we evaluated the role of Runx3 in human pancreatic cancer. MATERIALS AND METHODS Runx3 was retrospectively assessed by immunohistochemistry in seventy-eight tumor samples of patients who underwent surgical resection for PDCA and were followed at least for 24 months. RESULTS Thirty-two cases resulted completely negative for Runx3; forty-six showed highly variable expression. We established an optimal cut-off value of Runx3 in predicting distant metastasis equal to 0.04. The odds ratio (ORs) for development of distant metastases at multivariate analysis for patients having Runx3 ≥0.04 was 4.26 (p=0.043) and 4.68 (p=0.032) after adjusting for residual tumor and treatment, respectively; OR for development of metastases in multiple sites was 4.28 (p=0.025) for Runx3 ≥0.04. CONCLUSION Our results support the ability of Runx3 to contribute to the dissemination of human PDAC thus confirming the observations from murine models.
Collapse
Affiliation(s)
- Ernesto Rossi
- Department of Medical Oncology, Fondazione Policlinico "A.Gemelli", Largo A. Gemelli, Rome, Italy
| | - Cinzia Bagalà
- Department of Medical Oncology, Fondazione Policlinico "A.Gemelli", Largo A. Gemelli, Rome, Italy
| | - Frediano Inzani
- Institute of Anatomic Pathology, Fondazione Policlinico "A.Gemelli", Largo A. Gemelli, Rome, Italy
| | - Emanuele Leoncini
- Section of Hygiene, Institute of Public Health, Fondazione Policlinico "A.Gemelli", Largo A. Gemelli, Rome, Italy
| | - Chiara Brunelli
- Institute of Anatomic Pathology, Fondazione Policlinico "A.Gemelli", Largo A. Gemelli, Rome, Italy
| | - Paola Lanza
- Institute of Anatomic Pathology, Fondazione Policlinico "A.Gemelli", Largo A. Gemelli, Rome, Italy
| | - Michele Basso
- Department of Medical Oncology, Fondazione Policlinico "A.Gemelli", Largo A. Gemelli, Rome, Italy
| | - Gian Carlo Mattiucci
- Department of Radiation Oncology, Fondazione Policlinico "A.Gemelli", Largo A. Gemelli, Rome, Italy
| | - Alessandra Cassano
- Department of Medical Oncology, Fondazione Policlinico "A.Gemelli", Largo A. Gemelli, Rome, Italy
| | - Guido Rindi
- Institute of Anatomic Pathology, Fondazione Policlinico "A.Gemelli", Largo A. Gemelli, Rome, Italy
| | - Carlo Barone
- Department of Medical Oncology, Fondazione Policlinico "A.Gemelli", Largo A. Gemelli, Rome, Italy
| | - Giovanni Schinzari
- Department of Medical Oncology, Fondazione Policlinico "A.Gemelli", Largo A. Gemelli, Rome, Italy
| |
Collapse
|
5
|
Lee Y, Lee H, Park H, Kim JW, Hwang JH, Kim J, Yoon YS, Han HS, Kim H. Combination immunohistochemistry for SMAD4 and Runt-related transcription factor 3 may identify a favorable prognostic subgroup of pancreatic ductal adenocarcinomas. Oncotarget 2017; 8:76699-76711. [PMID: 29100342 PMCID: PMC5652736 DOI: 10.18632/oncotarget.20815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/23/2017] [Indexed: 12/20/2022] Open
Abstract
Purposes SMAD4/DPC4 mutations have been associated with aggressive behavior in pancreatic ductal adenocarcinomas (PDAC), and it has recently been suggested that RUNX3 expression combined with SMAD4 status may predict the metastatic potential of PDACs. We evaluated the prognostic utility of SMAD4/RUNX3 status in human PDACs by immunohistochemistry. Materials and Methods Immunohistochemical stains were performed for SMAD4 and RUNX3 on 210 surgically resected PDACs, and the results were correlated with the clinicopathological features. Results Loss of SMAD4 expression was associated with poor overall survival (OS) (p = 0.015) and progression-free survival (PFS) (p = 0.044). Nuclear RUNX3 expression was associated with decreased OS (p = 0.010) and PFS (p = 0.009), and more frequent in poorly differentiated PDACs (p = 0.037). On combining RUNX3/SMAD4 status, RUNX3-/SMAD4+ PDACs demonstrated longer OS (p = 0.008, median time; RUNX3-/SMAD4+ 34 months, others 17 months) and PFS (p = 0.009, median time; RUNX3-/SMAD4+ 29 months, others 8 months) compared to RUNX3+/SMAD4+ and SMAD4- groups; RUNX3-/SMAD4+ was a significant independent predictive factor for both OS [p = 0.025, HR 1.842 (95% CI 1.079-3.143)] and PFS [p = 0.020, HR 1.850 (95% CI 1.100-3.113)]. Conclusions SMAD4-positivity with RUNX3-negativity was a significant independent predictive factor for favorable OS and PFS in PDAC. This is the first and large clinicopathological study of RUNX3/SMAD4 expression status in human PDAC. Combination immunohistochemistry for SMAD4 and RUNX3 may help identify a favorable prognostic subgroup of PDAC.
Collapse
Affiliation(s)
- Yangkyu Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Hyejung Lee
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyunjin Park
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin-Won Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Jin-Hyeok Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Jaihwan Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Yoo-Seok Yoon
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Ho-Seong Han
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Haeryoung Kim
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
6
|
Yao K, Wang Q, Jia J, Zhao H. A competing endogenous RNA network identifies novel mRNA, miRNA and lncRNA markers for the prognosis of diabetic pancreatic cancer. Tumour Biol 2017. [PMID: 28639886 DOI: 10.1177/1010428317707882] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Pancreatic cancer (PaC) is highly associated with diabetes mellitus (DM). However, the mechanisms are insufficient. The study aimed to uncover the underlying regulatory mechanism on diabetic PaC and find novel biomarkers for the disease prognosis. Two RNA-sequencing (RNA-seq) datasets, GSE74629 and GSE15932, as well as relevant data in TCGA were utilized. After pretreatment, differentially expressed genes (DEGs) or miRNAs (DEMs) or lncRNAs (DELs) between diabetic PaC and non-diabetic PaC patients were identified, and further examined for their correlations with clinical information. Prognostic RNAs were selected using KM curve. Optimal gene set for classification of different samples were recognized by support vector machine. Protein-protein interaction (PPI) network was constructed for DEGs based on protein databases. Interactions among three kinds of RNAs were revealed in the 'lncRNA-miRNA-mRNA' competing endogenous RNA (ceRNA) network. A group of 32 feature genes were identified that could classify diabetic PaC from non-diabetic PaC, such as CCDC33, CTLA4 and MAP4K1. This classifier had a high accuracy on the prediction. Seven lncRNAs were tied up with prognosis of diabetic PaC, especially UCA1. In addition, crucial DEMs were selected, such as hsa-miR-214 (predicted targets: MAP4K1 and CCDC33) and hsa-miR-429 (predicted targets: CTLA4). Notably, interactions of 'HOTAIR-hsa-miR-214-CCDC33' and 'CECR7-hsa-miR-429-CTLA4' were highlighted in the ceRNA network. Several biomarkers were identified for diagnosis of diabetic PaC, such as HOTAIR, CECR7, UCA1, hsa-miR-214, hsa-miR-429, CCDC33 and CTLA4. 'HOTAIR-hsa-miR-214-CCDC33' and 'CECR7-hsa-miR-429-CTLA4' regulations might be two important mechanisms for the disease progression.
Collapse
Affiliation(s)
- Kanyu Yao
- 1 Department of Emergency Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, People's Republic of China
| | - Qi Wang
- 1 Department of Emergency Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, People's Republic of China
| | - Jianhua Jia
- 2 The 253th Hospital of P.L.A., Hohhot, People's Republic of China
| | - Haiping Zhao
- 3 Department of Hepatobiliary Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, People's Republic of China
| |
Collapse
|
7
|
Runx3 and Cell Fate Decisions in Pancreas Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 962:333-352. [PMID: 28299667 DOI: 10.1007/978-981-10-3233-2_21] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The RUNX family transcription factors are critical regulators of development and frequently dysregulated in cancer. RUNX3, the least well characterized of the three family members, has been variously described as a tumor promoter or suppressor, sometimes with conflicting results and opinions in the same cancer and likely reflecting a complex role in oncogenesis. We recently identified RUNX3 expression as a crucial determinant of the predilection for pancreatic ductal adenocarcinoma (PDA) cells to proliferate locally or promulgate throughout the body. High RUNX3 expression induces the production and secretion of soluble factors that support metastatic niche construction and stimulates PDA cells to migrate and invade, while simultaneously suppressing proliferation through increased expression of cell cycle regulators such as CDKN1A/p21 WAF1/CIP1 . RUNX3 expression and function are coordinated by numerous transcriptional and post-translational inputs, and interactions with diverse cofactors influence whether the resulting RUNX3 complexes enact tumor suppressive or tumor promoting programs. Understanding these exquisitely context-dependent tumor cell behaviors has the potential to inform clinical decision-making including the most appropriate timing and sequencing of local vs. systemic therapies.
Collapse
|
8
|
Abstract
In this chapter we summarize the pros and cons of the notion that Runx3 is a major tumor suppressor gene (TSG). Inactivation of TSGs in normal cells provides a viability/growth advantage that contributes cell-autonomously to cancer. More than a decade ago it was suggested that RUNX3 is involved in gastric cancer development, a postulate extended later to other epithelial cancers portraying RUNX3 as a major TSG. However, evidence that Runx3 is not expressed in normal gastric and other epithelia has challenged the RUNX3-TSG paradigm. In contrast, RUNX3 is overexpressed in a significant fraction of tumor cells in various human epithelial cancers and its overexpression in pancreatic cancer cells promotes their migration, anchorage-independent growth and metastatic potential. Moreover, recent high-throughput quantitative genome-wide studies on thousands of human samples of various tumors and new investigations of the role of Runx3 in mouse cancer models have unequivocally demonstrated that RUNX3 is not a bona fide cell-autonomous TSG. Importantly, accumulating data demonstrated that RUNX3 functions in control of immunity and inflammation, thereby indirectly influencing epithelial tumor development.
Collapse
|
9
|
Kurklu B, Whitehead RH, Ong EK, Minamoto T, Fox JG, Mann JR, Judd LM, Giraud AS, Menheniott TR. Lineage-specific RUNX3 hypomethylation marks the preneoplastic immune component of gastric cancer. Oncogene 2014; 34:2856-66. [PMID: 25088199 DOI: 10.1038/onc.2014.233] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 06/20/2014] [Indexed: 12/22/2022]
Abstract
Runt domain transcription factor 3 (RUNX3) is widely regarded as a tumour-suppressor gene inactivated by DNA hypermethylation of its canonical CpG (cytidine-phosphate-guanidine) island (CGI) promoter in gastric cancer (GC). Absence of RUNX3 expression from normal gastric epithelial cells (GECs), the progenitors to GC, coupled with frequent RUNX3 overexpression in GC progression, challenge this longstanding paradigm. However, epigenetic models to better describe RUNX3 deregulation in GC have not emerged. Here, we identify lineage-specific DNA methylation at an alternate, non-CGI promoter (P1) as a new mechanism of RUNX3 epigenetic control. In normal GECs, P1 was hypermethylated and repressed, whereas in immune lineages P1 was hypomethylated and widely expressed. In human GC development, we detected aberrant P1 hypomethylation signatures associated with the early inflammatory, preneoplastic and tumour stages. Aberrant P1 hypomethylation was fully recapitulated in mouse models of gastric inflammation and tumorigenesis. Cell sorting showed that P1 hypomethylation reflects altered cell-type composition of the gastric epithelium/tumour microenvironment caused by immune cell recruitment, not methylation loss. Finally, via long-term culture of gastric tumour epithelium, we revealed that de novo methylation of the RUNX3 canonical CGI promoter is a bystander effect of oncogenic immortalization and not likely causal in GC pathogenesis as previously argued. We propose a new model of RUNX3 epigenetic control in cancer, based on immune-specific, non-CGI promoter hypomethylation. This novel epigenetic signature may have utility in early detection of GC and possibly other epithelial cancers with premalignant immune involvement.
Collapse
Affiliation(s)
- B Kurklu
- 1] Infection and Immunity, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia [2] Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - R H Whitehead
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - E K Ong
- Infection and Immunity, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - T Minamoto
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - J G Fox
- Division of Comparative Medicine, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - J R Mann
- 1] Infection and Immunity, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia [2] Department of Zoology, University of Melbourne, Melbourne, Victoria, Australia
| | - L M Judd
- 1] Infection and Immunity, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia [2] Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - A S Giraud
- 1] Infection and Immunity, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia [2] Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - T R Menheniott
- 1] Infection and Immunity, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia [2] Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
10
|
Barnie PA, Zhang P, Lu P, Chen X, Su Z, Wang S, Xu H. CpG-oligodeoxynucleotides suppress the proliferation of A549 lung adenocarcinoma cells via toll-like receptor 9 signaling and upregulation of Runt-related transcription factor 3 expression. Biomed Rep 2014; 2:374-377. [PMID: 24748977 PMCID: PMC3990193 DOI: 10.3892/br.2014.257] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 03/07/2014] [Indexed: 12/25/2022] Open
Abstract
The aim of the present study was to investigate the effect of CpG-oligodeoxynucelotides (CpG-ODN) on the proliferation of the A549 human lung adenocarcinoma cell line and the expression of Runt-related transcription factor 3 (Runx3) and investigate the association between the toll-like receptor 9 (TLR9) signaling pathway and Runx3 expression during A549 cell proliferation. Different concentrations of CpG-ODN were used in this study to stimulate A549 cells and the expression of Runx3 at the mRNA or protein level was detected by reverse transcription-polymerase chain reaction or western blot analysis. Moreover, Runx3 siRNA was synthesized and transiently transfected into the A549 cells and the MTT assay was used to detect the effects of CpG-ODN on transfected cell growth. Our data demonstrated that CpG-ODN significantly inhibited the proliferation of A549 cells. The expression of Runx3 in the mRNA and protein level was increased in A549 cells stimulated by CpG-ODN. The CpG-ODN-stimulated cell proliferation was significantly inhibited in Runx3 siRNA-transfected A549 cells. In conclusion, CpG-ODN may bind to TLR9, inhibit the proliferation of A549 cells and upregulate the expression of Runx3.
Collapse
Affiliation(s)
- Prince Amoah Barnie
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Pan Zhang
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Ping Lu
- Division of Nephrology, Department of Internal Medicine, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Xiaobo Chen
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Zhaoliang Su
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Shengjun Wang
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Huaxi Xu
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
11
|
Avci CB, Dodurga Y, Susluer SY, Sıgva ZOD, Yucebas M, Caglar HO, Akalin T, Dalbasti T, Oktar N, Gunduz C. Promoter hypermethylation-mediated down-regulation of RUNX3 gene in human brain tumors. Ir J Med Sci 2013; 183:259-64. [PMID: 23934435 DOI: 10.1007/s11845-013-1001-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 08/02/2013] [Indexed: 01/12/2023]
Abstract
BACKGROUND The Runx family proteins, including RUNX3, are tissue-restricted transcription factors and play role in neuronal development and tumorigenesis. RUNX3 has an important role in glioblastoma (GBM) tumorigenesis because of its promoter hypermethylation. AIM We aimed to evaluate the methylation-mediated expression regulation of RUNX3 gene in brain tumors. PATIENTS AND METHODS Cases of meningiomas WHO grade III (3), anaplastic astrocytomas (3), diffuse astrocytoma (3), and GBM (12) were recruited into this study. Real-time quantitative PCR was performed for analyses of DNA promoter methylation and analyses of methylation-mediated expression status of RUNX3 gene was performed by real-time quantitative RT-PCR. RESULTS There was no significant difference between methylated and unmethylated quantitative ratio of RUNX3 gene promoter region and also no significant difference in relative ratio of RUNX3 gene expression in brain tumor groups. Methylated and unmethylated ratio in anaplastic astrocytoma, diffuse astrocytoma, GBM, meningioma (WHO grade III) and in all groups were; 1.44, 1.09, 1.51, 1.52 and 1.43, respectively. One allele was found methylated necessarily. No methylation was detected in one case of GBM group and one case of anaplastic astrocytoma group. There was no unmethylated promoter in one of the GBM cases. There were significant differences between relative ratio of RUNX3 gene expression and methylated/unmethylated ratio rates for all cases (p = 0.001) and GBM groups (p = 0.041). CONCLUSION This study overemphasized the RUNX3 gene importance in brain tumors, due to the existence of at least one methylated allele.
Collapse
Affiliation(s)
- C B Avci
- Department of Medical Biology, Ege University Medical Faculty, Bornova, 35100, Izmir, Turkey,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Horiguchi S, Shiraha H, Nagahara T, Kataoka J, Iwamuro M, Matsubara M, Nishina S, Kato H, Takaki A, Nouso K, Tanaka T, Ichimura K, Yagi T, Yamamoto K. Loss of runt-related transcription factor 3 induces gemcitabine resistance in pancreatic cancer. Mol Oncol 2013; 7:840-849. [PMID: 23664167 PMCID: PMC5528422 DOI: 10.1016/j.molonc.2013.04.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 03/27/2013] [Accepted: 04/15/2013] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND & AIM Runt-related transcription factor 3 (RUNX3) is a tumor suppressor gene that is expressed in gastric and other cancers including pancreatic cancer. However, the precise function of RUNX3 in pancreatic cancer has not been fully elucidated. In this study, we aimed to determine the effect of decreased RUNX3 expression in pancreatic cancer. METHODS This study included 36 patients with primary pancreatic cancer, who had undergone pancreaticoduodenectomy. All patients were treated with 1000 mg/m2 gemcitabine after the surgery. The pancreatic cancer cell lines PANC-1, MIAPaCa-2, BxPC-3, SUIT-2, and KLM-1 were used for immunoblotting analysis of RUNX3 and multidrug resistance protein (MRP) expressions. Ectopic RUNX3 expression was achieved by cDNA transfection of the cells, and small interfering RNA (siRNA) against RUNX3 was used to knock down endogenous RUNX3. Cell growth in the presence of gemcitabine was assessed using the MTT assay. RESULTS Patients with RUNX3-positive and RUNX3-negative pancreatic cancer had a median survival of 1006 and 643 days, respectively. Exogenous RUNX3 expression reduced the expression of MRP1, MRP2, and MRP5 in endogenous RUNX3-negative cells, whereas RUNX3 siRNA increased the expressions of these genes in endogenous RUNX3-positive cells. Exogenous RUNX3 expression decreased gemcitabine IC50 in RUNX3-negative cells. CONCLUSION Loss of RUNX3 expression contributes to gemcitabine resistance by inducing MRP expression, thereby resulting in poor patient survival.
Collapse
Affiliation(s)
- Shigeru Horiguchi
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Hidenori Shiraha
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Teruya Nagahara
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Jyunnro Kataoka
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Masaya Iwamuro
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Minoru Matsubara
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Shinichi Nishina
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Hironari Kato
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Akinobu Takaki
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Kazuhiro Nouso
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Takehiro Tanaka
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Koichi Ichimura
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Takahito Yagi
- Department of Gastroenterological Surgery, Transplant, and Surgical Oncology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Kazuhide Yamamoto
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| |
Collapse
|
13
|
Jin Z, Han YX, Han XR. Loss of RUNX3 expression may contribute to poor prognosis in patients with chondrosarcoma. J Mol Histol 2013; 44:645-52. [DOI: 10.1007/s10735-013-9511-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 05/06/2013] [Indexed: 12/20/2022]
|
14
|
Strong N, Millena AC, Walker L, Chaudhary J, Khan SA. Inhibitor of differentiation 1 (Id1) and Id3 proteins play different roles in TGFβ effects on cell proliferation and migration in prostate cancer cells. Prostate 2013; 73:624-33. [PMID: 23060149 PMCID: PMC4018743 DOI: 10.1002/pros.22603] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 09/17/2012] [Indexed: 12/30/2022]
Abstract
BACKGROUND In prostate cancer cells, transforming growth factor β (TGFβ) inhibits proliferation in earlier stages of the disease; however, the cancer cells become refractory to growth inhibitory effects in advanced stages where TGFβ promotes cancer progression and metastasis. Inhibitor of differentiation (Id) family of closely related proteins (Id1-Id4) are dominant negative regulators and basic helix loop helix (bHLH) transcription factors and in general promote proliferation, and inhibit differentiation. In the present study, we have investigated the role of Id1 and Id3 proteins in the growth inhibitory effects of TGFβ on prostate cancer cells. METHODS The effect of TGF β on proliferation and Id1 and Id3 expression were investigated in PZ-HPV7, DU145, and PC3 cells. Id1 silencing through siRNA was also used in DU145 and PC3 cells to examine its role in anti-proliferative and migratory effects of TGFβ. RESULTS TGFβ increased expression of Id1 and Id3 in all cell lines followed by a later down regulation of Id1 in PZ-HPV7 expression and DU145 cells but not in PC3 cells. Id3 expression remained elevated in all three cell lines. This loss of Id1 protein correlated with an increase of CDKNI p21. Id1 knockdown in both DU145 and PC3 cells resulted in decreased proliferation. However, while TGFβ caused a further decrease in proliferation of DU145, but had no further effects in PC3 cells. Knockdown of Id1 or Id3 inhibited TGFβ1induced migration in PC3 cells. CONCLUSIONS These findings suggest an essential role of Id1 and Id3 in TGFβ1 effects on proliferation and migration in prostate cancer cells.
Collapse
Affiliation(s)
| | | | | | | | - Shafiq A. Khan
- Correspondence to: Shafiq A. Khan, PhD, Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr, SW, Atlanta, GA 30314.
| |
Collapse
|
15
|
Lotem J, Levanon D, Negreanu V, Groner Y. The False Paradigm of RUNX3 Function as Tumor Suppressor in Gastric Cancer. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/jct.2013.41a003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
16
|
Ko HJ, Kim BY, Jung CH, Chun SW, Mok JO, Kim YJ, Park HK, Kim CH, Kim SJ, Byun DW, Suh KI, Yoo MH, Kang SG. DNA methylation of RUNX3 in papillary thyroid cancer. Korean J Intern Med 2012; 27:407-10. [PMID: 23269881 PMCID: PMC3529239 DOI: 10.3904/kjim.2012.27.4.407] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 11/03/2011] [Accepted: 11/21/2011] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND/AIMS The relationship between Runt-related transcription factor 3 (RUNX3) gene inactivation and various solid tumors has been reported; however, little information is available about RUNX3 in thyroid cancers. METHODS We evaluated the DNA methylation of RUNX3 in 13 papillary thyroid cancer tissues and four thyroid cancer cell lines. Additionally, using reverse transcriptase-polymerase chain reaction, we analyzed RUNX3 gene expression in several thyroid cancer cell lines after treating with the demethylating agent 5-aza-2'-deoxycytidine (DAC). RESULTS RUNX3 was hypermethylated in many thyroid cancer cell lines and in 10 of the 12 papillary thyroid cancer tissues. Treatment with DAC increased the expression of RUNX3 in some thyroid cancer cell lines. CONCLUSIONS We suggest that RUNX3 is associated with thyroid carcinogenesis, and RUNX3 methylation is a potentially useful diagnostic marker for papillary thyroid cancer.
Collapse
Affiliation(s)
- Hee Ja Ko
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Bo Yeon Kim
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Chan Hee Jung
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Sung Wan Chun
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Ji Oh Mok
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Yeo Joo Kim
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Hyeong Kyu Park
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Chul Hee Kim
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Sang Jin Kim
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Dong Won Byun
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Kyo Il Suh
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Myung Hi Yoo
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Sung Gu Kang
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea
| |
Collapse
|
17
|
Tanaka S, Shiraha H, Nakanishi Y, Nishina SI, Matsubara M, Horiguchi S, Takaoka N, Iwamuro M, Kataoka J, Kuwaki K, Hagihara H, Toshimori J, Ohnishi H, Takaki A, Nakamura S, Nouso K, Yagi T, Yamamoto K. Runt-related transcription factor 3 reverses epithelial-mesenchymal transition in hepatocellular carcinoma. Int J Cancer 2012; 131:2537-2546. [PMID: 22488108 DOI: 10.1002/ijc.27575] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 03/19/2012] [Indexed: 01/17/2023]
Abstract
Loss or decreased expression of runt-related transcription factor 3 (RUNX3), a tumor suppressor gene involved in gastric and other cancers, has been frequently observed in hepatocellular carcinoma (HCC). The objective of this study was to identify the regulatory mechanism of the epithelial-mesenchymal transition (EMT) by RUNX3 in HCC. Human HCC cell lines, Hep3B, Huh7, HLF and SK-Hep1, were divided into low- and high-EMT lines, based on their expression of TWIST1 and SNAI2, and were used in this in vitro study. Ectopic RUNX3 expression had an anti-EMT effect in low-EMT HCC cell lines characterized by increased E-cadherin expression and decreased N-cadherin and vimentin expression. RUNX3 expression has previously been reported to reduce jagged-1 (JAG1) expression; therefore, JAG1 ligand peptide was used to reinduce EMT in RUNX3-expressing low-EMT HCC cells. Immunohistochemical analyses were performed for RUNX3, E-cadherin, N-cadherin and TWIST1 in 33 human HCC tissues, also divided into low- and high-EMT HCC, based on TWIST1 expression. E-cadherin expression was correlated positively and N-cadherin expression was correlated negatively with RUNX3 expression in low-EMT HCC tissues. Correlations between EMT markers and RUNX3 mRNA expression were analyzed using Oncomine datasets. Similarly, mRNA expression of E-cadherin was also significantly correlated with that of RUNX3 in low-EMT HCC, while mRNA expression of JAG1 was negatively correlated with that of RUNX3. These results suggest a novel mechanism by which loss or decreased expression of RUNX3 induces EMT via induction of JAG1 expression in low-EMT HCC.
Collapse
Affiliation(s)
- Shigetomi Tanaka
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Huynh KT, Hoon DSB. Epigenetics of regional lymph node metastasis in solid tumors. Clin Exp Metastasis 2012; 29:747-56. [DOI: 10.1007/s10585-012-9491-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 05/20/2012] [Indexed: 01/01/2023]
|
19
|
Lenoir O, Flosseau K, Ma FX, Blondeau B, Mai A, Bassel-Duby R, Ravassard P, Olson EN, Haumaitre C, Scharfmann R. Specific control of pancreatic endocrine β- and δ-cell mass by class IIa histone deacetylases HDAC4, HDAC5, and HDAC9. Diabetes 2011; 60:2861-71. [PMID: 21953612 PMCID: PMC3198089 DOI: 10.2337/db11-0440] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Class IIa histone deacetylases (HDACs) belong to a large family of enzymes involved in protein deacetylation and play a role in regulating gene expression and cell differentiation. Previously, we showed that HDAC inhibitors modify the timing and determination of pancreatic cell fate. The aim of this study was to determine the role of class IIa HDACs in pancreas development. RESEARCH DESIGN AND METHODS We took a genetic approach and analyzed the pancreatic phenotype of mice lacking HDAC4, -5, and -9. We also developed a novel method of lentiviral infection of pancreatic explants and performed gain-of-function experiments. RESULTS We show that class IIa HDAC4, -5, and -9 have an unexpected restricted expression in the endocrine β- and δ-cells of the pancreas. Analyses of the pancreas of class IIa HDAC mutant mice revealed an increased pool of insulin-producing β-cells in Hdac5(-/-) and Hdac9(-/-) mice and an increased pool of somatostatin-producing δ-cells in Hdac4(-/-) and Hdac5(-/-) mice. Conversely, HDAC4 and HDAC5 overexpression showed a decreased pool of insulin-producing β-cells and somatostatin-producing δ-cells. Finally, treatment of pancreatic explants with the selective class IIa HDAC inhibitor MC1568 enhances expression of Pax4, a key factor required for proper β-and δ-cell differentiation and amplifies endocrine β- and δ-cells. CONCLUSIONS We conclude that HDAC4, -5, and -9 are key regulators to control the pancreatic β/δ-cell lineage. These results highlight the epigenetic mechanisms underlying the regulation of endocrine cell development and suggest new strategies for β-cell differentiation-based therapies.
Collapse
Affiliation(s)
- Olivia Lenoir
- Institut National de la Santé et de la Recherche Médicale (INSERM) U845, Research Center Growth and Signalling, Paris Descartes University, Sorbonne Paris Cité, Necker Hospital, Paris, France
| | - Kathleen Flosseau
- Institut National de la Santé et de la Recherche Médicale (INSERM) U845, Research Center Growth and Signalling, Paris Descartes University, Sorbonne Paris Cité, Necker Hospital, Paris, France
| | - Feng Xia Ma
- Institut National de la Santé et de la Recherche Médicale (INSERM) U845, Research Center Growth and Signalling, Paris Descartes University, Sorbonne Paris Cité, Necker Hospital, Paris, France
| | - Bertrand Blondeau
- INSERM Unité Mixte de Recherche (UMR)-S 872, Cordeliers Research Center, Paris, France
| | - Antonello Mai
- Pasteur Institute-Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Philippe Ravassard
- Institute of Brain and Spinal Cord Research Center, Centre National de la Recherche Scientifique (CNRS) UMR 7225, INSERM UMR-S 975, Pierre and Marie Curie University, Pitié Salpêtrière Hospital, Paris, France
| | - Eric N. Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Cécile Haumaitre
- Institut National de la Santé et de la Recherche Médicale (INSERM) U845, Research Center Growth and Signalling, Paris Descartes University, Sorbonne Paris Cité, Necker Hospital, Paris, France
- Corresponding author: Cécile Haumaitre, , or Raphaël Scharfmann,
| | - Raphaël Scharfmann
- Institut National de la Santé et de la Recherche Médicale (INSERM) U845, Research Center Growth and Signalling, Paris Descartes University, Sorbonne Paris Cité, Necker Hospital, Paris, France
- Corresponding author: Cécile Haumaitre, , or Raphaël Scharfmann,
| |
Collapse
|
20
|
Detection of deleted in malignant brain tumors 1 and runt-related transcription factor 3 gene expressions in bladder carcinoma. Mol Biol Rep 2011; 39:4691-5. [DOI: 10.1007/s11033-011-1261-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Accepted: 09/14/2011] [Indexed: 10/17/2022]
|
21
|
Shiraha H, Nishina SI, Yamamoto K. Loss of runt-related transcription factor 3 causes development and progression of hepatocellular carcinoma. J Cell Biochem 2011; 112:745-9. [PMID: 21328447 DOI: 10.1002/jcb.22973] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Runt-related transcription factor 3 (RUNX3) is reported as a tumor suppressor gene for gastric cancer, and may be important in the development of hepatocellular carcinoma (HCC). RUNX3 expression is frequently lost or decreased by hemizygous deletion or hypermethylation of its promoter lesion in HCC. The significance of decreased expression of RUNX3 in HCC has not been fully elucidated, but is likely related to dysfunction of cell cycle regulation, decrement of apoptosis, enhancement of angiogenesis, and development of epithelial-mesenchymal transition. RUNX3 is a promising candidate as a tumor suppressor gene for HCC.
Collapse
Affiliation(s)
- Hidenori Shiraha
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine and Dentistry, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | | | | |
Collapse
|
22
|
Lee JH, Pyon JK, Kim DW, Lee SH, Nam HS, Kang SG, Kim CH, Lee YJ, Chun JS, Cho MK. Expression of RUNX3 in skin cancers. Clin Exp Dermatol 2011; 36:769-74. [PMID: 21623876 DOI: 10.1111/j.1365-2230.2011.04069.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Expression of Runt-related transcription factor 3 (RUNX3) is reduced in a large number of cancers. However, a few studies have reported higher expression of RUNX3 in several cancers, including basal cell carcinoma (BCC). In light of this, we explored the expression of RUNX3 in skin cancers generally, to determine whether it acts as an oncogene or a tumour-suppressor gene in skin tumours. AIM To investigate the expression of RUNX3 in normal skin and malignant skin tumours. METHODS RUNX3 expression was evaluated by western blotting in 24 specimens, comprising 6 malignant melanoma (MM), 6 squamous cell carcinoma (SCC), 6 BCC and 6 normal skin specimens. Immunohistochemical staining was carried out to analyse RUNX3 expression in 16 MM, 16 SCC and 16 BCC specimens. To identify where the protein was expressed, the cytoplasmic and nuclear protein expression of RUNX3 in skin cancer tissues was determined. A cell-proliferation study was performed on an MM line (G361) by small interfering (si)RNA transfection. RESULTS The western blotting experiments showed that RUNX3 was not expressed in normal skin tissues, but it was overexpressed in all MM and SCC samples, and in five of the six BCC samples. Using immunochemistry, RUNX3 was found to be overexpressed in all cancer tissues analysed. Subcellular fraction analysis revealed that RUNX3 was expressed in the nuclei but not the cytoplasm of all the skin cancer tissues analysed, and RUNX3 silencing by siRNA in G361 cells resulted in a decrease in proliferation. CONCLUSIONS Based on these results, we suggest that RUNX3 has an oncogenic potential and does not act as a tumour suppressor in skin cancers.
Collapse
Affiliation(s)
- J H Lee
- Department of Plastic and Reconstructive Surgery, Hanyang University Guri Hospital, Guri, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Kudo Y, Tsunematsu T, Takata T. Oncogenic role of RUNX3 in head and neck cancer. J Cell Biochem 2011; 112:387-93. [PMID: 21268058 DOI: 10.1002/jcb.22967] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cumulative evidences show that Runt-related transcription factor 3 (RUNX3) has a tumor suppressive role in various cancers. In particular, RUNX3 appears to be an important component of the transforming growth factor-β (TGF-β)-induced tumor suppression pathway. Contrary to reports on this tumor suppressive role of RUNX3, RUNX3 can also function as an oncogene when overexpressed. Recently, we found that RUNX3 overexpression was frequently observed and was well correlated with malignant behaviors in head and neck cancer, which is one of the most common types of human cancer. Moreover, it has been revealed that RUNX3 overexpression promoted cell growth and inhibited apoptosis in head and neck cancer cells. This review introduces the oncogenic role of RUNX3 in certain types of cancer including head and neck cancer.
Collapse
Affiliation(s)
- Yasusei Kudo
- Division of Frontier Medical Science, Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan.
| | | | | |
Collapse
|
24
|
Nakanishi Y, Shiraha H, Nishina SI, Tanaka S, Matsubara M, Horiguchi S, Iwamuro M, Takaoka N, Uemura M, Kuwaki K, Hagihara H, Toshimori J, Ohnishi H, Takaki A, Nakamura S, Kobayashi Y, Nouso K, Yagi T, Yamamoto K. Loss of runt-related transcription factor 3 expression leads hepatocellular carcinoma cells to escape apoptosis. BMC Cancer 2011; 11:3. [PMID: 21205319 PMCID: PMC3022884 DOI: 10.1186/1471-2407-11-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Accepted: 01/04/2011] [Indexed: 01/27/2023] Open
Abstract
Background Runt-related transcription factor 3 (RUNX3) is known as a tumor suppressor gene for gastric cancer and other cancers, this gene may be involved in the development of hepatocellular carcinoma (HCC). Methods RUNX3 expression was analyzed by immunoblot and immunohistochemistry in HCC cells and tissues, respectively. Hep3B cells, lacking endogenous RUNX3, were introduced with RUNX3 constructs. Cell proliferation was measured using the MTT assay and apoptosis was evaluated using DAPI staining. Apoptosis signaling was assessed by immunoblot analysis. Results RUNX3 protein expression was frequently inactivated in the HCC cell lines (91%) and tissues (90%). RUNX3 expression inhibited 90 ± 8% of cell growth at 72 h in serum starved Hep3B cells. Forty-eight hour serum starvation-induced apoptosis and the percentage of apoptotic cells reached 31 ± 4% and 4 ± 1% in RUNX3-expressing Hep3B and control cells, respectively. Apoptotic activity was increased by Bim expression and caspase-3 and caspase-9 activation. Conclusion RUNX3 expression enhanced serum starvation-induced apoptosis in HCC cell lines. RUNX3 is deleted or weakly expressed in HCC, which leads to tumorigenesis by escaping apoptosis.
Collapse
Affiliation(s)
- Yutaka Nakanishi
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine and Dentistry, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Levkovitz L, Yosef N, Gershengorn MC, Ruppin E, Sharan R, Oron Y. A novel HMM-based method for detecting enriched transcription factor binding sites reveals RUNX3 as a potential target in pancreatic cancer biology. PLoS One 2010; 5:e14423. [PMID: 21203558 PMCID: PMC3008686 DOI: 10.1371/journal.pone.0014423] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 09/10/2010] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma (PAC) is one of the most intractable malignancies. In order to search for potential new therapeutic targets, we relied on computational methods aimed at identifying transcription factor binding sites (TFBSs) over-represented in the promoter regions of genes differentially expressed in PAC. Though many computational methods have been implemented to accomplish this, none has gained overall acceptance or produced proven novel targets in PAC. To this end we have developed DEMON, a novel method for motif detection. METHODOLOGY DEMON relies on a hidden Markov model to score the appearance of sequence motifs, taking into account all potential sites in a promoter of potentially varying binding affinities. We demonstrate DEMON's accuracy on simulated and real data sets. Applying DEMON to PAC-related data sets identifies the RUNX family as highly enriched in PAC-related genes. Using a novel experimental paradigm to distinguish between normal and PAC cells, we find that RUNX3 mRNA (but not RUNX1 or RUNX2 mRNAs) exhibits time-dependent increases in normal but not in PAC cells. These increases are accompanied by changes in mRNA levels of putative RUNX gene targets. CONCLUSIONS The integrated application of DEMON and a novel differentiation system led to the identification of a single family member, RUNX3, which together with four of its putative targets showed a robust response to a differentiation stimulus in healthy cells, whereas this regulatory mechanism was absent in PAC cells, emphasizing RUNX3 as a promising target for further studies.
Collapse
Affiliation(s)
- Liron Levkovitz
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Nir Yosef
- School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Marvin C. Gershengorn
- Clinical Endocrinology Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Eytan Ruppin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Roded Sharan
- School of Computer Science, Tel Aviv University, Tel Aviv, Israel
- * E-mail: (RS); (YO)
| | - Yoram Oron
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- * E-mail: (RS); (YO)
| |
Collapse
|
26
|
Molecular pathology of RUNX3 in human carcinogenesis. Biochim Biophys Acta Rev Cancer 2009; 1796:315-31. [PMID: 19682550 DOI: 10.1016/j.bbcan.2009.07.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Accepted: 07/31/2009] [Indexed: 12/12/2022]
Abstract
A major goal of molecular biology is to elucidate the mechanisms underlying cancer development and progression in order to achieve early detection, better diagnosis and staging and novel preventive and therapeutic strategies. We feel that an understanding of Runt-related transcription factor 3 (RUNX3)-regulated biological pathways will directly impact our knowledge of these areas of human carcinogenesis. The RUNX3 transcription factor is a downstream effector of the transforming growth factor-beta (TGF-beta) signaling pathway, and has a critical role in the regulation of cell proliferation and cell death by apoptosis, and in angiogenesis, cell adhesion and invasion. We previously identified RUNX3 as a major gastric tumor suppressor by establishing a causal relationship between loss of function and gastric carcinogenesis. More recently, we showed that RUNX3 functions as a bona fide initiator of colonic carcinogenesis by linking the Wnt oncogenic and TGF-beta tumor suppressive pathways. Apart from gastric and colorectal cancers, a multitude of epithelial cancers exhibit inactivation of RUNX3, thereby making it a putative tumor suppressor in human neoplasia. This review highlights our current understanding of the molecular mechanisms of RUNX3 inactivation in the context of cancer development and progression.
Collapse
|
27
|
Kitago M, Martinez SR, Nakamura T, Sim MS, Hoon DSB. Regulation of RUNX3 tumor suppressor gene expression in cutaneous melanoma. Clin Cancer Res 2009; 15:2988-94. [PMID: 19336521 DOI: 10.1158/1078-0432.ccr-08-3172] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE RUNX3 is a known tumor suppressor gene in several carcinomas. Aberration in RUNX3 expression has not been described for cutaneous melanoma. Therefore, we assessed the expression of RUNX3 in cutaneous melanoma and its regulatory mechanisms relative to tumor progression. EXPERIMENTAL DESIGN The expression of RUNX3 mRNA and miR-532-5p (microRNA) was assessed in melanoma lines and in primary and metastatic melanoma tumors. RESULTS RUNX3 mRNA expression was down-regulated in 11 of 11 (100%) metastatic melanoma lines relative to normal melanocytes (P < 0.001). Among 123 primary and metastatic melanoma tumors and 12 normal skin samples, RUNX3 expression was significantly down-regulated in primary melanomas (n = 82; P = 0.02) and in melanoma metastasis (n = 41; P < 0.0001) versus normal skin (n = 12). This suggested that RUNX3 down-regulation may play a role in the development and progression of melanoma. RUNX3 promoter region hypermethylation was assessed as a possible regulator of RUNX3 expression using methylation-specific PCR. Assessment of RUNX3 promoter region methylation showed that only 5 of 17 (29%) melanoma lines, 2 of 52 (4%) primary melanomas, and 5 of 30 (17%) metastatic melanomas had hypermethylation of the promoter region. A microRNA (miR-532-5p) was identified as a target of RUNX3 mRNA sequences. miR-532-5p expression was shown to be significantly up-regulated in melanoma lines and metastatic melanoma tumors relative to normal melanocytes and primary melanomas, respectively. To investigate the relation between RUNX3 and miR-532-5p, anti-miR-532-5p was transfected into melanoma lines. Inhibition of miR-532-5p up-regulated both RUNX3 mRNA and protein expression. CONCLUSIONS RUNX3 is down-regulated during melanoma progression and miR-532-5p is a regulatory factor of RUNX3 expression.
Collapse
Affiliation(s)
- Minoru Kitago
- Department of Molecular Oncology, John Wayne Cancer Institute at Saint John's Health Center, Santa Monica, California, USA
| | | | | | | | | |
Collapse
|
28
|
Robertson AJ, Coluccio A, Knowlton P, Dickey-Sims C, Coffman JA. Runx expression is mitogenic and mutually linked to Wnt activity in blastula-stage sea urchin embryos. PLoS One 2008; 3:e3770. [PMID: 19020668 PMCID: PMC2582955 DOI: 10.1371/journal.pone.0003770] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2008] [Accepted: 11/01/2008] [Indexed: 11/25/2022] Open
Abstract
Background The Runt homology domain (Runx) defines a metazoan family of sequence-specific transcriptional regulatory proteins that are critical for animal development and causally associated with a variety of mammalian cancers. The sea urchin Runx gene SpRunt-1 is expressed throughout the blastula stage embryo, and is required globally during embryogenesis for cell survival and differentiation. Methodology/Principal Findings Depletion of SpRunt-1 by morpholino antisense-mediated knockdown causes a blastula stage deficit in cell proliferation, as shown by bromodeoxyuridine (BrdU) incorporation and direct cell counts. Reverse transcription coupled polymerase chain reaction (RT-PCR) studies show that the cell proliferation deficit is presaged by a deficit in the expression of several zygotic wnt genes, including wnt8, a key regulator of endomesoderm development. In addition, SpRunt-1-depleted blastulae underexpress cyclinD, an effector of mitogenic Wnt signaling. Blastula stage cell proliferation is also impeded by knockdown of either wnt8 or cyclinD. Chromatin immunoprecipitation (ChIP) indicates that Runx target sites within 5′ sequences flanking cyclinD, wnt6 and wnt8 are directly bound by SpRunt-1 protein at late blastula stage. Furthermore, experiments using a green fluorescent protein (GFP) reporter transgene show that the blastula-stage operation of a cis-regulatory module previously shown to be required for wnt8 expression (Minokawa et al., Dev. Biol. 288: 545–558, 2005) is dependent on its direct sequence-specific interaction with SpRunt-1. Finally, inhibitor studies and immunoblot analysis show that SpRunt-1 protein levels are negatively regulated by glycogen synthase kinase (GSK)-3. Conclusions/Significance These results suggest that Runx expression and Wnt signaling are mutually linked in a feedback circuit that controls cell proliferation during development.
Collapse
Affiliation(s)
- Anthony J. Robertson
- Mount Desert Island Biological Laboratory, Salisbury Cove, Maine, United States of America
| | - Alison Coluccio
- Mount Desert Island Biological Laboratory, Salisbury Cove, Maine, United States of America
| | - Peter Knowlton
- Mount Desert Island Biological Laboratory, Salisbury Cove, Maine, United States of America
| | - Carrie Dickey-Sims
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - James A. Coffman
- Mount Desert Island Biological Laboratory, Salisbury Cove, Maine, United States of America
- * E-mail:
| |
Collapse
|
29
|
Abstract
Alteration in transforming growth factor-β signalling pathway is one of the main causes of pancreatic cancer. The human runt-related transcription factor 3 gene (RUNX3) is an important component of this pathway. RUNX3 locus 1p36 is commonly deleted in a variety of human cancers, including pancreatic cancer. Therefore, we examined genetic and epigenetic alterations of RUNX3 in human pancreatic cancer. Thirty-two patients with pancreatic cancer were investigated in this study. We examined the methylation status of RUNX3 promoter region, loss of heterozygosity (LOH) at 1p36, and conducted a mutation analysis. The results were compared with clinicopathological data. Promoter hypermethylation was detected in 20 (62.5%) of 32 pancreatic cancer tissues, confirmed by sequence of bisulphite-treated DNA. Loss of heterozygosity was detected in 11 (34.3%) of 32 pancreatic cancers. In comparison with clinicopathological data, hypermethylation showed a relation with a worse prognosis (P=0.0143). Hypermethylation and LOH appear to be common mechanisms for inactivation of RUNX3 in pancreatic cancer. Therefore, RUNX3 may be an important tumour suppressor gene related to pancreatic cancer.
Collapse
|
30
|
Ni Z, Bao MX, Liu NZ, Zhao Q, Qin H, Yang Y, Qiu YJ, Wang TT. Relationship between tumor suppressor gene RUNX3 expression and cell proliferation and apoptosis in colonic cancer cell line Lovo. Shijie Huaren Xiaohua Zazhi 2008; 16:711-715. [DOI: 10.11569/wcjd.v16.i7.711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression and methylation status of tumor suppressor gene RUNX3 in human colon cancer cell line Lovo and explore the effects of 5-aza-2'-deoxycytidine (5-Aza-CdR) on the proliferation and apoptosis of Lovo cells and the expression of RUNX3 gene.
METHODS: Human colon cancer cell line Lovo was treated with 5-Aza-CdR, a specific methyltransferase inhibitor, at the concentrations of 0.4, 4 and 40 μmol/L for 3 d, and then cultured in RPMI 1640 medium for 5 d. The activation of Lovo cells was respectively observed by Tetrazolium salt colorimetric (MTT) assay before and after 5-Aza-CdR treatment. The change in expression of RUNX3 mRNA was observed by semi-quantitative reverse transcription-polymerase chain reaction (RT-PCR). The apoptosis was analyzed by flow cytometry. The methylation status of gene promoter was determined by methylation-specific PCR (MSP).
RESULTS: Lovo cells treated with 5-Aza-CdR (0.4, 4, 40 μmol/L) displayed a slowed growth rate in different degrees in contrast with those in the control group and their growth rates decreased accordingly with the increase of 5-Aza-CdR concentration. There were significant increases in RUNX3 mRNA expression (0.46 ± 0.06, 0.71 ± 0.06, 0.84 ± 0.07 vs 0, P < 0.01) and apoptotic rates of Lovo cells (10.95% ± 2.09%, 17.61% ± 1.51%, 26.60% ± 1.89% vs 2.92% ± 0.93%, P < 0.01) after 5-Aza-CdR treatment in comparison with those in the control group. The level of RUNX3 mRNA expression and the apoptotic rates of Lovo cells were increased in correlation with 5-Aza-CdR concentration (F = 168.4, F = 145.7, P < 0.01). Methylation of RUNX3 promoter region was confirmed in Lovo cells of control group and detected partly in 5-Aza-CdR-treated group.
CONCLUSION: 5-Aza-CdR is able to reverse the methylation status of RUNX3 promoter region. The re-expression of RUNX3 gene can inhibit Lovo cell growth and partly induce Lovo cell apoptosis.
Collapse
|
31
|
Kayed H, Jiang X, Keleg S, Jesnowski R, Giese T, Berger MR, Esposito I, Löhr M, Friess H, Kleeff J. Regulation and functional role of the Runt-related transcription factor-2 in pancreatic cancer. Br J Cancer 2007; 97:1106-15. [PMID: 17876328 PMCID: PMC2360444 DOI: 10.1038/sj.bjc.6603984] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Recent evidence suggests that Runt-related transcription factors play a role in different human tumours. In the present study, the localisation of the Runt-related transcription factor-2 (Runx2), its transcriptional activity, as well as its regulation of expression was analysed in human pancreatic ductal adenocarcinoma (PDAC). Quantitative real-time PCR and immunohistochemistry were used for Runx2 expression and localisation analysis. Runt-related transcription factor-2 expression was silenced using specific siRNA oligonucleotides in pancreatic cancer cells (Panc-1) and immortalised pancreatic stellate cells (IPSCs). Overexpression of Runx2 was achieved using a full-length expression vector. TGF-β1, BMP2, and other cytokines were assessed for their potential to regulate Runx2 expression. There was a 6.1-fold increase in median Runx2 mRNA levels in PDAC tissues compared to normal pancreatic tissues (P<0.0001). Runt-related transcription factor-2 was localised in pancreatic cancer cells, tubular complexes, and PanIN lesions of PDAC tissues as well as in tumour-associated fibroblasts/stellate cells. Coculture of IPSCs and Panc-1 cells, as well as treatment with TGF-β1 and BMP2, led to increased Runx2 expression in Panc-1 cells. Runt-related transcription factor-2 overexpression was associated with decreased MMP1 release as well as decreased growth and invasion of Panc-1 cells. These effects were reversed by Runx2 silencing. In conclusion, Runx2 is overexpressed in PDAC, where it is regulated by certain cytokines such as TGF-β1 and BMP2 in an auto- and paracrine manner. In addition, Runx2 has the potential to regulate the transcription of extracellular matrix modulators such as SPARC and MMP1, thereby influencing the tumour microenvironment.
Collapse
Affiliation(s)
- H Kayed
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - X Jiang
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - S Keleg
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - R Jesnowski
- Molecular Gastroenterology Unit, German Cancer Research Centre, Heidelberg, Germany
- Department of Medicine II, University of Heidelberg, Mannheim, Germany
| | - T Giese
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - M R Berger
- Unit of Toxicology and Chemotherapy, German Cancer Research Centre, Heidelberg, Germany
| | - I Esposito
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - M Löhr
- Molecular Gastroenterology Unit, German Cancer Research Centre, Heidelberg, Germany
- Department of Medicine II, University of Heidelberg, Mannheim, Germany
| | - H Friess
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - J Kleeff
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
- Department of Surgery, Technical University of Munich, Ismaningerstrasse 22, Munich 81675, Germany. E-mail:
| |
Collapse
|
32
|
Hwang KT, Han W, Bae JY, Hwang SE, Shin HJ, Lee JE, Kim SW, Min HJ, Noh DY. Downregulation of the RUNX3 gene by promoter hypermethylation and hemizygous deletion in breast cancer. J Korean Med Sci 2007; 22 Suppl:S24-31. [PMID: 17923751 PMCID: PMC2694388 DOI: 10.3346/jkms.2007.22.s.s24] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The RUNX3 gene is regarded as a tumor suppressor gene in many human solid tumors, and its inactivation is believed to be related with solid tumor carcinogenesis. As little information is available about the role of the RUNX3 gene in breast cancer, we investigated the relationship between the RUNX3 gene and breast cancer. We performed reverse transcriptase-polymerases chain reaction (RT-PCR), methylation specific PCR, and bicolor fluorescent in situ hybridization analysis in an effort to reveal related mechanisms. Forty breast tissue samples and 13 cell lines were used in this study. Eighty-five percent of breast cancer tissues showed downregulated RUNX3 gene expression, whereas it was downregulated in only 25% of normal breast tissues by RT-PCR assay. Sixty-seven percent of breast cancer cell lines showed downregulated RUNX3 expression, but the RUNX3 gene was not expressed in two normal breast cell lines. Hypermethylation was observed in 53% of breast cancer tissues and 57% of breast cancer cell lines. Hemizygous deletion was observed in 43% of breast cancer cell lines. Hypermethylation and/or hemizygous deletion was observed in 5 of 7 breast cancer cell lines, and the four of these five examined showed no RUNX3 gene expression. We suggest that various mechanisms, including methylation and hemizygous deletion, could contribute to RUNX3 gene inactivation.
Collapse
Affiliation(s)
- Ki-Tae Hwang
- Department of Surgery, Seoul National University Boramae Hospital, Seoul, Korea
| | - Wonshik Han
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Ji-Yeon Bae
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Eun Hwang
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Hyuk Jai Shin
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Jeong Eon Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Sung-Won Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Jung Min
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Dong-Young Noh
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
33
|
Hartel M, di Mola FF, Selvaggi F, Mascetta G, Wente MN, Felix K, Giese NA, Hinz U, Di Sebastiano P, Büchler MW, Friess H. Vanilloids in pancreatic cancer: potential for chemotherapy and pain management. Gut 2006; 55:519-28. [PMID: 16174661 PMCID: PMC1856157 DOI: 10.1136/gut.2005.073205] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Success of chemotherapy and alleviation of pain are frequently less than optimal in pancreatic cancer patients, leading to increasing interest in new pharmacological substances, such as vanilloids. Our study addressed the question of whether vanilloids influence pancreatic cancer cell growth, and if vanilloids could be used for pain treatment via the vanilloid 1 receptor (VR1) in pancreatic cancer patients. METHODS In vitro, the effect of resiniferatoxin (vanilloid analogue) on apoptosis and cell growth in pancreatic cancer cells--either alone, combined with 5-fluorouracil (5-FU), or combined with gemcitabine--was determined by annexin V staining, FACS analysis, and MTT assay, respectively. VR1 expression was evaluated on RNA and protein level by quantitative polymerase chain reaction and immunohistochemistry in human pancreatic cancer and chronic pancreatitis. Patient characteristics--especially pain levels--were registered in a prospective database and correlated with VR1 expression. RESULTS Resiniferatoxin induced apoptosis by targeting mitochondrial respiration and decreased cell growth in pancreatic cancer cells without showing synergistic effects with 5-FU or gemcitabine. Expression of VR1 was significantly upregulated in human pancreatic cancer and chronic pancreatitis. VR1 expression was related to the intensity of pain reported by cancer patients but not to the intensity of pain reported by patients with chronic pancreatitis. CONCLUSIONS Resiniferatoxin induced apoptosis in pancreatic cancer cells indicates that vanilloids may be useful in the treatment of human pancreatic cancer. Furthermore, vanilloid might be a novel and effective treatment option for neurogenic pain in patients with pancreatic cancer.
Collapse
Affiliation(s)
- M Hartel
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Puig-Kröger A, Corbí A. RUNX3: A new player in myeloid gene expression and immune response. J Cell Biochem 2006; 98:744-56. [PMID: 16598764 DOI: 10.1002/jcb.20813] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
RUNX transcription factors function as scaffolds for interaction with various coregulatory proteins during developmental processes such as hematopoiesis, neurogenesis, and osteogenesis. The current view places RUNX proteins within the TGF-beta signaling pathway, although each one exhibits cell- and tissue-specific functions. In the case of RUNX3, recent data have suggested its function as a tumor suppressor factor and highlighted its involvement in immune cell differentiation and activation. The molecular mechanisms for the pleiotropic effects of Runx3 deficiency are not completely understood. The present article will summarize the known functional activities of RUNX3, emphasizing its role in myeloid cell gene expression and its potential contribution to the migratory and adhesive capabilities of this cell lineage.
Collapse
|
35
|
Abstract
Pancreatic ductal adenocarcinoma is one of the most fatal malignancies. Intensive investigation of molecular pathogenesis might lead to identifying useful molecules for diagnosis and treatment of the disease. Pancreatic ductal adenocarcinoma harbors complicated aberrations of alleles including losses of 1p, 6q, 9p, 12q, 17p, 18q, and 21q, and gains of 8q and 20q. Pancreatic cancer is usually initiated by mutation of KRAS and aberrant expression of SHH. Overexpression of AURKA mapping on 20q13.2 may significantly enhance overt tumorigenesity. Aberrations of tumor suppressor genes synergistically accelerate progression of the carcinogenic pathway through pancreatic intraepithelial neoplasia (PanIN) to invasive ductal adenocarcinoma. Abrogation of CDKN2A occurs in low-grade/early PanIN, whereas aberrations of TP53 and SMAD4 occur in high-grade/late PanIN. SMAD4 may play suppressive roles in tumorigenesis by inhibition of angiogenesis. Loss of 18q precedes SMAD4 inactivation, and restoration of chromosome 18 in pancreatic cancer cells results in tumor suppressive phenotypes regardless of SMAD4 status, indicating the possible existence of a tumor suppressor gene(s) other than SMAD4 on 18q. DUSP6 at 12q21-q22 is frequently abrogated by loss of expression in invasive ductal adenocarcinomas despite fairly preserved expression in PanIN, which suggests that DUSP6 works as a tumor suppressor in pancreatic carcinogenesis. Restoration of chromosome 12 also suppresses growths of pancreatic cancer cells despite the recovery of expression of DUSP6; the existence of yet another tumor suppressor gene on 12q is strongly suggested. Understanding the molecular mechanisms of pancreatic carcinogenesis will likely provide novel clues for preventing, detecting, and ultimately curing this life-threatening disease.
Collapse
Affiliation(s)
- Toru Furukawa
- Department of Molecular Pathology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | | | | |
Collapse
|
36
|
Guweidhi A, Kleeff J, Adwan H, Giese NA, Wente MN, Giese T, Büchler MW, Berger MR, Friess H. Osteonectin influences growth and invasion of pancreatic cancer cells. Ann Surg 2005; 242:224-34. [PMID: 16041213 PMCID: PMC1357728 DOI: 10.1097/01.sla.0000171866.45848.68] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE We sought to examine the expression and functional role of osteonectin in primary and metastatic pancreatic ductal adenocarcinoma (PDAC). BACKGROUND The glycoprotein osteonectin plays a vital role in cell-matrix interactions and is involved in various biologic processes. Overexpression of osteonectin is present in malignant tumors and correlates with disease progression and poor prognosis. METHODS Expression of osteonectin was analyzed by quantitative polymerase chain reaction and immunohistochemistry in pancreatic tissues and by enzyme-linked immunosorbent assay in the serum of patients and donors. Recombinant osteonectin and specific antisense oligonucleotides were used to examine the effects of osteonectin on induction of target genes, and on proliferation and invasiveness of pancreatic cancer cells. RESULTS There was a 31-fold increase in osteonectin mRNA levels in PDAC and a 16-fold increase in chronic pancreatitis as compared with the normal pancreas (P < 0.01). By immunohistochemistry, faint immunoreactivity was detected in the normal pancreas. In contrast, strong staining of the cancer cells was observed in addition to extensive osteonectin immunoreactivity in surrounding fibroblasts and in the extracellular matrix. In metastatic tissues, strong immunoreactivity was observed in fibroblasts and in extracellular matrix surrounding metastatic cancer cells, whereas the signal was absent in most tumor cells. In vitro studies showed that osteonectin was able to inhibit cancer cell growth while promoting invasiveness of pancreatic tumor cells. CONCLUSION Osteonectin is markedly overexpressed in pancreatic cancer and has the potential to increase the invasiveness of pancreatic cancer cells.
Collapse
Affiliation(s)
- Ahmed Guweidhi
- Department of General Surgery, University of Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Li J, Kleeff J, Esposito I, Kayed H, Felix K, Giese T, Büchler MW, Friess H. Expression analysis of PMP22/Gas3 in premalignant and malignant pancreatic lesions. J Histochem Cytochem 2005; 53:885-93. [PMID: 15995147 DOI: 10.1369/jhc.4a6546.2005] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
PMP22 is a structural protein of Schwann cells, but it also influences cell proliferation. In the present study, quantitative RT-PCR (QRT-PCR) and immunohistochemistry were used to determine PMP22 mRNA levels and to localize PMP22 in the normal pancreas (n=20), chronic pancreatitis (CP) (n=22), pancreatic ductal adenocarcinoma (PDAC) (n=31), intraductal papillary mucinous neoplasms (IPMN) (n=9), mucinous cystic tumors (MCN) (n=4), and in a panel of PanIN lesions (n=29). PMP22 mRNA levels were significantly higher in CP (3-fold) and PDAC (2.5-fold), compared to normal pancreatic tissues. PMP22 expression was restricted to nerves in the normal pancreas, while in CP and PDAC PMP22 was also expressed in PanIN lesions and in a small percentage of pancreatic cancer cells. PMP22 was weak to absent in the tumor cells of IPMNs and MCNs. PMP22 mRNA was present at different levels in cultured pancreatic cancer cells and up-regulated by transforming growth factor (TGF)-beta1 in 2 of 8 of these cell lines. In conclusion, PMP22 expression is present in both CP and PDAC tissues. Its expression in PanIN lesions and some pancreatic cancer cells in vitro and in vivo suggests a role of PMP22 in the neoplastic transformation process from the normal pancreas to pre-malignant lesions to pancreatic cancer.
Collapse
Affiliation(s)
- Junsheng Li
- Department of General Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Guo C, Ding J, Yao L, Sun L, Lin T, Song Y, Sun L, Fan D. Tumor suppressor gene Runx3 sensitizes gastric cancer cells to chemotherapeutic drugs by downregulating Bcl-2, MDR-1 and MRP-1. Int J Cancer 2005; 116:155-60. [PMID: 15756676 DOI: 10.1002/ijc.20919] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The Runx3 gene is a member of the runt domain family transcription factors, key regulators of development and differentiation in metazoan. Recently, Runx3 was identified as a tumor suppressor gene. Loss of Runx3 was found to be associated with genesis and progression of gastric cancer. In this study, we transfected the gastric cancer cell line SGC7901 with eukaryotic expression vector of Runx3. In vitro drug sensitivity assay suggested that SGC7901/Runx3 cells were more sensitive to various chemotherapeutic drugs. Blocking Runx3 expression in immortalized stomach mucosal cells (GES-1) or gastric cancer cells (SGC7901) by Runx3-specific small interfering RNA conferred the cells resistance to chemotherapeutic drugs. Flow cytometry examination suggested that expression of Runx3 in gastric cancer cells increased the intracellular accumulation and retention of adriamycin. Semiquantitative RT-PCR and Western blot suggested that Runx3 downregulated expression of Bcl-2, MDR-1 (P-gp) and MRP-1. Binding of Runx3 to promoter sequences of Bcl-2, MDR-1 and MRP-1 gene was detected by eletrophoretic mobility shift assay (EMSA) and supershift EMSA. We cloned the MDR-1 and MRP-1 gene promoters containing Runx binding sites and constructed the luciferase reporter vectors of these 2 promoters. Luciferase reporter assay suggested that Runx3 inhibited the promoter activity of the MDR-1 and MRP-1 promoter in SGC7901 cells. Taken together, our findings suggested that overexpression of Runx3 could sensitize gastric cancer cells to chemotherapeutic drugs by downregulating the Bcl-2, MDR-1 and MRP-1.
Collapse
Affiliation(s)
- Changcun Guo
- Department of Gastroenterology and Key State Laboratory of Cancer Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
van der Meulen T, Kranenbarg S, Schipper H, Samallo J, van Leeuwen JL, Franssen H. Identification and characterisation of two runx2 homologues in zebrafish with different expression patterns. ACTA ACUST UNITED AC 2005; 1729:105-17. [PMID: 15894389 DOI: 10.1016/j.bbaexp.2005.03.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2004] [Revised: 03/22/2005] [Accepted: 03/25/2005] [Indexed: 01/14/2023]
Abstract
Genome and gene duplications are considered to be the impetus to generate new genes, as the presence of multiple copies of a gene allows for paralogues to adopt novel function. After at least two rounds of genome/gene duplication, the Runt gene family consists of three members in vertebrates, instead of one in invertebrates. One of the family members, Runx2, plays a key role in the development of bone, a tissue that first occurs in vertebrates. The family has thus gained new gene function in the course of evolution. Two Runx2 genes were cloned in the vertebrate model system the zebrafish (Danio rerio). The expression patterns of the two genes differ and their kinetics differ up to four fold. In addition, splice forms exist that are novel when compared with mammals. Together, these findings comprise opportunities for selection and retention of the paralogues towards divergent and possibly new function.
Collapse
Affiliation(s)
- T van der Meulen
- Experimental Zoology group, Wageningen University, Marijkeweg 40, 6709 PG, Wageningen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
The RUNX genes have come to prominence recently because of their roles as essential regulators of cell fate in development and their paradoxical effects in cancer, in which they can function either as tumour-suppressor genes or dominant oncogenes according to context. How can this family of transcription factors have such an ambiguous role in cancer? How and where do these genes impinge on the pathways that regulate growth control and differentiation? And what is the evidence for a wider role for the RUNX genes in non-haematopoietic cancers?
Collapse
Affiliation(s)
- Karen Blyth
- Molecular Oncology Laboratory, Institute of Comparative Medicine, University of Glasgow Veterinary School, Glasgow, G61 1QH, UK
| | | | | |
Collapse
|
41
|
Li J, Kleeff J, Abiatari I, Kayed H, Giese NA, Felix K, Giese T, Büchler MW, Friess H. Enhanced levels of Hsulf-1 interfere with heparin-binding growth factor signaling in pancreatic cancer. Mol Cancer 2005; 4:14. [PMID: 15817123 PMCID: PMC1087876 DOI: 10.1186/1476-4598-4-14] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2005] [Accepted: 04/07/2005] [Indexed: 12/15/2022] Open
Abstract
UNLABELLED Hsulf-1 is a newly identified enzyme, which has the ability to decrease the growth of hepatocellular, ovarian, and head and neck squamous cell carcinoma cells by interfering with heparin-binding growth factor signaling. Since pancreatic cancers over-express a number of heparin-binding growth factors and their receptors, the expression and function of this enzyme in pancreatic cancer was analyzed. RESULTS Pancreatic cancer samples expressed significantly (22.5-fold) increased Hsulf-1 mRNA levels compared to normal controls, and Hsulf-1 mRNA was localized in the cancer cells themselves as well as in peritumoral fibroblasts. 4 out of 8 examined pancreatic cancer cell lines expressed Hsulf-1, whereas its expression was below the level of detection in the other cell lines. Stable transfection of the Hsulf-1 negative Panc-1 pancreatic cancer cell line with a full length Hsulf-1 expression vector resulted in increased sulfatase activity and decreased cell-surface heparan-sulfate proteoglycan (HSPG) sulfation. Hsulf-1 expression reduced both anchorage-dependent and -independent cell growth and decreased FGF-2 mediated cell growth and invasion in this cell line. CONCLUSION High expression of Hsulf-1 occurs in the stromal elements as well as in the tumor cells in pancreatic cancer and interferes with heparin-binding growth factor signaling.
Collapse
Affiliation(s)
- Junsheng Li
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
- Department of General Surgery, Zhong-Da Hospital, Southeast University, Nanjing, China
| | - Jörg Kleeff
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Ivane Abiatari
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Hany Kayed
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Nathalia A Giese
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Klaus Felix
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Thomas Giese
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Markus W Büchler
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Helmut Friess
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|