1
|
Fuse H, Zheng Y, Alzoubi I, Graeber MB. TAMing Gliomas: Unraveling the Roles of Iba1 and CD163 in Glioblastoma. Cancers (Basel) 2025; 17:1457. [PMID: 40361384 PMCID: PMC12070867 DOI: 10.3390/cancers17091457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/19/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Gliomas, the most common type of primary brain tumor, are a significant cause of morbidity and mortality worldwide. Glioblastoma, a highly malignant subtype, is particularly common, aggressive, and resistant to treatment. The tumor microenvironment (TME) of gliomas, especially glioblastomas, is characterized by a distinct presence of tumor-associated macrophages (TAMs), which densely infiltrate glioblastomas, a hallmark of these tumors. This macrophage population comprises both tissue-resident microglia as well as macrophages derived from the walls of blood vessels and the blood stream. Ionized calcium-binding adapter molecule 1 (Iba1) and CD163 are established cellular markers that enable the identification and functional characterization of these cells within the TME. This review provides an in-depth examination of the roles of Iba1 and CD163 in malignant gliomas, with a focus on TAM activation, migration, and immunomodulatory functions. Additionally, we will discuss how recent advances in AI-enhanced cell identification and visualization techniques have begun to transform the analysis of TAMs, promising unprecedented precision in their characterization and providing new insights into their roles within the TME. Iba1 and CD163 appear to have both unique and shared roles in glioma pathobiology, and both have the potential to be targeted through different molecular and cellular mechanisms. We discuss the therapeutic potential of Iba1 and CD163 based on available preclinical (experimental) and clinical (human tissue-based) evidence.
Collapse
Affiliation(s)
- Haneya Fuse
- School of Medicine, Sydney Campus, University of Notre Dame, 160 Oxford Street, Sydney, NSW 2010, Australia;
| | - Yuqi Zheng
- Ken Parker Brain Tumor Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia;
| | - Islam Alzoubi
- School of Computer Science, The University of Sydney, J12/1 Cleveland St, Sydney, NSW 2008, Australia;
| | - Manuel B. Graeber
- Ken Parker Brain Tumor Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia;
- University of Sydney Association of Professors (USAP), University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
2
|
Benitha G, Ramani P, Jayakumar S, Ramalingam K. Molecular expression of Forkhead Box C2 gene (FOXC2) and Prospero homeobox gene (PROX-1) in oral squamous carcinoma and their correlation with clinicopathological parameters: A prospective cohort study. J Oral Maxillofac Pathol 2024; 28:216-225. [PMID: 39157851 PMCID: PMC11329087 DOI: 10.4103/jomfp.jomfp_394_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 02/05/2024] [Accepted: 03/29/2024] [Indexed: 08/20/2024] Open
Abstract
Background Forkhead box C2 gene (FOXC2) acts as an epithelial-mesenchymal transition (EMT) inducer while Prospero homeobox 1 gene (PROX-1) function as a regulator of lymphangiogenesis and angiogenesis in oral squamous cell carcinoma (OSCC). It is presumed that PROX-1 has both tumour-suppressive and oncogenic effects. The main aim of this study is to evaluate the role of PROX-1 and FOXC2 in the invasion and progression of OSCC cases and to correlate their expression with various histopathological parameters. Materials and Methods A prospective cohort study was conducted in a total sample size of 52 OSCC tissues and histologically tumour-free margins of 20. mRNA expression and protein levels of FOXC2 and PROX-1 were evaluated using real-time PCR and sandwich enzyme-linked immunosorbent assay techniques. Chi-square analysis and correlation analysis were done. Kaplan-Meier analysis evaluated the survival rate. Results Mean Ct values of FOXC2 were 1.915 ± 0.519 and PROX-1 was 0.061 ± 0.173. There was a significant 2-fold increase in the FOXC2 expression and a 0.5-fold decrease in the PROX-1 expression in OSCC tissue. Increased levels of FOXC2 protein and decreased levels of PROX-1 with a mean difference of 1.64 ± 0.73 ng/ml and 1.27 ± 0.33 ng/ml were observed in OSCC compared to histologically tumour-free margins. A significant positive correlation was found between the FOXC2 expression and clinicopathological parameters such as staging, perineural invasion (PNI) and lymphovascular invasion (LVI) whereas PROX-1 showed a significant negative correlation with histopathological parameters such as staging, PNI, LVI and tumour staging. There was a significant positive correlation between the PROX-1 and histologically tumour-free margins in disease-free survival patients (P-value = 0.03). Conclusion FOXC2 and PROX-1 expressions were correlated with lymphovascular invasion, OSCC tumour staging and PNI. Thus, FOXC2 and PROX-1 could be possible therapeutic targets in the treatment of OSCC that can inhibit the EMT in OSCC and thereby favouring a better prognosis.
Collapse
Affiliation(s)
- Georgia Benitha
- Department of Oral and Maxillofacial Pathology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Pratibha Ramani
- Department of Oral and Maxillofacial Pathology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Selvaraj Jayakumar
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Karthikeyan Ramalingam
- Department of Oral and Maxillofacial Pathology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| |
Collapse
|
3
|
Hargadon KM, Goodloe TB, Lloyd ND. Oncogenic functions of the FOXC2 transcription factor: a hallmarks of cancer perspective. Cancer Metastasis Rev 2022; 41:833-852. [PMID: 35701636 DOI: 10.1007/s10555-022-10045-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/06/2022] [Indexed: 01/25/2023]
Abstract
Epigenetic regulation of gene expression is a fundamental determinant of molecular and cellular function, and epigenetic reprogramming in the context of cancer has emerged as one of the key enabling characteristics associated with acquisition of the core hallmarks of this disease. As such, there has been renewed interest in studying the role of transcription factors as epigenetic regulators of gene expression in cancer. In this review, we discuss the current state of knowledge surrounding the oncogenic functions of FOXC2, a transcription factor that frequently becomes dysregulated in a variety of cancer types. In addition to highlighting the clinical impact of aberrant FOXC2 activity in cancer, we discuss mechanisms by which this transcription factor becomes dysregulated in both tumor and tumor-associated cells, placing particular emphasis on the ways in which FOXC2 promotes key hallmarks of cancer progression. Finally, we bring attention to important issues related to the oncogenic dysregulation of FOXC2 that must be addressed going forward in order to improve our understanding of FOXC2-mediated cancer progression and to guide prognostic and therapeutic applications of this knowledge in clinical settings.
Collapse
Affiliation(s)
- Kristian M Hargadon
- Hargadon Laboratory, Department of Biology, Hampden-Sydney College, Hampden-Sydney, VA, 23943, USA.
| | - Travis B Goodloe
- Hargadon Laboratory, Department of Biology, Hampden-Sydney College, Hampden-Sydney, VA, 23943, USA
| | - Nathaniel D Lloyd
- Hargadon Laboratory, Department of Biology, Hampden-Sydney College, Hampden-Sydney, VA, 23943, USA
| |
Collapse
|
4
|
Recouvreux MS, Miao J, Gozo MC, Wu J, Walts AE, Karlan BY, Orsulic S. FOXC2 Promotes Vasculogenic Mimicry in Ovarian Cancer. Cancers (Basel) 2022; 14:4851. [PMID: 36230774 PMCID: PMC9564305 DOI: 10.3390/cancers14194851] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
FOXC2 is a forkhead family transcription factor that plays a critical role in specifying mesenchymal cell fate during embryogenesis. FOXC2 expression is associated with increased metastasis and poor survival in various solid malignancies. Using in vitro and in vivo assays in mouse ovarian cancer cell lines, we confirmed the previously reported mechanisms by which FOXC2 could promote cancer growth, metastasis, and drug resistance, including epithelial-mesenchymal transition, stem cell-like differentiation, and resistance to anoikis. In addition, we showed that FOXC2 expression is associated with vasculogenic mimicry in mouse and human ovarian cancers. FOXC2 overexpression increased the ability of human ovarian cancer cells to form vascular-like structures in vitro, while inhibition of FOXC2 had the opposite effect. Thus, we present a novel mechanism by which FOXC2 might contribute to cancer aggressiveness and poor patient survival.
Collapse
Affiliation(s)
- Maria Sol Recouvreux
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jiangyong Miao
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Maricel C. Gozo
- Women’s Cancer Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jingni Wu
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ann E. Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Beth Y. Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sandra Orsulic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90095, USA
| |
Collapse
|
5
|
Peres J, Damerell V, Chauhan J, Popovic A, Desprez PY, Galibert MD, Goding CR, Prince S. TBX3 Promotes Melanoma Migration by Transcriptional Activation of ID1, which Prevents Activation of E-Cadherin by MITF. J Invest Dermatol 2021; 141:2250-2260.e2. [PMID: 33744299 DOI: 10.1016/j.jid.2021.02.740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/22/2021] [Accepted: 02/09/2021] [Indexed: 01/22/2023]
Abstract
In melanoma, a phenotype switch from proliferation to invasion underpins metastasis, the major cause of melanoma-associated death. The transition from radial to vertical growth phase (invasive) melanoma is characterized by downregulation of both E-cadherin (CDH1) and MITF and upregulation of the key cancer-associated gene TBX3 and the phosphatidylinositol 3 kinase signaling pathway. Yet, whether and how these diverse events are linked remains poorly understood. Here, we show that TBX3 directly promotes expression of ID1, a dominant-negative regulator of basic helix-loop-helix transcription factors, and that ID1 decreases MITF binding and upregulation of CDH1. Significantly, we show that TBX3 activation of ID1 is necessary for TBX3 to enhance melanoma cell migration, and the mechanistic links between TBX3, ID1, MITF, and invasion revealed here are reflected in their expression in human melanomas. Our results reveal that melanoma migration is promoted through a TBX3-ID1-MITF-E-cadherin axis and that ID1-mediated repression of MITF activity may reinforce maintenance of an MITFLow phenotype associated with disease progression and therapy resistance.
Collapse
Affiliation(s)
- Jade Peres
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Victoria Damerell
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jagat Chauhan
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Oxford United Kingdom
| | - Ana Popovic
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Pierre-Yves Desprez
- California Pacific Medical Center, Research Institute, San Francisco, California, USA
| | - Marie-Dominique Galibert
- IGDR (Institut de Génétique et Développement de Rennes) - UMR6290, CNRS, University of Rennes, Rennes, France; Department of Molecular Genetics and Genomics, Hospital University of Rennes (CHU Rennes), Rennes, France
| | - Colin R Goding
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Oxford United Kingdom
| | - Sharon Prince
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
6
|
Khalili-Tanha G, Moghbeli M. Long non-coding RNAs as the critical regulators of doxorubicin resistance in tumor cells. Cell Mol Biol Lett 2021; 26:39. [PMID: 34425750 PMCID: PMC8381522 DOI: 10.1186/s11658-021-00282-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/17/2021] [Indexed: 12/16/2022] Open
Abstract
Resistance against conventional chemotherapeutic agents is one of the main reasons for tumor relapse and poor clinical outcomes in cancer patients. Various mechanisms are associated with drug resistance, including drug efflux, cell cycle, DNA repair and apoptosis. Doxorubicin (DOX) is a widely used first-line anti-cancer drug that functions as a DNA topoisomerase II inhibitor. However, DOX resistance has emerged as a large hurdle in efficient tumor therapy. Furthermore, despite its wide clinical application, DOX is a double-edged sword: it can damage normal tissues and affect the quality of patients’ lives during and after treatment. It is essential to clarify the molecular basis of DOX resistance to support the development of novel therapeutic modalities with fewer and/or lower-impact side effects in cancer patients. Long non-coding RNAs (lncRNAs) have critical roles in the drug resistance of various tumors. In this review, we summarize the state of knowledge on all the lncRNAs associated with DOX resistance. The majority are involved in promoting DOX resistance. This review paves the way to introducing an lncRNA panel marker for the prediction of the DOX response and clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Ghazaleh Khalili-Tanha
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Tsang ES, Topham JT, Karasinska JM, Lee MKC, Williamson LM, Mendis S, Denroche RE, Jang GH, Kalloger SE, Moore RA, Mungall AJ, Bathe OF, Tang PA, Notta F, Wilson JM, Laskin J, O'Kane GM, Knox JJ, Goodwin RA, Loree JM, Jones SJM, Marra MA, Gallinger S, Schaeffer DF, Renouf DJ. Delving into Early-onset Pancreatic Ductal Adenocarcinoma: How Does Age Fit In? Clin Cancer Res 2020; 27:246-254. [PMID: 32958704 DOI: 10.1158/1078-0432.ccr-20-1042] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 07/15/2020] [Accepted: 09/14/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE With the rising incidence of early-onset pancreatic cancer (EOPC), molecular characteristics that distinguish early-onset pancreatic ductal adenocarcinoma (PDAC) tumors from those arising at a later age are not well understood. EXPERIMENTAL DESIGN We performed bioinformatic analysis of genomic and transcriptomic data generated from 269 advanced (metastatic or locally advanced) and 277 resectable PDAC tumor samples. Patient samples were stratified into EOPC (age of onset ≤55 years; n = 117), intermediate (age of onset 55-70 years; n = 264), and average (age of onset ≥70 years; n = 165) groups. Frequency of somatic mutations affecting genes commonly implicated in PDAC, as well as gene expression patterns, were compared between EOPC and all other groups. RESULTS EOPC tumors showed significantly lower frequency of somatic single-nucleotide variant (SNV)/insertions/deletions (indel) in CDKN2A (P = 0.0017), and were more likely to achieve biallelic mutation of CDKN2A through homozygous copy loss as opposed to heterozygous copy loss coupled with a loss-of-function SNV/indel mutation, the latter of which was more common for tumors with later ages of onset (P = 1.5e-4). Transcription factor forkhead box protein C2 (FOXC2) was significantly upregulated in EOPC tumors (P = 0.032). Genes significantly correlated with FOXC2 in PDAC samples were enriched for gene sets related to epithelial-to-mesenchymal transition (EMT) and included VIM (P = 1.8e-8), CDH11 (P = 6.5e-5), and CDH2 (P = 2.4e-2). CONCLUSIONS Our comprehensive analysis of sequencing data generated from a large cohort of PDAC patient samples highlights a distinctive pattern of biallelic CDKN2A mutation in EOPC tumors. Increased expression of FOXC2 in EOPC, with the correlation between FOXC2 and EMT pathways, represents novel molecular characteristics of EOPC.See related commentary by Lou, p. 8.
Collapse
Affiliation(s)
- Erica S Tsang
- BC Cancer, Vancouver, British Columbia, Canada.,Pancreas Centre BC, Vancouver, British Columba, Canada
| | | | | | - Michael K C Lee
- BC Cancer, Vancouver, British Columbia, Canada.,Pancreas Centre BC, Vancouver, British Columba, Canada
| | - Laura M Williamson
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Shehara Mendis
- BC Cancer, Vancouver, British Columbia, Canada.,Pancreas Centre BC, Vancouver, British Columba, Canada
| | | | - Gun Ho Jang
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | - Richard A Moore
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Andrew J Mungall
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | | | | | - Faiyaz Notta
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Julie M Wilson
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | | | - Jennifer J Knox
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Rachel A Goodwin
- The Ottawa Hospital Cancer Centre, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Jonathan M Loree
- BC Cancer, Vancouver, British Columbia, Canada.,Pancreas Centre BC, Vancouver, British Columba, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | | | - David F Schaeffer
- Pancreas Centre BC, Vancouver, British Columba, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Daniel J Renouf
- BC Cancer, Vancouver, British Columbia, Canada. .,Pancreas Centre BC, Vancouver, British Columba, Canada
| |
Collapse
|
8
|
Yan M, Gao H, Lv Z, Liu Y, Zhao S, Gong W, Liu W. Circular RNA PVT1 promotes metastasis via regulating of miR-526b/FOXC2 signals in OS cells. J Cell Mol Med 2020; 24:5593-5604. [PMID: 32249539 PMCID: PMC7214167 DOI: 10.1111/jcmm.15215] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 12/18/2022] Open
Abstract
As a class of covalently closed non-coding RNAs, circular RNAs (circRNAs) are key regulators in various malignancies including osteosarcoma (OS). In the present study, we found that circular RNA PVT1 (circPVT1) was up-regulated in OS and correlated with poor prognosis of patients with OS. Functionally, we showed that knockdown of circPVT1 suppressed OS cells metastasis. In addition, we found that (forkhead box C2) FOXC2 was a downstream gene in circPVT1-mediated metastasis in OS cells. We demonstrated that circPVT1 promoted OS cells metastasis via post-transcriptionally regulating of FOXC2. Furthermore, we revealed that microRNA 526b (miR-526b) was a key bridge which connected circPVT1 and FOXC2. We showed that miR-526b was down-regulated in OS tissue and cell lines. Through a transwell assay, we found that miR-526b suppressed OS cells metastasis by targeting of FOXC2. We also showed that miR-526b targeted circPVT1 via similar mircoRNA response elements (MREs) as it did for FOXC2. Finally, we proved that circPVT1 decoyed miR-526b to promote FOXC2-mediated metastasis in OS cells. In brief, our current study demonstrated that circPVT1, functioning as an oncogene, promotes OS cells metastasis via regulation of FOXC2 by acting as a ceRNA of miR-526b. CircPVT1/miR-526b/FOXC2 axis might be a novel target in molecular treatment of OS.
Collapse
Affiliation(s)
- Ming Yan
- Department of Spinal Surgery, The First Hospital of Jilin University, Changchun, P.R.China
| | - Hang Gao
- Department of Bone and Joint Surgery, The First Hospital of Jilin University, Changchun, P.R.China
| | - Zhenshan Lv
- Department of Spinal Surgery, The First Hospital of Jilin University, Changchun, P.R.China
| | - Ying Liu
- Department of Spinal Surgery, The First Hospital of Jilin University, Changchun, P.R.China
| | - Song Zhao
- Department of Spinal Surgery, The First Hospital of Jilin University, Changchun, P.R.China
| | - Weiquan Gong
- Department of Spinal Surgery, The First Hospital of Jilin University, Changchun, P.R.China
| | - Wei Liu
- Department of Spinal Surgery, The First Hospital of Jilin University, Changchun, P.R.China
| |
Collapse
|
9
|
Li S, Pradhan L, Ashur S, Joshi A, Nam HJ. Crystal Structure of FOXC2 in Complex with DNA Target. ACS OMEGA 2019; 4:10906-10914. [PMID: 31460188 PMCID: PMC6648891 DOI: 10.1021/acsomega.9b00756] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/22/2019] [Indexed: 06/10/2023]
Abstract
Forkhead transcription factor C2 (FOXC2) is a transcription factor regulating vascular and lymphatic development, and its mutations are linked to lymphedema-distichiasis syndrome. FOXC2 is also a crucial regulator of the epithelial-mesenchymal transition processes essential for tumor metastasis. Here, we report the crystal structure of the FOXC2-DNA-binding domain in complex with its cognate DNA. The crystal structure provides the basis of DNA sequence recognition by FOXC2 for the T/CAAAC motif. Helix 3 makes the majority of the DNA-protein interactions and confers the DNA sequence specificity. The computational energy calculation results also validate the structural observations. The FOXC2 and DNA complex structure provides a detailed picture of protein and DNA interactions, which allows us to predict its DNA recognition specificity and impaired functions in mutants identified in human patients.
Collapse
|
10
|
Guha Majumdar A, Subramanian M. Hydroxychavicol from Piper betle induces apoptosis, cell cycle arrest, and inhibits epithelial-mesenchymal transition in pancreatic cancer cells. Biochem Pharmacol 2019; 166:274-291. [PMID: 31154000 DOI: 10.1016/j.bcp.2019.05.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/28/2019] [Indexed: 12/30/2022]
Abstract
Pancreatic cancer is a major cause of cancer-related mortality around the world. Currently, options for diagnosis and treatment are extremely limited, which culminates in a very high mortality rate. Intensive research spanning more than four decades has met several roadblocks in terms of improvement in overall survival. In this study, we have evaluated the effect of Hydroxychavicol (HC), a naturally occurring and abundantly isolatable allylarene from Piper betle leaves on pancreatic cancer cells. Our investigation reveals that HC inhibits proliferation and epithelial-mesenchymal transition (EMT) in pancreatic cancer cells. HC induces DNA damage, as evidenced by γ-H2AX, 53BP1 induction and comet assay, which further results in mitotic catastrophe and apoptosis. The apoptosis induced by HC is JNK pathway-dependent and caspase-mediated. HC also inhibits migration and invasion of pancreatic cancer cells via a generalized repression of genes involved in EMT. A quantitative real time PCR-based array revealed at least 14 different genes to be differentially expressed upon HC treatment in pancreatic cancer cells. These results show significant potential of HC as an anticancer agent against pancreatic cancer.
Collapse
Affiliation(s)
- Ananda Guha Majumdar
- Bio-Organic Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400 085, India
| | - Mahesh Subramanian
- Bio-Organic Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400 085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400 094, India.
| |
Collapse
|
11
|
Xu YJ, Li P, Zheng L, Guo FX, Kang CM, Ding L, Xu BM, Lu JB, Xiao L, Wu Q, Lu ZF, Bai HL, Hu YW, Wang Q. Forkhead Box C2 Attenuates Lipopolysaccharide-Induced Cell Adhesion via Suppression of Intercellular Adhesion Molecule-1 Expression in Human Umbilical Vein Endothelial Cells. DNA Cell Biol 2019; 38:583-591. [PMID: 30994379 DOI: 10.1089/dna.2019.4663] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Atherosclerosis is a chronic vascular inflammatory disease that involves diverse cell types and circulating regulatory factors, including intercellular adhesion molecule (ICAM)-1, a proinflammatory cytokine. Lipopolysaccharides (LPS) increase ICAM-1 expression and promote cell adhesion, but the mechanism is not clear. We found that LPS induced time- and dose-regulated upregulation of ICAM-1 expression and downregulation of forkhead box protein C2 (Foxc2) expression in human umbilical vein endothelial cells (HUVECs). Overexpression of Foxc2 significantly inhibited both LPS-induced ICAM-1 expression in HUVECs and LPS-induced adhesion of THP-1 cells to HUVECs. Foxc2 siRNA dramatically increased both LPS-induced ICAM-1 expression and LPS-induced adhesion of THP-1 human monocytes cells to HUVECs. We conclude that Foxc2 inhibited LPS-induced adhesion of THP-1 cells to HUVECs by suppressing ICAM-1 expression in HUVECs.
Collapse
Affiliation(s)
- Yuan-Jun Xu
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Pan Li
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Zheng
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Feng-Xia Guo
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chun-Min Kang
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Li Ding
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bang-Ming Xu
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing-Bo Lu
- 2 Department of Vascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Xiao
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qian Wu
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhi-Feng Lu
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Huan-Lan Bai
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yan-Wei Hu
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qian Wang
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
12
|
Abstract
INTRODUCTION Arachnoid cysts are commonly considered congenital lesions, but this has not been proven. With the development of neuroimaging and DNA testing technology, more cases of familial arachnoid cysts have been reported. Herein, we review such cases. MATERIALS AND METHODS The PubMed, Embase, and Web of Science databases were searched for case reports of arachnoid cysts published through April 2018. Case reports were included only if two or more related patients were diagnosed with an arachnoid cyst by neuroimaging or intraoperatively. For each report, the following data were extracted: first author name, date of publication, number of families, number of patients, location of the arachnoid cysts, patient age, patient sex, and genetic mutations and associated disease. RESULTS Our searches identified 33 case reports involving 35 families and 115 patients. The locations of arachnoid cysts were similar in 25 of the 35 families. Spinal extradural arachnoid cysts were reported most often, followed by arachnoid cysts in the middle fossa and posterior fossa. A left-sided predominance was noticed for arachnoid cysts of the middle fossa. Mutation of the FOXC2 gene was reported most often, and arachnoid cysts may be associated with mutations on chromosome 16. CONCLUSIONS Although the origin of arachnoid cysts is believed to have a genetic component by some researchers, the genes associated with arachnoid cysts remain unknown. Unfortunately, the evidence remains insufficient.
Collapse
|
13
|
Elie BT, Fernández-Gallardo J, Curado N, Cornejo MA, Ramos JW, Contel M. Bimetallic titanocene-gold phosphane complexes inhibit invasion, metastasis, and angiogenesis-associated signaling molecules in renal cancer. Eur J Med Chem 2018; 161:310-322. [PMID: 30368130 DOI: 10.1016/j.ejmech.2018.10.034] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/11/2018] [Accepted: 10/14/2018] [Indexed: 01/08/2023]
Abstract
Following promising recent in vitro and in vivo studies of the anticancer efficacies of heterometallic titanocene-gold chemotherapeutic candidates against renal cancer, we report here on the synthesis, characterization, stability studies and biological evaluation of a new titanocene complex containing a gold-triethylphosphane fragment [(η-C5H5)2TiMe(μ-mba)Au(PEt3)] (4) Titanofin. The compound is more stable in physiological fluid than those previously reported, and it is highly cytotoxic against a line of human clear cell renal carcinoma. We describe here preliminary mechanistic data for this compound and previously reported [(η-C5H5)2TiMe(μ-mba)Au(PPh3)] (2) Titanocref which displayed remarkable activity in an in vivo mouse model. Mechanistic studies were carried out in the human clear cell renal carcinoma Caki-1 line for the bimetallic compounds [(η-C5H5)2TiMe(μ-mba)Au(PR3)] (PR3 = PPh32 Titanocref and PEt34 Titanofin), the two monometallic gold derivatives [Au(Hmba)(PR3)] (PR3 = PPh31 cref; PEt33 fin), titanocene dichloride and Auranofin as controls. These studies indicate that bimetallic compounds Titanocref (2) and Titanofin (4) are more cytotoxic than gold monometallic derivatives (1 and 3) and significantly more cytotoxic than titanocene dichloride while being quite selective. Titanocref (2) and Titanofin (4) inhibit migration, invasion, and angiogenic assembly along with molecular markers associated with these processes such as prometastatic IL(s), MMP(s), TNF-α, and proangiogenic VEGF, FGF-basic. The bimetallic compounds also strongly inhibit the mitochondrial protein TrxR often overexpressed in cancer cells evading apoptosis and also inhibit FOXC2, PECAM-1, and HIF-1α whose overexpression is linked to resistance to genotoxic chemotherapy. In summary, bimetallic titanocene-gold phosphane complexes (Titanocref 2 and Titanofin 4) are very promising candidates for further preclinical evaluations for the treatment of renal cancer.
Collapse
Affiliation(s)
- Benelita T Elie
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA; Biology PhD Programs, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY, 10016, USA
| | - Jacob Fernández-Gallardo
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
| | - Natalia Curado
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
| | - Mike A Cornejo
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
| | - Joe W Ramos
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, USA
| | - María Contel
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA; Biology PhD Programs, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY, 10016, USA; Chemistry PhD Programs, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY, 10016, USA; Biochemistry PhD Programs, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY, 10016, USA; Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, USA.
| |
Collapse
|
14
|
Ahn J, Han KS, Heo JH, Bang D, Kang YH, Jin HA, Hong SJ, Lee JH, Ham WS. FOXC2 and CLIP4 : a potential biomarker for synchronous metastasis of ≤7-cm clear cell renal cell carcinomas. Oncotarget 2018; 7:51423-51434. [PMID: 27283491 PMCID: PMC5239485 DOI: 10.18632/oncotarget.9842] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/20/2016] [Indexed: 12/15/2022] Open
Abstract
Renal cell carcinomas (RCC) smaller than 7-cm are heterogeneous and exhibit metastatic potential in approximately 15% of cases. Although large-scale characterization of mutations in clear cell RCC (ccRCC), the most common RCC subtype, has been established, the genetic alterations related to ≤7-cm ccRCCs undergoing synchronous metastasis are poorly understood. To discover biomarkers that can be used to estimate the risk of synchronous metastasis in these ccRCC patients, we performed whole exome sequencing on the formalin-fixed paraffin-embedded (FFPE) samples of 10 ccRCC patients with ≤7-cm tumors and synchronous metastasis and expanded our study using The Cancer Genome Atlas (TCGA) ccRCC dataset (n = 201). Recurrent mutations were selected according to functional prediction and statistical significance. Mutations in three candidate genes, RELN (1 out of 10), FOXC2 (1 out of 10), and CLIP4 (2 out of 10) were found in expanded analysis using a TCGA cohort. Furthermore, siRNA-mediated target gene knockdown (FOXC2 and CLIP4) and overexpression (RELN) assays showed that FOXC2 and CLIP4 significantly increased cell migration and viability in ccRCCs. Our study demonstrated that FOXC2 and CLIP4 activity correlates to the presence of ≤7-cm ccRCCs with synchronous metastasis and may be potential molecular predictors of synchronous metastasis of ≤7-cm ccRCCs.
Collapse
Affiliation(s)
- Jinwoo Ahn
- Department of Chemistry, Yonsei University, Seoul, Korea
| | - Kyung Seok Han
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Jun Hyeok Heo
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Duhee Bang
- Department of Chemistry, Yonsei University, Seoul, Korea
| | - You Hyun Kang
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun A Jin
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Joon Hong
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hyun Lee
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Won Sik Ham
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
15
|
Wang T, Zheng L, Wang Q, Hu YW. Emerging roles and mechanisms of FOXC2 in cancer. Clin Chim Acta 2018; 479:84-93. [PMID: 29341903 DOI: 10.1016/j.cca.2018.01.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/12/2018] [Accepted: 01/12/2018] [Indexed: 12/20/2022]
Abstract
Forkhead box protein C2 (FOXC2), a transcription factor of the forkhead/winged-helix family, is required for embryonic and prenatal development. FOXC2 acts as a crucial modulator during both angiogenesis and lymphangiogenesis via multiple angiogenic and lymphangiogenic pathways, respectively. Although recent studies have shed light on the emerging role of FOXC2 in cancer, very little is known about the precise underlying mechanisms. The purpose of this review is to summarize the current understanding of FOXC2 and provide potential mechanistic explanations of the relationship between FOXC2 and cancer, as well as discuss the prospect for future research in the promising prognostic value of FOXC2 in cancer.
Collapse
Affiliation(s)
- Teng Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Lei Zheng
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Qian Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yan-Wei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
16
|
Wang J, Yue X. Role and importance of the expression of transcription factor FOXC2 in cervical cancer. Oncol Lett 2017; 14:6627-6631. [PMID: 29151910 PMCID: PMC5678244 DOI: 10.3892/ol.2017.7004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 06/08/2017] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to investigate the relationship between the expression of transcription factor forkhead box C2 (FOXC2) and the clinical features of cervical cancer. A total of 66 patients with cervical cancer, 42 patients with cervical intraepithelial neoplasia (CIN) and 25 patients with cervical inflammation were enrolled. The positive expression rates and expression levels of mRNA of FOXC2, E-cadherin, N-cadherin, vascular endothelial growth factor (VEGF), stromal cell-derived factor-1 (SDF-1), Notch protein and lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1) in cervical tissues were detected using immunohistochemistry and RT-PCR. The positive expression rates and expression levels of mRNA of FOXC2, N-cadherin, VEGF, SDF-1, Notch and LYVE-1 in cervical cancer were significantly higher than those in CIN, and those in the inflammatory tissues were the lowest, while the positive expression rate of E-cadherin in cervical cancer was lower than that in CIN, and that in the inflammatory tissues was the highest (P<0.05). The positive expression rates of FOXC2, N-cadherin, VEGF, SDF-1, Notch and LYVE-1 in patients with cervical cancer [human papillomavirus (HPV) positive, squamous cell carcinoma, Stages III–IV, maximal diameter ≥3.8 cm and low differentiation] were increased, and the positive expression rate of E-cadherin was decreased (P<0.05). Correlation analysis revealed that FOXC2 was positively correlated with the positive expression rates of N-cadherin, VEGF, SDF-1, Notch and LYVE-1, and negatively correlated with E-cadherin (P<0.05). In conclusion, the high expression of FOXC2 is correlated with the HPV infection, pathological pattern, clinical stage, tumor diameter and differentiation grade of cervical cancer, which may be involved in the epithelial-mesenchymal transition, vascular and matrix formation, Notch signaling pathway and lymphangiogenesis.
Collapse
Affiliation(s)
- Jing Wang
- Department of Obstetrics and Gynecology, Linyi Hospital of Τraditional Chinese Medicine, Linyi, Shandong 276000, P.R. China
| | - Xiujuan Yue
- Department of Obstetrics, Linyi Hospital of Τraditional Chinese Medicine, Linyi, Shandong 276000, P.R. China
| |
Collapse
|
17
|
Werden SJ, Sphyris N, Sarkar TR, Paranjape AN, LaBaff AM, Taube JH, Hollier BG, Ramirez-Peña EQ, Soundararajan R, den Hollander P, Powell E, Echeverria GV, Miura N, Chang JT, Piwnica-Worms H, Rosen JM, Mani SA. Phosphorylation of serine 367 of FOXC2 by p38 regulates ZEB1 and breast cancer metastasis, without impacting primary tumor growth. Oncogene 2016; 35:5977-5988. [PMID: 27292262 PMCID: PMC5114155 DOI: 10.1038/onc.2016.203] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 03/31/2016] [Accepted: 04/22/2016] [Indexed: 01/02/2023]
Abstract
Metastatic competence is contingent upon the aberrant activation of a latent embryonic program, known as the epithelial-mesenchymal transition (EMT), which bestows stem cell properties as well as migratory and invasive capabilities upon differentiated tumor cells. We recently identified the transcription factor FOXC2 as a downstream effector of multiple EMT programs, independent of the EMT-inducing stimulus, and as a key player linking EMT, stem cell traits and metastatic competence in breast cancer. As such, FOXC2 could serve as a potential therapeutic target to attenuate metastasis. However, as FOXC2 is a transcription factor, it is difficult to target by conventional means such as small-molecule inhibitors. Herein, we identify the serine/threonine-specific kinase p38 as a druggable upstream regulator of FOXC2 stability and function that elicits phosphorylation of FOXC2 at serine 367 (S367). Using an orthotopic syngeneic mouse tumor model, we make the striking observation that inhibition of p38-FOXC2 signaling selectively attenuates metastasis without impacting primary tumor growth. In this model, circulating tumor cell numbers are significantly reduced in mice treated with the p38 inhibitor SB203580, relative to vehicle-treated counterparts. Accordingly, genetic or pharmacological inhibition of p38 decreases FOXC2 protein levels, reverts the EMT phenotype and compromises stem cell attributes in vitro. We also identify the EMT-regulator ZEB1-known to directly repress E-cadherin/CDH1-as a downstream target of FOXC2, critically dependent on its activation by p38. Consistent with the notion that activation of the p38-FOXC2 signaling axis represents a critical juncture in the acquisition of metastatic competence, the phosphomimetic FOXC2(S367E) mutant is refractory to p38 inhibition both in vitro and in vivo, whereas the non-phosphorylatable FOXC2(S367A) mutant fails to elicit EMT and upregulate ZEB1. Collectively, our data demonstrate that FOXC2 regulates EMT, stem cell traits, ZEB1 expression and metastasis in a p38-dependent manner, and attest to the potential utility of p38 inhibitors as antimetastatic agents.
Collapse
Affiliation(s)
- S J Werden
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - N Sphyris
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - T R Sarkar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - A N Paranjape
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - A M LaBaff
- Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J H Taube
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - B G Hollier
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - E Q Ramirez-Peña
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - R Soundararajan
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - P den Hollander
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - E Powell
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - G V Echeverria
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - N Miura
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - J T Chang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - H Piwnica-Worms
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J M Rosen
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - S A Mani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Metastasis Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for Stem Cell and Developmental Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
18
|
Qiang Y, Chen Z. Epithelial mesenchymal transition related molecular markers and invasion and metastasis of cholangiocarcinoma. Shijie Huaren Xiaohua Zazhi 2015; 23:4051-4059. [DOI: 10.11569/wcjd.v23.i25.4051] [Citation(s) in RCA: 1] [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
Tumor metastasis is a major cause of death in patients with solid tumors. Epithelial mesenchymal transition (EMT) is a process in which the epithelial cells are transformed into the stroma cells. This process is accompanied by changes in gene expression and cell phenotype, which are often activated during tumor invasion and metastasis. Cholangiocarcinoma is a kind of malignancy originating from the bile duct epithelium, and its main biological characteristics are early invasion, metastasis and recurrence. The research of cholangiocarcinoma metastasis could provide a theoretical basis for the development of new treatment strategies to manage this malignancy. This paper reviews the roles of EMT related molecular markers metastasis in the invasion and metastasis of cholangiocarcinoma.
Collapse
|
19
|
Sun TY, Xie HJ, Li Z, Kong LF. Expression of FOXC2 in renal cell carcinoma and its relationship to clinical pathological features. Int J Clin Exp Med 2015; 8:13388-13392. [PMID: 26550271 PMCID: PMC4612956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 08/06/2015] [Indexed: 06/05/2023]
Abstract
OBJECTIVE This study aimed to investigate expression level of FOXC2 and its relationship to clinical pathological features of renal cell carcinoma (RCC). METHODS The expression levels of FOXC2 in RCC tissues and normal renal tissues (62 samples, respectively) were detected by immunohistochemistry and PCR Array. Statistics analyses were done with SPSS to compare the differences between RCC tissues and normal renal tissue, and to explore the relationship between the expression level of FOXC2 and the clinical pathological features of RCC. RESULTS Expression level of FOXC2 in RCC tissues was significantly higher than in normal renal tissues, and other related cancer genes also highly expressed in RCC tissues. FOXC2 expression was positively associated with clinical stage and pathological grade (P < 0.05), but not significantly related to the gender and age (P > 0.05). CONCLUSION The expression of FOXC2 in renal cell carcinoma was significantly higher than that in normal renal tissues. It is suggested that FOXC2 might play a crucial role in the occurrence and development of RCC and could be an important prognostic indicator for clinical therapy.
Collapse
Affiliation(s)
- Ting-Yi Sun
- Department of Pathology, Henan Provincial People's Hospital Zhengzhou, China
| | - Hong-Jian Xie
- Department of Pathology, Henan Provincial People's Hospital Zhengzhou, China
| | - Zhen Li
- Department of Pathology, Henan Provincial People's Hospital Zhengzhou, China
| | - Ling-Fei Kong
- Department of Pathology, Henan Provincial People's Hospital Zhengzhou, China
| |
Collapse
|
20
|
Zhu KP, Zhang CL, Shen GQ, Zhu ZS. Long noncoding RNA expression profiles of the doxorubicin-resistant human osteosarcoma cell line MG63/DXR and its parental cell line MG63 as ascertained by microarray analysis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:8754-8773. [PMID: 26464619 PMCID: PMC4583851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 07/21/2015] [Indexed: 06/05/2023]
Abstract
Long non-coding RNAs (lncRNAs) are emerging in molecular biology as crucial regulators of cancer. The efficacy of doxorubicin--based chemotherapy in osteosarcoma (OS) is usually limited by acquired drug resistance. To explore the mechanism of chemoresistance of OS in terms of lncRNA, using a human lncRNA-mRNA combined microarray, we identified 3,465 lncRNAs (1,761 up and 1,704 down) and 3,278 mRNAs (1,607 up and 1,671 down) aberrantly expressed in all three sets of doxorubicin-resistant MG63/DXR and their paired parental MG63 cells (fold-change >2.0, P<0.05 and FDR <0.05). Fifteen randomly selected lncRNAs were dysregulated in MG63/DXR cells relative to MG63 cells by qRT-PCR detection, which were consistent with our microarray data. Bioinformatics analysis identified that classical genes and pathways involved in cell proliferation, apoptosis, and drug metabolism were differently expressed in these cell lines. A lncRNA-mRNA co-expression network identified lncRNAs, including ENST00000563280 and NR-036444, may play a critical role in doxorubicin-resistance of OS by interacting with important genes such as ABCB1, HIF1A and FOXC2. Besides, we found that lncRNA ENST00000563280 was distinctly increased in specimens of OS patients with a poor chemoresponse compared to those with a good chemoresponse and the patients of lower expression of it may survive longer than those of higher expression, which suggest that it may serve as a biomarker to predict the chemoresponse and prognosis of osteosarcoma patients. These results provide important insights about the lncRNAs involved in osteosarcoma chemoresistance and lay a solid foundation for uncovering the mechanism ultimately.
Collapse
Affiliation(s)
- Kun-Peng Zhu
- Department of Orthopaedic Surgery, The Sixth People’s Hospital Affiliated to Shanghai Jiaotong UniversityShanghai 200233, PR China
| | - Chun-Lin Zhang
- Department of Orthopaedic Surgery, Shanghai Tenth People’s Hospital Affiliated to Tongji UniversityShanghai 200072, PR China
| | - Guo-Qi Shen
- Department of Orthopaedic Surgery, Shanghai Sixth People’s Hospital, Soochow UniversityShanghai 200233, PR China
| | - Zhong-Sheng Zhu
- Department of Orthopaedic Surgery, Shanghai Tenth People’s Hospital Affiliated to Tongji UniversityShanghai 200072, PR China
| |
Collapse
|
21
|
Zhao H, Zhou W, Yao Z, Wan Y, Cao J, Zhang L, Zhao J, Li H, Zhou R, Li B, Wei G, Zhang Z, French CA, Dekker JD, Yang Y, Fisher SE, Tucker HO, Guo X. Foxp1/2/4 regulate endochondral ossification as a suppresser complex. Dev Biol 2015; 398:242-54. [PMID: 25527076 PMCID: PMC4342236 DOI: 10.1016/j.ydbio.2014.12.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 12/03/2014] [Accepted: 12/04/2014] [Indexed: 12/16/2022]
Abstract
Osteoblast induction and differentiation in developing long bones is dynamically controlled by the opposing action of transcriptional activators and repressors. In contrast to the long list of activators that have been discovered over past decades, the network of repressors is not well-defined. Here we identify the expression of Foxp1/2/4 proteins, comprised of Forkhead-box (Fox) transcription factors of the Foxp subfamily, in both perichondrial skeletal progenitors and proliferating chondrocytes during endochondral ossification. Mice carrying loss-of-function and gain-of-function Foxp mutations had gross defects in appendicular skeleton formation. At the cellular level, over-expression of Foxp1/2/4 in chondroctyes abrogated osteoblast formation and chondrocyte hypertrophy. Conversely, single or compound deficiency of Foxp1/2/4 in skeletal progenitors or chondrocytes resulted in premature osteoblast differentiation in the perichondrium, coupled with impaired proliferation, survival, and hypertrophy of chondrocytes in the growth plate. Foxp1/2/4 and Runx2 proteins interacted in vitro and in vivo, and Foxp1/2/4 repressed Runx2 transactivation function in heterologous cells. This study establishes Foxp1/2/4 proteins as coordinators of osteogenesis and chondrocyte hypertrophy in developing long bones and suggests that a novel transcriptional repressor network involving Foxp1/2/4 may regulate Runx2 during endochondral ossification.
Collapse
Affiliation(s)
- Haixia Zhao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wenrong Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhengju Yao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yong Wan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jingjing Cao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lingling Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianzhi Zhao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hanjun Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Rujiang Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Gang Wei
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS), Shanghai 200032, China
| | - Zhenlin Zhang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated the Sixth People׳s Hospital, Shanghai, China
| | - Catherine A French
- Champalimaud Neuroscience Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Joseph D Dekker
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Yingzi Yang
- Developmental Genetics Section, National Human Genome Research Institute, NIH, MD 20892, USA
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Haley O Tucker
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Xizhi Guo
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
22
|
Imayama N, Yamada SI, Yanamoto S, Naruse T, Matsushita Y, Takahashi H, Seki S, Fujita S, Ikeda T, Umeda M. FOXC2 expression is associated with tumor proliferation and invasion potential in oral tongue squamous cell carcinoma. Pathol Oncol Res 2015; 21:783-91. [PMID: 25573594 DOI: 10.1007/s12253-014-9891-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 12/23/2014] [Indexed: 02/08/2023]
Abstract
Forkhead box protein C2 (FOXC2) is a gene encoding a transcription factor that controls the generation of mesodermal tissue including vascular and lymphatic tissues. FOXC2 has previously been associated with EMT and tumor angiogenesis in various cancers. Moreover, a relationship between the expression of FOXC2 and poor prognosis has been reported in various cancers. We herein examined the clinicopathological significance of FOXC2 in oral tongue squamous cell carcinoma (OTSCC) and attempted to clarify the function of FOXC2 in OTSCC cell lines in vitro. The overexpression of FOXC2 was more frequent in cancers with higher grades according to the pattern of invasion (grade 4 vs. 1-3; p < 0.05). A correlation was observed between the expression of FOXC2 and that of VEGF-A and -C (VEGF-A; p < 0.05, VEGF-C; p < 0.001). The high-FOXC2 expression group had a significantly poorer prognosis than that of the low-expression group (p < 0.001). Multivariate analysis indicated that the overexpression of FOXC2 may also be an independent prognostic factor, similar to N classification (N0 vs 1/2; p < 0.05), stage classification (stage I/II vs III/IV; p < 0.05), pattern of invasion (grade 1-3vs 4; p < 0.05), local recurrence (local recurrence (+) vs (-); p < 0.01), and the overexpression of FOXC2 (FOXC2 overexpression (-) vs.(+); p < 0.05). In the OTSCC cell line analysis, the expression of FOXC2 was also associated with proliferation and invasion potential. These results strongly suggest that the overexpression of FOXC2 may be a potent predictor of survival in OTSCC patients.
Collapse
Affiliation(s)
- Naomi Imayama
- Department of Clinical Oral Oncology, Unit of Translational Medicine, Course of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Cui YM, Jiao HL, Ye YP, Chen CM, Wang JX, Tang N, Li TT, Lin J, Qi L, Wu P, Wang SY, He MR, Liang L, Bian XW, Liao WT, Ding YQ. FOXC2 promotes colorectal cancer metastasis by directly targeting MET. Oncogene 2014; 34:4379-90. [PMID: 25381815 DOI: 10.1038/onc.2014.368] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 09/24/2014] [Accepted: 09/30/2014] [Indexed: 12/11/2022]
Abstract
Metastasis is the major cause of death in colorectal cancer (CRC). Although multiple genes have been identified to be responsible for the development of CRC, the molecular changes that enable CRC cells to undergo early local invasion and to form distant metastatic colonies still remain largely unknown. Herein, we investigated the role of Forkhead box protein C2 (FOXC2) and explored the underlying mechanisms in invasion and metastasis of CRC. We show that both high FOXC2 expression and nuclear localization of FOXC2 are significantly correlated with advanced TNM (T=primary tumor; N=regional lymph nodes; M=distant metastasis) stages. FOXC2 enhanced the invasive abilities of CRC cells in vitro and promoted local invasion and distant metastasis in an orthotopic mouse metastatic model of CRC. Microarray analysis revealed that overexpression of FOXC2 increased the proto-oncogene MET tyrosine kinase expression and activated the hepatocyte growth factor (HGF)-MET signaling pathway. Furthermore, luciferase reporter assays and chromatin immunoprecipitation assays revealed that FOXC2 directly associated with MET promoter to increase the transcriptional activity of MET. Inhibition of MET attenuates the invasive phenotype and metastatic potential of FOXC2-overexpressing CRC cells, indicating that MET is a major mediator of FOXC2-promoted metastasis. In addition, FOXC2 expression was positively correlated with MET expression in CRC tissue samples. Our findings suggest that FOXC2 has a crucial role in CRC metastasis by regulating HGF-MET signaling via inducing MET expression, highlighting FOXC2 as a potential therapeutic target for preventing or reducing metastasis in CRC.
Collapse
Affiliation(s)
- Y-M Cui
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - H-L Jiao
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Y-P Ye
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - C-M Chen
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - J-X Wang
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - N Tang
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - T-T Li
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - J Lin
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - L Qi
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - P Wu
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - S-Y Wang
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - M-R He
- Guangdong Provincial Key Laboratory of Gastroenterology and Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - L Liang
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - X-W Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - W-T Liao
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Y-Q Ding
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| |
Collapse
|
24
|
Bai X, Wang J, Guo Y, Pan J, Yang Q, Zhang M, Li H, Zhang L, Ma J, Shi F, Shu W, Wang Y, Leng J. Prostaglandin E2 stimulates β1-integrin expression in hepatocellular carcinoma through the EP1 receptor/PKC/NF-κB pathway. Sci Rep 2014; 4:6538. [PMID: 25289898 PMCID: PMC5377465 DOI: 10.1038/srep06538] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 09/05/2014] [Indexed: 02/07/2023] Open
Abstract
Prostaglandin E2 (PGE2) has been implicated in cell invasion in hepatocellular carcinoma (HCC), via increased β1-integrin expression and cell migration; however, the mechanism remains unclear. PGE2 exerts its effects via four subtypes of the E prostanoid receptor (EP receptor 1–4). The present study investigated the effect of EP1 receptor activation on β1-integrin expression and cell migration in HCC. Cell migration increased by 60% in cells treated with 17-PT-PGE2 (EP1 agonist), which was suppressed by pretreatment with a β1-integrin polyclonal antibody. PGE2 increased β1-integrin expression by approximately 2-fold. EP1 receptor transfection or treatment with 17-PT-PGE2 mimicked the effect of PGE2 treatment. EP1 siRNA blocked PGE2-mediated β1-integrin expression. 17-PT-PGE2 treatment induced PKC and NF-κB activation; PKC and NF-κB inhibitors suppressed 17-PT-PGE2-mediated β1-integrin expression. FoxC2, a β1-integrin transcription factor, was also upregulated by 17-PT-PGE2. NF-κB inhibitor suppressed 17-PT-PGE2-mediated FoxC2 upregulation. Immunohistochemistry showed p65, FoxC2, EP1 receptor and β1-integrin were all highly expressed in the HCC cases. This study suggested that PGE2 upregulates β1-integrin expression and cell migration in HCC cells by activating the PKC/NF-κB signaling pathway. Targeting PGE2/EP1/PKC/NF-κB/FoxC2/β1-integrin pathway may represent a new therapeutic strategy for the prevention and treatment of this cancer.
Collapse
Affiliation(s)
- Xiaoming Bai
- Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Jie Wang
- Department of Pathology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing 210029, P. R. China
| | - Yan Guo
- Institute of Pediatrics, Fourth Clinical Medical College, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Jinshun Pan
- The Center of Metabolic Disease Research, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Qinyi Yang
- Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Min Zhang
- Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Hai Li
- Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Li Zhang
- Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Juan Ma
- Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Feng Shi
- Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Wei Shu
- Department of Periodontal, Institute of Stomatology, The Stomatological Hospital Affiliated to Nanjing Medical University, Nanjing 210029, P. R. China
| | - Yipin Wang
- Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Jing Leng
- Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing 210029, P. R. China
| |
Collapse
|
25
|
Finley SD, Chu LH, Popel AS. Computational systems biology approaches to anti-angiogenic cancer therapeutics. Drug Discov Today 2014; 20:187-97. [PMID: 25286370 DOI: 10.1016/j.drudis.2014.09.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 08/05/2014] [Accepted: 09/29/2014] [Indexed: 01/06/2023]
Abstract
Angiogenesis is an exquisitely regulated process that is required for physiological processes and is also important in numerous diseases. Tumors utilize angiogenesis to generate the vascular network needed to supply the cancer cells with nutrients and oxygen, and many cancer drugs aim to inhibit tumor angiogenesis. Anti-angiogenic therapy involves inhibiting multiple cell types, molecular targets, and intracellular signaling pathways. Computational tools are useful in guiding treatment strategies, predicting the response to treatment, and identifying new targets of interest. Here, we describe progress that has been made in applying mathematical modeling and bioinformatics approaches to study anti-angiogenic therapeutics in cancer.
Collapse
Affiliation(s)
- Stacey D Finley
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Liang-Hui Chu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
26
|
Sasahira T, Ueda N, Yamamoto K, Kurihara M, Matsushima S, Bhawal UK, Kirita T, Kuniyasu H. Prox1 and FOXC2 act as regulators of lymphangiogenesis and angiogenesis in oral squamous cell carcinoma. PLoS One 2014; 9:e92534. [PMID: 24647631 PMCID: PMC3960274 DOI: 10.1371/journal.pone.0092534] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 02/24/2014] [Indexed: 12/22/2022] Open
Abstract
Prospero homeobox 1 (Prox1) and forkhead box (FOX) C2 regulate angiogenesis and/or lymphangiogenesis. However, the detailed role and function of Prox1 and FOXC2 in cancer remains controversial. In the present study, we examined the expression of Prox1 and FOXC2 proteins in specimens from 163 cases with oral squamous cell carcinoma (OSCC). Furthermore, the role of Prox1 and FOXC2 in cancer cell growth and invasion was evaluated in cultured OSCC cells. Prox1 expression was significantly associated with local progression of the tumor (P = 0.0023), clinical stage (P<0.0001), lymphovessel density (LVD) (P<0.0001), nodal metastasis (P<0.0001), and worse prognosis (P<0.0001). Immunoreactivity of FOXC2 was strongly correlated with microvessel density (MVD) (P<0.0001) and poor prognosis (P = 0.0076). In vitro analysis demonstrated that Prox1 regulates cell growth, proliferation, invasion, and lymphangiogenesis by activating vascular endothelial growth factor (VEGF)-C expression. Furthermore, FOXC2 enhanced the expression level of Prox1 and promoted angiogenesis by enhancement of VEGF-A expression. Our results suggested that Prox1 and FOXC2 play key roles in OSCC progression and that further studies focusing on these proteins may yield useful insights for diagnosis and therapy of OSCC.
Collapse
Affiliation(s)
- Tomonori Sasahira
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Nobuhiro Ueda
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| | - Kazuhiko Yamamoto
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| | - Miyako Kurihara
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| | - Sayako Matsushima
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Ujjal K. Bhawal
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Tadaaki Kirita
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
- * E-mail:
| |
Collapse
|
27
|
Xu Y, Shao QS, Yao HB, Jin Y, Ma YY, Jia LH. Overexpression of FOXC1 correlates with poor prognosis in gastric cancer patients. Histopathology 2014; 64:963-70. [PMID: 24329718 DOI: 10.1111/his.12347] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 12/08/2013] [Indexed: 01/14/2023]
Abstract
AIMS The aim of this study was to determine FOXC1 expression in gastric tissues, and the clinical significance of FOXC1 in the development, progression and metastasis of gastric cancer (GC). METHODS AND RESULTS We screened GCs for the expression of FOXC1 using the Affymetrix U133 plus 2.0 Gene Chip Array, and found that expression was significantly higher in GC tissues than in controls. Furthermore, we validated the expression levels of FOXC1 using real-time quantitative RT-PCR (qRT-PCR), and of FOXC1 using immunohistochemistry (IHC). Our study showed that expression levels of FOXC1 mRNA and FOXC1 in GC tissues were significantly higher than those in corresponding non-tumour tissues. High FOXC1 expression correlated with the degree of histological differentiation (P < 0.01), TNM stage (P < 0.001), invasive depth (P < 0.05), lymph node metastasis (P < 0.05), and distant metastasis (P < 0.01). Survival analysis revealed that patients with high FOXC1 expression had shorter overall survival than those with low expression (P < 0.001). Multivariate analysis showed that high FOXC1 expression was an independent prognostic factor for GC patients (P < 0.01). CONCLUSIONS Overexpression of FOXC1 may play a key role in the progression of GC, and FOXC1 expression may serve as a useful marker for predicting the outcome of patients with GC.
Collapse
Affiliation(s)
- Yuan Xu
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| | | | | | | | | | | |
Collapse
|
28
|
Watanabe A, Suzuki H, Yokobori T, Altan B, Kubo N, Araki K, Wada S, Mochida Y, Sasaki S, Kashiwabara K, Hosouchi Y, Kuwano H. Forkhead box protein C2 contributes to invasion and metastasis of extrahepatic cholangiocarcinoma, resulting in a poor prognosis. Cancer Sci 2013; 104:1427-32. [PMID: 23919841 DOI: 10.1111/cas.12249] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 07/24/2013] [Accepted: 07/25/2013] [Indexed: 01/25/2023] Open
Abstract
Extrahepatic cholangiocarcinoma (EHCC) is a cancer with a poor prognosis, and the postoperative survival of patients depends on the existence of invasion and metastasis. The epithelial-to-mesenchymal transition (EMT) is an important step in EHCC invasion and metastasis. Forkhead box protein C2 (FOXC2) is a transcription factor that has been reported to induce the EMT. Therefore we examined the correlation between FOXC2 expression and clinical pathological factors, and analysed the function of FOXC2. The expression of FOXC2 in 77 EHCC cases was investigated by immunohistochemical staining, and the relationship between FOXC2 expression and clinicopathological factor was assessed. Knockdown by small interfering RNA (siRNA) was performed to determine the roles of FOXC2 in EHCC cell line. FOXC2 expression correlated with lymph node metastasis (P = 0.0205). Patients in the high FOXC2 expression group had a poorer prognosis than the patients in the low FOXC2 expression group. Moreover, FOXC2 knockdown inhibited cell motility and invasion, and decreased the expression of EMT markers (N-cadherin, and matrix metalloproteinase (MMP) -2) and Angiopietin-2 (Ang-2). The EMT inducer FOXC2 contributes to a poor prognosis and cancer progression. FOXC2 may be a promising molecular target for regulating EHCC metastasis.
Collapse
Affiliation(s)
- Akira Watanabe
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
A novel 99mTc-labeled molecular probe for tumor angiogenesis imaging in hepatoma xenografts model: a pilot study. PLoS One 2013; 8:e61043. [PMID: 23573294 PMCID: PMC3616001 DOI: 10.1371/journal.pone.0061043] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 03/05/2013] [Indexed: 02/07/2023] Open
Abstract
Introduction Visualization of tumor angiogenesis using radionuclide targeting provides important diagnostic information. In previous study, we proved that an arginine-arginine-leucine (RRL) peptide should be a tumor endothelial cell specific binding sequence. The overall aim of this study was to evaluate whether 99mTc-radiolabeled RRL could be noninvasively used for imaging of malignant tumors in vivo, and act as a new molecular probe targeting tumor angiogenesis. Methods The RRL peptide was designed and radiosynthesized with 99mTc by a one-step method. The radiolabeling efficiency and radiochemical purity were then characterized in vitro. 99mTc-RRL was injected intravenously in HepG2 xenograft-bearing BALB/c nude mice. Biodistribution and in vivo imaging were performed periodically. The relationship between tumor size and %ID uptake of 99mTc-RRL was also explored. Results The labeling efficiencies of 99mTc-RRL reached 76.9%±4.5% (n = 6) within 30–60 min at room temperature, and the radiochemical purity exceeded 96% after purification. In vitro stability experiment revealed the radiolabeled peptide was stable. Biodistribution data showed that 99mTc-RRL rapidly cleared from the blood and predominantly accumulated in the kidneys and tumor. The specific uptake of 99mTc-RRL in tumor was significantly higher than that of unlabeled RRL blocking and free pertechnetate control test after injection (p<0.05). The ratio of the tumor-to-muscle exceeded 6.5, tumor-to-liver reached 1.98 and tumor-to-blood reached 1.95. In planar gamma imaging study, the tumors were imaged clearly at 2–6 h after injection of 99mTc-RRL, whereas the tumor was not imaged clearly in blocking group. The tumor-to-muscle ratio of images with 99mTc-RRL was comparable with that of 18F-FDG PET images. Immunohistochemical analysis verified the excessive vasculature of tumor. There was a linear relationship between the tumor size and uptake of 99mTc-RRL with R2 = 0.821. Conclusion 99mTc-RRL can be used as a potential candidate for visualization of tumor angiogenesis in malignant carcinomas.
Collapse
|
30
|
Sasman A, Nassano-Miller C, Shim KS, Koo HY, Liu T, Schultz KM, Millay M, Nanano A, Kang M, Suzuki T, Kume T. Generation of conditional alleles for Foxc1 and Foxc2 in mice. Genesis 2012; 50:766-74. [PMID: 22522965 DOI: 10.1002/dvg.22036] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 04/10/2012] [Accepted: 04/14/2012] [Indexed: 12/28/2022]
Abstract
The Forkhead box transcription factors, Foxc1 and Foxc2, are crucial for development of the eye, cardiovascular network, and other physiological systems, but their cell-type specific and postdevelopmental functions are unknown, in part because conventional (i.e., whole-organism) homozygous-null mutations of either factor result in perinatal death. Here, we describe the generation of mice with conditional-null Foxc1(flox) and Foxc2(flox) mutations that are induced via Cre-mediated recombination. Mice homozygous for the unrecombined alleles are viable and fertile, indicating that the conditional alleles retain their wild-type function. The embryos of Foxc1(flox) or Foxc2(flox) mice crossed with Cre-deleter mice that are homozygous for the recombined allele (i.e., Foxc1(Δ/Δ) or Foxc2(Δ/Δ) embryos) lack expression of the corresponding gene and show the same developmental defects observed in conventional homozygous mutant embryos. We expect these conditional mutations to enable characterization of the cell-type specific functions of Foxc1 and Foxc2 in development, disease, and adult animals.
Collapse
Affiliation(s)
- Amy Sasman
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|