1
|
Bai H, Zhu X, Gao L, Feng S, Li H, Gu X, Xu J, Zong C, Hou X, Yang X, Jiang J, Zhao Q, Wei L, Zhang L, Han Z, Liu W, Qian J. ERG mediates the differentiation of hepatic progenitor cells towards immunosuppressive PDGFRα + cancer-associated fibroblasts during hepatocarcinogenesis. Cell Death Dis 2025; 16:26. [PMID: 39827226 PMCID: PMC11743139 DOI: 10.1038/s41419-024-07270-9] [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: 09/05/2024] [Revised: 11/03/2024] [Accepted: 11/27/2024] [Indexed: 01/22/2025]
Abstract
Cancer-associated fibroblasts (CAFs) play important roles in the occurrence and development of hepatocellular carcinoma (HCC) and are a key component of the immunosuppressive microenvironment. However, the origin of CAFs has not been fully elucidated. We employed single-cell sequencing technology to identify the dynamic changes in different subsets of fibroblasts at different time points in rat primary HCC model. Inflammation-associated CAFs (Pdgfrα+ CAFs) were subsequently identified, which demonstrated a significant correlation with the survival duration of HCC patients and a dual role in the tumour microenvironment (TME). On the one hand, they secrete the chemokines CCL3 and CXCL12, which recruit macrophages to the tumour site. On the other hand, they produce TGFβ, inducing the polarization of these macrophages towards an immunosuppressive phenotype. According to the in vitro and in vivo results, hepatic progenitor cells (HPCs) can aberrantly differentiate into PDGFRα+ CAFs upon stimulation with inflammatory cytokine. This differentiation is mediated by the activation of the MAPK signaling pathway and the downstream transcription factor ERG via the TLR4 receptor. Downregulating the expression of ERG in HPCs significantly reduces the number of PDGFRα+ CAFs and the infiltration of tumour-associated macrophages in HCC, thereby suppressing hepatocarcinogenesis. Collectively, our findings elucidate the distinct biological functions of PDGFRα+ cancer-associated fibroblasts (PDGFRα+ CAFs) within the TME. These insights contribute to our understanding of the mechanisms underlying the establishment of an immunosuppressive microenvironment in HCC, paving the way for the exploration of novel immunotherapeutic strategies tailored for HCC treatment.
Collapse
Affiliation(s)
- Haoran Bai
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinyu Zhu
- Changhai Clinical Research Unit, Changhai Hospital of Naval Medical University, Shanghai, China
| | - Lu Gao
- National Center for Liver Cancer, Shanghai, China
| | - Shiyao Feng
- Department of Urology, Chaohu Hospital of Anhui Medical University, HeFei, Anhui, China
| | - Hegen Li
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoqiang Gu
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiahua Xu
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Zong
- National Center for Liver Cancer, Shanghai, China
| | - Xiaojuan Hou
- National Center for Liver Cancer, Shanghai, China
| | - Xue Yang
- National Center for Liver Cancer, Shanghai, China
| | | | - Qiudong Zhao
- National Center for Liver Cancer, Shanghai, China
| | - Lixin Wei
- National Center for Liver Cancer, Shanghai, China
| | - Li Zhang
- Changhai Clinical Research Unit, Changhai Hospital of Naval Medical University, Shanghai, China.
| | - Zhipeng Han
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- National Center for Liver Cancer, Shanghai, China.
| | - Wenting Liu
- National Center for Liver Cancer, Shanghai, China.
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Jianxin Qian
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
2
|
Saito K, van der Garde M, Umemoto T, Miharada N, Sjöberg J, Sigurdsson V, Shirozu H, Kamei S, Radulovic V, Suzuki M, Nakano S, Lang S, Hansson J, Olsson ML, Minami T, Gouras G, Flygare J, Miharada K. Lipoprotein metabolism mediates hematopoietic stem cell responses under acute anemic conditions. Nat Commun 2024; 15:8131. [PMID: 39284836 PMCID: PMC11405780 DOI: 10.1038/s41467-024-52509-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 09/06/2024] [Indexed: 09/20/2024] Open
Abstract
Hematopoietic stem cells (HSCs) react to various stress conditions. However, it is unclear whether and how HSCs respond to severe anemia. Here, we demonstrate that upon induction of acute anemia, HSCs rapidly proliferate and enhance their erythroid differentiation potential. In severe anemia, lipoprotein profiles largely change and the concentration of ApoE increases. In HSCs, transcription levels of lipid metabolism-related genes, such as very low-density lipoprotein receptor (Vldlr), are upregulated. Stimulation of HSCs with ApoE enhances their erythroid potential, whereas HSCs in Apoe knockout mice do not respond to anemia induction. VldlrhighHSCs show higher erythroid potential, which is enhanced after acute anemia induction. VldlrhighHSCs are epigenetically distinct because of their low chromatin accessibility, and more chromatin regions are closed upon acute anemia induction. Chromatin regions closed upon acute anemia induction are mainly binding sites of Erg. Inhibition of Erg enhanced the erythroid differentiation potential of HSCs. Our findings indicate that lipoprotein metabolism plays an important role in HSC regulation under severe anemic conditions.
Collapse
Affiliation(s)
- Kiyoka Saito
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Mark van der Garde
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
- Department of Medicine III, Hematology and Oncology, Technical University of Munich, Munich, Germany
| | - Terumasa Umemoto
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Natsumi Miharada
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Julia Sjöberg
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Valgardur Sigurdsson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Haruki Shirozu
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shunsuke Kamei
- Division of Molecular and Vascular Biology, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Visnja Radulovic
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Mitsuyoshi Suzuki
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
- Department of Pediatrics, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Satoshi Nakano
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
- Department of Pediatrics, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Stefan Lang
- StemTherapy Bioinformatics Core facility, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Jenny Hansson
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Martin L Olsson
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Takashi Minami
- Division of Molecular and Vascular Biology, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Gunnar Gouras
- Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Johan Flygare
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Kenichi Miharada
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan.
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden.
| |
Collapse
|
3
|
Mumtaz S, Usman Rashid M, Khan RU, Malkani N. miR-4482 and miR-3912 aim for 3'UTR of ERG mRNA in prostate cancer. PLoS One 2023; 18:e0286996. [PMID: 37310937 DOI: 10.1371/journal.pone.0286996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/29/2023] [Indexed: 06/15/2023] Open
Abstract
Ets-related gene (ERG) is overexpressed as a fusion protein in prostate cancer. During metastasis, the pathological role of ERG is associated with cell proliferation, invasion, and angiogenesis. Here, we hypothesized that miRNAs regulate ERG expression through its 3'UTR. Several bioinformatics tools were used to identify miRNAs and their binding sites on 3'UTR of ERG. The selected miRNAs expression was analyzed in prostate cancer samples by qPCR. The miRNAs overexpression was induced in prostate cancer cells (VCaP) to analyze ERG expression. Reporter gene assay was performed to evaluate the ERG activity in response to selected miRNAs. The expression of ERG downstream target genes was also investigated through qPCR after miRNAs overexpression. To observe the effects of selected miRNAs on cell proliferation and migration, scratch assay was performed to calculate the cell migration rate. miR-4482 and miR-3912 were selected from bioinformatics databases. miR-4482 and -3912 expression were decreased in prostate cancer samples, as compared to controls (p<0.05 and p<0.001), respectively. Overexpression of miR-4482 and miR-3912 significantly reduced ERG mRNA (p<0.001 and p<0.01), respectively) and protein (p<0.01) in prostate cancer cells. The transcriptional activity of ERG was significantly reduced (p<0.01) in response to miR-4482 and-3912. ERG angiogenic targets and cell migration rate was also reduced significantly (p<0.001) after miR-4482 and -3912 over-expression. This study indicates that miR-4482 and -3912 can suppress the ERG expression and its target genes, thereby, halt prostate cancer progression. These miRNAs may be employed as a potential therapeutic target for the miRNA-based therapy against prostate cancer.
Collapse
Affiliation(s)
- Sidra Mumtaz
- Department of Zoology, GC University, Lahore, Pakistan
| | - Muhammad Usman Rashid
- Department of Basic Sciences Research, Shaukat Khanum Memorial Cancer Hospital and Research Centre (SKMCH&RC), Lahore, Pakistan
| | | | - Naila Malkani
- Department of Zoology, GC University, Lahore, Pakistan
| |
Collapse
|
4
|
Mullen D, Nowak K, Chetty R. Gene of the month: ERG. J Clin Pathol 2022; 75:577-580. [PMID: 35738886 DOI: 10.1136/jcp-2022-208350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2022] [Indexed: 11/04/2022]
Abstract
The ERG gene belongs to the erythroblastosis transformation specific family of transcription factors and encodes for the transcription regulator protein ERG. It is located on chromosome 22q22 and is a nuclear transcription factor. In normal physiology, ERG protein is expressed in endothelial cells and is involved in processes including, but not limited to, angiogenesis and haematopoiesis. Of diagnostic value in clinical practice, ERG immunohistochemistry is a useful marker of endothelial differentiation for both benign and malignant vascular lesions. It is also reliable for identifying ERG gene translocated malignancies such as EWS/FUS::ERG Ewing's sarcoma and TMPSSR2::ERG prostatic carcinoma.
Collapse
Affiliation(s)
- Dorinda Mullen
- Department of Pathology, University Health Network Laboratory Medicine Program, University of Toronto, Toronto, Ontario, Canada
| | - Klaudia Nowak
- Department of Pathology, University Health Network Laboratory Medicine Program, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
5
|
Yamada Y, Ichiki T, Susuki Y, Yamada-Nozaki Y, Tateishi Y, Furue M, Oda Y. Diagnostic utility of ERG immunostaining in dermatofibroma. J Clin Pathol 2022:jclinpath-2022-208158. [PMID: 35318257 DOI: 10.1136/jclinpath-2022-208158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/07/2022] [Indexed: 01/25/2023]
Abstract
AIMS Dermatofibroma/fibrous histiocytoma (DF/FH) is a common cutaneous mesenchymal neoplasm exhibiting benign biological behaviour. However, the immunohistochemical utility of erythroblast transformation-specific-related gene (ERG) for diagnosing DF remains unknown. The authors reviewed the immunohistochemical status of ERG in different subtypes of DF and in its differential diagnoses. METHODS Overall, 97 cases of ordinary DF/FH, 6 cases of aneurysmal FH, 10 cases of cellular FH, 5 cases of angiomatoid FH, 2 cases of epithelioid FH, 64 cases of dermatofibrosarcoma protuberans (DFSP) and 52 cases of fibrous scar were retrieved. As the other histological types of cutaneous neoplasms, 6 cases of myxofibrosarcoma, 4 cases of undifferentiated pleomorphic sarcoma, 11 cases of atypical fibroxanthoma, 19 cases of malignant melanoma, 20 cases of nevocellular nevus, 20 cases of neurofibroma, 19 cases of schwannoma, 8 cases of angioleiomyoma and 1 case of pilar leiomyoma were included. RESULTS Immunohistochemical positivity for ERG was demonstrated in 87 of 97 cases (89.6%) of ordinary DF/FH, 7 of 10 cases (70%) of cellular FH, 3 of 6 cases (50%) of aneurysmal FH, 1 of 5 cases (20%) of angiomatoid FH and 1 of 52 cases (0.1%) of fibrous scar. All cases of DFSP, epithelioid FH and other types of cutaneous neoplasms included in the current investigation were negative for ERG. The intensity of ERG immunohistochemical staining in spindle-shaped cells appeared weaker than that in endothelial cells. CONCLUSIONS DF/FH was frequently positive for ERG immunostaining. ERG immunostaining may thus be useful to distinguish DF/FH from DFSP.
Collapse
Affiliation(s)
- Yuichi Yamada
- Department of Anatomic Pathology, Kyushu University, Fukuoka, Japan
| | - Toshio Ichiki
- Department of Anatomic Pathology, Kyushu University, Fukuoka, Japan
| | - Yosuke Susuki
- Department of Anatomic Pathology, Kyushu University, Fukuoka, Japan
| | | | - Yuki Tateishi
- Department of Anatomic Pathology, Kyushu University, Fukuoka, Japan
| | - Masutaka Furue
- Department of Dermatology, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Kyushu University, Fukuoka, Japan
| |
Collapse
|
6
|
Tharakan B, Hunter FA, Muthusamy S, Randolph S, Byrd C, Rao VN, Reddy ESP, Childs EW. ETS-Related Gene Activation Preserves Adherens Junctions and Permeability in Microvascular Endothelial Cells. Shock 2022; 57:309-315. [PMID: 34907119 DOI: 10.1097/shk.0000000000001899] [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] [Indexed: 11/26/2022]
Abstract
ABSTRACT ERG (ETS-related gene) is a member of the ETS (Erythroblast-transformation specific) family of transcription factors abundantly present in vascular endothelial cells. Recent studies demonstrate that ERG has important roles in blood vessel stability and angiogenesis. However, it is unclear how ERG is potentially involved in microvascular barrier functions and permeability. A wide variety of diseases and clinical conditions including trauma-hemorrhagic shock and burn injury are associated with microvascular dysfunctions, which causes excessive microvascular permeability, tissue edema and eventually, multiple organ dysfunction and death. The main purpose of this study was to determine the specific role of ERG in regulating microvascular permeability in human lung microvascular endothelial cells (HLMEC) and to evaluate if exogenous ERG will protect the barrier. The HLMECs were grown on Transwell inserts as monolayers and were transfected with ERG CRISPR/cas9 knockdown plasmid, ERG CRISPR activation plasmid, recombinant ERG protein or their respective controls. Recombinant vascular endothelial growth factor (VEGF) was used as an inducer of permeability for evaluating the effect of ERG activation on permeability. Changes in barrier integrity and permeability were studied using monolayer permeability assay and immunofluorescence of adherens junction proteins (VE-cadherin and β-catenin) respectively. CRISPR/cas9-based ERG knockdown as well as VEGF treatment induced monolayer hyperpermeability, VE-cadherin, and β-catenin junctional relocation and cytoskeletal F-actin stress fiber formation. CRISPR based ERG activation and recombinant ERG transfection attenuated VEGF-induced monolayer hyperpermeability. ERG activation preserved the adherens junctions and cytoskeleton. These results demonstrate that ERG is a potent regulator of barrier integrity and permeability in human lung microvascular endothelial cells and endogenously or exogenously enhancing ERG provides protection against barrier dysfunction and hyperpermeability.
Collapse
Affiliation(s)
- Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, Georgia
| | - Felicia A Hunter
- Department of Surgery, Morehouse School of Medicine, Atlanta, Georgia
| | | | - Sonya Randolph
- Department of Surgery, Morehouse School of Medicine, Atlanta, Georgia
| | - Crystal Byrd
- Department of Surgery, Morehouse School of Medicine, Atlanta, Georgia
| | - Veena N Rao
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, Atlanta, Georgia
| | - E Shyam P Reddy
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, Atlanta, Georgia
| | - Ed W Childs
- Department of Surgery, Morehouse School of Medicine, Atlanta, Georgia
| |
Collapse
|
7
|
Eldhose B, Pandrala M, Xavier C, Mohamed AA, Srivastava S, Sunkara AD, Dobi A, Malhotra SV. New Selective Inhibitors of ERG Positive Prostate Cancer: ERGi-USU-6 Salt Derivatives. ACS Med Chem Lett 2021; 12:1703-1709. [PMID: 34790292 PMCID: PMC8591719 DOI: 10.1021/acsmedchemlett.1c00308] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022] Open
Abstract
![]()
Prostate
cancer is among the leading causes of cancer related death
of men in the United States. The ERG gene fusion
leading to overexpression of near full-length ERG transcript and protein represents most prevalent (50–65%)
prostate cancer driver gene alterations. The ERG oncoprotein overexpression
persists in approximately 35% of metastatic castration resistant prostate
cancers. Due to the emergence of eventual refractoriness to second-
and third-generation androgen axis-based inhibitors, there remains
a pressing need to develop drugs targeting other validated prostate
cancer drivers such as ERG. Here we report the new and more potent
ERG inhibitor ERGi-USU-6 developed by structure–activity studies
from the parental ERGi-USU. We have developed an improved procedure
for the synthesis of ERGi-USU-6 and identified a salt formulation
that further improves its activity in biological assays for selective
targeting of ERG harboring prostate cancer cells.
Collapse
Affiliation(s)
- Binil Eldhose
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland 20817, United States
| | - Mallesh Pandrala
- Division of Radiation & Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Charles Xavier
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland 20817, United States
| | - Ahmed A. Mohamed
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland 20817, United States
| | - Shiv Srivastava
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889, United States
| | - Anu D. Sunkara
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889, United States
- Washington Adventist University, Takoma Park, Maryland 20912, United States
| | - Albert Dobi
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland 20817, United States
| | - Sanjay V. Malhotra
- Division of Radiation & Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California 94305, United States
| |
Collapse
|
8
|
Moradi Tabriz H, Aghapour Sabaghi L, Nabighadim A, Elham E, Aghamir SMK. Evaluation of ERG Expression in Prostate Adenocarcinoma and Its Prognostic Impact in Patients Survival Rate. IRANIAN JOURNAL OF PATHOLOGY 2021; 16:411-417. [PMID: 34567190 PMCID: PMC8463748 DOI: 10.30699/ijp.20201.530515.2644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 06/29/2021] [Indexed: 11/07/2022]
Abstract
Background & Objective: Some certain markers, including prostatic specific antigen (PSA), are being used to screen prostate cancer (PC), but none of them have sufficient sensitivity and specificity for evaluation of prognosis. Currently, genetic variants have found their place in the prognosis of PC. ETS-related gene (ERG) expression and its intensity have contradictory evidence regarding ERG expression with PC incidence or associating outcome. Our purpose was to survey the relationship of ERG expression and its intensity with PC and relative clinical outcome. Methods: We studied the immunohistochemichal (IHC) expression of ERG in 101 radical prostatectomy specimens with PC of different histologic grades. All samples were chosen from pathology department of Sina hospital in Tehran-Iran from 2011 to 2018. Positive ERG expression and its association with Gleason score, preoperative PSA, metastasis status, stage and grade of tumors was evaluated. Results: In total, ERG expression was observed in 42 cases (41.58%) and of these, 7 (16.66%) were categorized as weak, 13 (30.95%) moderate and 22(52.38%) as strong. There was no significant correlation between ERG expression and age, preoperative PSA, Gleason score, lymph node involvement, metastatic pattern, stage, and grade of the tumor (P>0.05). ERG expression frequency in the two groups of survived and expired patients was 42.85% and 0%, respectively; despite the noticeable difference, it was not statistically significance (P=0.264). Conclusion: Evaluation of ERG expression and its intensity may have no essential role as an acceptable prognostic factor in Iranian’s population for anticipating whether PC itself or the outcomes accompanied. This relation is vigorously under the influence of geographical/ethnical features.
Collapse
Affiliation(s)
- Hedieh Moradi Tabriz
- Department of Pathology, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Aghapour Sabaghi
- Department of Pathology, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirreza Nabighadim
- Pediatric Urology and Regenerative Medicine Research Center, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Elham
- Department of Pathology, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
9
|
Chen B, Zhu Y, Chen J, Feng Y, Xu Y. Activation of TC10-Like Transcription by Lysine Demethylase KDM4B in Colorectal Cancer Cells. Front Cell Dev Biol 2021; 9:617549. [PMID: 34249900 PMCID: PMC8260841 DOI: 10.3389/fcell.2021.617549] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 04/06/2021] [Indexed: 12/19/2022] Open
Abstract
Malignant colorectal cancers (CRCs) are characterized by enhanced migration and invasion thus acquiring the ability to metastasize. We have previously shown that the small GTPase TC10-like (TCL) contributes to aggressive migration and invasion in malignant CRC cells. TCL expression is differentially expressed in CRC cells and can be upregulated by hypoxia although the underlying epigenetic mechanism is not fully appreciated. Here, we report that differential TCL expression in CRC cells appeared to be associated with histone H3K9 methylation. RNAi screening revealed that the lysine demethylase KDM4B was essential for TCL transcription in CRC cells. KDM4B interacted with and was recruited by the sequence-specific transcription factor ETS-related gene 1 (ERG1) to the TCL promoter to activate transcription. Mechanistically, KDM4B mediated H3K9 demethylase facilitated the assembly of pre-initiation complex (PIC) on the TCL promoter. KDM4B knockdown attenuated migration and invasion of CRC cells. Importantly, KDM4B expression was upregulated in human CRC specimens of advanced stages compared to those of lower grades and associated with poor prognosis. Together, these data uncover a novel epigenetic mechanism underlying malignant transformation of CRC cells and suggest that KDM4B may be considered as a therapeutic target in CRC intervention.
Collapse
Affiliation(s)
- Baoyu Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yuwen Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Junliang Chen
- Department of Pathophysiology, Wuxi Medical School, Jiangnan University, Wuxi, China
| | - Yifei Feng
- Department of Colorectal Surgery, The First Hospital Affiliated With Nanjing Medical University, Nanjing, China.,Department of General Surgery, The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| |
Collapse
|
10
|
Panagopoulos I, Heim S. Interstitial Deletions Generating Fusion Genes. Cancer Genomics Proteomics 2021; 18:167-196. [PMID: 33893073 DOI: 10.21873/cgp.20251] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/16/2022] Open
Abstract
A fusion gene is the physical juxtaposition of two different genes resulting in a structure consisting of the head of one gene and the tail of the other. Gene fusion is often a primary neoplasia-inducing event in leukemias, lymphomas, solid malignancies as well as benign tumors. Knowledge about fusion genes is crucial not only for our understanding of tumorigenesis, but also for the diagnosis, prognostication, and treatment of cancer. Balanced chromosomal rearrangements, in particular translocations and inversions, are the most frequent genetic events leading to the generation of fusion genes. In the present review, we summarize the existing knowledge on chromosome deletions as a mechanism for fusion gene formation. Such deletions are mostly submicroscopic and, hence, not detected by cytogenetic analyses but by array comparative genome hybridization (aCGH) and/or high throughput sequencing (HTS). They are found across the genome in a variety of neoplasias. As tumors are increasingly analyzed using aCGH and HTS, it is likely that more interstitial deletions giving rise to fusion genes will be found, significantly impacting our understanding and treatment of cancer.
Collapse
Affiliation(s)
- Ioannis Panagopoulos
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway;
| | - Sverre Heim
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
11
|
Brossa A, Buono L, Fallo S, Fiorio Pla A, Munaron L, Bussolati B. Alternative Strategies to Inhibit Tumor Vascularization. Int J Mol Sci 2019; 20:E6180. [PMID: 31817884 PMCID: PMC6940973 DOI: 10.3390/ijms20246180] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/26/2019] [Accepted: 12/04/2019] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells present in tumors show different origin, phenotype, and genotype with respect to the normal counterpart. Various mechanisms of intra-tumor vasculogenesis sustain the complexity of tumor vasculature, which can be further modified by signals deriving from the tumor microenvironment. As a result, resistance to anti-VEGF therapy and activation of compensatory pathways remain a challenge in the treatment of cancer patients, revealing the need to explore alternative strategies to the classical anti-angiogenic drugs. In this review, we will describe some alternative strategies to inhibit tumor vascularization, including targeting of antigens and signaling pathways overexpressed by tumor endothelial cells, the development of endothelial vaccinations, and the use of extracellular vesicles. In addition, anti-angiogenic drugs with normalizing effects on tumor vessels will be discussed. Finally, we will present the concept of endothelial demesenchymalization as an alternative approach to restore normal endothelial cell phenotype.
Collapse
Affiliation(s)
- Alessia Brossa
- Department of Molecular Biotechnology and Health Sciences, Universitty of Torino, 10126 Torino, Italy; (A.B.); (L.B.); (S.F.)
| | - Lola Buono
- Department of Molecular Biotechnology and Health Sciences, Universitty of Torino, 10126 Torino, Italy; (A.B.); (L.B.); (S.F.)
| | - Sofia Fallo
- Department of Molecular Biotechnology and Health Sciences, Universitty of Torino, 10126 Torino, Italy; (A.B.); (L.B.); (S.F.)
| | - Alessandra Fiorio Pla
- Department of Life Science and Systems Biology, University of Torino, 10126 Torino, Italy; (A.F.P.); (L.M.)
| | - Luca Munaron
- Department of Life Science and Systems Biology, University of Torino, 10126 Torino, Italy; (A.F.P.); (L.M.)
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, Universitty of Torino, 10126 Torino, Italy; (A.B.); (L.B.); (S.F.)
| |
Collapse
|
12
|
Fry EA, Mallakin A, Inoue K. Translocations involving ETS family proteins in human cancer. INTEGRATIVE CANCER SCIENCE AND THERAPEUTICS 2018; 5:10.15761/ICST.1000281. [PMID: 30542624 PMCID: PMC6287620 DOI: 10.15761/icst.1000281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The ETS transcription factors regulate expression of genes involved in normal cell development, proliferation, differentiation, angiogenesis, and apoptosis, consisting of 28 family members in humans. Dysregulation of these transcription factors facilitates cell proliferation in cancers, and several members participate in invasion and metastasis by activating certain gene transcriptions. ETS1 and ETS2 are the founding members of the ETS family and regulate transcription by binding to ETS sequences. Three chimeric genes involving ETS genes have been identified in human cancers, which are EWS-FLI1 in Ewing's sarcoma, TMPRSS2-ERG in prostate cancer, and ETV6-RUNX1 in acute lymphocytic leukemia. Although these fusion transcripts definitely contribute to the pathogenesis of the disease, the impact of these fusion transcripts on patients' prognosis is highly controversial. In the present review, the roles of ETS protein translocations in human carcinogenesis are discussed.
Collapse
Affiliation(s)
- Elizabeth A. Fry
- Dept. of Pathology, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157 USA
| | | | - Kazushi Inoue
- Dept. of Pathology, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157 USA
| |
Collapse
|
13
|
Ullman D, Dorn D, Rais-Bahrami S, Gordetsky J. Clinical Utility and Biologic Implications of Phosphatase and Tensin Homolog (PTEN) and ETS-related Gene (ERG) in Prostate Cancer. Urology 2017; 113:59-70. [PMID: 29225123 DOI: 10.1016/j.urology.2017.11.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/15/2017] [Accepted: 11/15/2017] [Indexed: 12/13/2022]
Abstract
Phosphatase and tensin homolog (PTEN) and ETS-related gene (ERG) mutations are commonly found in prostate cancer. Although mouse studies have demonstrated that PTEN and ERG cooperatively interact during tumorigenesis, human studies examining these genes have been inconclusive. A systematic PubMed search including original articles assessing the pathogenesis of PTEN and ERG in prostate cancer was performed. Studies examining ERG's prognostic significance have conflicting results. Studies examining PTEN and ERG simultaneously found these genes are likely to occur together, but cooperative tumorigenesis functions have not been conclusively established. PTEN mutations are associated with a range of prognostic features. However, the practical clinical utility of this information remains to be determined.
Collapse
Affiliation(s)
- David Ullman
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - David Dorn
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - Soroush Rais-Bahrami
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL; Department of Radiology, University of Alabama at Birmingham, Birmingham, AL
| | - Jennifer Gordetsky
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL; Department of Urology, University of Alabama at Birmingham, Birmingham, AL.
| |
Collapse
|
14
|
Abou-Ouf H, Zhao L, Bismar TA. ERG expression in prostate cancer: biological relevance and clinical implication. J Cancer Res Clin Oncol 2016; 142:1781-93. [PMID: 26711283 DOI: 10.1007/s00432-015-2096-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 12/10/2015] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Screening for increased levels of prostate-specific antigen (PSA) has allowed early detection of a large majority of prostate cancer (PCa) cases. However, the relative lack of specificity of PSA has resulted in significant over-diagnosis and unnecessary treatment for indolent tumors. The fusion of the transmembrane protease serine 2 with E26 transformation-specific family genes, particularly ERG, is the most widespread genetic alteration in prostate cancer, and data suggest that it is more specific for neoplastic prostate disease and may be of added prognostic value and point toward molecular subtype of PCa. METHODS In this review, retrospective studies and clinical trials were analyzed to highlight the recent advances in our understanding of the cellular consequence of ERG rearrangement, describe its interactions with other genetic and molecular pathways, and discuss its potential diagnostic and prognostic value. CONCLUSION ERG over-expression has an emerging role in the diagnosis of PCa pathology, although there is still debate about its prognostic value. Elucidation of the mechanisms of ERG gene rearrangements and expression promises novel therapeutic and diagnostic avenues for prostate cancer.
Collapse
Affiliation(s)
- Hatem Abou-Ouf
- Department of Pathology and Laboratory Medicine, University of Calgary and Calgary Laboratory Services, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute and Tom Baker Cancer Center, Calgary, AB, Canada
| | - Liena Zhao
- Department of Pathology and Laboratory Medicine, University of Calgary and Calgary Laboratory Services, Calgary, AB, Canada
| | - Tarek A Bismar
- Department of Pathology and Laboratory Medicine, University of Calgary and Calgary Laboratory Services, Calgary, AB, Canada.
- Departments of Oncology, Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada.
- Arnie Charbonneau Cancer Institute and Tom Baker Cancer Center, Calgary, AB, Canada.
| |
Collapse
|
15
|
Liu M, Chen L, Ma NF, Chow RKK, Li Y, Song Y, Chan THM, Fang S, Yang X, Xi S, Jiang L, Li Y, Zeng TT, Li Y, Yuan YF, Guan XY. CHD1L promotes lineage reversion of hepatocellular carcinoma through opening chromatin for key developmental transcription factors. Hepatology 2016; 63:1544-59. [PMID: 27100146 DOI: 10.1002/hep.28437] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 01/07/2016] [Indexed: 01/16/2023]
Abstract
UNLABELLED High-grade tumors with poor differentiation usually show phenotypic resemblance to their developmental ancestral cells. Cancer cells that gain lineage precursor cell properties usually hijack developmental signaling pathways to promote tumor malignant progression. However, the molecular mechanisms underlying this process remain unclear. In this study, the chromatin remodeler chromodomain-helicase-DNA-binding-protein 1-like (CHD1L) was found closely associated with liver development and hepatocellular carcinoma (HCC) tumor differentiation. Expression of CHD1L decreased during hepatocyte maturation and increased progressively from well-differentiated HCCs to poorly differentiated HCCs. Chromatin immunoprecipitation followed by high-throughput deep sequencing found that CHD1L could bind to the genomic sequences of genes related to development. Bioinformatics-aided network analysis indicated that CHD1L-binding targets might form networks associated with developmental transcription factor activation and histone modification. Overexpression of CHD1L conferred ancestral precursor-like properties of HCC cells both in vitro and in vivo. Inhibition of CHD1L reversed tumor differentiation and sensitized HCC cells to sorafenib treatment. Mechanism studies revealed that overexpression of CHD1L could maintain an active "open chromatin" configuration at promoter regions of estrogen-related receptor-beta and transcription factor 4, both of which are important regulators of HCC self-renewal and differentiation. In addition, we found a significant correlation of CHD1L with developmental transcriptional factors and lineage differentiation markers in clinical HCC patients. CONCLUSION Genomic amplification of chromatin remodeler CHD1L might drive dedifferentiation of HCC toward an ancestral lineage through opening chromatin for key developmental transcriptional factors; further inhibition of CHD1L might "downgrade" poorly differentiated HCCs and provide novel therapeutic strategies.
Collapse
Affiliation(s)
- Ming Liu
- Department of Clinical Oncology, University of Hong Kong, Hong Kong.,Center for Cancer Research, University of Hong Kong, Hong Kong.,State Key Laboratory for Liver Research, University of Hong Kong, Hong Kong
| | - Leilei Chen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | - Raymond Kwok Kei Chow
- Department of Clinical Oncology, University of Hong Kong, Hong Kong.,Center for Cancer Research, University of Hong Kong, Hong Kong.,State Key Laboratory for Liver Research, University of Hong Kong, Hong Kong
| | - Yan Li
- Department of Clinical Oncology, University of Hong Kong, Hong Kong.,Center for Cancer Research, University of Hong Kong, Hong Kong.,State Key Laboratory for Liver Research, University of Hong Kong, Hong Kong
| | - Yangyang Song
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Tim Hon Man Chan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Shuo Fang
- Department of Clinical Oncology, University of Hong Kong, Hong Kong.,Center for Cancer Research, University of Hong Kong, Hong Kong.,State Key Laboratory for Liver Research, University of Hong Kong, Hong Kong
| | - Xiaodong Yang
- Department of Clinical Oncology, University of Hong Kong, Hong Kong.,Center for Cancer Research, University of Hong Kong, Hong Kong.,State Key Laboratory for Liver Research, University of Hong Kong, Hong Kong
| | - Shaoyan Xi
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lingxi Jiang
- Department of Clinical Oncology, University of Hong Kong, Hong Kong.,Center for Cancer Research, University of Hong Kong, Hong Kong.,State Key Laboratory for Liver Research, University of Hong Kong, Hong Kong
| | - Yun Li
- Department of Clinical Oncology, University of Hong Kong, Hong Kong.,Center for Cancer Research, University of Hong Kong, Hong Kong.,State Key Laboratory for Liver Research, University of Hong Kong, Hong Kong
| | - Ting-Ting Zeng
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yan Li
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yun-Fei Yuan
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, University of Hong Kong, Hong Kong.,Center for Cancer Research, University of Hong Kong, Hong Kong.,State Key Laboratory for Liver Research, University of Hong Kong, Hong Kong.,State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
16
|
Yang Z, Yu L, Wang Z. PCA3 and TMPRSS2-ERG gene fusions as diagnostic biomarkers for prostate cancer. Chin J Cancer Res 2016; 28:65-71. [PMID: 27041928 DOI: 10.3978/j.issn.1000-9604.2016.01.05] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The incidence of prostate cancer (PCa) is rising steadily among males in many countries. Serum prostate-specific antigen (PSA) is widely applied to clinical diagnosis and screening of PCa. However, the so-called grey area of PSA levels 4.0-10.0 ng/mL has a low specificity of 25-40% resulting in a high rate of negative biopsy and overtreatment. So in order to treat PCa patients in early stage, there is an urgent need for new biomarkers in PCa diagnosis. The PCA3 gene, a non-coding RNA (ncRNA) that is highly expressed in prostate cancer (PCa) cells, has been identified as a molecular biomarkers to detect PCa, of which PCA3 has already under clinical application. PCA3 is strongly overexpressed in malignant prostate tissue compared to benign or normal adjacent one. Newly, PCA3 is considered to be a promising biomarker in clinical diagnosis and targeted therapy. The diagnostic significance of PCA3, however, is awaiting further researches. Moreover, it has been demonstrated recently that TMPRSS2-ERG gene fusion is identified as the predominant genetic change in patients diagnosed with PCa. Recent study revealed that combination of the PCA3 and TMPRSS2-ERG gene fusion test optimizes PCa detection compared with that of single biomarker, which would lead to a considerable reduction of the number of prostate biopsies. In this review, we focused on the potential use of PCA3 and TMPRSS2-ERG gene fusion detection in the diagnosis of PCa.
Collapse
Affiliation(s)
- Zheng Yang
- 1 State Key Laboratory of Cancer Biology, Department of Pathology, Xi Jing Hospital, Xi'an 710032, China ; 2 The First Cadet Brigade, Fourth Military Medical University, Xi'an 710032, China
| | - Lu Yu
- 1 State Key Laboratory of Cancer Biology, Department of Pathology, Xi Jing Hospital, Xi'an 710032, China ; 2 The First Cadet Brigade, Fourth Military Medical University, Xi'an 710032, China
| | - Zhe Wang
- 1 State Key Laboratory of Cancer Biology, Department of Pathology, Xi Jing Hospital, Xi'an 710032, China ; 2 The First Cadet Brigade, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
17
|
Hoesel B, Malkani N, Hochreiter B, Basílio J, Sughra K, Ilyas M, Schmid JA. Sequence-function correlations and dynamics of ERG isoforms. ERG8 is the black sheep of the family. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:205-218. [PMID: 26554849 PMCID: PMC4716293 DOI: 10.1016/j.bbamcr.2015.10.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/27/2015] [Accepted: 10/28/2015] [Indexed: 12/31/2022]
Abstract
The transcription factor ERG is known to have divergent roles. On one hand, it acts as differentiation factor of endothelial cells. On the other hand, it has pathological roles in various cancers. Genomic analyses of the ERG gene show that it gives rise to several isoforms. However, functional differences between these isoforms, representing potential reasons for distinct effects in diverse cell types have not been addressed in detail so far. We set out to investigate the major protein isoforms and found that ERG8 contains a unique C-terminus. This isoform, when expressed as GFP-fusion protein, localized mainly to the cytosol, whereas the other major isoforms (ERG1-4) were predominantly nuclear. Using site directed mutagenesis and laser scanning microscopy of live cells, we could identify nuclear localization (NLS) and nuclear export sequences (NES). These analyses indicated that ERG8 lacks a classical NLS and the DNA-binding domain, but holds an additional NES within its distinctive C-terminus. All the tested isoforms were shuttling between nucleus and cytosol and showed a high degree of mobility. ERG’s 1 to 4 were transcriptionally active on ERG-promoter elements whereas ERG8 was inactive, which is in line with the absence of a DNA-binding domain. Fluorescence resonance energy transfer (FRET) microscopy revealed that ERG8 can bind to the transcriptionally active ERG’s. Knockdown of ERG8 in endothelial cells resulted in upregulation of endogenous ERG-transcriptional activity implying ERG8 as an inhibitor of the active ERG isoforms. Quantitative PCR revealed a different ratio of active ERG’s to ERG8 in cancer- versus non-transformed cells.
Collapse
Affiliation(s)
- Bastian Hoesel
- Dept. of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Naila Malkani
- Dept. of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Bernhard Hochreiter
- Dept. of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - José Basílio
- Dept. of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Kalsoom Sughra
- Dept. of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Muhammad Ilyas
- Dept. of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Johannes A Schmid
- Dept. of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| |
Collapse
|
18
|
Morsalin S, Yang C, Fang J, Reddy S, Kayarthodi S, Childs E, Matthews R, Rao VN, Reddy ESP. Molecular Mechanism of β-Catenin Signaling Pathway Inactivation in ETV1-Positive Prostate Cancers. JOURNAL OF PHARMACEUTICAL SCIENCES AND PHARMACOLOGY 2015; 2:208-216. [PMID: 28497076 PMCID: PMC5423671 DOI: 10.1166/jpsp.2015.1069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In the United States of America, prostate cancer is the second most common age-related cancer among men. African-American men have the highest incidence of, and mortality rate from this disease in the United States. According to the American Cancer Society, 29% of all cancer cases and 9% of all cancer deaths are a result of prostate cancer. Individuals who are at highest risk include African-American men, men over 60 years of age, and those with a family history of the disease. African-Americans also have twice the risk of developing prostate cancer as compared to Caucasians. Erythroblastosis virus E26 transformation-specific (ETS) factors play an important role in human cancers. ETS Variant 1 (ETV1), an ETS factor, is notable for its association in prostate cancers, where truncated ETV1 (dETV1) or its full length counterpart is overexpressed in approximately 10% of the prostate cancer patients. Prostate cancer tumorigenesis may be initiated by deregulation of the Wnt/β-catenin pathway. Mutations that stabilize β-catenin were shown to contribute to the loss of cell-growth control in tumorigenesis. We hypothesized that ETV1's interaction with components of the Wnt/β-catenin pathway may alter β-catenin's interaction with downstream tumor-suppressor genes, which are critical in regulating apoptosis and cell-growth properties of prostate cells. Our results demonstrate for the first time that ETV1 alters β-catenin activity by activating kinases that regulate Wnt/β-catenin activity through post-translational modification in prostate cancer cells. We further demonstrate that therapeutic agents such as PD98059, that reverse effect of ETV1 on Wnt/β-catenin signaling pathway, can be used to target ETV1-positive prostate cancer cells. These therapeutic agents could have a profound impact on prevention and treatment of prostate cancer which may help to reduce health disparity seen in minority patients. Understanding the role of ETV1 in Wnt/β-catenin pathway will also allow us to develop better diagnostic tools, which can be used as a biomarker for ETV1-positive prostate cancers.
Collapse
Affiliation(s)
- Sharif Morsalin
- Cancer Biology Program, Department of OB/GYN, Georgia Cancer Center for Excellence, RM 10C009, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Chunshu Yang
- Cancer Biology Program, Department of OB/GYN, Georgia Cancer Center for Excellence, RM 10C009, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Jinbo Fang
- Cancer Biology Program, Department of OB/GYN, Georgia Cancer Center for Excellence, RM 10C009, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Sampreet Reddy
- Cancer Biology Program, Department of OB/GYN, Georgia Cancer Center for Excellence, RM 10C009, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Shubhalaxmi Kayarthodi
- Cancer Biology Program, Department of OB/GYN, Georgia Cancer Center for Excellence, RM 10C009, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Ed Childs
- Department of Surgery, Morehouse School of Medicine, Georgia Cancer Center for Excellence, RM 10C009, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Roland Matthews
- Cancer Biology Program, Department of OB/GYN, Georgia Cancer Center for Excellence, RM 10C009, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Veena N. Rao
- Cancer Biology Program, Department of OB/GYN, Georgia Cancer Center for Excellence, RM 10C009, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - E. Shyam P. Reddy
- Cancer Biology Program, Department of OB/GYN, Georgia Cancer Center for Excellence, RM 10C009, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| |
Collapse
|
19
|
Du J, Zhang L. Integrated analysis of DNA methylation and microRNA regulation of the lung adenocarcinoma transcriptome. Oncol Rep 2015; 34:585-94. [PMID: 26035298 PMCID: PMC4487669 DOI: 10.3892/or.2015.4023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/20/2015] [Indexed: 11/06/2022] Open
Abstract
Lung adenocarcinoma, as a common type of non-small cell lung cancer (40%), poses a significant threat to public health worldwide. The present study aimed to determine the transcriptional regulatory mechanisms in lung adenocarcinoma. Illumina sequence data GSE 37764 including expression profiling, methylation profiling and non-coding RNA profiling of 6 never-smoker Korean female patients with non-small cell lung adenocarcinoma were obtained from the Gene Expression Omnibus (GEO) database. Differentially methylated genes, differentially expressed genes (DEGs) and differentially expressed microRNAs (miRNAs) between normal and tumor tissues of the same patients were screened with tools in R. Functional enrichment analysis of a variety of differential genes was performed. DEG-specific methylation and transcription factors (TFs) were analyzed with ENCODE ChIP-seq. The integrated regulatory network of DEGs, TFs and miRNAs was constructed. Several overlapping DEGs, such as v-ets avian erythroblastosis virus E26 oncogene homolog (ERG) were screened. DEGs were centrally modified by histones of tri-methylation of lysine 27 on histone H3 (H3K27me3) and di-acetylation of lysine 12 or 20 on histone H2 (H2BK12/20AC). Upstream TFs of DEGs were enriched in different ChIP-seq clusters, such as glucocorticoid receptors (GRs). Two miRNAs (miR-126-3p and miR-30c-2-3p) and three TFs including homeobox A5 (HOXA5), Meis homeobox 1 (MEIS1) and T-box 5 (TBX5), played important roles in the integrated regulatory network conjointly. These DEGs, and DEG-related histone modifications, TFs and miRNAs may be important in the pathogenesis of lung adenocarcinoma. The present results may indicate directions for the next step in the study of the further elucidation and targeted prevention of lung adenocarcinoma.
Collapse
Affiliation(s)
- Jiang Du
- Department of Thoracic Surgery, Chinese Medical University Affiliated No. 1 Hospital, Shenyang, Liaoning 110001, P.R. China
| | - Lin Zhang
- Department of Thoracic Surgery, Chinese Medical University Affiliated No. 1 Hospital, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
20
|
The oncogene ERG: a key factor in prostate cancer. Oncogene 2015; 35:403-14. [PMID: 25915839 DOI: 10.1038/onc.2015.109] [Citation(s) in RCA: 179] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/05/2015] [Accepted: 03/06/2015] [Indexed: 12/20/2022]
Abstract
ETS-related gene (ERG) is a member of the E-26 transformation-specific (ETS) family of transcription factors with roles in development that include vasculogenesis, angiogenesis, haematopoiesis and bone development. ERG's oncogenic potential is well known because of its involvement in Ewing's sarcoma and leukaemia. However, in the past decade ERG has become highly associated with prostate cancer development, particularly as a result of a gene fusion with the promoter region of the androgen-induced TMPRRSS2 gene. We review ERG's structure and function, and its role in prostate cancer. We discuss potential new therapies that are based on targeting ERG.
Collapse
|
21
|
He J, Schepmoes AA, Shi T, Wu C, Fillmore TL, Gao Y, Smith RD, Qian WJ, Rodland KD, Liu T, Camp DG, Rastogi A, Tan SH, Yan W, Mohamed AA, Huang W, Banerjee S, Kagan J, Srivastava S, McLeod DG, Srivastava S, Petrovics G, Dobi A, Srinivasan A. Analytical platform evaluation for quantification of ERG in prostate cancer using protein and mRNA detection methods. J Transl Med 2015; 13:54. [PMID: 25889691 PMCID: PMC4342100 DOI: 10.1186/s12967-015-0418-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 01/23/2015] [Indexed: 11/17/2022] Open
Abstract
Background The established methods for detecting prostate cancer (CaP) are based on tests using PSA (blood), PCA3 (urine), and AMACR (tissue) as biomarkers in patient samples. The demonstration of ERG oncoprotein overexpression due to gene fusion in CaP has thus provided ERG as an additional biomarker. Based on this, we hypothesized that ERG protein quantification methods can be of use in the diagnosis of prostate cancer. Methods An antibody-free assay for ERG3 protein detection was developed based on PRISM (high-pressure high-resolution separations with intelligent selection and multiplexing)-SRM (selected reaction monitoring) mass spectrometry. We utilized TMPRSS2-ERG positive VCaP and TMPRSS2-ERG negative LNCaP cells to simulate three different sample types (cells, tissue, and post-DRE urine sediment). Enzyme-linked immunosorbent assay (ELISA), western blot, NanoString, and qRT-PCR were also used in the analysis of these samples. Results Recombinant ERG3 protein spiked into LNCaP cell lysates could be detected at levels as low as 20 pg by PRISM-SRM analysis. The sensitivity of the PRISM-SRM assay was approximately 10,000 VCaP cells in a mixed cell population model of VCaP and LNCaP cells. Interestingly, ERG protein could be detected in as few as 600 VCaP cells spiked into female urine. The sensitivity of the in-house ELISA was similar to the PRISM-SRM assay, with detection of 30 pg of purified recombinant ERG3 protein and 10,000 VCaP cells. On the other hand, qRT-PCR exhibited a higher sensitivity, as TMPRSS2-ERG transcripts were detected in as few as 100 VCaP cells, in comparison to NanoString methodologies which detected ERG from 10,000 cells. Conclusions Based on this data, we propose that the detection of both ERG transcriptional products with RNA-based assays, as well as protein products of ERG using PRISM-SRM assays, may be of clinical value in developing diagnostic and prognostic assays for prostate cancer given their sensitivity, specificity, and reproducibility.
Collapse
Affiliation(s)
- Jintang He
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Athena A Schepmoes
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Tujin Shi
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Chaochao Wu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Thomas L Fillmore
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Yuqian Gao
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Richard D Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Karin D Rodland
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Tao Liu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - David G Camp
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Anshu Rastogi
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | - Shyh-Han Tan
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | - Wusheng Yan
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | - Ahmed A Mohamed
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | - Wei Huang
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | - Sreedatta Banerjee
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | - Jacob Kagan
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA.
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA.
| | - David G McLeod
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA. .,Urology Service, Walter Reed National Military Medical Center, Bethesda, MD, USA.
| | - Shiv Srivastava
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | - Gyorgy Petrovics
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | - Albert Dobi
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | - Alagarsamy Srinivasan
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
22
|
Nannini M, Astolfi A, Urbini M, Indio V, Santini D, Heinrich MC, Corless CL, Ceccarelli C, Saponara M, Mandrioli A, Lolli C, Ercolani G, Brandi G, Biasco G, Pantaleo MA. Integrated genomic study of quadruple-WT GIST (KIT/PDGFRA/SDH/RAS pathway wild-type GIST). BMC Cancer 2014; 14:685. [PMID: 25239601 PMCID: PMC4181714 DOI: 10.1186/1471-2407-14-685] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 09/17/2014] [Indexed: 12/24/2022] Open
Abstract
Background About 10-15% of adult gastrointestinal stromal tumors (GIST) and the vast majority of pediatric GIST do not harbour KIT or platelet-derived growth factor receptor alpha (PDGFRA) mutations (J Clin Oncol 22:3813–3825, 2004; Hematol Oncol Clin North Am 23:15–34, 2009). The molecular biology of these GIST, originally defined as KIT/PDGFRA wild-type (WT), is complex due to the existence of different subgroups with distinct molecular hallmarks, including defects in the succinate dehydrogenase (SDH) complex and mutations of neurofibromatosis type 1 (NF1), BRAF, or KRAS genes (RAS-pathway or RAS-P). In this extremely heterogeneous landscape, the clinical profile and molecular abnormalities of the small subgroup of WT GIST suitably referred to as quadruple wild-type GIST (quadrupleWT or KITWT/PDGFRAWT/SDHWT/RAS-PWT) remains undefined. The aim of this study is to investigate the genomic profile of KITWT/PDGFRAWT/SDHWT/RAS-PWT GIST, by using a massively parallel sequencing and microarray approach, and compare it with the genomic profile of other GIST subtypes. Methods We performed a whole genome analysis using a massively parallel sequencing approach on a total of 16 GIST cases (2 KITWT/PDGFRAWT/SDHWT and SDHBIHC+/SDHAIHC+, 2 KITWT/PDGFRAWT/SDHAmut and SDHBIHC-/SDHAIHC- and 12 cases of KITmut or PDGFRAmut GIST). To confirm and extend the results, whole-genome gene expression analysis by microarray was performed on 9 out 16 patients analyzed by RNAseq and an additional 20 GIST patients (1 KITWT/PDGFRAWTSDHAmut GIST and 19 KITmut or PDGFRAmut GIST). The most impressive data were validated by quantitave PCR and Western Blot analysis. Results We found that both cases of quadrupleWT GIST had a genomic profile profoundly different from both either KIT/PDGFRA mutated or SDHA-mutated GIST. In particular, the quadrupleWT GIST tumors are characterized by the overexpression of molecular markers (CALCRL and COL22A1) and of specific oncogenes including tyrosine and cyclin- dependent kinases (NTRK2 and CDK6) and one member of the ETS-transcription factor family (ERG). Conclusion We report for the first time an integrated genomic picture of KITWT/PDGFRAWT/SDHWT/RAS-PWT GIST, using massively parallel sequencing and gene expression analyses, and found that quadrupleWT GIST have an expression signature that is distinct from SDH-mutant GIST as well as GIST harbouring mutations in KIT or PDGFRA. Our findings suggest that quadrupleWT GIST represent another unique group within the family of gastrointestintal stromal tumors. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-685) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Maria A Pantaleo
- Department of Specialized, Experimental and Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| |
Collapse
|
23
|
Fang J, Xu H, Yang C, Morsalin S, Kayarthodi S, Rungsrisuriyachai K, Gunnal U, Mckenzie B, Rao VN, Reddy ESP. Ets Related Gene and Smad3 Proteins Collaborate to Activate Transforming Growth Factor-Beta Mediated Signaling Pathway in ETS Related Gene-Positive Prostate Cancer Cells. ACTA ACUST UNITED AC 2014; 1:175-181. [PMID: 25745638 DOI: 10.1166/jpsp.2014.1022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
TGF-β/Smads signaling plays a significant role in the regulation of growth of normal and prostate cancer cells. Smad proteins function as important mediators of intracellular signal transduction of transforming growth factor-β (TGF-β). TGF-β signaling pathway is known to regulate cell proliferation, differentiation, apoptosis and play a major role in some human diseases and cancers. Following their phosphorylation by TGF-β receptor-I, Receptor-regulated Smads (including Smad2 and Smad3 proteins) form a heteromeric complex with co-Smad (Smad4) and then translocate into the nucleus where they bind and regulate the expression of target genes. ERG (Ets Related Gene) belongs to the ETS family of transcriptional factors. Chromosomal rearrangement of TMPRSS2 gene and ERG gene has been found in majority of prostate cancers. Over-expression of full length or truncated ERG proteins have been shown to associate with a higher rate of recurrent and unfavorable prognosis of prostate cancer. In order to understand how ERG oncoprotein regulates TGF-β/Smads signaling pathway, we have studied the effect of ERG on TGF-β/Smad3 signaling pathway. In this study, we demonstrate that ERG oncoprotein physically interacts with Smad3 protein and stabilizes phospho-Smad3 protein and thereby enhance TGF-β/Smad3 signaling pathway in prostate cells. Thus, ERG oncoprotein plays an important role in prostate tumorigenesis by using a novel mechanism to activate TGF-β/Smad3 signaling pathway.
Collapse
Affiliation(s)
- Jinbo Fang
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Huali Xu
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Chunshu Yang
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Sharif Morsalin
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Shubhalaxmi Kayarthodi
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Kunchala Rungsrisuriyachai
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Ujwala Gunnal
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA ; Department of Medicine, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Brittany Mckenzie
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Veena N Rao
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - E Shyam P Reddy
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| |
Collapse
|
24
|
Erg cooperates with TGF-β to control mesenchymal differentiation. Exp Cell Res 2014; 328:410-8. [PMID: 25139621 DOI: 10.1016/j.yexcr.2014.08.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 08/08/2014] [Accepted: 08/09/2014] [Indexed: 01/26/2023]
Abstract
Transforming growth factor β (TGF-β) signaling plays an integral role in skeletal development. Conditional deletion of the TGF-β type II receptor (Tgfbr2) from type II Collagen (Col2a) expressing cells results in defects in development of the annulus fibrosus (AF) of the intervertebral disc (IVD). We previously used microarray analysis to search for marker genes of AF as well as transcription factors regulated by TGF-β during AF development. The transcription factor avian erythroblastosis virus E-26 (v-ets) oncogene related (Erg) was identified in the microarray screen as a candidate regulator of AF development. To study the effects of TGF-β on AF differentiation and the role of Erg in this process, we used mouse sclerotome grown in micromass cultures. At 0.5ng TGF-β/ml, sclerotome cells started to express markers of AF. Regulation of Erg by TGF-β was confirmed in these cells. In addition, TGF-β soaked Affi-gel beads implanted into the axial skeleton of stage HH 25 chick embryos showed that TGF-β could induce expression of Erg mRNA in vivo. Next, an adenovirus to over-express Erg in primary sclerotome micromass cultures was generated. Over-expression of Erg led to a change in cell morphology and inhibition of differentiation into hyaline cartilage as seen by reduced Alcian blue staining and decreased Sox9 and c-Maf expression. Erg was not sufficient to induce expression of AF markers and expression of Sca1, a marker of pluripotent progenitor cells, was up-regulated in Erg expressing cells. When cells that ectopically expressed Erg were treated with TGF-β, enhanced expression of specific differentiation markers was observed suggesting Erg can cooperate with TGF-β to regulate differentiation of the sclerotome. Furthermore, we showed using co-immunopreciptiation that Erg and Smad3 bind to each other suggesting a mechanism for their functional interaction.
Collapse
|
25
|
Rastogi A, Tan SH, Mohamed AA, Chen Y, Hu Y, Petrovics G, Sreenath T, Kagan J, Srivastava S, McLeod DG, Sesterhenn IA, Srivastava S, Dobi A, Srinivasan A. Functional antagonism of TMPRSS2-ERG splice variants in prostate cancer. Genes Cancer 2014; 5:273-84. [PMID: 25221645 PMCID: PMC4162137 DOI: 10.18632/genesandcancer.25] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 08/08/2014] [Indexed: 12/14/2022] Open
Abstract
The fusion between ERG coding sequences and the TMPRSS2 promoter is the most prevalent in prostate cancer (CaP). The presence of two main types of TMPRSS2-ERG fusion transcripts in CaP specimens, Type I and Type II, prompted us to hypothesize that the cumulative actions of different ERG variants may impact CaP development/progression. Using TMPRSS2-ERG3 (Type I) and TMPRSS2-ERG8 (Type II) expression vectors, we determined that the TMPRSS2- ERG8 encoded protein is deficient in transcriptional regulation compared to TMPRSS2-ERG3. Co-transfection of vectors resulted in decreased transcriptional regulation compared to TMPRSS2-ERG3 alone, suggesting transdominance of ERG8. Expression of exogenous ERG8 protein resulted in a decrease in endogenous ERG3 protein levels in TMPRSS2-ERG positive VCaP cells, with a concomitant decrease in C-MYC. Further, we showed a physical association between ERG3 and ERG8 in live cells by the bimolecular fluorescence complementation assay, providing a basis for the observed effects. Inhibitory effects of TMPRSS2-ERG8 on TMPRSS2- ERG3 were also corroborated by gene expression data from human prostate cancers, which showed a positive correlation between C-MYC expression and TMPRSS2-ERG3/TMPRSS2- ERG8 ratio. We propose that an elevated TMPRSS2-ERG3/TMPRSS2-ERG8 ratio results in elevated C-MYC in CaP, providing a strong rationale for the biomarker and therapeutic utility of ERG splice variants, along with C-MYC.
Collapse
Affiliation(s)
- Anshu Rastogi
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Shyh-Han Tan
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Ahmed A. Mohamed
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Yongmei Chen
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Ying Hu
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Gyorgy Petrovics
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Taduru Sreenath
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Jacob Kagan
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - David G. McLeod
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Urology Service, Department of Surgery, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | | | - Shiv Srivastava
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Albert Dobi
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Alagarsamy Srinivasan
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
26
|
Fillmore RA, Kojima C, Johnson C, Kolcun G, Dangott LJ, Zimmer WE. New concepts concerning prostate cancer screening. Exp Biol Med (Maywood) 2014; 239:793-804. [PMID: 24928864 DOI: 10.1177/1535370214539091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Prostate Cancer (CaP) is rapidly becoming a worldwide health issue. While CaP mortality has decreased in recent years, coincident with the widespread use of Prostate-Specific Antigen (PSA) screening, it remains the most common solid tumor in men and is the second leading cause of cancer death in the United States. The frequency of CaP is growing not only in western cultures, but also its incidence is dramatically increasing in eastern nations. Recently, examination of data from long-term trials and follow up has cast a shadow on the effectiveness of employing PSA as a primary screening tool for CaP. In this review, we not only summarize opinions from this examination and synthesize recommendations from several groups that suggest strategies for utilizing PSA as a tool, but also call for research into biomarkers for CaP diagnosis and disease progression. We also describe our recent work that identified a smooth muscle contractile protein in prostate epithelia, namely smooth muscle gamma actin, and indicate the potential for this molecule as a new unique footprint and as a CaP marker.
Collapse
Affiliation(s)
- Rebecca A Fillmore
- Department of Biological Sciences, University of Southern Mississippi Gulf Coast, Long Beach MS 39560, USA
| | - Chinatsu Kojima
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, TX 77843-1114, USA
| | - Chevaun Johnson
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, TX 77843-1114, USA
| | - Georgina Kolcun
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, TX 77843-1114, USA
| | - Lawrence J Dangott
- Department of Biochemistry and Biophysics, College of Agriculture and Life Sciences, Texas A&M University, College of Medicine, TX 77843, USA
| | - Warren E Zimmer
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, TX 77843-1114, USA Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, Texas, 77843 Faculty of Genetics, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
27
|
Wang ZB, Yuan J, Chen W, Wei LX. Transcription factor ERG is a specific and sensitive diagnostic marker for hepatic angiosarcoma. World J Gastroenterol 2014; 20:3672-3679. [PMID: 24707153 PMCID: PMC3974537 DOI: 10.3748/wjg.v20.i13.3672] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 02/09/2014] [Accepted: 03/10/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression of ERG, CD34, CD31 (PECAM-1, platelet/endothelial cell adhesion molecule 1) and factor VIII-related antigen (FVIIIRAg) in the diagnosis of hepatic angiosarcoma patients.
METHODS: Patient samples were collected from January 1986 to December 2012 from the People’s Liberation Army General Hospital in Beijing, China. We obtained twenty-four samples of hepatic angiosarcoma (HAS) that were confirmed by two pathologist. The samples were the result of three autopsy cases, eight biopsy cases and 13 patients who underwent surgical tumor removal. The HAS cases accounted for 2.23% (24/1075) of all hepatic vascular tumors at the hospital during the same time period. Patient histories including age, gender, clinical manifestations, medical treatments, laboratory tests, radiological images, histological observations and outcomes for each case were analyzed in detail. All samples were evaluated histologically with hematoxylin and eosin staining. Using immunohistochemistry, the expression and localization of ERG was examined in all HAS specimens and compared to the known endothelial markers CD34, CD31 and FVIIIRAg. The endothelial markers were also evaluated in a panel of non-HAS tumors.
RESULTS: This cohort of 24 HAS cases is, to the best of our knowledge, currently the largest cohort in the world in the publicly available literature. Hepatic angiosarcoma tissue samples were obtained from 14 males and 10 females with a mean age of 50.6 years (range: 7-86 years). The patients presented with the following clinical manifestations: abdominal pain (16/24), back pain (3/24), heart palpitations (1/20), cough (1/24) or no clinical symptoms (3/24). Tumors were predominantly localized in the right hepatic lobe (15/24) or left hepatic lobe (6/24), or a diffuse growth on the right and left hepatic lobes (3/24). Eleven patients underwent surgical resection (45.8%), two patients received a liver transplant (8.3%), eight patients received interventional therapy (33.3%) and three patients received no treatment (lesions discovered at autopsy, 12.5%). Postoperative follow-up of patients revealed that 87.5% (21/24) of patients had died and three cases were not able to be tracked. In all cases, the mean survival time was 12.1 mo. While 100% of the HAS samples were positive for ERG expression, expression of the other markers was more variable. CD31 was expressed in 79.2% (19/24) of samples, CD34 was expressed in 87.5% (21/24) of samples and FVIIIRAg was expressed in 41.7% (10/24) of samples.
CONCLUSION: ERG is a more sensitive and specific diagnostic marker for hepatic angiosarcoma in comparison to CD31, CD34 and FVIIIRAg.
Collapse
|
28
|
Fang J, Xu H, Yang C, Kayarthodi S, Matthews R, Rao VN, Reddy ESP. Molecular Mechanism of Activation of Transforming Growth Factor Beta/Smads Signaling Pathway in Ets Related Gene-Positive Prostate Cancers. ACTA ACUST UNITED AC 2014; 1:82-85. [PMID: 25679011 DOI: 10.1166/jpsp.2014.1008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Transforming growth factor beta (TGF-β) signaling pathway is involved in diverse cellular processes, including cell proliferation, differentiation, adhesion, apoptosis, and some human diseases including cancer. Smad proteins function as mediators of intracellular signal transduction of TGF-β. Following their phosphorylation by TGF-β receptor I, Smad2 and Smad3 form a heteromeric complex with Smad4 and then are translocated into the nucleus where they bind to other co-factors and regulate the expression of target genes. ERG (Ets Related Gene) belongs to the ETS family of transcriptional factors. Chromosomal rearrangement of TMPRSS2 gene and ERG gene has been found in the majority of prostate cancers. Over-expression of full length or truncated ERG proteins is associated with a higher rate of recurrence and unfavorable prognosis. In this review, we focus on recent understanding of regulation of TGF-β/Smads signaling pathway by ERG proteins in prostate cancer.
Collapse
Affiliation(s)
- Jinbo Fang
- Georgia Cancer Center for Excellence, Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Grady Health System, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Huali Xu
- Georgia Cancer Center for Excellence, Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Grady Health System, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Chunshu Yang
- Georgia Cancer Center for Excellence, Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Grady Health System, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Shubha Kayarthodi
- Georgia Cancer Center for Excellence, Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Grady Health System, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Roland Matthews
- Georgia Cancer Center for Excellence, Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Grady Health System, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Veena N Rao
- Georgia Cancer Center for Excellence, Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Grady Health System, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - E Shyam P Reddy
- Georgia Cancer Center for Excellence, Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Grady Health System, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| |
Collapse
|
29
|
Thangapazham R, Saenz F, Katta S, Mohamed AA, Tan SH, Petrovics G, Srivastava S, Dobi A. Loss of the NKX3.1 tumorsuppressor promotes the TMPRSS2-ERG fusion gene expression in prostate cancer. BMC Cancer 2014; 14:16. [PMID: 24418414 PMCID: PMC3897978 DOI: 10.1186/1471-2407-14-16] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 01/08/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In normal prostate epithelium the TMPRSS2 gene encoding a type II serine protease is directly regulated by male hormones through the androgen receptor. In prostate cancer ERG protooncogene frequently gains hormonal control by seizing gene regulatory elements of TMPRSS2 through genomic fusion events. Although, the androgenic activation of TMPRSS2 gene has been established, little is known about other elements that may interact with TMPRSS2 promoter sequences to modulate ERG expression in TMPRSS2-ERG gene fusion context. METHODS Comparative genomic analyses of the TMPRSS2 promoter upstream sequences and pathway analyses were performed by the Genomatix Software. NKX3.1 and ERG genes expressions were evaluated by immunoblot or by quantitative Real-Time PCR (qRT-PCR) assays in response to siRNA knockdown or heterologous expression. QRT-PCR assay was used for monitoring the gene expression levels of NKX3.1-regulated genes. Transcriptional regulatory function of NKX3.1 was assessed by luciferase assay. Recruitment of NKX3.1 to its cognate elements was monitored by Chromatin Immunoprecipitation assay. RESULTS Comparative analysis of the TMPRSS2 promoter upstream sequences among different species revealed the conservation of binding sites for the androgen inducible NKX3.1 tumor suppressor. Defects of NKX3.1, such as, allelic loss, haploinsufficiency, attenuated expression or decreased protein stability represent established pathways in prostate tumorigenesis. We found that NKX3.1 directly binds to TMPRSS2 upstream sequences and negatively regulates the expression of the ERG protooncogene through the TMPRSS2-ERG gene fusion. CONCLUSIONS These observations imply that the frequently noted loss-of-function of NKX3.1 cooperates with the activation of TMPRSS2-ERG fusions in prostate tumorigenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Albert Dobi
- Center for Prostate Disease Research, Uniform Services University of the Health Sciences, 1530 East Jefferson Street, Rockville, Maryland 20852, USA.
| |
Collapse
|
30
|
Abstract
Oncogenic activation of ERG resulting from gene fusion is present in over half of all patients with prostate cancer in Western countries. Although the underlying genetic mechanisms have been extensively studied, evaluation of the ERG oncoprotein--the translational product of ERG gene fusions--has just begun. The robust correlation between ERG oncoprotein detection and gene fusion status enables rapid characterization of this protein in large patient cohorts. Recent studies have focused on characterizing the ERG oncoprotein and determining its potential role in the diagnosis and biological stratification of prostate cancer.
Collapse
|
31
|
Squire JA, Park PC, Yoshimoto M, Alami J, Williams JL, Evans A, Joshua AM. Prostate cancer as a model system for genetic diversity in tumors. Adv Cancer Res 2012; 112:183-216. [PMID: 21925305 DOI: 10.1016/b978-0-12-387688-1.00007-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This chapter will summarize novel understandings of the early molecular events in prostatic carcinogenesis that may underlie both the genetic and clinical heterogeneity. Areas covered include preneoplasia, stem cell concepts, telomere abnormalities, and the nature of tumor-stromal interactions. The oncogenomics of prostate cancer is reviewed with emphasis on androgen signaling, ETS gene family aberrations, and PTEN deletion. The notion that "field cancerization," coupled with genomic instability may explain both the occurrence of multifocal disease, and the recent observations of genetic diversity of ERG alteration in individual tumors are discussed. Collectively, genomic studies are rapidly moving human prostate cancer closer to the promise of personalized medicine, so that specific genetic profiles of individual tumors will determine the best therapeutic approaches.
Collapse
Affiliation(s)
- Jeremy A Squire
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | | | | | | | | | | | | |
Collapse
|
32
|
Sreenath TL, Dobi A, Petrovics G, Srivastava S. Oncogenic activation of ERG: A predominant mechanism in prostate cancer. J Carcinog 2011; 10:37. [PMID: 22279422 PMCID: PMC3263025 DOI: 10.4103/1477-3163.91122] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 11/10/2011] [Indexed: 12/15/2022] Open
Abstract
Prevalent gene fusions involving regulatory sequences of the androgen receptor (AR) regulated genes (primarily TMPRSS2) and protein coding sequences of nuclear transcription factors of the ETS gene family (predominantly ERG) result in unscheduled androgen dependent ERG expression in prostate cancer (CaP).Cumulative data from a large number of studies in the past six years accentuate ERG alterations in more than half of all CaP patients in Western countries. Studies underscore that ERG functions are involved in the biology of CaP. ERG expression in normal context is selective to endothelial cells, specific hematopoetic cells and pre-cartilage cells. Normal functions of ERG are highlighted in hematopoetic stem cells. Emerging data continues to unravel molecular and cellular mechanisms by which ERG may contribute to CaP. Herein, we focus on biological and clinical aspects of ERG oncogenic alterations, potential of ERG-based stratification of CaP and the possibilities of targeting the ERG network in developing new therapeutic strategies for the disease.
Collapse
Affiliation(s)
- Taduru L Sreenath
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | | | | |
Collapse
|
33
|
Fortson WS, Kayarthodi S, Fujimura Y, Xu H, Matthews R, Grizzle WE, Rao VN, Bhat GK, Reddy ESP. Histone deacetylase inhibitors, valproic acid and trichostatin-A induce apoptosis and affect acetylation status of p53 in ERG-positive prostate cancer cells. Int J Oncol 2011; 39:111-9. [PMID: 21519790 PMCID: PMC3329756 DOI: 10.3892/ijo.2011.1014] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Accepted: 03/08/2011] [Indexed: 01/24/2023] Open
Abstract
An ETS family member, ETS Related Gene (ERG) is involved in the Ewing family of tumors as well as leukemias. Rearrangement of the ERG gene with the TMPRSS2 gene has been identified in the majority of prostate cancer patients. Additionally, overexpression of ERG is associated with unfavorable prognosis in prostate cancer patients similar to leukemia patients. Histone acetyltransferases (HATs) and histone deacetylases (HDACs) regulate transcription as well as epigenetic status of genes through acetylation of both histones and transcription factors. Deregulation of HATs and HDACs is frequently seen in various cancers, including prostate cancer. Many cellular oncogenes as well as tumor viral proteins are known to target either or both HATs and HDACs. Several studies have demonstrated that there are alterations of HDAC activity in prostate cancer cells. Recently, we found that ERG binds and inhibits HATs, which suggests that ERG is involved in deregulation of protein acetylation. Additionally, it has been shown that ERG is associated with a higher expression of HDACs. In this study, we tested the effect of the HDAC inhibitors valproic acid (VPA) and trichostatin-A (TSA) on ERG-positive prostate cancer cells (VCaP). We found that VPA and TSA induce apoptosis, upregulate p21/Waf1/CIP1, repress TMPRSS2-ERG expression and affect acetylation status of p53 in VCaP cells. These results suggest that HDAC inhibitors might restore HAT activity through two different ways: by inhibiting HDAC activity and by repressing HAT targeting oncoproteins such as ERG.
Collapse
Affiliation(s)
- Wendell S Fortson
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Health System, 80 Jessie Hill Jr. Drive, Atlanta, GA 30303, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Erg is required for self-renewal of hematopoietic stem cells during stress hematopoiesis in mice. Blood 2011; 118:2454-61. [PMID: 21673349 DOI: 10.1182/blood-2011-03-344739] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Hematopoietic stem cells (HSCs) are rare residents of the bone marrow responsible for the lifelong production of blood cells. Regulation of the balance between HSC self-renewal and differentiation is central to hematopoiesis, allowing precisely regulated generation of mature blood cells at steady state and expanded production at times of rapid need, as well as maintaining ongoing stem cell capacity. Erg, a member of the Ets family of transcription factors, is deregulated in cancers; and although Erg is known to be required for regulation of adult HSCs, its precise role has not been defined. We show here that, although heterozygosity for functional Erg is sufficient for adequate steady-state HSC maintenance, Erg(+/Mld2) mutant mice exhibit impaired HSC self-renewal after bone marrow transplantation or during recovery from myelotoxic stress. Moreover, although mice functionally compromised for either Erg or Mpl, the receptor for thrombopoietin, a key regulator of HSC quiescence, maintained sufficient HSC activity to sustain hematopoiesis, Mpl(-/-) Erg(+/Mld2) compound mutant mice displayed exacerbated stem cell deficiencies and bone marrow failure. Thus, Erg is a critical regulator of adult HSCs, essential for maintaining self-renewal at times of high HSC cycling.
Collapse
|
35
|
Ellett F, Kile BT, Lieschke GJ. The role of the ETS factor erg in zebrafish vasculogenesis. Mech Dev 2009; 126:220-9. [PMID: 19027849 PMCID: PMC2844499 DOI: 10.1016/j.mod.2008.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Revised: 10/30/2008] [Accepted: 11/02/2008] [Indexed: 10/21/2022]
Abstract
Erg, a member of the ETS family of transcription factors, has been implicated by previous studies in endothelial and haematopoietic development. Deregulation of the human ERG locus is associated with acute myeloid leukaemia, prostate cancer and Ewing's sarcoma. To better understand the role of Erg during early development, we utilised the zebrafish as a model amenable to descriptive and functional studies in vivo. Zebrafish have a single erg gene that is expressed in mesoderm and its vascular derivatives during angioblast migration, vasculogenesis and early angiogenesis. Mutant and morphant expression analyses placed erg in a genetic pathway downstream of cloche, tal1/scl and etsrp during early angioblast migration. Furthermore, a combination of gain-of-function and loss-of-function studies suggested a redundant yet specific role for erg in both angioblast specification/proliferation and early angiogenesis, and a synergistic interaction with the critical ETS factor etsrp.
Collapse
Affiliation(s)
- Felix Ellett
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3050, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Benjamin T. Kile
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3050, Australia
| | - Graham J. Lieschke
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3050, Australia
| |
Collapse
|
36
|
|
37
|
Abstract
Almost half of adult acute myelogenous leukemia (AML) is normal cytogenetically, and this subgroup shows a remarkable heterogeneity of genetic mutations at the molecular level and an intermediate response to therapy. The finding of recurrent cytogenetic abnormalities has influenced, in a primary way, the understanding and treatment of leukemias. Yet "normal karyotype AML" lacks such obvious abnormalities, but has a variety of prognostically important genetic abnormalities. Thus, the presence of a FLT3-ITD (internal tandem duplication), MLL-PTD (partial tandem duplication), or the increased expression of ERG or EVI1 mRNAs confer a poor prognosis, and an increased risk of relapse. In contrast, the presence of cytoplasmic nucleophosmin or C/EBPA mutations is associated with lower relapse rates and improved survival. Although resistance to treatment is associated with specific mutations, the degree to which the leukemia resembles a stem cell in its functional properties may provide greater protection from the effects of treatment. Although usually all of the circulating leukemia cells are cleared following treatment, a small residual population of leukemic cells in the bone marrow persists, making this disease hard to eradicate. Increased understanding of the biological consequences of at least some of these mutations in "normal karyotype AML" is leading to more targeted approaches to develop more effective treatments for this disease.
Collapse
Affiliation(s)
- Stephen D Nimer
- Division of Hematologic Oncology, Memorial Sloan Kettering Cancer Center, NY 1275 York Avenue, New York, NY 10021, USA.
| |
Collapse
|
38
|
Moore SDP, Offor O, Ferry JA, Amrein PC, Morton CC, Dal Cin P. ELF4 is fused to ERG in a case of acute myeloid leukemia with a t(X;21)(q25-26;q22). Leuk Res 2005; 30:1037-42. [PMID: 16303180 DOI: 10.1016/j.leukres.2005.10.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2005] [Indexed: 11/27/2022]
Abstract
We report a novel chromosomal translocation in AML, t(X;21)(q25-26;q22), resulting in a fusion transcript between two ETS domain family members, ELF4 (at Xq25) and ERG (at 21q22). ERG has been associated previously with other fusion partners, specifically FUS and EWSR1, and implicated in both AML and Ewing's sarcoma. RT-PCR analysis of RNA isolated from bone marrow samples from the patient demonstrates that the translocation occurs within intron 1 of ERG isoform 1 (ERG-1) and intron 2 of ELF4 resulting in an in-frame fusion joining exon 2 from ELF4 with exon 2 of ERG. This is the first reported case of an ELF4-ERG fusion and identification of the specific ERG exon involved in the fusion that differentiates ERG isoforms. In addition, this case also directly implicates a new role for ELF4 in cancer.
Collapse
Affiliation(s)
- Steven D P Moore
- Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
39
|
Petrovics G, Liu A, Shaheduzzaman S, Furusato B, Furasato B, Sun C, Chen Y, Nau M, Ravindranath L, Chen Y, Dobi A, Srikantan V, Sesterhenn IA, McLeod DG, Vahey M, Moul JW, Srivastava S. Frequent overexpression of ETS-related gene-1 (ERG1) in prostate cancer transcriptome. Oncogene 2005; 24:3847-52. [PMID: 15750627 DOI: 10.1038/sj.onc.1208518] [Citation(s) in RCA: 264] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Transcription factors encoded by the ETS family of genes are central in integrating signals that regulate cell growth and differentiation, stress responses, and tumorigenesis. This study, analysing laser microdissected paired benign and malignant prostate epithelial cells from prostate cancer (CaP) patients (n=114; 228 specimen) by GeneChip and quantitative real-time RT-PCR, identifies ETS-related gene (ERG), a member of the ETS transcription factor family, as the most frequently overexpressed proto-oncogene in the transcriptome of malignant prostate epithelial cells. Combined quantitative expression analysis of ERG with two other genes commonly overexpressed in CaP, AMACR and DD3, revealed overexpression of at least one of these three genes in virtually all CaP specimen (54 of 55). Comprehensive evaluation of quantitative ERG1 expression with clinicopathological features also suggested that ERG1 expression level in prostate tumor cells relative to benign epithelial cells is indicator of disease-free survival after radical prostatectomy.
Collapse
Affiliation(s)
- Gyorgy Petrovics
- Center for Prostate Disease Research (CPDR), Department of Surgery and US Military Cancer Institute, Uniformed Services University, Rockville, MD 20852, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Papas TS, Watson DK, Sacchi N, Fujiwara S, Seth AK, Fisher RJ, Bhat NK, Mavrothalassitis G, Koizumi S, Jorcyk CL. ETS family of genes in leukemia and Down syndrome. AMERICAN JOURNAL OF MEDICAL GENETICS. SUPPLEMENT 2005; 7:251-61. [PMID: 2149958 DOI: 10.1002/ajmg.1320370751] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The human ETS2 and ERG genes are members of the ETS gene family, with sequence homology to the viral ets gene of the avian erythroblastosis retrovirus, E26. These genes are located on chromosome 21 and molecular genetic analysis of Down syndrome (DS) patients with partial trisomy 21 suggested that ETS2 may be a gene within the minimal DS genetic region. We have, in fact, been able to confirm the presence of the ETS2 gene dosage in triplicate occurring in occult human 21 chromosome abnormalities. It is known that ERG and ETS2 gene translocations occur in certain specific leukemias associated with defined chromosome rearrangements [e.g., t(8;21)]. Moreover, it is known that DS individuals are at greater risk for leukemic disease than their normal familial cohorts, implying that trisomy of that region of human chromosome 21 may play a role in the development of this type of neoplasia. The human ETS genes, first identified in our laboratory, are highly conserved, being found from lower organisms, like Drosophila and sea urchin, to humans. In mammals, the ETS genes are structurally distinct, located on separate chromosomes; they are transcriptionally active and differentially regulated. The ETS2 protein is phosphorylated and turns over with a half-life of approximately 20 min. After activation with the tumor promoter, TPA, the level of ETS2 elevates 5- to 20-fold. The properties of the ETS2 protein, such as nuclear localization, phosphorylation, rapid turnover, and response to protein kinase C, indicate that this protein belongs to a group of oncogene proteins thought to have regulatory functions in the nucleus. In the mouse thymus ets-1 and ets-2 are 8-10-fold higher, respectively, in the CD4+ subset than in other subsets examined, suggesting a role in T-cell development for these genes. Cells transfected with the cellular ets-2 gene, expressing higher levels of ets-2 products, showed a stimulated proliferation response, abolished their serum requirement and formed colonies in soft agar that could induce tumors in nude mice. Collectively, these data suggest that this family of genes might play a role in controlling specific steps of the signaling transduction pathway. Thus, the ETS genes, as other genes with homology to viral oncogenes, might be instrumental in regulating cellular growth and differentiation, as well as organismal development.
Collapse
Affiliation(s)
- T S Papas
- Laboratory of Molecular Oncology, National Cancer Institute, Frederick, Maryland 21701-1013
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Davidson B, Goldberg I, Tell L, Vigdorchik S, Baekelandt M, Berner A, Kristensen GB, Reich R, Kopolovic J. The clinical role of the PEA3 transcription factor in ovarian and breast carcinoma in effusions. Clin Exp Metastasis 2004; 21:191-9. [PMID: 15387369 DOI: 10.1023/b:clin.0000037703.37275.35] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Ets transcription factors play a central role in invasion and metastasis through regulation of synthesis of proteolytic enzymes and angiogenic molecules. The objective of this study was to investigate the role of PEA3 in tumor progression of ovarian and breast carcinoma metastatic to effusions, and to evaluate the expression of Ets-2 and Erg in ovarian carcinoma. Ovarian (83 malignant effusions, 102 corresponding solid lesions) and breast (33 malignant effusions, 40 corresponding solid lesions) carcinomas were evaluated for expression of PEA3 using mRNA in situ Hybridization (ISH). Expression of Ets-2 and Erg mRNA was analyzed in 50 ovarian carcinoma effusions using the same method. PEA3 mRNA expression was comparable at all sites in ovarian carcinoma (44 out of 83; 53% of effusions, 48 out of 102; 47% of solid tumors). PEA3 mRNA expression in effusions correlated with mRNA expression of the previously studied alphav (P = 0.022), alpha6 (P < 0.001) and beta1 (P < 0.001) integrin subunits, the matrix metalloproteinase (MMP) inducer EMMPRIN (P = 0.015) and interleukin-8 (IL-8) (P = 0.033). Erg and Ets-2 mRNA was expressed in 15 out of 50 (30%) and 18 out of 50 (36%) effusions, respectively, and co-localized with PEA3 (P = 0.017 for Erg, P = 0.004 for Ets-2). In breast carcinoma, PEA3 expression was seen in 19/40 (48%) of solid lesions, with a significant upregulation in corresponding effusions compared to primary tumors (24 out of 33; 73%, P = 0.038). PEA3 mRNA expression in effusions obtained prior to the institution of chemotherapy predicted significantly shorter overall survival in univariate analysis (24 vs 37 months, P = 0.03), with a similar trend for Erg (13 vs 30 months, P = 0.1). In conclusion, PEA3 is expressed at all anatomic sites in serous ovarian cancer and co-localizes with Erg, Ets-2 and several metastasis-associated molecules. PEA3 mRNA expression is a novel marker for tumor progression to malignant effusion in breast carcinoma, and predicts poor outcome in effusions sampled prior to therapeutic intervention in ovarian carcinoma. These findings support a biological role for Ets transcription factors in these malignancies and suggests that they may be targets for therapeutic intervention.
Collapse
Affiliation(s)
- Ben Davidson
- Department of Pathology, The Norwegian Radium Hospital, Montebello, University of Oslo, Oslo, Norway.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Ramakrishnan R, Fujimura Y, Zou JP, Liu F, Lee L, Rao VN, Reddy ESP. Role of protein-protein interactions in the antiapoptotic function of EWS-Fli-1. Oncogene 2004; 23:7087-94. [PMID: 15273724 DOI: 10.1038/sj.onc.1207927] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In the majority of Ewing's family tumors, chromosomal translocation t(11;22) leads to aberrant fusion of RNA-binding protein EWS with DNA-binding ETS transcriptional factor Fli-1. EWS-Fli-1 has altered the transcriptional activity and modulating its downstream target genes through this transcriptional activity is thought to be responsible for this tumor. We have previously shown that both EWS-Fli-1 and Fli-1 have antiapoptotic activity against several apoptotic inducers. Here, we show that the transcriptional activity of EWS-Fli-1 and Fli-1 is not essential for its antiapoptotic activity. We also demonstrate that EWS-Fli-1 and Fli-1 interact with CBP through its amino-terminal region and inhibit the CBP-dependent transcriptional activity of RXR. This activity appears to be independent of DNA-binding activity of EWS-Fli-1. Introduction of the dominant-negative form of CBP into Ewing's sarcoma cells sensitizes these cells against genotoxic or retinoic-acid induced apoptosis. These results suggest that the ability of EWS-Fli-1/Fli-1 to target transcriptional cofactor(s) and modulate apoptotic pathways may be responsible for its antiapoptotic and tumorigenic activities.
Collapse
|
43
|
Mackereth CD, Schärpf M, Gentile LN, MacIntosh SE, Slupsky CM, McIntosh LP. Diversity in structure and function of the Ets family PNT domains. J Mol Biol 2004; 342:1249-64. [PMID: 15351649 DOI: 10.1016/j.jmb.2004.07.094] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2004] [Revised: 06/23/2004] [Accepted: 07/16/2004] [Indexed: 10/26/2022]
Abstract
The PNT (or Pointed) domain, present within a subset of the Ets family of transcription factors, is structurally related to the larger group of SAM domains through a common tertiary arrangement of four alpha-helices. Previous studies have shown that, in contrast to the PNT domain from Tel, this domain from Ets-1 contains an additional N-terminal helix integral to its folded structure. To further investigate the structural plasticity of the PNT domain, we have used NMR spectroscopy to characterize this domain from two additional Ets proteins, Erg and GABPalpha. These studies both define the conserved and variable features of the PNT domain, and demonstrate that the additional N-terminal helix is also present in GABPalpha, but not Erg. In contrast to Tel and Yan, which self-associate to form insoluble polymers, we also show that the isolated PNT domains from Ets-1, Ets-2, Erg, Fli-1, GABPalpha, and Pnt-P2 are monomeric in solution. Furthermore, these soluble PNT domains do not associate in any pair-wise combination. Thus these latter Ets family PNT domains likely mediate interactions with additional components of the cellular signaling or transcriptional machinery.
Collapse
Affiliation(s)
- Cameron D Mackereth
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada V6T 1Z3
| | | | | | | | | | | |
Collapse
|
44
|
Ferrando-Miguel R, Cheon MS, Yang JW, Lubec G. Overexpression of transcription factor BACH1 in fetal Down syndrome brain. JOURNAL OF NEURAL TRANSMISSION. SUPPLEMENTUM 2003:193-205. [PMID: 15068251 DOI: 10.1007/978-3-7091-6721-2_17] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
There is a series of about 12 transcription factors expressed on chromosome 21. These transcription factors (TFs) are major candidates for playing a pathogenetic role for the abnormal wiring of the brain in fetal Down Syndrome (DS) as approximately 5,000 TFs are developmentally involved in the complex architecture of the human brain. TF derangement in DS has been already reported and we decided to contribute to the problem by studying four TFs encoded on chromosome 21 in fetal DS brain. We used fetal cortex of 8 DS fetuses and 6 controls (females) from the 18-19th week of gestation. Brain homogenates were subject to immunoblotting using goat-anti-BACH1, rabbit anti-heme oxygenase 1 (HO1), rabbit anti-ERG, rabbit anti-RUNX1 and goat anti-SIM2 l. Antibodies against beta-actin were used to normalise cell loss and antibodies against neuron-specific enolase were used to compensate neuronal loss. BACH1 was significantly overexpressed in fetal DS (p < 0.008) as compared to controls whereas RUNX1 and ERG proteins were comparable between groups, and SIM2 l was not detectable in any specimen. BACH1 was even significantly increased in the DS panel when normalised versus the housekeeping protein beta-actin (p < 0.01) or the neuron specific enolase (p < 0.01). HO-1 was found comparable between groups. BACH1, a member of the family of BTB-basic leucine zipper transcription factors, regulates gene expression through the NF-E2 site. More specifically, BACH1 suppresses expression of HO1. Increased BACH1, however, did not lead to decreased HO1, which would have explained oxidative stress observed in fetal DS.
Collapse
|
45
|
Fujimura Y, Siddique H, Lee L, Rao VN, Reddy ES. EWS-ATF-1 chimeric protein in soft tissue clear cell sarcoma associates with CREB-binding protein and interferes with p53-mediated trans-activation function. Oncogene 2001; 20:6653-9. [PMID: 11709699 DOI: 10.1038/sj.onc.1204684] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2001] [Revised: 05/23/2001] [Accepted: 05/31/2001] [Indexed: 11/09/2022]
Abstract
The recurrent t(12;22) (q13;q12) chromosomal translocation associated with soft tissue clear cell sarcoma results in a chimeric protein EWS-ATF-1 that acts as a constitutive transcriptional activator. The CBP/p300 transcriptional coactivator, which links various transcriptional factors to basal transcription apparatus, participates in transcriptional activation, growth and cell cycle control and differentiation. In this study, we show that EWS-ATF-1 associates constitutively with CBP both in vitro and in vivo. Both EWS and ATF-1 fusion domains are needed for this interaction. Here, we demonstrate that EWS-ATF-1 represses p53/CBP-mediated trans-activation function. Overexpression of CBP can counteract this repressive effect of EWS-ATF-1. Taken together, these findings suggest that one of the mechanisms by which EWS-ATF-1 may cause tumors is through targeting CBP/p300 resulting in the loss of function of p53. This novel mechanism may be responsible for the development of these and other related solid tumors.
Collapse
MESH Headings
- Blotting, Western
- Dose-Response Relationship, Drug
- Genes, p53
- Glutathione Transferase/metabolism
- Humans
- Models, Genetic
- Nuclear Proteins/metabolism
- Oncogene Proteins, Fusion/metabolism
- Plasmids/metabolism
- Precipitin Tests
- Protein Binding
- Protein Structure, Tertiary
- Sarcoma, Clear Cell/genetics
- Sarcoma, Clear Cell/metabolism
- Soft Tissue Neoplasms/genetics
- Soft Tissue Neoplasms/metabolism
- Trans-Activators/metabolism
- Transcription Factors
- Transcription, Genetic
- Transcriptional Activation
- Transfection
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Y Fujimura
- Program of Cancer Genetics, Department of Biochemistry, School of Medicine, MCP Hahnemann University, Broad and Vine, Philadelphia, PA 19102, USA
| | | | | | | | | |
Collapse
|
46
|
Abstract
Chromosomal translocations involving transcription factors and aberrant expression of transcription factors are frequently associated with leukemogenesis. Transcription factors are essential in maintaining the regulation of cell growth, development, and differentiation in the hematopoietic system. Alterations in the mechanisms that normally control these functions can lead to hematological malignancies. Further characterization of the molecular biology of leukemia will enhance our ability to develop disease-specific treatment strategies, and to develop effective methods of diagnosis and prognosis.
Collapse
Affiliation(s)
- H N Crans
- Department of Pediatrics, UCLA School of Medicine and Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | | |
Collapse
|
47
|
Remy P, Baltzinger M. The Ets-transcription factor family in embryonic development: lessons from the amphibian and bird. Oncogene 2000; 19:6417-31. [PMID: 11175358 DOI: 10.1038/sj.onc.1204044] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This chapter reviews the expression and role of Ets-genes during embryogenesis of amphibians and birds. In addition to overlapping expression domains, some of them exhibit cell type-specific expression. Many of them are expressed in migratory cells: neural crest, endothelial, and pronephric duct cells for instance. They are also transcribed in embryonic areas affected by epithelio-mesenchymal transitions. Both processes involve modifications of cellular adhesion. Ets-family genes appear to coordinate changes in the expression of adhesion molecules and degradation of the extracellular matrix upon regulation of matrix metalloproteinases and their specific inhibitors. These functions are essential for physiological processes like tissue remodelling during embryogenesis or wound healing. Unfortunately they also play a harmful role in metastasis. Recent studies in the nervous system showed that Ets-genes contribute to the establishment of a cellular identity. This identity could rely on definite cell-surface determinants, among which cadherins could play an important role. In addition to cell-type specific expression, other factors contribute to the specificity of function of Ets-genes. These genes have a broad specificity of recognition of target sequences in gene promoters, insufficient for accurate control of gene expression. A fine tuning could arise from combinatorial interactions with other Ets- or accessory proteins.
Collapse
Affiliation(s)
- P Remy
- FRE 2168 du CNRS, IPCB, 21 rue René Descartes, 67084 Strasbourg cedex, France
| | | |
Collapse
|
48
|
Maroulakou IG, Bowe DB. Expression and function of Ets transcription factors in mammalian development: a regulatory network. Oncogene 2000; 19:6432-42. [PMID: 11175359 DOI: 10.1038/sj.onc.1204039] [Citation(s) in RCA: 157] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The Ets transcription factor family is involved in a variety of mammalian developmental processes at the cellular, tissue and organ levels. They are implicated in cellular proliferation, differentiation, migration, apoptosis and cell - cell interactions. This article reviews recent studies that demonstrate the integral importance of Ets in the dosage dependent regulation of development. The expression of many Ets genes is associated with mesenchymal - epithelial interactions and changes in extracellular matrix proteins. These inductive processes contribute to tissue remodeling and integrity, particularly during embryonic development. Overlapping as well as unique patterns of Ets expression are evident in developing tissues, including development of the lymphoid and myeloid lineages, brain and central nervous system, bone and mammary gland. Integration of these data will allow the development of predictive models for the regulation of complex developmental processes.
Collapse
Affiliation(s)
- I G Maroulakou
- Center for Molecular and Structural Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, SC 29403, USA
| | | |
Collapse
|
49
|
Kim J, Pelletier J. Molecular genetics of chromosome translocations involving EWS and related family members. Physiol Genomics 1999; 1:127-38. [PMID: 11015571 DOI: 10.1152/physiolgenomics.1999.1.3.127] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Many types of sarcomas are characterized by specific chromosomal translocations that appear to result in the production of novel, tumor-specific chimeric transcription factors. Many of these show striking similarities: the emerging picture is that the amino-terminal domain of the fusion product is donated by the Ewing's sarcoma gene (EWS) or a related member from the same gene family, whereas the carboxy-terminal domain often consists of a DNA-binding domain derived from one of a number of transcription factors. Given the observation that the different translocation partners of the EWS protooncogene are associated with distinct types of sarcomas, the functional consequence of fusing EWS (or a related family member) to a different DNA-binding domain can only be understood in the context of functional studies that define the specificity of action of the different fusion products. An understanding of the molecular structure and function of these translocations provides new methods for diagnosis and novel targets for therapeutics.
Collapse
Affiliation(s)
- J Kim
- Department of Biochemistry, Department of Oncology, McGill University, Montreal, Quebec, Canada H3G 1Y6
| | | |
Collapse
|
50
|
Ichikawa H, Shimizu K, Katsu R, Ohki M. Dual transforming activities of the FUS (TLS)-ERG leukemia fusion protein conferred by two N-terminal domains of FUS (TLS). Mol Cell Biol 1999; 19:7639-50. [PMID: 10523652 PMCID: PMC84797 DOI: 10.1128/mcb.19.11.7639] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The FUS (TLS)-ERG chimeric protein associated with t(16;21)(p11;q22) acute myeloid leukemia is structurally similar to the Ewing's sarcoma chimeric transcription factor EWS-ERG. We found that both FUS-ERG and EWS-ERG could induce anchorage-independent proliferation of the mouse fibroblast cell line NIH 3T3. However, only FUS-ERG was able to inhibit the differentiation into neutrophils of a mouse myeloid precursor cell line L-G and induce its granulocyte colony-stimulating factor-dependent growth. We constructed several deletion mutants of FUS-ERG lacking a part of the N-terminal FUS region. A deletion mutant lacking the region between amino acids 1 and 173 (exons 1 to 5) lost the NIH 3T3-transforming activity but retained the L-G-transforming activity. On the other hand, a mutant lacking the region between amino acids 174 and 265 (exons 6 and 7) lost the L-G-transforming activity but retained the NIH 3T3-transforming activity. These results indicate that the N-terminal region of FUS contains two independent functional domains required for the NIH 3T3 and L-G transformation, which we named TR1 and TR2, respectively. Although EWS intrinsically possessed the TR2 domain, the EWS-ERG construct employed lacked the EWS sequence containing this domain. Since the TR2 domain is always found in chimeric proteins identified from t(16;21) leukemia patients but not in chimeric proteins from Ewing's sarcoma patients, it seems that the TR2 function is required only for the leukemogenic potential. In addition, we identified three cellular genes whose expression was altered by ectopic expression of FUS-ERG and found that these are regulated in either a TR1-dependent or a TR2-dependent manner. These results suggest that FUS-ERG may activate two independent oncogenic pathways during the leukemogenic process by modulating the expression of two different groups of genes simultaneously.
Collapse
MESH Headings
- 3T3 Cells
- Acute Disease
- Animals
- Base Sequence
- Cell Transformation, Neoplastic/genetics
- Chromosomes, Human, Pair 16
- Chromosomes, Human, Pair 21
- DNA Primers
- Exons
- Gene Expression Regulation
- Hematopoietic Stem Cells
- Heterogeneous-Nuclear Ribonucleoproteins
- Humans
- Leukemia, Myeloid/etiology
- Leukemia, Myeloid/genetics
- Mice
- Models, Genetic
- Molecular Sequence Data
- Oncogene Proteins, Fusion/genetics
- Peptide Fragments/genetics
- RNA-Binding Protein EWS
- RNA-Binding Protein FUS
- Ribonucleoproteins/genetics
- Translocation, Genetic
Collapse
Affiliation(s)
- H Ichikawa
- Radiobiology Division, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045, Japan.
| | | | | | | |
Collapse
|